Economic Evaluation of Cancer Drugs

Download as pdf or txt
Download as pdf or txt
You are on page 1of 442
At a glance
Powered by AI
The document discusses economic evaluation of cancer drugs using clinical trial and real-world data. It covers statistical methods for analyzing survival data and extrapolating results.

The document discusses advanced modelling techniques for survival data as well as methods like proportional hazards models, transition probabilities, and Monte-Carlo sampling for extrapolating survival rates. It also discusses issues in fitting survival models.

The document mentions challenges in valuing health for healthcare decisions, including uncertainty, variability, and determining willingness to pay thresholds and their impact.

Economic Evaluation of

Cancer Drugs
Using Clinical Trial and
Real-World Data
Chapman & Hall/CRC
Biostatistics Series
Shein-Chung Chow, Duke University School of Medicine
Byron Jones, Novartis Pharma AG
Jen-pei Liu, National Taiwan University
Karl E. Peace, Georgia Southern University
Bruce W. Turnbull, Cornell University

Recently Published Titles


Bayesian Methods for Repeated Measures
Lyle D. Broemeling
Modern Adaptive Randomized Clinical Trials
Statistical and Practical Aspects
Oleksandr Sverdlov
Medical Product Safety Evaluation
Biological Models and Statistical Methods
Jie Chen, Joseph Heyse, Tze Leung Lai
Statistical Methods for Survival Trial Design
With Applications to Cancer Clinical Trials Using R
Jianrong Wu
Bayesian Applications in Pharmaceutical Development
Satrajit Roychoudhury, Soumi Lahiri
Platform Trials in Drug Development
Umbrella Trials and Basket Trials
Zoran Antonjevic and Robert Beckman
Innovative Strategies, Statistical Solutions and Simulations for Modern
Clinical Trials
Mark Chang, John Balser, Robin Bliss and Jim Roach
Cost-effectiveness Analysis of Medical Treatments
A Statistical Decision Theory Approach
Elias Moreno, Francisco Jose Vazquez-Polo and Miguel Angel Negrin-Hernandez
Analysis of Incidence Rates
Peter Cummings
Mixture Modelling for Medical and Health Sciences
Shu-Kay Ng, Liming Xiang, Kelvin Kai Wing Yau
Economic Evaluation of Cancer Drugs
Using Clinical Trial and Real-World Data
Iftekhar Khan, Ralph Crott, Zahid Bashir
Economic Evaluation of
Cancer Drugs
Using Clinical Trial and
Real-World Data

Iftekhar Khan,
Ralph Crott, and Zahid Bashir
CRC Press
Taylor & Francis Group
6000 Broken Sound Parkway NW, Suite 300
Boca Raton, FL 33487-2742

© 2020 by Taylor & Francis Group, LLC


CRC Press is an imprint of Taylor & Francis Group, an Informa business

No claim to original U.S. Government works

Printed on acid-free paper

International Standard Book Number-13: 978-1-498-76130-7 (Hardback)

This book contains information obtained from authentic and highly regarded sources. Reasonable
efforts have been made to publish reliable data and information, but the author and publisher cannot
assume responsibility for the validity of all materials or the consequences of their use. The authors
and publishers have attempted to trace the copyright holders of all material reproduced in this
publication and apologize to copyright holders if permission to publish in this form has not been
obtained. If any copyright material has not been acknowledged please write and let us know so we
may rectify in any future reprint.

Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced,
transmitted, or utilized in any form by any electronic, mechanical, or other means, now known
or hereafter invented, including photocopying, microfilming, and recording, or in any information
storage or retrieval system, without written permission from the publishers.

For permission to photocopy or use material electronically from this work, please access www.copy-
right.com (http://www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC), 222
Rosewood Drive, Danvers, MA 01923, 978-750-8400. CCC is a not-for-profit organization that pro-
vides licenses and registration for a variety of users. For organizations that have been granted a
photocopy license by the CCC, a separate system of payment has been arranged.

Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and
are used only for identification and explanation without intent to infringe.

Visit the Taylor & Francis Web site at


http://www.taylorandfrancis.com

and the CRC Press Web site at


http://www.crcpress.com
These are our works, these works our souls display

Behold our works, when we have passed away

For Suhailah, Yohanis, Hanzalah, my father, and all those affected by cancer

For Jon and Fiona, my children

For Saima, Hassan, and Hibba


Contents

Preface......................................................................................................................xv
Acknowledgments.............................................................................................. xvii
About the Authors............................................................................................... xix
Acronyms and Abbreviations............................................................................ xxi

1 Introduction to Cancer....................................................................................1
1.1 Cancer......................................................................................................1
1.2 Epidemiology of Cancer........................................................................ 1
1.2.1 Cancer Trends............................................................................2
1.3 Prognostic Factors Associated with Cancer Outcomes ................... 5
1.4 Economic Burden of Cancer.................................................................6
1.4.1 Health Expenditure.................................................................. 6
1.4.2 Healthcare Expenditure on Drugs......................................... 7
1.5 Treatments for Cancer......................................................................... 10
1.6 Important Economic Concepts for Cost-Effectiveness of
Cancer Interventions�������������������������������������������������������������������������� 12
1.6.1 Economics, Health Economics, Economic Evaluation,
and Pharmacoeconomics������������������������������������������������������ 12
1.6.1.1 Value ......................................................................... 13
1.6.1.2 Allocative Efficiency............................................... 14
1.6.1.3 Technical Efficiency................................................ 15
1.6.1.4 Opportunity Cost.................................................... 16
1.6.1.5 Discounting.............................................................. 17
1.6.1.6 The Incremental Cost-Effectiveness Ratio .......... 18
1.6.1.7 The Cost-Effectiveness Plane................................. 19
1.6.1.8 Quality-Adjusted Life-Years (QALY) ...................22
1.7 Health Economic Evaluation and Cancer Drug
Development in Practice�������������������������������������������������������������������� 23
1.7.1 The Modern Paradigm........................................................... 24
1.8 Efficacy versus Effectiveness ............................................................. 26
1.9 Real-World Data .................................................................................. 27
1.10 Economic versus Clinical Hypotheses ............................................. 29
1.11 Summary............................................................................................... 32
1.12 Exercises for Chapter 1........................................................................ 33
2 Important Outcomes for Economic Evaluation in Cancer Studies...... 35
2.1 Introduction ......................................................................................... 35
2.2 Important Common, Surrogate, and Novel Cancer Endpoints ����� 36
2.2.1 Overall Survival...................................................................... 36
2.2.1.1 OS and Economic Evaluation ............................... 41
2.2.2 Surrogate Endpoints............................................................... 46

vii
viii Contents

2.3 HTAs with Surrogate Endpoints....................................................... 53


2.4 Emerging Tumor-Centered Endpoints............................................. 55
2.5 Demonstrating Value from Other Cancer Endpoints..................... 57
2.6 Summary............................................................................................... 58
2.7 Exercises for Chapter 2........................................................................ 58

3 Health-Related Quality of Life for Cost-Effectiveness.......................... 59


3.1 Health-Related Quality of Life (HRQoL) in Cancer Patients......... 59
3.1.1 Limitations of Anti-Cancer Treatments............................... 59
3.1.2 Why Collect HRQoL Data?.................................................... 60
3.1.3 Challenges with HRQoL in Cancer Studies........................ 61
3.2 Measuring Health-Related Quality of Life Outcomes for
Common Cancer Types���������������������������������������������������������������������� 62
3.2.1 Condition-Specific Measures of HRQoL ............................ 62
3.2.2 Common General Condition-Specific Measures of
HRQoL in Cancer....................................................................63
3.3 Measuring HRQoL for Economic Evaluation ................................. 67
3.3.1 EuroQol EQ-5D-3L and 5L.................................................... 68
3.3.2 EuroQol EQ-5D-5L................................................................. 69
3.4 Constructing Utilities.......................................................................... 70
3.5 Quality-Adjusted Life-Years (QALYs)................................................ 72
3.5.1 QALY Calculation in Cancer Trials...................................... 73
3.6 Economic Evaluation in the Absence of Utility Data:
Mapping and Utility Studies������������������������������������������������������������� 74
3.7 Sensitivity and Responsiveness of EQ-5D versus QLQ-C30
HRQoL for Detecting Improvement in Cancer Patients��������������� 76
3.8 Measuring Post-Progression (PP) Utility: Some Approaches .......77
Why Is Estimation of Utility between Disease Progression
and Death Relevant?............................................................... 78
The Behavior of Utility in Cancer Patients between
Progression and Death?......................................................... 79
3.8.1 Plausible Post-Progression Utility Behavior ......................80
3.8.2 Non-Linear Models................................................................. 82
3.9 HRQoL issues in Health Technology Appraisals of Cancer
Drugs���������������������������������������������������������������������������������������������������� 87
3.10 Summary............................................................................................... 89
3.11 Exercises for Chapter 3........................................................................ 89

4 Introductory Statistical Methods for Economic Evaluation in


Cancer............................................................................................................... 91
4.1 Introduction.......................................................................................... 91
4.2 Uncertainty and Variability................................................................ 91
4.2.1 Uncertainty.............................................................................. 92
4.2.2 Variability................................................................................. 92
4.2.2.1 Hypothesis Testing................................................. 93
Contents ix

4.3 Distributions: Cost, Utility, and Survival Data ............................... 93


4.4 Important Measures Used in Cancer Trials..................................... 95
4.4.1 Time-to-Event Endpoints....................................................... 95
4.4.2 Median Survival..................................................................... 96
4.4.3 Hazard Rate and Hazard Ratio............................................ 98
4.4.4 Hazard Ratio............................................................................ 99
4.4.5 Survival Rates and Proportions.......................................... 101
4.4.6 Relationship between Hazard Rate and Survival Rate......102
4.4.7 Transition Probability and Matrix...................................... 103
4.4.8 Relation between Transition Probability and
Survival Rates����������������������������������������������������������������������� 104
4.4.9 Proportional Hazards.......................................................... 106
4.4.10 Mean Survival and Restricted Mean ................................ 106
4.5 Simulation: Bootstrapping and Monte-Carlo Simulation............ 109
4.5.1 Simulating Using Monte-Carlo Sampling......................... 111
4.6 Analyzing Data from Cancer Trials................................................ 111
4.6.1 Semi-Parametric Methods: The Cox PH Model............... 111
4.6.1.1 Adjusting for Covariates with the Cox Model......112
4.6.1.2 Using Hazard Ratios to Predict Survival Rates.....113
4.6.2 Parametric Methods: Modeling Survival Data for
Extrapolation ����������������������������������������������������������������������� 114
4.6.3 Advanced Modeling Techniques for Survival Data ....... 118
4.6.3.1 Flexible Parametric Survival Models................. 118
4.6.3.2 Applications in Cancer Surveillance ................. 119
4.7 Issues in Fitting Models.................................................................... 122
4.8 Handling Crossover, Treatment Switching, and
Subsequent Anti-Cancer Therapy�������������������������������������������������� 123
4.8.1 Introduction to Treatment Switching................................. 123
4.8.2 Types of Switching ............................................................... 124
4.8.3 Implications of Switching.................................................... 124
4.8.4 Handling Switching in Statistical Analyses .................... 126
4.8.4.1 Intent-to-Treat (ITT)............................................... 127
4.8.4.2 Per Protocol Analysis............................................ 128
4.8.4.3 IPCW....................................................................... 128
4.8.4.4 RPFSTM.................................................................. 129
4.8.4.5 Two-Stage Adjustment Model ............................ 131
4.8.4.6 Other Approaches: Structural
Nested Mean Models (SNNM)��������������������������� 131
4.9 Data Synthesis and Network Meta-Analyses................................ 132
4.9.1 Mixed Treatment Comparisons ......................................... 132
4.9.1.1 Direct Comparison................................................ 133
4.9.1.2 Indirect Treatment Comparison (ITC) .............. 133
4.9.1.3 Meta-Analysis ....................................................... 134
4.9.1.4 Network of Evidence ........................................... 134
4.9.2 Assumptions for Carrying Out MTCs .............................. 134
x Contents

4.10 Summary............................................................................................. 138


4.11 Exercises for Chapter 4...................................................................... 140

5 Collecting and Analysis of Costs from Cancer Studies...................... 141


5.1 Types of Costs Typical of Cancer Trials.......................................... 141
5.1.1 Categorization of Health Resource Use............................. 142
5.1.2 Resource Use Monitoring.................................................... 142
5.1.3 Baseline Characteristics and Health Resource Use......... 143
5.1.4 Costs Determined by a Study Protocol.............................. 144
5.2 Perspective of Analysis and Costs Collection................................ 145
5.3 Collecting Health Resource Use across the Treatment
Pathway ��������������������������������������������������������������������������������������������� 146
5.3.1 Time Horizon ....................................................................... 148
5.4 Costing Methods: Micro versus Macro Approach........................ 150
5.4.1 Average versus Marginal and Incremental Cost.............. 151
5.4.2 Inflation.................................................................................. 152
5.4.3 Time Preference and Discounting...................................... 153
5.5 Charges................................................................................................ 154
5.5.1 Cost-to-Charge Ratios.......................................................... 155
5.5.2 Other Non-Medical Costs (e.g. Societal Costs)................. 155
5.6 Distribution of Costs.......................................................................... 155
5.6.1 Transforming Cost Data....................................................... 157
5.7 Handling Censored and Missing Costs ........................................ 158
5.7.1 Strategies for Avoiding Missing Resource Data .............. 160
5.7.2 Strategies for Analyzing Cost Data
When Data Are Missing or Censored ������������������������������ 160
5.7.3 Imputation Methods............................................................. 161
5.8 Handling Future Costs...................................................................... 162
5.9 Case Report Forms and Health Resource Use............................... 164
5.10 Statistical Analyses of Costs ............................................................ 165
5.11 Summary............................................................................................. 172
5.12 Exercises for Chapter 5...................................................................... 173

6 Designing Cost-Effectiveness into Cancer Trials................................. 175


6.1 Introduction and Reasons for Collecting Economic
Data in a Clinical Trial �������������������������������������������������������������������� 175
6.2 Clinical Trial Designs for Cancer Studies...................................... 178
6.2.1 Clinical Trial Designs........................................................... 178
6.2.2 Interim Analyses and Data Monitoring Committees
(DMC)..................................................................................... 188
6.3 Planning a Health Economic Evaluation in a Clinical Trial ....... 191
6.3.1 Important Considerations When Designing
a Cancer Study for Economic Evaluation ������������������������ 191
6.3.2 Integrating Economic Evaluation in a Clinical Trial:
Considerations���������������������������������������������������������������������� 194
Contents xi

6.3.3 Endpoints and Outcomes.................................................... 196


6.3.3.1 Timing of Measurements..................................... 198
6.3.3.2 Trial Design............................................................ 198
6.3.3.3 CRF Design............................................................ 199
6.3.3.4 Sample Size Methods for Cost-Effectiveness....... 199
6.3.3.5 Sample Size Formulae for
Cost-Effectiveness: Examples����������������������������� 201
6.3.4 Treatment Pathways............................................................. 204
6.3.5 Time of Generic/Competition Entry.................................. 204
6.3.6 Treatment Compliance......................................................... 205
6.3.7 Identify Subgroups/Heterogeneity.................................... 206
6.3.8 Early ICER/INMB................................................................. 206
6.3.9 Multicenter Trials.................................................................. 207
6.4 Case Study of Economic Evaluation of Cancer Trials................... 210
6.4.1 TA516 Cabozanitib + Vandetanib....................................... 210
6.5 Summary............................................................................................. 210
6.6 Exercises for Chapter 6...................................................................... 213

7 Models for Economic Evaluation of Cancer........................................... 215


7.1 Types of Health Economic Models.................................................. 215
7.2 Decision Tree Models........................................................................ 215
7.2.1 Further Possible Improvements to the
Decision Model �������������������������������������������������������������������� 224
7.3 Markov Models.................................................................................. 226
7.4 Continuous Time Markov Models................................................... 230
7.5 The Partitioned Survival Model...................................................... 231
7.5.1 Developing an Economic Model Using Patient-Level
Data Using a Partitioned Survival Model Approach������ 231
7.5.1.1 Modeling the Efficacy Data (Survival Data)...... 231
7.5.2 Case Study of an Economic Model Using Patient-
Level Data: A Partitioned Survival Model ���������������������� 232
7.5.3 Crossover............................................................................... 236
7.6 Summary of Cost-Effectiveness Models for Cancer
Used in HTA Submissions��������������������������������������������������������������� 239
7.7 Summary............................................................................................. 243
7.8 Exercises for Chapter 7...................................................................... 243

8 Real-World Data in Cost-Effectiveness


Studies on Cancer ....................................................................................... 249
8.1 Introduction to Real-World Data..................................................... 249
8.2 Using RWD to Support Cost-Effectiveness Analysis ................... 251
8.3 Strengths and Limitations of Using RWD to Support
Cost-Effectiveness Analysis ������������������������������������������������������������ 253
8.3.1 Limitations............................................................................. 255
8.3.2 Internal Validity versus Generalizability.......................... 256
xii Contents

8.4 Sources for RWD Generation........................................................... 257


8.4.1 Registries ............................................................................... 260
8.4.2 Audits .................................................................................... 261
8.4.3 Primary Care Databases: CPRD, THIN, QResearch........ 262
8.4.4 Insurance Claims Databases............................................... 263
8.4.5 Digital Data Sources, Social Media and Applications....... 263
8.4.6 Commercial Data Sources................................................... 264
8.4.7 Pragmatic Clinical Trials..................................................... 264
8.4.8 Prospective Observational Research Studies................... 265
8.4.9 Case Control Studies............................................................ 265
8.5 Using Cancer Registries.................................................................... 265
8.5.1 Examples of Registries in the UK for RWE ...................... 267
8.6 Statistical Analyses of RWD: Addressing Selection Bias............. 268
8.6.1 Propensity Score Modeling................................................. 268
8.6.2 Instrumental Variable Methods.......................................... 274
Results.................................................................................................. 277
8.7 Summary and Conclusion................................................................ 279
8.8 Exercises for Chapter 8...................................................................... 281

9 Reporting and Interpreting Results of Cost-Effectiveness


Analyses from Cancer Trials..................................................................... 283
9.1 Interpreting Incremental Costs and QALYs................................... 283
9.1.1 Informative Censoring.........................................................284
9.2 Interpreting Incremental QALYs..................................................... 287
9.3 Relationship between Costs and QALYs........................................ 290
9.4 Interpreting the ICER and the Cost-Effectiveness Plane.............. 292
9.4.1 Uncertainty............................................................................ 292
9.5 Presenting and Interpreting Results from Uncertainty
Analysis ��������������������������������������������������������������������������������������������� 296
9.6 Bayesian Sensitivity Analysis........................................................... 306
9.6.1 Limitations of the ICER and Using the INMB.................. 307
9.7 Presenting and Interpreting Results from Value of
Information Analyses ����������������������������������������������������������������������308
9.8 Challenges of VOI Analysis in Healthcare Decisions................... 316
9.9 Summary ............................................................................................ 317
9.10 Exercises for Chapter 9...................................................................... 317
Technical Appendix for Chapter 9.............................................................. 318
A9.1 Simulation.............................................................................. 318
A9.2 Bayesian PSA......................................................................... 319
A9.3 Value of Information............................................................ 320
Before Any Data Is Observed........................................................... 321
After Data Have Been Observed...................................................... 321
Contents xiii

10 Factors Predictive of HTA Success and the Global Landscape.......... 323


10.1 Introduction........................................................................................ 323
10.2 Cancer Drugs Rejected by NICE...................................................... 323
10.3 Summary of Criticisms of Economic Models of Cancer.............. 324
10.4 Factors Predictive of Successful HTAs in Cancer.......................... 335
10.5 The Changing Pace of the Reimbursement Environment .......... 341
10.6 Reimbursement and Payer Evidence Requirements
Across Different Countries��������������������������������������������������������������344
10.6.1 Canada....................................................................................345
10.6.2 France......................................................................................345
10.6.3 Germany.................................................................................346
10.6.4 Italy......................................................................................... 347
10.6.5 Spain....................................................................................... 347
10.6.6 Australia.................................................................................348
10.6.7 United Kingdom................................................................... 349
10.7 Pricing and Reimbursement Environment in the
United States....................................................................................... 349
10.8 Value-Based Pricing (VBP) for Cancer Drugs................................ 350
10.9 Risk-Sharing Scheme ........................................................................ 352
10.10 The Future of Cost-Effectiveness of Cancer Treatments.............. 356
10.10.1 Future Research: Methodology.......................................... 356
10.10.2 Future Reimbursement Landscape.................................... 358
Budget Impact Threshold................................................... 359
10.10.2.1 Automatic Funding for Highly Specialized
Drugs for Rare Diseases������������������������������������ 359
10.10.2.2 Fast-Track Appraisals......................................... 359
10.11 Summary............................................................................................. 360
10.12 Exercises for Chapter 10.................................................................... 360
References............................................................................................................ 361
Additional Bibliography.............................................................................. 394
Chapter 1............................................................................................. 394
Chapter 3............................................................................................. 395
Chapter 4............................................................................................. 395
Chapter 5............................................................................................. 399
Chapter 7............................................................................................. 399
Chapter 9............................................................................................. 399
Index...................................................................................................................... 401
Preface

The cost of cancer care has increased hugely and put pressure on health-
care systems around the world. An important part of this cost is the cost
of cancer medicines. Economic evaluation of cancer drugs is an extremely
important area that affects health policy and access to cancer treatment. The
economic evaluation of cancer drugs can involve careful trial design, robust
economic modeling, and sound statistical analysis and research methodol-
ogy, drawing together the disciplines of medical statistics, clinical research,
and economics.
Several books already exist that address theoretical or practical aspects of
cost-effectiveness analysis. However, no unified text on the cost-effective-
ness of cancer medicines is currently available. This book attempts to deal
with the matter within a practical framework, focusing on key concepts and
drawing on the experiences of health technology appraisals (HTA) of cancer
drugs – either approved or rejected by government reimbursement agencies
(the payers). This book also offers an insight into how health economic evalu-
ation of cancer interventions has been carried out in practice, with many
examples throughout, where data are collected in clinical trials or in real-
world settings.
This book is not just about performing cost-effectiveness analyses of
cancer drugs using clinical trial and real-world data, but also emphasizes
the strategic importance of economic evaluation through the drug devel-
opment process. It also offers guidance and advice on the complex factors
at play before, during, and after an economic evaluation for a cancer inter-
vention. In addition, this book bridges the gap between industry (phar-
maceutical) applications of economic evaluation and what students may
learn on university courses. The book is suitable for statisticians, health
economists, cancer researchers, oncologists, and anyone with an interest in
the cost-effectiveness of interventions for cancer using clinical trial and/or
real-world data. It would also be a valuable book for a postgraduate course
in health economics.
We candidly admit that our objectives have been set high when structur-
ing this book. Economic evaluation covers several disciplines and address-
ing all of these has been challenging. We hope that the material in this book
is suitable for a range of researchers of varying abilities, so that some will
find the entire book useful whereas, for others, particular chapters will be

xv
xvi Preface

useful. As a student textbook, this book can be complemented by additional


reading material suggested in the bibliography.

Iftekhar Khan
Centre for Statistics in Medicine, University of Oxford
Ralph Crott
Consultant Health Economist, Belgium
Zahid Bashir
Clinical Consultant in Cancer Trials
Acknowledgments

We would like to gratefully acknowledge helpful reviews by Dr. Noan-Min


Chau from the Licensing Division of UK Medicines and Health Regulator
Agency (MHRA) and also Tarita Murray-Thomas from the Clinical Practice
Research Data Link (CPRD) within the MHRA. Their comments have
resulted in a much-improved text. We also acknowledge the reviews by Dr.
Suhailah Khan and Uzma Ikramullah for proofreading parts of the text. We
graciously acknowledge the National Institute for Health and Care Excellence
(NICE), which has made available to the public such excellent material, as
­demonstrated in this book. Finally, we would like to thank the anonymous
reviewers of earlier chapters. Their comments have been extremely helpful
enabling us to deliver a much-improved text.

xvii
About the Authors

Dr. Iftekhar Khan is a medical statistician and health economist by quali-


fication and training. Dr. Khan has extensive experience in cancer trials in
industry, academia, and regulatory environments, spanning over 18 years.
He was formerly associate professor in Medical Statistics and Methodology
at King’s College, London, and lead statistician at UCL CRUK Cancer Trials
Centre and Oxford University’s Center for Statistics in Medicine. Professor
Khan is also a Senior Research Fellow in Health Economics at the University
of Warwick and a Senior Statistical Assessor within the Licensing Division
of the UK Medicine and Health Regulation Agency where he regularly eval-
uates oncologic and other drugs and devices for licensing.

Dr. Ralph Crott is a former professor in Pharmacoeconomics at the University


of Montreal in Quebec, Canada and former head of the EORTC Health
Economics Unit (Brussels, Belgium) and former senior health economist at
the Belgian HTA organization (KCE). Dr. Crott has been active in economic
evaluation of new medical technology since 1984. He also held research and/
or teaching positions at York University in the United Kingdom and the
Catholic University of Louvain, Belgium. He received his PhD in applied
economics at the Catholic University of Louvain (UCL, Belgium) and holds
additional masters degrees in econometrics (Flemish University of Brussels
(VUB, Belgium), biostatistics (Limburg University, Belgium), public health
with a major in clinical research (Catholic University of Louvain, Belgium),
and technology assessment (Aston University, United Kingdom).

Dr. Zahid Bashir has over twelve years of experience working in the phar-
maceutical industry, specifically in medical affairs and oncology drug devel-
opment, where he is involved in the design and execution of oncological
clinical trials and development of reimbursement dossiers for HTA submis-
sion. Dr. Bashir also has extensive clinical experience in teaching oncology
and haematology in the UK NHS hospital.

xix
Acronyms and Abbreviations

ACD advanced consultation document


AE adverse events
AEMPS Agencia Española de Medicamentos y Productos Sanitarios
AFT accelerated failure time
AHRQ Agency for Healthcare Research and Quality
AIC Akaike Information Criterion
ALK anaplastic lymphoma kinase
ASCO American Society of Clinical Oncology
ASMR medical improvement score
ATC average total cost
AUC area under the curve
BB beta binomial
BEV bevacizumab
BIM budget impact model
BNF British National Formulary
BRCA breast cancer gene
BSC best supportive care
CADTH Canadian Agency for Drugs and Technologies in Health
CAP chemotherapy for advanced prostate cancer
CCG clinical commissioning group
CCyR complete cytogenic response
CDF Cancer Drug Fund
CDR Common Drug Review
CE cost-effectiveness
CEA cost-effectiveness analysis
CEAC cost-effectiveness acceptability curve
CEESP Commission d’Évaluation Économique et de Santé
Publique
CESP Comité Economique des Produits de Santé
CHEERS Consolidated Health Economic Evaluation Reporting
chemo-RT chemoradiotherapy
CI confidence interval
CMA cost minimization analysis
CML chronic myeloid leukemia
CMS Center for Medicare Services
CPRD Clinical Practice Research Datalink
CPT current procedural terminology
CR complete response
CRA clinical research associate
CRC colorectal cancer

xxi
xxii Acronyms and Abbreviations

CRF case report form


CRUK Cancer Research UK
CSM condition-specific measures
CSRI client services receipt inventory
CT computed (axial) tomography
CUA cost-utility analysis
CV cabozanitib and vandetanib
DALY disability-adjusted life-years
DAPA dementia and physical activity
DCF data collection form
DES discrete event simulation
DFS disease-free survival
DICE discretely integrated condition event
DLBCL diffuse large B-cell lymphoma
DMC data monitoring committee
DoR duration of response
DRG diagnostic-related group
EAMS early access to medicine
ECOG Eastern Cooperative Oncology Group
eCRF electronic case report form (CRF)
EFS event-free survival
EGFR epidermal growth factor receptor
EHR electronic health records
EINB expected incremental net benefit
EMA European Medicines Agency
ENB expected net benefit
ENMB expected net monetary benefit
ENBS expected net benefit of sampling
EoL end-of-life
EORTC European Organization for Research and Treatment of
Cancer
ERG evidence review group
ESMO European Society of Medical Oncology
EUnetHTA European Network of Health Technology assessment
EVPI expected value of perfect information
EVPPI expected value of partially perfect information
EVSI expected value of sample information
FACT Functional Assessment of Cancer Therapy
FDA Food and Drug Administration
FDAAA FDA Amendments Act
FDG-PET 2-fluoro-2-deoxyglucose positron emission tomography
FISH fluorescence in situ hybridization
GCP Good Clinical Practice
GDP gross domestic product
GDPR General Data Protection Regulation
Acronyms and Abbreviations xxiii

GLM general / generalized linear model


GLOBOCAN Global Cancer Observatory Database
HAS Haute Autorité de Santé
HCCor half-cycle correction
HCC hepatocellular carcinoma
HEAP health economic analysis plan
HES Hospital Episode Statistics
HR hazard ratio
HRQoL health-related quality of life
HTA health technology assessment
HUI health utility index
ICD international classification of diseases
ICD-O international classification of diseases for oncology
ICER incremental cost-effectiveness ratio
ICERev Institute for Clinical and Economic Review
IDMC independent data monitoring committee
INB incremental net benefit
INFORMED Information Exchange and Data Transformation
INMB incremental net monetary benefit
IPCW inverse probability-of-censoring weighting
IQWiG Institute for Quality and Efficiency
irRC immune-related response criteria
ISPOR International Society of Pharmacoeconomics and
Outcomes Research
ITC indirect treatment comparison
ITT intention-to-treat
IV instrumental variable
IV intravenous
KM Kaplan-Meier
LCS lung cancer scale
LMG life-month gained (as unit)
LOCF last observation carried forward
LSmean least squares mean
LT liver transplantation
LYG life-years gained
MAMS multi-arm, multi-stage
MAR missing at random
MCAR missing completely at random
MCID minimum clinical important difference
MCO managed care organization
MDS myelodysplastic syndrome
MDT multidisciplinary team
MED minimum effective dose
MHRA Medicines and Healthcare products Regulatory Agency
MI multiple imputation
xxiv Acronyms and Abbreviations

mITT modified ITT


MLM multilevel models
MM multiple myeloma
MMR major molecular response
MNAR missing not at random
MPFS Medicare Physician Fee Schedule
MRD minimal residual disease
MSM marginal structural model
MTA multiple technology appraisal
MTC mixed treatment comparison
MTD maximum tolerated dose
NBM negative binomial model
NCCN National Comprehensive Cancer Network
NCI National Cancer Institute
NCLA National Lung Cancer Audit
NCR National Cancer Registry
NCRS National Cancer Registration Service
NHB net health benefit
NHS National Health Service
NICE National Institute of Health and Care Excellence
NIHR National Institute for Health Research
NMA network meta-analysis
NMB net monetary benefit
NPV net present value
NSCLC non-small-cell lung cancer/carcinoma
OCS ovarian cancer-specific
QD once daily
OLE open label extension
OLS ordinary least square
ORR objective response rate
ORR overall response rate
OS overall survival
PAE potential adverse event
PAS patient-access scheme
PBAC Pharmaceutical Benefits Advisory Committee
PBM preference-based measures
PBS Pharmaceutical Benefits Scheme
pCODR pan-Canadian Oncology Drug Review
pCR pathological complete response
PCS prostate cancer subscale
PD progressive disease
PFS progression-free survival
PFS1 progression-free survival 1
PH proportional hazards
PIM promising innovative medicine
Acronyms and Abbreviations xxv

PP post-progression
PP per protocol
PRCT pragmatic RCT
PPRS Pharmaceutical Price Regulation Scheme
PPS post-progression survival
PR partial response
PS propensity score
PSA probabilistic sensitivity analysis
PSM propensity score model
PSSRU Personal Social Services Research Unit
QALY quality-adjusted life-year
QLQ Quality of Life Questionnaire
QoL quality of life
QTWiST Quality of Time Spent Without Symptoms of Disease and
Toxicity
RBRVS resource-based relative value system
RCC renal cell cancer
RCT random clinical trial
REC research ethics committees
RECIST Response Evaluation Criteria in Solid Tumors
RFA radiofrequency ablation
RP Royston-Parmar
RPSFTM rank-preserving structural failure time model
RT radiotherapy (see chemo-RT)
RWD real-world data
RWE real-world evidence
SACT systemic anti-cancer therapy
SAPs statistical analysis plan
SBRT stereotactic body radiation therapy
SD stable disease
STDev standard deviation
SDiff standardized difference
SE standard error
SG standard gamble
SMC Scottish Medicines Consortium
SNM structural nested model
SNNM structural nested mean model
SNS Sistema Nacional de Salud
SR surgical resection
STA single technology appraisal
TA technology appraisal
TACE transarterial chemoembolization
TEAE treatment emergent adverse events
THIN The Health Improvement Network
TKI tyrosine kinase inhibitor
xxvi Acronyms and Abbreviations

TOI Trial Outcome Index


TPS tumor proportion score
TSD technical support document
TTBT time to next treatment
TTF time-to-treatment failure
TTO time trade-off
TTP time to progression
UCLH University College London Hospitals Foundation Trust
VAS visual analog scale
VBP value-based pricing
VOI value of information
VSI value of sample information
WHO World Health Organization
WTP willingness to pay
YLL years of life lost
ZIN Zorginstituut Nederland
1
Introduction to Cancer

1.1 Cancer
The term ‘carcinoma’ is derived from the Greek word ‘karkinos,’ meaning
crab. Hippocrates associated cancer with the shape of a crab, because of the
way it spreads through the body and its persistent nature (Long, 1999).
Cancer is prevalent worldwide and impacts not only millions of people
but also their families, carers, health systems, and even employers. Cancer
impacts people’s physical, cognitive, and functional ability as well as their
health-related quality of life (HRQoL) and economic well-being. The National
Cancer Institute’s Dictionary of Cancer Terms (NCI, 2015) defines cancer as:

A term for diseases in which abnormal cells divide without control and
can invade nearby tissues. Cancer cells can also spread to other parts of
the body through the blood and lymph systems. There are several main
types of cancer. Carcinoma is cancer that begins in the skin or in tissues
that line or cover internal organs. Sarcoma is cancer that begins in bone,
cartilage, fat, muscle, blood vessels, or other connective or supportive
tissue. Leukaemia is cancer that starts in blood-forming tissue, such as
the bone marrow and causes large numbers of abnormal blood cells to
be produced and enter the blood. Lymphoma and multiple myelomas
are cancers that begin in the cells of the immune system. Central ner-
vous system cancers are cancers that begin in the tissues of the brain
and spinal cord.

1.2 Epidemiology of Cancer


An estimated 14.1 million new cases of cancer occurred across the world in
2018. The four most common types of cancers are lung, female breast, colorec-
tal, and prostate cancer (Bray et al., 2018). According to the Global Cancer
Incidence, Mortality and Prevalence study (GLOBOCAN) (Bray et al., 2018),
prostate cancer is the most commonly diagnosed cancer among males from

1
2 Economic Evaluation of Cancer Drugs

87 countries, especially in North and South America and northern, western,


and southern Europe. Lung cancer is the most commonly diagnosed cancer
among males in eastern Europe. Among females, breast cancer is the most
common cancer in North America, Europe, and Oceania. Breast and cervi-
cal cancers are the most frequently diagnosed cancers in Latin America and
the Caribbean, Africa, and most of Asia. However, the most common female
cancers in Asia also include lung, liver, and thyroid.
Due to more screening, earlier detection, and improved treatment, cancer
mortality rates are either plateauing or decreasing, particularly in the high-
income regions.
Table 1.1 summarizes the types of cancers and some key symptoms and
features, along with the common clinical and economic outcomes collected
in clinical cancer research.
These endpoints will be discussed in more detail in Chapter 2. For indo-
lent malignancies with long survival, other endpoints such as cytogenetic
response and minimal residual disease are used to assess the effectiveness
of new drugs, particularly in earlier lines of treatment. Surrogate endpoints
are also discussed in more detail in Chapter 2. For a single cancer type, there
are likely to be further subtypes (e.g. adenocarcinoma) for which some treat-
ments might work better for patients belonging to this subpopulation.

1.2.1 Cancer Trends
Mortality rates in several developing and low-income regions are increasing
for some of these cancers due to increases in smoking, excess body weight,
and physical inactivity. In 2011, there were nearly 8 million cancer-related
deaths. All cancers, taken together, are now a leading cause of disease-
related death worldwide, responsible for about 14% of the total of 55 million
deaths from all causes in 2011. Cancer incidence in the UK is reported to have
increased between 1993 and 2015 especially for females (Figure 1.1).
On the other hand, cancer incidence appears to be decreasing globally for
many cancers in the United States, Europe, and other high-income countries.
In low- to middle-income countries, the trend for cancers is unclear. Liver
cancer, however, is reported to be increasing globally. Table 1.2 provides a
summary of mortality trends for different cancer types between the years
2000 and 2019 (Hashim, 2016).

Example 1.1: Lung Cancer


Lung cancer is one of the leading causes of cancer-related deaths in the
world and accounts for nearly 1.4 million deaths per year worldwide,
with a yearly incidence of over 41,000 in the UK alone (Cancer Research
UK [CRUK] Statistics, 2012). More than 8 out of 10 lung cancer cases occur
in people aged 60 and over. Rates of lung cancer in Scotland are among
the highest in the world, owing to the high prevalence of smoking.
TABLE 1.1
Examples of Some Common Cancers Regarding Clinical and Economic Outcomes
Tumor Clinical Key Economic
Cancer Type Common Symptoms Key Features Outcomes Outcomes
Lung cancer Solid Cough, blood in sputum, pain, weight loss Short survival, PFS, OSa – Cost of treatment
diagnosed late – Radiotherapy
Melanoma Solid Changing mole, mass, symptoms due to Aggressive PFS, OS – Biomarker testing
distant disease spread – Palliative Care
Introduction to Cancer

– Quality of Life
Diffuse large B cell Solid Enlarged lymph nodes, pain, weight loss, Aggressive PFS, OS
– Nursing visits
lymphoma fever, night sweats, local symptoms due to lymphoma, short
– GP visits
enlarged mass e.g. intestinal obstruction survival without
– Hospital visits
treatment
– Physiotherapy
Chronic myeloid Blood Incidental diagnosis on routine blood tests, Indolent Cytogenetic aids/equipment
leukemia fatigue, bone pain, weight loss, sweats response – family support
Glioblastoma Solid Headache, vomiting, neurologic symptoms Aggressive, short PFS, OS – childcare costs
survival
Colorectal carcinoma Solid Blood in stools, pain, mass in abdomen, Aggressive, short PFS, OS
unexplained changes in bowel habits survival time
Hepatocellular Solid Nonspecific symptoms due to underlying Aggressive, short
carcinoma liver disease, mass in abdomen survival time
Gastric cancer Solid Mass in abdomen, pain, weight loss, Aggressive, short PFS, OS
vomiting, blood in vomit, symptoms due to survival time
obstruction
a Note:  OS: overall survival; PFS: progression-free survival.
3
4 Economic Evaluation of Cancer Drugs

FIGURE 1.1
All cancers excluding non-melanoma skin cancer, European age-standardized incidence rates,
UK, 1993–2015.

Source: CRUK Cancer Statistics.

TABLE 1.2
Summary of Countries by Cancer Type Showing Where Deaths from Each Type of
Cancer are Increasing/Decreasing
Cancer Increasinga Decreasing
All Brazil, Cuba, Latvia, Moldova, Serbia, Decreasing for other countries
and Malaysia
Stomach cancer Not increasing in any country Decreasing for all countries
Colorectal Latin America, Asia, South Africa, Decreasing for other countries
cancer Romania, Malaysia, Kuwait, and Latvia
Liver cancer North America, Asia, and Latin America Decreasing for other countries
Lung cancer Women: most countries: North America, Decreasing for: Ireland, Asian
Spain, Belgium, and Denmark countries, Lithuania, and
Men: Venezuela, Moldova, Malaysia, some Latin American
Serbia, Bulgaria, Portugal, and Romania countries
Breast cancer Japan/Korea, Malaysia, Philippines, Decreasing for other countries
South Africa, and Latin America
Uterine cancer Puerto Rico, Malaysia, and Philippines Decreasing for other countries
Prostate cancer Malaysia, Latvia, Serbia, Moldova, Decreasing for other countries
Ukraine, Belarus, USSR, and Korea
a Note:  See Hashim et al. (2016) for list of country studies.
Introduction to Cancer 5

Lung cancer incidence in a given country is directly linked with the


level of tobacco smoking in that country. Lung cancer-related deaths
occur approximately two to three decades after the widespread uptake
of smoking in any given country, with mortality trends approximating
the incidence trends. Among males, lung cancer mortality rates have
peaked and are now decreasing in many developed countries, reflecting
the uptake and subsequent decline in male smoking prevalence. Lung
cancer incidence in women lagged behind that in males because women
began smoking later.
In countries with the earliest uptake of smoking among women
(e.g. US, UK, and Australia), lung cancer mortality rates have peaked,
whereas they continue to climb in countries where women began smok-
ing later. Lung cancer is often diagnosed later in life, frequently with
aggressive disease progression leading to high mortality rates for this
cancer. In the 1950s, for every 1 lung cancer case diagnosed in women in
the UK, there were 6 in men. That ratio is now 3 cases in women for every
4 in men. The lowest lung cancer rates in the world for men and women
are in Northern, Western, and Middle African countries and South-
Central Asia; but this will also change if the current trends in the uptake
of smoking persist (Jemal et al., 2011; Toms, 2004; CRUK Statistics, 2012).

1.3 Prognostic Factors Associated with Cancer Outcomes


Prognostic factors are known or unknown factors that may be related to
either an increased or decreased chance (risk) of a (cancer-related) outcome
such as death (overall survival), disease progression, or any other outcome of
interest, including surrogate outcomes (Chapter 2). The relationship between
prognostic factors and cancer outcomes can influence the value of cancer
treatments. For example, if the survival benefit for patients with a poor prog-
nosis, defined by, say, an Eastern Cooperative Oncology Group (ECOG) per-
formance status of 3 or 4, is lower (in general, higher values of ECOG suggest
the patient has a worse prognosis) compared to patients with better progno-
ses (e.g. ECOG of 0 to 1), then ECOG would be considered an important prog-
nostic factor. The survival benefit may be greater in lower ECOG patients
(because patients might be relatively healthier or fitter for, say, surgery or
chemotherapy). Consequently, the treatment might be more cost-effective for
patients who are ECOG (0–1) compared to ECOG (2–3). Therefore, prognostic
factors play an important role in trial design considerations from both a clini-
cal and a health economic perspective.
One example of this is the use of a biomarker, a chemical test conducted
to determine the genetic disposition of a patient. The result of the test could
be related to a higher or lower clinical benefit. For example, the epidermal
growth factor receptor (EGFR) is one type of marker that a patient might
have. Patients with a known biomarker status (e.g. EGFR +ve for the drug
6 Economic Evaluation of Cancer Drugs

Erlotinib, used to treat lung cancer patients) are reported to have longer sur-
vival than those who are EGFR –ve. Despite the costs of the biomarker test
and other health resource use, including the cost of the drug, the targeted
treatment might still therefore offer greater value (be more cost-effective) for
future patients who test EGFR +ve.
Risk factors may not act independently but may be additive or multiplica-
tive in nature. That is, higher survival rates associated with ECOG status
might also depend on the ages of patients. It may be that better ECOG sta-
tus and age (younger patients might be relatively fitter) are associated with
higher survival rates compared to the survival rates of those who are older
and have poorer ECOG status. Hence treatment benefit may be dependent on
a combination of factors. These are called interactions. Such interactions can
play an important role in subgroup analyses from both a clinical and health
economic perspective.
Several risk factors common to different cancer types have been reported.
Some of these factors include age, genetic disposition (e.g. biomarker status),
smoking, lifestyle (e.g. insufficient physical activity, alcohol, diet), obesity,
and infections. These factors are associated with a high proportion of cancers
worldwide but may also vary by region or country. Smoking, in particular, is
the single most preventable cause of cancer death in the world; around a third
of tobacco-caused deaths are due to cancer. Excessive alcohol consumption is
reported to be associated with 13% of cancer-related deaths (Ferlay et al., 2010).

1.4 Economic Burden of Cancer


1.4.1 Health Expenditure
The constraint on healthcare resources, particularly during times of economic
turmoil and instability, may result in governments taking a hard look at all
public expenditure, including the medicine budget. Policymakers have a lim-
ited budget from which to decide how healthcare resources are provided for
its citizens. What this means in practice is that the ‘payers’ (i.e. governments
or health insurance providers who contribute toward the cost of healthcare
provision for their citizens or customers) are likely to be more selective and
choose with greater care from the healthcare options (i.e. new treatments)
available to patients (due to budget constraints). Just like most individuals
cannot have all the things they want, either because they do not have suf-
ficient resources (e.g. money, time), healthcare systems also have similar con-
straints when trying to meet the demands of its consumers (patients).
Figure 1.2 shows the worldwide healthcare annual expenditure between
2000 and 2013 as a percentage of gross domestic product (GDP) as reported by
the World Health Organization (WHO). It shows that health expenditure as a
Introduction to Cancer 7

12
10
8 %of GDP
6
4
2
0
1996
1997
1998
1999
2000
2001
2002
2003
2004
2005
2006
2007
2008
2009
2010
2011
2012
2013
year

FIGURE 1.2
Health expenditure as a fraction of GDP between 1996 and 2014.

Source: https ://da ta.wo rldba nk.or g/ind icato r/SH. XPD.C HEX.G D.ZS.

percentage of GDP has generally increased over the past 20 years, although
with some cyclical variation. Hence, the expenditure on health relative to
available resources (GDP) is increasing at a significant rate. An examination
of similar graphs per country shows an increasing expenditure trend for
developed and mid-sized undeveloped countries (WHO Report, 2014).

1.4.2 Healthcare Expenditure on Drugs


The economic burden of treating cancer is high worldwide. Cancer has the
most devastating economic impact in the world compared to other diseases.
The exact worldwide economic costs of cancer are unknown but are esti-
mated to be at least US $895 billion. With a growing and aging population,
prevention efforts are critical for reducing new cancer cases, human suffer-
ing, and economic costs. Since 1995, the cost of cancer drugs has increased
by an average of 10% annually (NCI, 2016). The largest economic burden for
cancer patients is often related to the direct cost of cancer treatment, namely
treatment drugs, and the costs to treat associated toxicities.
In 2014, the cost of every new cancer drug approved in the US exceeded
$120,000 per year of treatment. An American Society of Clinical Oncology
(ASCO) statement highlighted that in the decade from 2010 to 2020,
between $125 billion and $158 billion will have been spent on cancer care
(Lowell et al., 2015). Drug cost is a major (but not the only) cost component
associated with treating cancer. Other costs include the costs of treating
side effects, surgery, or radiotherapy, all of which can be significant. Given
that one in three people will be diagnosed with cancer in their lifetime,
the future cost of cancer treatment and care is likely to be a serious eco-
nomic burden.
In the UK, the annual economic burden of cancer is estimated to be
£15 billion. The National Health Service (NHS) increased its budget for
8 Economic Evaluation of Cancer Drugs

cancer drugs from £200 million in 2013 to an expected £340 million in


2015 (NHS Statistics, 2015), a 70% increase in the so-called Cancer Drug
Fund (CDF, NHS Statistics 2013, 2014). This represents the cost of cancer
drugs alone.
The CDF was set up in 2011 by the UK government to make funds available
for paying for cancer drugs. It was changed in 2016 as its form had become
economically unsustainable. One critical change was an explicit reference to
cost-effectiveness (CE) and resolving its uncertainty, which underlines the
importance of the costs of treating cancer:

Managed access agreements between NHS England and pharmaceutical


companies, setting out the terms of a drug’s entry into the CDF and the
means by which data will be collected to resolve any uncertainty relat-
ing to a drug’s clinical and cost-effectiveness. (NHS Statistics 2013, 2014)

One objective of government health departments is the desire to opti-


mize the use of cancer drugs by a combination of negotiated price reduc-
tions (of drugs) and improved clinical effectiveness. Some cancer drugs were
removed from the CDF list due to their lack of cost-effectiveness. The National
Institute of Health and Care Excellence (NICE), the UK body that publishes
guidelines on the value of new health technologies, has defined acceptable
cost-effectiveness thresholds as high as £50,000 per quality-adjusted life-year
(QALY). A QALY (see Section 1.6.1.8) is a composite measure of the length of
life and the quality of life experienced during this period. Hence, a survival
time of 1 year in perfect quality of life is 1 QALY, but if the quality of life was
scored at 0.5 (using a 0 to 1 scale, 0 being death and 1 being full health), the
QALY would be 0.5 (6 months). In 2016 the CDF underwent a review of which
one key objective was to ensure cancer drugs (in fact all drugs) offer strong
value for money (Cohen, 2017).
As an example, in the UK the total annual cost of treating lung cancer in
2012 was about £3 billion (20% of cancer costs) – the yearly average cost per
patient was £9,071. This was comparable to £2,756 for bowel cancer, £1,584
for prostate cancer and £1,076 for breast cancer (ACS, 2016). Therefore, the
costs associated with treating and managing lung cancer can be three
times higher compared to the other types of cancer. In the US, the mean
monthly cost of treating lung cancer patients was estimated at £1,669 (no
active treatment) and £5,814 for chemoradiotherapy (exchange rate of £1 =
$1.61).

Example 1.2 Costs and QALYs Reported in


Some Published Cost-Utility Studies
Table 1.3 shows the key results from 47 published cost-effectiveness
analyses in a lung cancer setting. About 20% did not report QALYs. Of
the 80% that did report the cost per QALY (36 out of 47), only 13 out
of 36 (36%) reported this to be below £30,000 (see Chapter 2 on ICERs).
Introduction to Cancer 9

TABLE 1.3
Published Costs and QALYs in Lung Cancer
Cost/QALY Source (See
Treatment Cost (£) QALY (£) Year Bibliography)
Paclitaxel 28,210 0.53 53,227 2011 Goulart et al.
27,902 0.923 30,230 2010 Brown et al.
21,967 NR NR 2000 Berthelot et al.
24,216 NR NR 2000 Berthelot et al.
26,228 NR NR 2000 Berthelot et al.
33,685 0.4513 74,639 2009 Klein, R.
Gemcitabine 27,837 0.934 29,804 2010 Brown et al.
27,401 0.966 28,365 2010 Brown et al.
18,129 NR NR 2000 Berthelot et al.
47,876 1.96 24,427 2013 Wang et al.
38,859 0.4676 83,102 2009 Klein, R.
Vinorelbine 23,516 0.888 26,482 2010 Brown et al.
16,678 NR NR 2000 Berthelot et al.
17,482 NR NR 2000 Berthelot et al.
6,901 NR NR 2010 Maniadakis
Docetaxel 4,129 0.1606 25,712 2012 Thongprasert et al.
13,956 0.206 67,748 2010 Lewis et al.
27,409 0.42 65,260 2010 Asukai et al.
24,798 0.225 110,215 2008 Araujo et al.
24,904 0.42 59,296 2008 Carlson
11,622 0.42 27,672 2011 Vergnenge et al.
20,903 NR NR 2011 Cromwell et al.
Pemetrexed 5,791 0.1715 33,767 2012 Thongprasert et al.
29,387 0.52 56,514 2010 Asukai et al.
27,764 0.241 115,205 2008 Araujo et al.
37,119 0.41 90,533 2008 Carlson
14,239 0.41 34,729 2011 Vergnenge et al.
17,455 0.97 17,995 2010 Greenhalgh et al.
41,731 0.5016 83,195 2009 Klein, R.
8,905 0.41 21,720 2012 Fragoulakis
Gefitinib 3,973 0.1745 22,766 2012 Thongprasert et al.
NR 1.111 NR 2010 Brown et al.
19,787 0.79 250,47 2013 Zhu
7,704 0.79 9,752 2013 Zhu
28,471 0.91 31,287 2012 Gilberto de Lima Lopez
8,980 0.2881 31,170 2010 Ontario Health
10,536 0.3188 33,048 2010 Ontario Health
Erlotinib 13,730 0.238 57,689 2010 Lewis et al.
22,439 0.25 89,756 2008 Araujo et al.
(Continued)
10 Economic Evaluation of Cancer Drugs

TABLE 1.3 (CONTINUED)


Published Costs and QALYs in Lung Cancer
Cost/QALY Source (See
Treatment Cost (£) QALY (£) Year Bibliography)
23,567 0.42 56,112 2008 Carlson
5,286 0.1745 30,292 2012 Thongprasert et al.
25,546 1.4 18,247 2013 Wang et al.
23,503 0.51 46,085 2012 Chouaid et al.
12,909 0.33 39,119 2013 Chouaid
8,104 0.42 19,296 2012 Fragoulakis
22,744 NR NR 2011 Cromwell
7,488 NR NR 2010 Bradbury et al.

In the UK, the cost-effectiveness threshold is set to between £20,000


and £30,000. The data in Table 1.3 shows the difficulties and challenges
involved in finding cost-effective lung cancer drugs. The variation
in costs and QALYs in Table 1.3 may be due to any number of factors,
including variations in populations, geography, clinical trial design (e.g.
patient follow-up), how costs were collected, and so forth. We will dis-
cuss these aspects in later chapters.

1.5 Treatments for Cancer


Common approaches for treating cancer (solid tumors) include (i) surgery,
followed by (ii) chemotherapy, and (iii) radiotherapy (though not neces-
sarily in that order). Despite treatment with chemotherapy, cancer recur-
rence is not uncommon. Recent novel chemotherapy (e.g. immunotherapy)
treatments use the body’s immune system to fight and kill cancer cells.
Although some of these drugs have proved to be cost-effective, others
have not (see later in Chapter 10 for examples). For blood cancers (e.g. leu-
kemia), surgery is not an option and chemotherapy is often the first choice
of treatment.
An example of treatment options for a patient diagnosed with non-
small-cell lung cancer (NSCLC) in the UK is shown in Figure 1.3. Often, a
particular treatment may be used for several different tumor types, often
resulting in similar side effects across tumors, with a marked impact on
HRQoL.
HRQoL and patient-reported outcomes are a central part of economic eval-
uation. (HRQoL will be discussed more fully in Chapter 3.)
Introduction to Cancer

FIGURE 1.3
Example of common treatment options for NSCLC: advanced or metastatic NSCLC treatment pathway based on NICE guidance CG121.
11
12 Economic Evaluation of Cancer Drugs

1.6 Important Economic Concepts for


Cost-Effectiveness of Cancer Interventions
Economic evaluation is the process of systematic identification, measure-
ment, and evaluation of the inputs and outcomes of two (or more) alternative
activities (health interventions) and their subsequent comparative analy-
sis (Drummond, 2002, 2015). Economic evaluation in the context of cancer
involves assessing the value of various cancer treatments, often through a
metric that combines the quantity (length) of life and the quality of life expe-
rienced during that time, called a QALY. This is particularly valuable when
some expensive cancer drugs demonstrate modest or small improvements in
survival (e.g. 1 or 2 weeks). In later chapters of this book, we will discuss the
importance of HRQoL for economic evaluation in a cancer context.
Concerns have been raised as to whether healthcare should be considered
an economic good (Morris et al., 2012; Santerre & Neun, 2000). Some think
these concerns are unfounded because, ultimately, healthcare resource is
finite and scarce. There is a limited supply of doctors, nurses, and healthcare
staff, so it is unlikely that any healthcare system in the world can achieve a
level of spending on the health of its citizens that would meet all their health-
care needs. In some cases, access to cancer treatments might be more readily
available in one region of a country and not in another, creating regional
equity issues (Chamberlain et al., 2015). It is reported, using real-world data
from registries, that women and those living in economically deprived areas
have less access to cancer treatment compared to other groups (Chamberlain
et al., 2015). Some have controversially argued that we can achieve health-
care for most individuals by not treating criminals, smokers, and alcoholics,
whereas others have argued that healthcare is a basic human right and that
it is not appropriate to treat it like an ‘economic good,’ as if it were any con-
sumer good or service (Morris et al., 2015). We will leave these questions to
the economic philosophers and policymakers because they involve compli-
cated judgments, which need not concern us here.

1.6.1 Economics, Health Economics, Economic


Evaluation, and Pharmacoeconomics
Economics is the science of scarcity and choice. Health economics is related
to the supply and demand for healthcare. Although health itself is not an
economic good, healthcare resource is an economic good. Healthcare
resource refers to items such as hospital beds, treatments, drugs, surger-
ies, GP time, and so on. These resources usually have costs associated with
them. For example, a visit to the doctor might be valued at £100 for one
hour. Government institutions might make decisions at the national level on
how the total healthcare demand and supply can be met for a given budget
constraint.
Introduction to Cancer 13

However, what is usually of concern in the pharmaceuticals context for


comparing new treatments is pharmacoeconomics. Pharmacoeconomics
uses certain principles of health economics for making policy decisions on
the supply and demand for medicines – particularly in clinical trials. The
methods (analysis techniques) used in pharmacoeconomics involve the
description and analysis of the costs of drug therapy to healthcare systems
and society. It identifies, measures, and compares the costs and consequences
of pharmaceutical products and services (Rascati, 2009). The process of using
these methods is called ‘economic evaluation.’ Economic evaluation applies
mathematical and statistical methods to compare the costs and consequences
of alternative healthcare options (Drummond 2002, 2005).
The term pharmacoeconomics might be considered as nothing more than
a generic term for economic evaluation of medical interventions (specifically
drugs). Rather than the cumbersome ‘economic evaluation of drugs,’ phar-
macoeconomics is a simpler term. However, pharmacoeconomics might also
address questions on affordability (and not just efficiency) of a new drug,
using budget impact models (BIMs). A BIM measures the net cumulative cost
of a particular treatment for a given number of patients in a specific popula-
tion. This is accomplished by implementing comparative cost-determination
analyses for competing scenarios, both including and excluding the product
of interest (Sullivan et al., 2014).
For over a decade, economic evaluation has become an increasingly impor-
tant component of clinical trials. The number of Phase III clinical trials with
a health economic component has also increased substantially. In particular,
submissions to reimbursement authorities – e.g. government-led subgroups
that assess the evidence for the ‘value’ a new treatment offers, such as the
Scottish Medicines Consortium (SMC) and the National Institute for Clinical
Health Excellence (NICE) in the UK, have also increased by more than 45%
over the same period.
In this book, we consider economic evaluation in the context of pharmaco-
economics, i.e. evaluating the costs and benefits of treatments from clinical
trials where data are collected prospectively in the trial. However, we also
extend this to determining costs and effects outside a controlled clinical or
experimental setting (in the real world). Economic evaluation uses various
techniques to assess the value of pharmaceutical interventions and health
strategies. In short, the terms ‘pharmacoeconomics,’ ‘cost-effectiveness,’
and ‘economic evaluation’ may be used interchangeably when describing
approaches to estimate the economic value of new cancer interventions.

1.6.1.1 Value
When it comes to healthcare, the average person has no idea about the price
for surgery or a treatment plan, especially when healthcare is considered to
be ‘free of charge,’ such as with the NHS in the UK. We might all be aware of
the price of diamonds or footballs, and most of us can value them (in relative
14 Economic Evaluation of Cancer Drugs

terms), or at least we are likely to put the price of diamonds higher than that
of footballs. Consequently, people may pay a premium price for diamonds
and similar goods. However, it is likely that, in a severe drought, someone
could well exchange diamonds for a cup of water (due to its scarcity).
Health products (or services) are not items we can buy ‘off the shelf.’
Therefore, it is harder to value and subsequently put a price on them. This
is true whether the health item is a new treatment or something as complex
as surgery. Some economists have suggested that the problem of value could
be determined by how much one was prepared to pay for a certain good,
assuming certain market conditions were satisfied. Economic theory was
then formulated to explain how value could be determined by the demand
and supply for goods (for a useful introduction, see Morris, 2012; Santerre &
Neun, 2000). Allocation of goods was determined simply by how much one
was prepared to pay (i.e. price paid) for that item – leading to French econo-
mist Jules Dupuit (Ekelund, 1999) to surmise in his 1844 paper that ‘the value
of a good is the amount someone is prepared to pay for it’ – or how much one
is willing to pay (WTP) for it.
In the context of health, the buyer is not necessarily the same as the con-
sumer. The buyer of the health product is likely to be a government institu-
tion or an entity responsible for healthcare provision. The consumer is the
patient. In other words, allocation of healthcare goods is based on the price
that governments (taxpayers) are prepared to pay. This is not necessarily true
in all countries however, and it will depend on the structure of the health-
care system of each country.
Valuing health, or for that fact, any product, by how much people are pre-
pared to pay does not take into account the impact on the wider society –
or the welfare of everyone. For example, richer people are less likely to suffer
from prices going up than the poor. This was not considered equitable, there-
fore, to address this, a further development in economics: (extra) welfare eco-
nomics took place, which need not concern us here (see bibliography for more
details). What is important for our purposes is to appreciate that the economic
evaluation techniques encountered in this book are tools for decision-making
to determine which treatments or health technologies offer the greater rela-
tive value in terms of a price (or cost) people (i.e. society) are prepared to pay,
termed the cost-effectiveness threshold.

1.6.1.2 Allocative Efficiency
Allocative efficiency is where health resources are deployed across an econ-
omy in the most efficient manner to match patient needs and preferences.
Allocative efficiency is where decision-makers use the evidence (results)
from economic evaluation to determine the best set of allocations of treat-
ments that gives an optimum for a given medicines budget. For example,
if only £100,000 were available for providing two cancer treatment options,
drug A and drug B, the question might be how best to spend the £100,000 on
Introduction to Cancer 15

these two treatments. If the price of drug A is £2,000 per year and B is £5,000
per year, one could treat 50 patients with treatment A or 20 patients with
treatment B. The other option is to have some patients take treatment A (so
long as it works) and some take treatment B. The exact mixture of treatments
A and B that will make up the £100,000 is for the decision-maker to deter-
mine. The comparative value that A and B offer will influence this decision.
Following the above example further, with £100 million to spend, treat-
ment A could be effective in patients with a particular genetic disposition
(biomarker). An optimal allocation might be to treat some patients who have
the presence of the biomarker (e.g. EGFR +ve) with the new treatment and
treat those that are EGFR-ve with the current treatment.
One technique for such comparative economic evaluation is called cost–util-
ity analysis (CUA), which seeks to address the question of (optimal) allocative
efficiency within the health sector. In the assessment of the cost-effectiveness
of cancer drugs, CUA is the one that is most pertinent. By using such methods,
it is hoped patients will have access to ‘value for money’ treatments through
an efficient allocation of various medicines subject to a ‘budget constraint.’

1.6.1.3 Technical Efficiency
Technical efficiency is when the minimum amount of resource (e.g. lowest
dose, or shortest duration of dosing) is used to elicit a given level of response,
i.e. when one produces a certain level of output with the least amount of
input – for example, the lowest dose that achieves a 20% reduction in lipid
levels. Economic efficiency occurs when the production cost of a given out-
put is as low as possible: for example, the least costly way of resolving a pep-
tic ulcer. Cost-effectiveness analysis (CEA) is one such tool used to address
the issue of economic efficiency.
In health economics, we distinguish between efficacy, e.g. whether a drug
or intervention actually works in a technical sense, which is mainly assessed
through randomized clinical trials (RCT) when feasible. Effectiveness is
whether the same intervention actually works in everyday operating condi-
tions, e.g. after market authorization or a license is given. Generally, (clinical)
effectiveness will be lower than, or at best equal to trial effectiveness due to
the inherent limitations of clinical trials.

Reimbursement
In the context of pharmaceuticals, once a drug has been approved for licensing
by the relevant regulatory body, such as the Food and Drug Administration
(FDA), European Medicines Agency (EMA), or some other national agency,
the pharmaceutical company will seek a price for its newly licensed drug.
Reimbursement, in simple terms, means the price the pharmaceutical com-
pany would like to obtain from the decision-maker (payer) for the new drug
it has produced. For example, the pharmaceutical company might want £120
per tablet, but the payer might want to pay only £95 per tablet, based on the
16 Economic Evaluation of Cancer Drugs

assessment of evidence of ‘value’ presented by the company. The price could


be on a per tablet basis or for a supply of 28 days – such as £50 per tablet,
or for 28 days £1,400. The price of, say, £50 is what the payer has agreed to
pay for each tablet. The price set is usually agreed between the payer (e.g.
the Department of Health in the UK) and the pharmaceutical company. For
example, the price per tablet of lenolidamide (in 2011) was agreed at £249.60
for a single 25 mg tablet. This price is recorded in publications such as the
British National Formulary (BNF, 2017).
In some countries, a dual-price system exists, whereby a market price is
set at a certain level and then, at a second stage, a reimbursement price is
set at a fraction of the market price. The difference between the two is the
price charged to patients; it constitutes the so-called patient’s ‘out-of-pocket
expense.’ The market price is typically calculated to cover the research and
development costs of the pharmaceutical company as a minimum, and to
make a profit to sustain future R&D activities. From the payer perspective,
the lower the price, the better. However, the price should not be set so low that
innovation is discouraged. A premium price is usually a price higher than
the current market price for similar existing products (for example when
there is a similar reference price set for all drugs within a therapeutic group).
A premium price may be awarded if the drug demonstrates improved ‘value
for money’ through economic evaluation techniques.
It is for this reason that, when a health economic evaluation is undertaken,
the payer perspective is considered carefully. That is, who is the economic
evaluation for? Is it from the perspective of a health insurance company (in
the US and some other countries, there is no equivalent of the NHS) or for the
local or national government (as in Spain where local provinces can influ-
ence decisions)? In short, who will be reimbursed?
In the UK, the Pharmaceutical Price Regulation Scheme (PPRS) is a volun-
tary agreement between the payer (the government) and industry with the
objective that (effective) medicines are available on reasonable terms to the
NHS, and this in turn maintains a strong, efficient, and profitable pharma-
ceutical industry. The workings of these groups are complex, but details can
be found in the official publications, such as those of the UK Department of
Health and Healthcare. Similar schemes exist for other countries. In France,
for example, the Health Products Pricing Committee (Comité Economique
des Produits de Santé) and drug manufacturer sign a number of agreements
allowing a variety of flexible means to monitor prices and drug use while
ensuring that public resources are properly allocated.

1.6.1.4 Opportunity Cost
A very important concept in health economics, and economics in general, is
the notion of opportunity cost. Opportunity costs are defined as “the value
of the next best alternative” (Polley, 2015; Folland et al., 1997). This applies
Introduction to Cancer 17

especially for health resources without market prices, such as informal care.
A shadow price is then derived from alternative marketed resources (for
example the cost of hiring a home-visiting nurse) to approximate the social
value of the non-marketed resource. However, market prices are only con-
sidered as adequate under ideal market conditions in perfect competition.
This may not apply to many resources in the healthcare sector. (For a recent
discussion on the application of opportunity costs to hospital bed days see
Sandmann et al., 2017.)
For some activities, like childcare, alternative market prices exist that yield
an upper price limit for these services; for others (e.g. the market price for
studying) one could use an opportunity cost approach by valuing the activ-
ity performed by the cost of forgone leisure time (this implicitly assumes
that everyone prefers leisure to work, or ‘work as punishment’). In this case,
a ‘proxy price’ needs to be established using contingent valuation methods,
such as willingness-to-pay or willingness-to-accept elicitation for non-mar-
ket, or some other stated preference method (Ryan, 2008; McIntosh et al.,
2010). Opportunity costs also arise in fixed-budget constraints.
Economic evaluation is often set against the background of an opportunity
cost when comparing treatments. In the context of medicines, for example,
assuming a fixed budget of £100 million, the payer may have the difficult
decision of allocating all £100 million to pay for drug A for 10,000 patients,
which might improve survival by 1 year (cost of £10,000 per year per patient).
The opportunity cost might be spending the £100 million on treatment B
for 20,000 patients, which might improve survival on average by 6 months
(£5,000 per year per patient). In practice, a combination of treatments A and
B may give an optimal allocation of available funds.

1.6.1.5 Discounting
Most people prefer to receive benefits sooner and pay costs later, rather
than sooner. For example, people prefer to enjoy smoking now and give less
importance to their future health. As someone becomes older, his or her time
preference may change.
In health economic evaluation, future costs and benefits are discounted so
that their value can be judged in present terms. This is achieved by applying
an annaul constant discount rate, e.g. 3.5% in the UK (based on the Treasury
Department’s so-called ‘Green Book’). The consequence of the discount
rate is that less weight is given to later costs than to the present costs. We
use the discounted values of future costs and benefits in cost-effectiveness
calculations.
Some cancer trials run for a long time and costs of any health resource
used at the beginning of a 7-year trial starting in 2015 may be different to
costs at the end of the trial, finishing in 2022. If the trial stops following up
patients after 4 years, costs might be determined at 2019 prices. However, for
18 Economic Evaluation of Cancer Drugs

the remaining 3 years (2020, 2021, and 2022), in each year, the future costs
would be discounted.
For example, the future costs of treatment for a single patient who experi-
ences disease progression in 2015 (and withdraws from the trial) in each of
years 2020, 2021, and 2022 are expected to be: £3,000, £5,000, and £7,000 (total
£15,000 over 3 years) – because the patient may have other subsequent treat-
ment or care. After discounting at 3% per year, the future costs are valued as:

Year 2020  £3,000 × 1/(1 + 0.03)1 = £2,912.62 1 year after withdrawal


Year 2021  £5,000 × 1/(1 + 0.03)2 = £4,712.98 2 years after withdrawal
Year 2022  £7,000 × 1/(1 + 0.03)3 = £6,405.99 3 years after withdrawal

The total future costs of £15,000 for this patient after discounting are
£14,031.59. In this example, only costs are discounted. In practice, health
benefits are also discounted. The debate about whether or not we should
discount costs only and not benefits or discount them at different rates, is
discussed elsewhere (e.g. see Drummond, 2002) and not considered further.
The current practice, however, is to discount both future health benefits and
costs. Note that if a trial is 7 years long (1-year recruitment plus a further
6 years follow-up) then discounting is important. However, if in a 7-year
trial where recruitment is over 6 years with a 1-year follow-up (e.g. a very
rare tumor), then a key concern is the application of a consistent price year.
Discounting is applied on expected future costs and effects beyond the first
year of follow-up.

1.6.1.6 The Incremental Cost-Effectiveness Ratio


The incremental cost-effectiveness ratio (ICER) is the basis of most economic
evaluations. It is a numerical quantity that expresses the relative (mean) dif-
ferences in costs between two or more treatments compared to the relative
effects. It is often written as a simple equation:

Mean Costs A - Mean Costs B


ICER = (1.1)
Mean Effect A - Mean Effect B
The numerator is called the mean incremental cost. The denominator is
called the mean incremental effect. The higher the value of the ICER, the
less cost-effective a treatment is (A vs. B). As can be seen from the denom-
inator, when two treatments are similar regarding their effectiveness, the
ICER is likely to be large. Hence cancer treatments that are cost-effective are
expected to show mean differences in effectiveness to be somewhat larger
than zero. The ICER is judged against a willingness-to-pay (WTP) threshold,
a term introduced earlier. The WTP is a value that is also referred to as the
cost-effectiveness threshold expressed by the term λ, or the shadow price.
The value of λ represents the cost in terms of health forgone elsewhere when
Introduction to Cancer 19

resources are reallocated within the healthcare system. In the UK, λ is set at
£20,000 to £30,000. If the ICER is <λ, then it is considered evidence the new
treatment is cost-effective. More recently, lower values of λ (e.g. £15,000) have
been used for promising treatments (Claxton et al., 2015).
As noted, when the denominator is very small, or zero, the ICER is very
large or undefined. When the ICER is negative, its interpretation is ambigu-
ous. A more useful approach might be to convert the formula in (1.1) into a
difference expressed in money value whatever the size of the denominator
(whether it is small or large). This could be used when drugs might be ‘simi-
lar’ (e.g. biosimilars):

Mean Difference Costs A − B


Expressing the ICER as = < λ (1.2)
Mean Difference Effects A − B
This can be rewritten as an incremental net monetary benefit (INMB):

INMB = mean Difference Costs A − B − λ ∗ Mean Difference Effects A − B (1.3)

If the INMB is >0, it means that the new treatment supports a hypothesis of
cost-effectiveness. Rewriting the ICER this way gets around the problem of
very small ICERs when treatments are similar, as is the case with a class of can-
cer drugs called biosimilars. Ideally, we would like a high chance or probability
that the INMB is positive (>0). This means the net benefit from a new treatment
(after taking into account differences in costs and how much one wishes to pay
for a new treatment) should have a high chance (e.g. 80%), that it is >0.

1.6.1.7 The Cost-Effectiveness Plane


In economic evaluation, the main results are usually reported in one of two
ways:

(i) The incremental cost-effectiveness ratio (ICER) and


(ii) The incremental net monetary benefit (INMB).

There is a relationship between the two that we identified above. In the previ-
ous section, the ICER was informally introduced as relative costs to benefits.
We now formally present the ICER in the context of the cost-effectiveness
plane, which is how the results of an economic evaluation are often reported
and interpreted.
The ICER is defined as:

Mean Costs (A) − Mean Costs (S)


(1.4)
Mean Effect (A) − Mean Effect (S)

= µ A − µ S / ε A − ε S = λµ A − S / ∆ε A − S = ∆ c / ∆ e
20 Economic Evaluation of Cancer Drugs

where D c = D mA – S and D e = D e A – S , the mean difference in costs and mean


difference in effects between treatments A and S, respectively.

Parameter Interpretation

µA Mean costs of treatment A

µS Mean costs of treatment S

εA Mean effect of treatment A

εS Mean effect of treatment S

∆µ A−S = ∆ c Mean difference in costs between A and S

∆ε A−S = ∆ e Mean difference in effects between A and S

The numerator in (1.4) expressed as ∆µ A−S , is called the incremental cost and
the denominator, ∆ε A −S , is termed incremental effectiveness; A and S are two
treatments (A is typically the new drug and S is the standard). It is this ratio
quantity ( ∆ c / ∆ e ) and the uncertainty around it that lies at the heart of eco-
nomic evaluation in clinical trials. This ratio is displayed on the cost-effec-
tiveness plane shown in Figure 1.4.
In Figure 1.4, the X-axis represents incremental effectiveness, or the mean
difference in effects between the treatments A versus S ( ∆ e ) . For example,
positive values of ∆ e ( ∆ e > 0) exist where the new drug is more effective.
Effective does not necessarily mean efficacy in equation (1.4). It could be a
measure of efficacy (e.g. survival time) combined with quality of life to get a
QALY (or life-years gained/saved – see Chapter 3). Negative values ( ∆ e < 0)
indicate the new treatment has worse effectiveness.
The Y-axis in Figure 1.4 is the mean difference in costs (∆c), measured in
some unit of currency (£s in this case). For example, if the new drug (treat-
ment A) costs £2,000 more than the standard (treatment S), the value of ∆c is
+£2,000. If ∆c is < 0 then the negative value means that treatment A costs less
than S, on average.
In quadrants 2 and 4 in, the decision as to which treatment is more or less
cost-effective is relatively easy. If the value of the ICER from equation (1.4)
lies in quadrant 2, the new treatment is cheaper and more effective. This
is the ideal scenario where pharmaceutical companies would like to have
their new drugs positioned. On the other hand, a very much less desirable
scenario is where the new treatment is worse, but also costlier (quadrant 4).
Values of the ICER can, however, be altered by changing parameters, such as
the price of the new treatment. Reducing the price (or increasing efficacy, if
possible) might be a strategy adopted so that the ICER can move into a differ-
ent quadrant in order to show a more favorable ICER – possibly at a reduced
profit. A new treatment that has an ICER falling into quadrant 4 is unlikely
to be considered as having a high chance of demonstrating value. Even if the
price was changed, the fact that the new treatment has poorer efficacy still
needs to be addressed.
Introduction to Cancer 21

New more costly


Increased efficacy
not worth increase 1
4 + c In cost
Trade Off
Old Dominates
= WTP

− e
New less effective New more effective
+ e

3 2
Trade Off Q (2, – , New Dominates
Reduced efficacy
worth the reduction
In cost
New less costly

FIGURE 1.4
The cost-effectiveness plane.

Example 1.3
Referring to Figure 1.4, we note that in quadrant 2, the point Q (2, –£10,000)
shows that a new drug is more effective (an improved effect of 2) and
cheaper by £10,000, on average. Hence the ICER is –£10,000/2 = –£5,000
per unit of effect (e.g. the unit could be QALY). The new treatment is said
to dominate the standard treatment. Most decision problems relating to
the value of the ICER are concerned with quadrants 1 and 3, and in par-
ticular quadrant 1 where justification of value is often sought.
The line that passes through the origin in Figure 1.4, denoted λ, is
called the willingness-to-pay or cost-effectiveness threshold. This is the
threshold ratio or amount in £ (or other currency) that a payer would be
prepared to pay for a new drug. Any ICER values calculated from data
that are to the right of this line (e.g. in quadrant 1) show that the new
treatment is cost-effective. In this example (Figure 1.4), the incremen-
tal effect value (∆e) is +2 and the incremental cost value (∆c) is –£10,000,
resulting in an ICER = –£5,000, shown as the point Q(+2, £–10,000) in
quadrant 2. Had the new treatment showed poorer efficacy compared
to the standard (e.g. a value of –2.8), the ICER would be –£10,000/–2.8
= £3,571. The ICER is now positive and has shifted from quadrant 2 to
quadrant 3 (Figure 1.4).

Example 1.4: Changing the Cost-Effectiveness Threshold


Figure 1.5 shows two slopes λ (dashed line) and λ* (solid line) – the cost-
effectiveness (CE) thresholds. The value of the CE threshold has changed
from λ = £30,000 (dashed line) to λ* = £12,000 (solid line). Initially, the new
22 Economic Evaluation of Cancer Drugs

New more costly


+∆c (£)
λ = WTP = £30,000

λ∗ = WTP = £12,000
Ζ (2, £28,000)

−∆e
+∆e
New less effective New more effective

−∆c (£)
New less costly

FIGURE 1.5
The cost-effectiveness plane: changing WTP/CE threshold.

treatment (not the same as in Example 1.1) showed a treatment benefit of


2 units, but it was more expensive (£28,000) – the point Z (2, £28,000) in
Figure 1.5. The point Z initially lies below the line λ = £30,000, but as the
value of λ = £30,000 changes to λ* = £12,000, the point Z is above the new
CE threshold. The observed slope, the ICER, is £28,000/2 = £14,000, which
lies to the left and above the new CE threshold. At this new threshold,
treatment A is no longer considered cost-effective (because the point Z
is above the line).
In general, in quadrant 1, if ∆c/∆e < λ, and so long as ∆e > 0, the new
treatment is cost-effective; values of ∆e > 0 suggest a benefit with the new
treatment. In quadrant 3, ∆c/∆e is always ≥0 (for ∆e ≠ 0), so the ratio is <λ,
and the new treatment is considered cost-effective.
In the CE plane, the value of the CE ratio (the ICER) needs to be com-
pared to various values of λ, and the number of points above or below
the line is difficult to visualize for changing values of λ.

1.6.1.8 Quality-Adjusted Life-Years (QALY)


A QALY is used as a generic measure of the utility of a (new) intervention on
the disease, which involves both the quality and the quantity (length of life)
lived. This is a key measure for assessing the cost-effectiveness of health-
care interventions. One QALY is interpreted as equivalent to one year of ‘full
health.’ If a patient’s HRQoL over the course of a year is less than in ‘full
health,’ the QALY will be less than 1. QALYs are often accumulated at a rate
of less than (or equal to) 1 per year. The EQ-5D, a generic HRQoL instru-
ment completed by patients (or carers), is often used to construct QALYs.
Introduction to Cancer 23

The EQ-5D is an important HRQoL measure and merits a separate discus-


sion (see Chapter 3).
In the economic evaluation of cancer, the QALY is a composite measure of
HRQoL and survival time. If the OS is the survival time of a patient mea-
sured from randomization (or the start of treatment) until death, or until the
last date the patient was assessed, the QALY combines the survival experi-
ence with the HRQoL. The HRQoL is often measured on a scale of 0 to 1,
where 0 represents a health state equivalent to death and 1 as ‘full health.’ In
some cases, a health state worse than death is also possible and a value less
than 0 is used. As a simplistic example, if the HRQoL for a patient who lives
for 6 months is 0.60, on average, then the QALY is 0.60 × 6 = 3.6 months. This
is like saying that 6 months of living in a less than full health state (i.e. an
HRQoL of 0.60), is equivalent to living 3.6 months in ‘full health’ or 3.6/12 =
0.30 years. In practice, the HRQoL is measured at multiple time points, and
an area under the curve (AUC) (time HRQoL curve) is constructed to derive
the QALY. This will be described further in later chapters.

1.7 Health Economic Evaluation and Cancer


Drug Development in Practice
Figure 1.6 displays the traditional drug development process and the
approaches to reimbursement and providing patient access. The square box
with broken lines in Figure 1.6 shows how the role of economic evaluation
has reshaped this process. In the past, less effort would be planned for dem-
onstrating the value of a new treatment. The traditional route was to perform
Phase I to Phase III trials, obtain a market authorization license and then
agree with each country separately a price for the new treatment. Evidence of

R
E
Clinical Trials Demonstrate Value I
A
M
Phase I P Cost-Effectiveness B
P U
R R Market
Phase II O Other studies S Access
V (synthesis) E
A M
L E
Phase III -IV N
T

FIGURE 1.6
Drug development and reimbursement.
24 Economic Evaluation of Cancer Drugs

‘value’ would not have been formally requested. Only efficacy concerns were
considered important at the time of pricing (not relative efficacy or costs).
Therefore, one approach to providing patient access to new drugs would be
to agree at the local (country) level, a price at which they would buy the
new drug – often agreed relatively shortly after the drug was approved. The
evidence for informing a pricing decision is often based primarily using the
Phase III clinical trial data submitted for market authorization.
In Germany, for example, the concept of ‘free pricing’ allowed innovator
companies to exert greater control over their prices and set them with con-
siderable flexibility. However, the AMNOG law in 2011 (Neuordnung des
Arzneimittelmarktes, see Bundesministerium für Gesundheit website) effec-
tively restricted free pricing for a one-year period only, with the pharmaceutical
company being required to assess value for money of the new treatment within
the first year. German payers no longer found it acceptable to pay for expensive
drugs that were seen to offer little value for money. In particular, oncology drugs
are likely to feel the impact of the Institut für Qualität und Wirtschaftlichkeit im
Gesundheitswesen (Institute for Quality and Efficiency, also known as IQWIG)
decisions more sharply because some of these drugs are particularly expensive
and have been reviewed judiciously from the perspective of demonstrating
value. Previously, approaches to providing patient (market) access to treatments
were not influenced by the concept of ‘value for money.’ There was a lesser need
both to formalize the health economic argument and to package the data in a
way that demonstrated the uncertainties of value for money.
In the US, drug manufacturers may be free to negotiate prices with payers
and insurance companies; however, recently some organizations have raised
concerns about ‘financial toxicity’ associated with cancer care in the US. In
June 2015, the American Society of Clinical Oncology unveiled its ‘concep-
tual framework’ to assess the value of new cancer treatment options, noting
that cancer care is one of the fastest-growing components of US healthcare
costs and the growth in healthcare spending, and stating that costs have not
been “accompanied by commensurate improvements in health outcomes”
(Lowell et al., 2015). This framework assigns a ‘net health benefit’ to oncol-
ogy therapies that take into account efficacy, toxicity, and cost. Indeed, in
addition to ASCO, the National Comprehensive Cancer Network (NCCN), a
not-for-profit alliance of 26 leading cancer centers, also recently decided to
include cost as a parameter in its guidelines. In addition to these professional
organizations, the Institute for Clinical and Economic Review (ICERev), a
non-profit organization, started publishing their own ‘value-based’ prices
for newly approved prescription drugs entering the market.

1.7.1 The Modern Paradigm


The modern drug development paradigm requires formal evidence of the cost–
benefit/value relationship. A multidisciplinary team (MDT) is set up that con-
siders as early as possible what is needed from the clinical trials data to form a
Introduction to Cancer 25

‘value’ argument. Although the analysis of data for economic evaluation occurs
after the Phase III trial results are finalized, the design and planning for both
efficacy and showing value must be considered well before then. The MDT
bridges the working relationship between individuals from clinical research,
biostatistics health economics, and other disciplines, to formalize the evidence
from clinical trials to obtain access for patients by demonstrating value.
The implication of Figure 1.6 is that if the innovator drug cannot demon-
strate value for money to the payer, the price desired may not be achieved.
This does not mean that a drug is not efficacious, but the decision-making
process should utilize all available information to minimize uncertainty.

Example 1.5: What Might Happen When Economic Evaluation


is not Considered in the Study Design Planning Process
A drug to treat myelodysplastic syndrome (MDS) was developed by a
manufacturer to reduce the need for blood transfusion in patients with
a specific subtype of the disease. Despite this benefit, the manufacturer
was unable to secure reimbursement for its drug. The reason for this
was because NICE determined that the price requested by the manufac-
turer did not offer sufficient value compared with patients who receive a
blood transfusion. Also, the health resource utilization related to blood
transfusion, e.g. hospital admissions for transfusion, day unit or clinic
chair times, long-term side effects of blood transfusion (e.g. iron over-
load and its side effects), etc., were not collected during the trial. This
routinely collected information may have added useful information to
the economic evaluation of the drug by showing not only a reduction in
need for blood transfusion but also a reduction in the health resources
utilization cost associated with blood transfusion.
In this example, the clinical research and manufacturing teams were
focused on producing a clinically excellent drug that reduced the need
for blood transfusion. However, the value argument was overlooked.
Had the company considered the reimbursement argument earlier the
story might have been different.
The relationship between the drug development program over time,
market authorization (getting a license), access for patients (as measured
by sales volume), and the importance of reimbursement is shown in
Figure 1.7. After license approval, there is a period between approval
and a decision for reimbursement. For most drugs, sales are usually
somewhat flat during the period between market authorization and
reimbursement. This is because reimbursement authorities are either
undecided as to whether the new treatment offers value, or a decision is
pending. However, when a decision has been made by a reimbursement
agency that the new treatment does offer value for money, the sales are
likely to increase (the difference between K and W is shown in Figure
1.7) because the new treatment has more value and would be recom-
mended for use. The premium price agreed would also influence prof-
its. The loss in revenue can be substantial if preparations have not been
made adequately for reimbursement.
26 Economic Evaluation of Cancer Drugs

Sales Time to reimbursement Reimbursement


Revenue should be minimized. Decision
(£) Phase III should consider all
plausible questions to
minimize further studies to
answer reimbursement
queries

Marketing
Authorization

Loss in revenue = £K – £W

Drug Development Process Time

FIGURE 1.7
Relationship between drug licensing, patient access, and reimbursement. W: Sales at some
amount £W are flat until £K is achieved (after reimbursement). Consequently, the loss of rev-
enue is £K – £W.

1.8 Efficacy versus Effectiveness


The concept of efficacy is well understood in the context of an RCT. The RCT
with its restricted inclusion criteria and very controlled monitoring of effi-
cacy and safety outcomes gives the RCT framework a ‘gold standard’ status
regarding internal validity. The possibility of selection bias is considered to
be well accounted for (Pockock, 1983). However, the RCT provides less exter-
nal validity because any inferences are restricted to the population under
investigation and lack generalizability (Sculpher, 2006). In most cancer trials,
efficacy is of paramount importance. Even if a new cancer treatment is not
cost-effective in the sense that a health system may not wish to pay for it, this
does not mean that the treatment does not work. The treatment may still be
bought privately, or health insurers may offer to pay for it.
Effectiveness, on the other hand, is where a new treatment is tested in
less restrictive (real-world) conditions. One objective of a clinical trial where
cost-effectiveness is a key component should be to measure some aspect of
the clinical effects of the new treatment in a real-world setting (if feasible).
Measuring efficacy in patients with comorbidities, longer-term follow-up,
alternative dosing regimens, and poorer patient compliance, may not have
been considered in the Phase III RCT. The target population of patients may
Introduction to Cancer 27

be broadly similar to those in the clinical trial, but additional questions need
to be addressed, such as “How well does the drug work in real practice?”;
“How well does the new treatment perform over a longer duration?”
If carefully designed, the RCT offers a good opportunity to collect such
data. For example, a clinical trial follow-up period might be suggested as
12 months; however, a follow-up of 24 months might offer an opportunity
to collect data in order to gain a sense of how the new treatment is working
in a real-world setting when double-blind, and some of the other restrictive
conditions, are relaxed. Also, a measure of compliance over a longer period
would provide valuable information on the use of the new treatment in prac-
tice – especially maintenance therapy, which, in cancer trials, can be particu-
larly expensive causing the ICER to be very large.
In RCTs, compliance is often closely monitored and highly protocol-driven
compliance rates may be artificial. True compliance may be as low as 60%, well
below some commonly stated compliance rates of 80% (often suggested for per
protocol analyses). Although the impact of a per protocol population (a popu-
lation of patients who are deemed to have complied with the protocol as far as
possible) is made on the efficacy endpoints, the importance of the protocol vio-
lators on costs are often not considered (Ordaz, 2013; Briggs, 2001; Noble, 2012).
The intention-to-treat (ITT) population, which usually includes all patients
randomized (as a minimum), is not always useful for assessing effectiveness.
If a patient is randomized but does not receive any randomized treatment,
should this patient be evaluable for treatment-related costs? For efficacy eval-
uation, the ITT principle is that analyses should be conducted based on the
randomized treatment (even if patients took the alternative treatment or did
not take any treatment). For cost-effectiveness analyses, costs (and effects)
incurred from a randomized patient, who did not take the medication (e.g.
costs associated with side effects of other drugs), may not reflect the true
cost-effectiveness of an intervention. Hence, a modified ITT (mITT) could
be defined whereby patients included for analysis must be randomized and
have received at least one dose or exposure to the randomized intervention.
In cancer trials, it can happen that the period between randomization and
treatment is long (e.g. several tests, biomarker status, radiotherapy planning,
illness). During this ‘waiting’ period a patient could deteriorate or progress.
How such issues of dropout and missing responses are handled can result
in biased estimates of treatment effects. This should be no less a concern
regarding estimates of cost-effectiveness.

1.9 Real-World Data
A more recent development related to effectiveness that has come about
recently is ‘real-world data’ (RWD). RWD often refers to data collected in real
health practice and not just later phase clinical trials with relaxed inclusion/
28 Economic Evaluation of Cancer Drugs

TABLE 1.4
Relationship between Types of Study and Design Features
Clinical Trial Clinical Trial (Phase
(Phases I–III) IV) Real-World Data
Objective Efficacy Effectiveness Longer-term effectiveness/
value
Design RCT Observational studies Observational and RCT
RCTs Retrospective
Retrospective Electronic health records
National/local cancer
registries
Population Protocol defined Broader population not Patients in routine clinical
in the main protocol practice
Measures Survival Survival Patient-based outcomes
Tumor response Tumor response Resource use
HRQoL HRQoL Impact on health economy
Patient-reported
Health resource use
Time frame Short term or long Long term Long term
term
Based on Ideal clinical Normal clinical practice Routine healthcare, hospital
practice (wider population) setting
(restricted
population)

exclusion criteria. Data are often collected in electronic medical records once
patients leave the clinical trial. For example, a cancer patient completes 12
months of follow-up in a RCT and then is monitored outside a clinical trial
through routine visits (e.g. for scans). These data are often held in scattered
local hospital records or possibly located centrally through a national reg-
istry – such as the National Cancer Registry (NCR). The NCR may collect
rich data on systemic anti-cancer therapies (SACT), and there may be an
opportunity to link this data with other routine data (general practice data,
hospital visits). These can be crucial for evaluating longer-term (real-world)
effectiveness of new cancer treatment – especially when a cancer treatment
has been given an accelerated approval using data based on a single Phase
II trial. There will still be uncertainty around longer-term effects and RWD
may help to reduce this.
Table 1.4 shows the relationship between study objectives and key features
for particular types of studies. The ‘gold standard’ to address confirmatory
efficacy is the RCT with primary efficacy and safety outcomes; the time
frame can be long term or short term – although longer trials (e.g. cancer,
cardiovascular, mortality endpoint) can become expensive to run. A study
with a primary objective of effectiveness (including economic evaluation)
may use a combination of evidence from RCTs or observational studies: out-
comes such as resource use (costs), quality of life (QoL), and compliance are
Introduction to Cancer 29

examples of the type of data collected in such studies to assess efficiency. In


practice, there may be a hybrid type of approach that optimizes the potential
to do as much as possible in a single trial.

1.10 Economic versus Clinical Hypotheses


Table 1.5 shows the relationship between the potential clinical advantage
envisaged in a clinical trial and how this can translate into a potential eco-
nomic hypothesis to demonstrate value. It is unlikely that an economic
hypothesis can be postulated unless some form of clinical advantage is plau-
sible. In some clinical trials, no clinical advantage is possible – such as ‘equiv-
alence’ trials where treatment benefits are considered to be ‘similar’ to, or not
worse than, a standard treatment. Bioequivalence trials are also equivalence
trials, and even though there might be a change in the mode of administra-
tion (e.g. where absolute bioavailability is required for intravenous vs. oral
dosing), this type of trial, usually with very few subjects, is unsuitable for
any economic evaluation because healthy volunteers are used, and clinical
benefit is not assessed.

(a) Hypotheses of Superiority

A superiority trial is when a new treatment is clinically better than the usual
treatment or current standard of care. The average treatment difference,
∆A–S, where A is the new treatment and S is the standard treatment. The
symbol ∆A–S represents a numerical value for the difference between treat-
ment A versus treatment S; this could be a mean difference, the difference
in proportions, or a hazard ratio. For a hazard ratio, used for time-to-event
outcomes (commonly for survival times), the lower (or upper 95% confidence

TABLE 1.5
Relationship between Clinical Objective and Plausible
Economic Hypotheses
Clinical Advantage Possible Economic Hypotheses
Superior efficacy Saves life years
Averts disease
Improved QoL/QALY gain
Better side-effect profile Improved QoL
Change in half-life More convenient administration
Improved compliance
Improved QoL/QALY gain
Improved delivery Better compliance
Improved QoL/QALY gain
30 Economic Evaluation of Cancer Drugs

limit) excludes the value of 1. When this happens, a new treatment is said to
be ‘superior’ to the comparator. For example, a 95% confidence interval (CI)
for a hazard ratio (HR) of 0.7 (30% less risk of death on treatment A compared
to S) of 0.41 to 0.95 is statistically significant because the value of 1 is not in
the interval (0.41, 0.95).
The value of ∆A–S (e.g. 0.70) should be large enough to postulate a cost-
effectiveness hypothesis. The value argument may depend on observed
differences in mean costs between treatment A and B relative to the mean
difference in costs. For example, an HR of 0.95 reported from a large trial
(n = 2,000 patients) might be statistically significant with a 95% CI of (0.89,
0.99). The value of ∆A–S = 0.95 suggests only 5% of patients more likely to
survive with the new treatment, on average. Whether this difference is large
enough to demonstrate cost-effectiveness is a separate question. This is an
example of a large trial with a small treatment benefit that is statistically
significant but that may not necessarily yield a clinical benefit that is cost-
effective. On the other hand, even if ∆A–S was large, but the costs associated
with this benefit were also high, then a cost-effectiveness argument may still
not exist, because the difference in costs may be too high relative to clinical
benefits. Table 1.6 gives a summary of how clinical hypotheses can be trans-
lated into cost-effectiveness statements.

TABLE 1.6
Summary of Hypotheses for the Primary Endpoint of a Trial
Average Difference Example of Possible
Hypothesis in (new vs. standard) at Cost-Effectiveness
Hypothesis Clinical Terms the End of Trial Argument
Superiority New treatment Improved with new Improved efficacy and
is better than (∆N–S > 0) possibly better safety
standard
Non-inferiority New treatment Improved with new On average, efficacy a little
is not worse (∆N–S > 0) better with new, safety
than standard better with new:
consequently new is more
cost-effective
Non-inferiority New treatment New is worse (∆N–S < 0) New is worse on average, but
is not worse not clinically worse; safety
than standard profile is much better with
new: cost-effectiveness
driven by better safety profile
Equivalence New treatment New is neither better or A variation of the above is
is not worse or worse possible
better than
standard
Note:  N, new; S, standard.
Introduction to Cancer 31

Example 1.6
In the above example, a difference between treatments in terms of sur-
vival reported an HR = 0.95. If drug A is $10,000 more expensive than S
(difference in costs of $10,000), the HR might translate into an average
survival difference of 1 week, or about 0.02 years (1 year divided by 52
weeks). The cost for each life-year gained here is $10,000/0.02 = $500,000.
That is, the relative cost of treating patients with a new cancer drug A
compared to the standard of care, S, costs $500,000 per year (for only a
1-week improvement in mortality). A payer may decide that $500,000 is
better spent elsewhere (for example, treating 100 dementia sufferers at
the cost of $500 per patient).

(b) Hypotheses of Equivalence: Biosimilars

Several cancer drugs, e.g. Herceptin, are termed ‘biologics,’ which are con-
sidered to be very expensive. Biologics are prepared through complex manu-
facturing processes (cells, DNA, proteins, tissue), which makes them difficult
to copy. Many chemical medicines are manufactured using a predictable
chemical process from which we can get an exact copy – these are called
generics. At the time of writing, a number of these drugs will come off pat-
ent and competition is underway to prepare generic versions of these, more
correctly termed ‘biosimilars.’ In this sense, biosimilars are not the same as
generics. To develop a biosimilar, a clinical trial is often needed with the
objective to demonstrate ‘similar’ or ‘equivalent’ efficacy and safety. The
biosimilar market is worth more than $11 billion. The European Medicines
Agency estimates savings to the health economy >1.5 billion (year 2009 esti-
mate) annually through the use of biosimilars.
If two treatments are equivalent regarding efficacy, then it would appear
that price and costs are the only driving force behind determining cost-effec-
tiveness. Given that some biosimilars are also considered to be expensive,
cost-effectiveness is a particular challenge when the clinical and statistical
hypothesis of interest is likely to be one of equivalence. The budget impact
on the health economy is likely to be important when considering a cost-
effectiveness argument between a choice of biosimilars.

Example 1.7
Consider the use of a biosimilar for Trastuzumab for breast cancer in
a Croatian population (Cesarec, 2017). The approach to demonstrating
cost-effectiveness was not made regarding improvements in efficacy,
but on the basis that the price of the biosimilar (test product) was 15%
below the reference (branded product). This led to the conclusion that
the Croatian health economy could save between €0.26 to €0.69 million
euros. In contrast, Brito et al. (2016) compared the drug Nivestim, with
a biosimilar for chemotherapy-induced neutropenia. They reported the
32 Economic Evaluation of Cancer Drugs

potential for greater cost-effectiveness in a secondary endpoint (febrile


neutropenia) using a cost-effectiveness model.
One approach here could be to use the method described in section 1.5
using equation (1.3) to derive the INMB since the denominator of equa-
tion (1.1) (the ICER) will be small in this scenario.

(c) The Hypothesis of Non-Inferior Equivalence

In this situation, the objective is to demonstrate that the new cancer treat-
ment is, on average, not worse than the current standard. In cancer trials,
such hypotheses would be rare, since patients are unlikely to enroll in trials
where there is an acceptance that a new treatment would result in worse
clinical outcomes. In this situation, as far as the new treatment is concerned,
a clinical advantage is unlikely or does not exist, and therefore cost-effective-
ness is unlikely – unless perhaps other secondary endpoints come into play,
or enhanced safety is observed (e.g. lower dose, leading to slightly lower
efficacy, but better safety). If there is a value argument, it is likely to be based
on ‘equivalent’ treatment benefit and lower costs, or improved safety.

Example 1.8
In this example, for treatment of infection, a twice-a-day regimen is
currently standard. A new once-a-day modified release formulation
is developed, which is a more convenient form of administration. The
value argument might be based on showing that ‘once-a-day versus
twice-a-day’ is likely to lead to better compliance and that it is cheaper.
The manufacturer would seek a premium price as a result of this added
value. The treatment effects might be similar or perhaps even worse
(although unlikely) with the once-a-day regimen. Since the costs associ-
ated with the new (once-a-day) regimen are likely to be lower, the formu-
lation with the lowest cost is likely to be more efficient (efficacy assumed
similar). An example of this situation might be a twice-a-day form of
clarithromycin (an anti-infective drug) versus a modified (once-a-day)
formulation.

1.11 Summary
In this chapter, we have discussed the importance of cancer from an epide-
miological and economic perspective. We have shown that expenditure on
cancer care is a challenge for almost any health economy. We also introduced
some important health economic concepts that we will refer to again in this
book. We have also shown that the old paradigm of obtaining a license from
the FDA, EMA, or other agency is unlikely to be sufficient and the value
Introduction to Cancer 33

of the new treatment needs to be demonstrated. Finally, we have shown


the relationship between clinical, statistical, and economic hypotheses and
how these need to be aligned so that the tools of economic evaluation can
be used appropriately. In the next chapter, we identify the key outcomes in
cancer trials, how they are derived, and their relevance and use in economic
evaluation.

1.12  Exercises for Chapter 1


1. A biosimilar drug is unlikely to demonstrate cost-effectiveness. Do
you agree?
2. What is a risk factor? Explain how a risk factor might influence the
cost-effective argument in each of breast, lung, and prostate cancers;
compare and contrast your findings. Are there any risk factors in
common?
3. The primary endpoint from a clinical trial is the only outcome that is
important for determining the value of cancer treatments. Discuss.
4. Biosimilar drugs are drugs that are aimed to show similar effective-
ness and yet remain expensive. Therefore, it is not possible to dem-
onstrate an economic advantage for these types of drugs. Do you
agree?
5. How would you decide on whether a new cancer treatment was cost-
effective in the following situations (assume two treatments being
compared against each other)?
a. The primary endpoint was very positive (i.e. a good outcome for
the new treatment) and all secondary outcomes were also better
for the new treatment.
b. The primary endpoint was very positive (i.e. a good outcome)
and all secondary outcomes were worse for the new treatment.
c. The primary endpoint was negative and all secondary outcomes
were worse for the new treatment.
d. The primary endpoint was no different between treatments but
all secondary outcomes were superior for the new treatment.
6. Is there a limitation in the way current economic evaluations of can-
cer drugs are performed based on your answers to the above?
2
Important Outcomes for Economic
Evaluation in Cancer Studies

2.1 Introduction
Cancer is a global health problem. There is an increased focus on oncology
research to discover and develop safer, more efficacious treatment options.
Well-designed clinical trials play an essential role in research and develop-
ment activities. A fundamental component of clinical trials research is iden-
tification of a measurable outcome to delineate clinical benefit of new cancer
treatments and further estimate the value they offer for patients and the
healthcare system.
Different types of clinical trial endpoints serve different purposes over
the phases of drug development. In early phase trials, the focus is to evalu-
ate safety and identify the maximum tolerated dose (MTD) or the minimum
effective dose (MED). In Phase I cancer trials, evidence of anti-tumor activity
is also investigated, followed by further trials that investigate preliminary
evidence of efficacy for designing later confirmatory trials.
Endpoints for confirmatory trials for drug registration (when a new
drug becomes available for general patient use by being issued with a
license) often define clinical benefit in terms of prolongation of overall sur-
vival (OS), progression-free survival (PFS), or an improvement in symp-
toms (FDA Guidance to Industry, 2018). In this chapter, we discuss the
importance of cancer endpoints and their relevance for economic evalua-
tion. Such endpoints can be grouped into two broad categories: (i) patient-
centered endpoints and (ii) tumor-centered endpoints. We start with a
discussion of common, surrogate, and emerging novel endpoints used in
oncology trials.

35
36 Economic Evaluation of Cancer Drugs

2.2 Important Common, Surrogate, and Novel Cancer


Endpoints
The two general categories of common cancer outcomes in clinical trials can
be grouped into are: patient-centered endpoints and tumor-centered end-
points (Fiteni et al., 2014). These are identified below and summarized in
Table 2.1.
Patient-centered endpoints

(i) Overall survival (OS)


(ii) Health-related quality of life (HRQoL)
Tumor-Centered Endpoints (Surrogate Endpoints/Intermediate Endpoints)
(iii) Progression-free survival (PFS)
(iv) Disease-free survival (DFS)
(v) Time to progression (TTP)
(vi) Time-to-treatment failure (TTF)
(vii) Event-free survival (EFS)
(viii) Time to next treatment (TTNT)
(ix) Objective response rate (ORR)
(x) Duration of response (DoR)
(xi) Tumor measurements

Tumor-centered outcomes may not always reflect the ultimate goal of the
therapy; that is, to increase life expectancy. In the case of an incurable dis-
ease, the objective may be to improve the HRQoL during survival as much
as possible (increase the QALY).

2.2.1 Overall Survival
OS is measured from the date of either randomization, registration (if not
an RCT), or start of first dose until the date of death (due to any cause). For
patients still alive by the time the trial has finished (or follow-up could not
be completed because the patient withdrew or was lost to follow-up), the
survival time is said to be ‘right censored.’ Hence, a patient’s survival time
may be censored at the date the patient was last known to be alive. This
also means the survival time for a patient that is censored is the minimal
survival time. Had the patient been followed up, survival time might have
been longer.
Survival data are often presented using Kaplan-Meier (KM) curves for
one or more groups. Figure 2.1 shows an example of a KM plot with several
types of endpoints, OS, PFS, and post-progression survival (PPS). The Y-axis
TABLE 2.1
Commonly Used Patient-Centered and Tumor-Centered Endpoints in Oncology Clinical Trials
Endpoint Definition Comments/ Issues and Relevance to Economic Evaluation
(i) Overall Time from randomization* until death from • Primary measure for estimating QALYs
survival (OS) any cause (or date of censoring) • Long-term survival often unknown, which is critical for longer-term evaluation
of cost-effectiveness
• Also considered gold standard by regulators for the purpose of drug registration
and approval
• Easily and precisely measured
• Affected by crossover and subsequent therapies
• May require large trial population or longer follow-up in case of less aggressive
cancer types
• Includes deaths unrelated to cancer
(ii) Health- HRQoL end-points measure physical and • Generic HRQoL used (may not be sensitive) for cost-effectiveness
related psychological status, participation in social • Rarely used as a primary endpoint
quality-of-life activities, and other indicators of well-being, • Tend to supplement other patient-centered or tumor-centered endpoints by
(HRQoL) such as the ability to work describing patient treatment experience
(iii) Progression- Time from randomization* until disease • Less important for cost-effectiveness although is needed to compute the
free survival progression or death post-progression survival period
(PFS)† • May be a marker of treatment duration and/or duration of benefit from
treatment (related to cost)
• Progression defined by several types of independent criteria such as RECISTa
• Smaller sample sizes and shorter follow-up time compared with OS
• Not affected by crossover or subsequent therapies
• Less influenced than OS by competing causes of death
Important Outcomes for Economic Evaluation in Cancer Studies

• Not influenced by treatments administered after progression


• No international consensus standard for the definition of PFS and DFS
• Requires frequent radiologic or other assessments
(iv) Disease-free Time from randomization until tumor • Requires balanced timing of assessment among treatment arms
survival (DFS) recurrence or any-cause death after • Less important for cost-effectiveness evaluation
treatments given with
37

curative intent
(Continued)
38
TABLE 2.1 (CONTINUED)
Commonly Used Patient-Centered and Tumor-Centered Endpoints in Oncology Clinical Trials
Endpoint Definition Comments/ Issues and Relevance to Economic Evaluation
(vi) Time-to- Time from randomization* to discontinuation • Useful in settings in which toxicity is potentially as serious as disease
treatment of treatment for any reason, including progression (e.g. allogeneic stem cell transplant)
failure (TTF) disease progression, treatment toxicity, and • Does not adequately distinguish efficacy from other variables, such as toxicity,
death therefore not used in the cost-effectiveness assessment
(vii) Event-free Time from randomization* to disease • Initiation of next therapy is subjective. Generally, not encouraged by regulatory
survival (EFS) progression, death, or discontinuation of agencies because it combines efficacy, toxicity, and patient withdrawal, therefore
treatment for any reason (e.g. toxicity, not used in the cost-effectiveness assessment
patient preference, or initiation of a new
treatment without documented progression)
(viii) Time-to- Time from end of primary treatment to • For indolent or incurable diseases, TTNT may provide a meaningful endpoint for
next treatment institution of next therapy patients.
(TTNT) Rarely used as primary endpoint as TTNT is subject to variability depending on
subsequent treatment options available for patient and physician
(ix) Objective Proportion of patients with reduction in • Measures direct effect of drug in objective fashion
response rate tumor burden of a predefined amount • Earlier assessment compared with survival endpoints
(ORR) • RECISTa or other relevant criteria applied
(x) Duration of Time from documentation of tumor response • Response to treatment may not result in better survival, therefore not a
response (DoR) to disease progression comprehensive measure of drug activity
• Commonly used in Phase I or Phase 2 trials
• Extrapolation of response rate and duration of response to survival is required,
however, due to single arm design of most of these trials, indirect comparison
with either historical control and/or best supportive care is performed
(xi) Tumor Often by RECIST or similar criteria • Used for solid tumors (RECISTa criteria)
measurements • Not used for confirmatory trials; often used in Phase I or II trials
• Useful for identifying anti-tumor activity
a Eisenhauer et al., 2009. RECIST: Response Evaluation Criteria in Solid Tumors. This is a criterion which determines how much a tumour has shrunk.
This criterion is used for solid tumours and not blood/haematological tumours. The criteria are shown in Appendix I based on RECIST version 1.1.
Economic Evaluation of Cancer Drugs

*  In nonrandomized trials, time from study enrollment or treatment initiation is used.


†  TTP and PFS are similar, with the exception that TTP does not include patients that die from other causes (e.g. cardiovascular events)
Important Outcomes for Economic Evaluation in Cancer Studies 39

FIGURE 2.1
Example of time-to-event curves.

shows the proportion of patients alive at a time point. In Figure 2.1, about
70% of patients are still alive at around 9 months. One important statistic
used to measure clinical benefit is the median survival time. The median OS
in Figure 2.1 is about 20 months (draw a horizontal line starting at 0.5 on the
Y-axis, until it meets the OS curve). This means that by 20 months, half (50%)
of the patients are still alive and 50% have died. The median PFS (brown line)
is about 4 months.
Comparing median survival times between treatments is a common way
of showing clinical benefit in cancer trials and is useful when such effects
are unambiguous. An alternative measure of treatment benefit might be to
compare the proportion of patients alive at a fixed time point. In Figure 2.1,
at 9 months, around 70% of patients are still alive. This value could be com-
pared with patients in a control treatment group. However, for comparison
of the survival rates over the entire KM curve (i.e. comparing the curves) an
alternative, more complicated, statistic called the hazard ratio (HR) is often
reported. An HR of 1 implies there is no difference between treatments in
terms of the event of interest. When the HR is either <1 or >1, then the sur-
vival (event) rates for one treatment, on average, are either higher or lower
compared to the other. One difficulty involved in interpreting such effects
occurs when the KM curves cross (Figure 2.2). This is called a nonpropor-
tional hazard and essentially means that treatment differences are not con-
stant across time and may depend on other factors.
40 Economic Evaluation of Cancer Drugs

FIGURE 2.2
Survival curves for comparing concurrent versus sequential chemoradiotherapy.

Source: Maguire et al., 2014.

In Figure 2.2, treatment consisting of either concurrent or sequential


chemoradiotherapy (chemo-RT) shows that prior to 12 months the sequential
treatment group showed higher survival. After 12 months, however, due to
the intensity of treatment, survival became worse than concurrent chemo-RT
(see Maguire et al., 2014). This is an example where the area under the KM
curve(s) can be used to estimate the mean OS for economic evaluation – the
so-called restricted mean.
The approach to measuring treatment benefit described here can also be
used for most time-to-event endpoints such as PFS, DFS, TTP, TTF, EFS, and
TTNT. An important point to mention here is that whereas clinicians use the
median OS as a measure of clinical benefit, for cost-effectiveness the mean
OS is used. This can be calculated as the area under the KM curve in Figure
2.1. It is sometimes referred to as the restricted mean because the final time
point at which all patients die might be unknown by the end of the trial,
and hence the area is calculated for a restricted set of survival times. We
will discuss further statistical issues for analyzing survival data for cost-
effectiveness analyzes purposes in later chapters.
One final point to mention on the use of OS is that the required sample
size can be large. Sample sizes in cancer trials (for time-to-event endpoints)
depend on the number of events. The events here are deaths (due to any
cause). Clearly if we had a trial with 1,000 patients per group and only
5 deaths after 1 year, the statistic of interest would not be of much use if
Important Outcomes for Economic Evaluation in Cancer Studies 41

patients died mostly after 2 years. In this case there would be substantial
censoring after 1 year making interpretation of the KM curve less useful,
and estimates of the median (and mean) OS may not be calculable. The lack
of events also has implications for estimating survival patterns over a much
longer time horizon. Using OS as an endpoint may therefore require wait-
ing for a long time to achieve the required number of events, resulting in lit-
tle or no information on longer-term effectiveness (because if death events
take a long time anyway, further longer-term effects will take even longer
to evaluate). The consequence of this might be that decisions on provid-
ing access may be delayed due to uncertainty around the longer-term cost-
effectiveness of the intervention. This is neither beneficial to patients nor
to industry. In this case special statistical methods can be used to estimate
(extrapolate) long-term survival using complex models (parametric survival
models). Moreover, such estimates of long-term survival can be confounded
by the effects of additional or subsequent treatments taken after disease
progression.

2.2.1.1 OS and Economic Evaluation


There are several factors relevant to economic evaluation when using OS in
cancer trials:

(i) Whether the OS benefit is achievable

OS is often the main endpoint that is of interest for drug licensing and for
payers. Since cancer often results in early death (shortening lifetime), the
value of a new treatment must be demonstrated in terms of extending OS.
OS is readily accepted by patients and oncologists as evidence for improving
patient benefit. In addition, payers of drugs, whether through health insur-
ance or through local or national health systems, value OS as an endpoint of
importance for assessing whether to pay for any given cancer drug. About
one-third of approved cancer drugs come to market on the basis of reporting
improvements in OS through randomized controlled trials (Kim & Prasad,
2016). However, OS is not so straightforward a measure when it comes to
assessing cost-effectiveness, even if some form of clinical benefit has been
demonstrated.
One limitation of OS is the low likelihood of showing large improvements
in OS, especially in elderly patients where some cancers are diagnosed at a
much later age (and stage). Fojo et al. (2014) reported median improvements
in overall survival from confirmatory trials to be just 2.1 months (Kumar,
Fojo, & Mailankody, 2016); colleagues examined 47 consecutive approvals
for cancer drugs and found that only 9% showed an absolute increase in OS
by 2.5 months (91% showed increases of less than 2.5 months). Even if an OS
difference was shown to be statistically significant, this ‘significance’ does
not imply it is a clinically meaningful benefit and moreover it may not have
42 Economic Evaluation of Cancer Drugs

high economic value, especially if the price for a drug is high. In the UK,
value thresholds are commonly set at £20,000–£30,000 per QALY (McCabe,
2008; NICE Guidelines, 2013). In some cases, reaching this hurdle is unlikely,
as shown in Example 2.1.

Example 2.1: QALYs Reported in Some Published Cost-


Effectiveness Analyses for Lung Cancer Using OS
In Chapter 1, Table 1.3 showed that among the 47 ICERs identified from
lung cancer trials, about 70% did not yield QALYs below the NICE
required thresholds of £20,000–£30,000 per QALY. The implication
of such thresholds (say £24,000) is that the new treatment should not
cost more than £2,000 per patient per month (or £2,500 per month for
a £30,000 cost-effectiveness threshold). This reinforces the challenge to
researchers and clinical trialists when designing cancer trials for both
efficacy and cost-effectiveness.

(ii) Clinical Trial and Real-World Setting

Clinical trials for registration (trials that demonstrate evidence for efficacy
for licensing of a drug) are often performed in highly selected populations,
unrepresentative of the general target population. Differences in the mag-
nitude of treatment benefit between experimental (protocol) conditions and
real-world settings can be explained in part by the type of (highly selected)
patients that present and the nonrandom choice of trial centers and physi-
cians. Economic evaluation of cancer drugs is of greatest interest when used
in a real-world or routine clinical practice context and often over a lifetime
horizon.
Data on survival from a real-world setting often means in tracking patients
well beyond trial follow-up. Such tracking might involve using national
(public) cancer registries and possible private data. A recent development
has been the use of private enterprises involved in working with public
sector institutions to help extract outcomes collected retrospectively from
routine hospital and/or clinical practice databases. One difficulty with data
collected outside clinical trials, such as cancer registries, is that there may be
little or no information on what other treatments were taken that might have
impacted the survival, nor what toxicity was experienced. Patient-reported
outcomes too, an essential data component for economic evaluation, may not
be available. It is better to plan for such real-world collection rather than ‘get
lucky’ from what may or may not be in scattered data registries. It is impor-
tant to note that regulatory agencies for marketing authorization may not
consider registry data as a basis for proof of efficacy, although this might be
acceptable for reimbursement agencies.
An important consideration is the recent General Data Protection
Regulation (GDPR) (EU) 2016/679 directive within the European Union,
which intends to primarily give control back to citizens and residents over
Important Outcomes for Economic Evaluation in Cancer Studies 43

their personal data. This includes the use of real-world clinical data. How
this law will impact access to outcomes needed for longer-term survival
effects is not immediately clear. Often though, by anonymizing the data, suf-
ficient privacy protection for patients is possible.

(iii) Incomplete Follow-Up Data

Complete information on OS may not be available until the last patient in the
trial dies. This might only happen for some cases where the life expectation
is not too long (see Wang & Li, 2012). As pointed out earlier, this may lead to
(right) censoring of patients who have not died at the time of analysis or end-
of-study follow-up, resulting in less statistical power to detect differences
between groups. This runs the risk of inconclusive results. For economic
evaluation, estimates of OS are required over the lifetime of patients, taking
into account those still alive at the end of the trial. Hence censoring impacts
both costs and effects in an economic evaluation. Methods are available to
adjust for censored costs (e.g. see those described in Khan, 2015; Menon et al.,
2017). The method of Lin (Lin et al., 1997) is one such method. This method
provides an estimate of the mean cost by taking into account patients who
are followed up to a particular time point and then are lost to follow-up (cen-
sored). The estimate of mean costs uses Kaplan-Meier methods (Chapter 4)
to generate weights that are multiplied by the mean costs for specified inter-
vals. A worked example is provided in Chapter 5 (Example 5.7).

(iv) Confounding from Subsequent Anti-Cancer Therapies

Estimates of long-term survival can be confounded by the effects of addi-


tional treatments taken after disease progression. In patients with advanced
or metastatic disease (not amenable to curative surgery), successions of dif-
ferent lines of treatment are employed. For example, initially a first-line treat-
ment will be given and after some time patients may progress (see Figure
2.3); this will be replaced at some point by different treatments. Figure 2.3
shows how some of the important outcomes relate to the various lines of
therapy. The second line (or choice) of therapy for example, starts when the
first line of therapy fails, often determined by when disease progression
occurs. It is the use of these subsequent lines of therapies that may confound
the estimates of OS.
In most, if not all cases, the subsequent treatment options will be less stan-
dardized (patients may receive different doses or regimens that impact OS in
different ways). For economic evaluation, this creates challenges in handling
the heterogeneity (variability) of these differing regimens in the way they
influence OS. This issue also extends to estimates of costs from taking other
anti-cancer treatments – specifically the costs associated with side effects
and the administration of later lines of therapies. When PFS is a primary
endpoint, the need for estimating longer-term OS becomes important for
44 Economic Evaluation of Cancer Drugs

FIGURE 2.3
Relationship of different surrogate endpoints and overall survival.

Source: Adapted and modified from Matuolonis et al., 2014.

estimating the ICER. Hence, although PFS might be acceptable for licensing,
OS is still needed to determine the value of a new cancer drug. OS, costs, and
QALY estimates can be biased if the impact of subsequent treatments is not
considered. This leads to the issue of adjusting effects for ‘crossover.’

(v) Crossover or Switching from the Control Drug to the Experimental


Drug

Slightly different to taking additional anti-cancer treatments after progres-


sive disease (PD) is the issue of patients being allowed to cross over to an
experimental treatment once the primary endpoint (e.g. after the target
number of events has been reached or disease progression has occurred) or
follow-up is complete. This may happen for ethical reasons to avoid delay-
ing potentially effective treatments for trial patients. If patients who crossed
over to the experimental treatment are excluded from the analyses, selection
bias may exist because those patients who switched from the control arm
to the experimental treatment may not be representative of patients in the
entire control arm. It has been noted that over half the health technology
assessments (HTAs) have been affected by some form of treatment switching
(Latimer, 2015).
Although crossover may confound and bias the treatment benefit, it may
be argued that since statistical analysis follows the ‘intent to treat’ (ITT) prin-
ciple (analyze patients according to what they were randomized to and not
Important Outcomes for Economic Evaluation in Cancer Studies 45

what they actually took), crossover is what is likely to happen in routine prac-
tice and may be a more realistic assessment of treatment benefit, despite the
potential biased estimate of treatment benefit. Moreover, several statistical
methods used in some cost-effectiveness analyses that attempt to adjust for
treatment switching have been rejected by decision-makers (Latimer, 2015).
The impact of switching on the ICER and QALY can be significant because
the OS benefit can be either over- or underestimated. As an example, the
NICE HTA TA269 (NICE, 2012) reveals how switching had a large impact on
the cost-effectiveness results in a melanoma trial. In this submission (vemu-
rafenib), adjusting for switching in 34% of control group patients, reduced
the ICER from £75,500 per QALY to £51,800 per QALY. The adjusted analy-
ses were considered acceptable resulting in vemurafenib’s recommendation
for reimbursement, despite the ICER being (marginally) above £50,000 per
QALY (used for end-of-life settings).
An example to the contrary involves an HTA of everolimus from the
RECORD-1 confirmatory trial in patients with advanced renal cell cancer
(RCC). The decision-makers felt that the estimates provided by the manu-
facturer’s economic model were overly optimistic and instead suggested a
smaller overall mortality benefit based on alternative statistical estimates –
adjusting for crossover. The decision-makers noted:

any estimate of OS obtained using statistical modeling would be subject


to some uncertainty because a number of assumptions would have to
be made.

Everolimus for advanced RCC was not considered to offer value because
the magnitude of its effect was highly uncertain, as were estimates of cost-
effectiveness, with several analyses showing cost-benefit ratios that exceeded
NICE’s standard recommended thresholds (£20,000–£30,000 per QALY).
In a further example, in TA381 (NICE, 2013) for the treatment of BRCA +ve
(a biomarker), platinum-sensitive, relapsed ovarian cancer, the hazard ratio
for PFS was 0.18 (95% CI 0.10–0.31, p < 0.0001) for olaparib versus placebo.
The OS was not significantly better (p = 0.19). There was some crossover after
disease progression and, after crossover-adjusted analysis, the treatment dif-
ference was statistically different (p = 0.039). However, although the analysis
adjusted for licensed treatments, it did not correct for unlicensed treatment
with an experimental drug (olaparib) beyond disease progression (patients
on the control arm switched to olaparib).
Additional work undertaken by the review group suggested that the incre-
mental cost-effectiveness ratio (ICER) for olaparib versus routine surveillance
in BRCA mutated platinum-sensitive, relapsed ovarian cancer patients who
received >2 lines of chemotherapy, was likely to be greater than £92,214 per
QALY gained. The manufacturer’s economic model produced a higher esti-
mate of effectiveness (1.43 QALYs) without adjustment for treatment cross-
over compared to that generated by NICE experts, found to be 0.52 QALYs
after adjusting for crossover (and hence the ICER exceeding £92,214/QALY).
46 Economic Evaluation of Cancer Drugs

Although OS has limitations, it is nevertheless a strong objective endpoint


that has clear meaning and value to both patients and clinicians. Where a
disease is known to shorten the lifetime of patients, an improvement in OS is
still the most important outcome when it can be measured.

2.2.2 Surrogate Endpoints
There is no agreed definition of surrogate endpoints. According to the National
Institute of Health (NIH) Biomarkers Definitions Working Group (NIH, 2001),
a clinical endpoint is a characteristic or variable that reflects how a patient feels,
functions, or survives, and a surrogate endpoint is defined as a biomarker or
intermediate endpoint intended to substitute and predict for a patient-relevant
final endpoint (Ciani et al., 2016).
In the absence of OS, surrogate or intermediate endpoints are used in the
majority of clinical trials as indirect measures of clinical benefit for several
reasons:

(i) One reason is because surrogate endpoints can generally be achieved


in a shorter time frame than OS. For example, progressive disease is
a prelude to death. An event of ‘disease progression’ will occur ear-
lier than death.

PFS and objective response rate (ORR) are the most commonly used tumor-
centered surrogate or intermediate endpoints in cancer trials. PFS is defined
as the time from randomization or patient enrolment (if not an RCT) until
first disease progression or death. Disease progression is determined by
either clinical signs and symptoms (which can be subjective) or objective
criteria such as those of the Response Evaluation Criteria in Solid Tumors
(RECIST) – (Eisenhauer et al., 2009). Other similar criteria exist for nonsolid
tumors.
The purpose of such criteria is to remove the possibility of bias when
judging patients to have disease progression. Typically, RECIST requires
measuring the tumor dimensions and calculating an approximate area. The
target or primary tumor of interest is measured at baseline (before treat-
ment starts) and post-baseline (after treatment is given). The difference
between the two measures is expressed as a percentage and the amount
of reduction is classified as either complete response (CR), partial response
(PR), or stable disease (SD).
If the tumor size increases or evidence of new lesions is observed in any
other part of the body (metastases), this is called progressive disease (PD).
PD is judged against the minimal (the nadir) of previous measures, whereas
response is always assessed when comparing post-treatment tumor mea-
sures with baseline measures. The exact timing of the progression is often
unknown. Discrete assessment points (e.g. every 3 months) for clinical or
radiological assessment are used in practice and therefore the PD is interval
Important Outcomes for Economic Evaluation in Cancer Studies 47

censored. The actual time of PD is therefore also design dependent – depen-


dent on the schedule of assessments and the growth rate of a cancer. Given
the open label design of many oncology clinical trials, PFS may be subject to
assessment bias. Discrepancies between investigator and independent adju-
dication of response need to be minimized.

(ii) Fewer Patients and Trials of Shorter Duration

An advantage of surrogate endpoints is that successful treatments can be


identified much earlier compared to OS and therefore reach patients and pro-
viders more quickly. Such intermediate endpoints are most frequently used
when it would be impractical to follow patients for a long time (until death),
as in for example indolent, less aggressive cancer types or patients in the early
stages of disease. This may also help to reduce the cost of the trial and be
commercially beneficial for the manufacturer (less costly trials). Depending
on factors such as effect size, duration of effects, and the comparative ben-
efits of other available treatments, surrogate endpoints can lead to accelerated
approval by regulatory authorities. An example of approved drugs using sur-
rogate outcomes is venetoclax for treating chronic lymphocytic leukemia.
In reporting the results of venetoclax, it was noted that:

The committee was concerned that the single-arm design of the trials
made it difficult to assess the efficacy of venetoclax (that is, there was no
comparator arm of patients having best supportive care)
The committee was aware that in M14-032, neither the median pro-
gression-free survival nor median overall survival had been reached,
and that because there was uncertainty associated with the efficacy of
venetoclax, the European Medicines Agency had granted the market-
ing authorization for venetoclax conditional on the company submitting
more mature data from M14-032, which is due to report in March 2018…

Hence, approval was based on a single-arm trial conditional on more


mature data being available. However, for cost-effectiveness, extrapolation
was used to predict future survival patterns. Consequently, even if market-
ing authorization is based on a combination of surrogate and other outcomes,
cost-effectiveness will still be evaluated over a lifetime horizon.

(iii) Translation into Final Outcomes

For economic evaluation, surrogate endpoints may need to be translated into


final patient-relevant outcomes (OS and HRQoL). The European Network of
Health Technology assessment (EUnetHTA, 2019) considers surrogate end-
points to be important and admissible for cost-effectiveness assessment,
provided these have been validated. The NICE Methods Guide (NICE, 2013)
acknowledges that when the use of a ‘final’ clinical endpoint is not possible
48 Economic Evaluation of Cancer Drugs

‘surrogate’ data on other outcomes can be used to infer the effect of treat-
ment on mortality and HRQoL. This would support the surrogate-to-final
endpoint outcome relationship so long as this relationship can be quanti-
fied and justified. Note that if a surrogate and final outcome do not generate
consistent results (e.g. hazard ratios for OS and PFS in different directions or
of vastly differing magnitudes), the uncertainty of treatment benefit is much
higher in terms of both market authorization and cost-effectiveness.
The usefulness of a surrogate endpoint for estimating QALYs will be great-
est when there is strong evidence that it accurately predicts HRQoL and/or
survival. However, it must be noted that in all cases, the association between
the surrogate endpoint, HRQoL and the final outcome (OS) may not be strong
and needs to be explored, quantified, and justified. Table 2.2 shows the OS
and the PFS for a number of clinical trials in glioblastoma:
The plot of OS versus PFS in Figure 2.4 shows the relationship is good (cor-
relation of around 0.78) but not perfect. Hence, one cannot be entirely certain
that the surrogate PFS outcome will lead to clinical benefit in OS in the case
of glioblastoma.

(iv) Not Affected by Treatment Crossover

PFS is central to understanding the effect of an intervention on tumor bur-


den and is not affected by treatment crossover and subsequent treatments. It
therefore potentially offers a direct (‘cleaner’) assessment of the effect of the
experimental anti-cancer treatment. For these reasons, regulatory authorities
around the world, including the US FDA and the EMA (European Medicines
Agency) (EMA, 2012) consider PFS to be an accepted regulatory endpoint to
support cancer drug approval (FDA, 2007), although not always in isolation.

(i) Relationship between OS and Response

Figure 2.4 shows the relationship between OS and PFS. A patient-access


(reimbursement) strategy might be to treat patients for their cancer so long
as they are responding (either complete or partial response). This could be
attractive to reimbursement authorities because if PFS is long and the costs
of treating until PD are also large, only patients who respond could be con-
sidered for treatment. For example, if the treatment duration is 6 cycles (1
cycle = 3 weeks), then treating beyond 6 cycles (18 weeks) might be expen-
sive if the vast majority of patients have a tumor response by say 8 weeks. In
other words, for patients who are responders or have stable disease (SD) by 8
weeks, their treatment would continue until progression. For those who do
not respond by say 12 weeks, future response is unlikely. This approach may
reduce the ICER.
Where objective response rates (ORR) or other intermediate endpoints are
secondary endpoints in clinical trials, these have been used by NICE to set
TABLE 2.2
OS and the PFS for a Number of Clinical Trials in Glioblastoma
OS (months) PFS (months)
Author* Year Phase Population Design N Treatments (median) (median)
Brandes 2016 II Recurrent 2:1 RCT 91 Bevcmb vs.fotemustine 7.3 vs. 8.7 3.4 vs. 3.5
Herrlinger 2016 II Newly diagnosed 2:1 RCT 182 TMZ vs. Bev+Irinotecan 17.5 vs. 16.6 6.0 vs. 9.8
Taphoon 2015 III Newly diagnosed RCT 1:1 921 BEV+RT/TmZ vs. pl+RT/TMZ 16.8 vs. 16.7 9.0 vs. 6.1
Gilbert 2014 III Newly diagnosed RCT 1:1 637 Bev vs.. pl vs. (TMZ/RT) 15.7 vs. 16.1 10.7 vs. 7.3
Gilbert 2013 II Newly diagnosed RCT 1:1 833 TMZ vs. Dense TMZ 16.6 vs. 15 5.5 vs. 6.7
Batchelor 2013 III Recurrent RCT 2:2:1 325 Cediranib 30mg 8.0 vs. 9.8 3.1 vs. 2.7
Cediranib 20mg vs. 9.4 vs. 9.8 4.25 vs. 2.7
Lomustine+placebo
Omuro 2013 II Recurrent Simon 2 stage 47 TMZ 50mg 7.0 2.0
Norden 2013 II Recurrent Single arm 58 TMZ 75-100mg 11.7 1.9
Vredenburgh 2010 II Newly diagnosed Single arm 75 TMZ –> Bev+irinotecan 19.6 13.6
Brada 2010 II Recurrent RCT 1:1 447 PCV vs. TMZ 6.7 vs. 7.2 3.6 vs. 4.7
Friedman 2009 II Recurrent RCT 1:1 167 BV vs. BV+Irinotecan 9.2 vs. 8.7 4.2 vs. 5.8
Fabrini 2009 II Recurrent Single arm 50 Fotemustine 24.5 9.1
Kong 2010 II Recurrent Single arm 38 TMZ metronomic 40mg 10.0 4.3
–>50mg
Important Outcomes for Economic Evaluation in Cancer Studies
49
50 Economic Evaluation of Cancer Drugs

FIGURE 2.4
Relationship between ORR and survival in glioblastoma trials in Table 2.2.

up performance-based patient-access schemes based on response to therapy


(see, for example, HTA, TA129, 2007).

(ii) Relationship between PFS and OS

Over the past decade, between 27% and 50% of HTA submissions to several
European and other reimbursement agencies (e.g. NICE, the Pharmaceutical
Benefits Advisory Committee (PBAC) in Australia, and the Common Drug
Review (CDR) in Canada) were based on surrogate endpoints, such as PFS
(Clement et al., 2009). However, several issues around PFS require further
consideration.
Despite its wide use in cancer trials, PFS is not a statistically validated
surrogate for OS in all settings (e.g. follicular lymphoma, ovarian cancer)
due to a variety of different challenges. For example, a change in tumor
burden with defined disease progression might be insufficient to affect the
time to death in all cancer types. A recent analysis by Kim and Prasad
(2016) showed that in a sample of 65 studies the correlation between sur-
rogate markers such as PFS and OS was ‘weak’ in 48% of these trials (31
of 65 studies). Although, as shown in glioblastoma trials, the correlation
between OS and PFS can be higher (Figure 2.4), in general the absence of a
strong relationship between OS and PFS is reported widely across several
tumor areas.
Important Outcomes for Economic Evaluation in Cancer Studies 51

FIGURE 2.5
Relationship between PFS and OS in glioblastoma trials.

Some statistical modeling indicates there is a strong concordance between


PFS and OS (Clement et al., 2009), but only when the median survival after
PD is short, i.e. <12 months. The relationship between post-progression sur-
vival (PPS) and OS is however much poorer when PPS is longer. This may
be explained by the fact that survival after PD is more variable because of
the number of available treatments after disease progression (see Figure 2.5).
Another possible reason for the poor correlation between PFS and OS may
be a lack of consistent definition of PD across studies and, indeed, different
tumor types (e.g. changes in RECIST criteria, blood cancers use different def-
initions for disease progression). In addition, in recent years with the advent
of immunotherapy, an increase in the size of the tumor on radiological scans
that are classed as PD may actually be due to the mobilization of patients’
immune systems by such immunotherapy drugs.

(iii) Prolonged Treatment until PD

Prolonged exposure (e.g. maintenance treatment or multiple sequential lines


of therapy) might lead to a different evolution of tumors, thus offsetting the
advantage from the treatment shown initially through delaying disease
progression (PFS1). This was noted by recent EMA anti-cancer guidelines
that highlighted the need for prolonged follow-up until the second progres-
sion (PFS2). PFS2 is the time between randomization and evidence of second
progression. However, treating beyond first progression is likely to make
the cost of treatment expensive and will reduce its value (higher ICER). One
would expect that by the time the second progression occurs, the HRQoL is
likely to be worse compared to the first progression with a much diminished
QALY.
52 Economic Evaluation of Cancer Drugs

Treatment until disease progression or maintenance treatment is a strat-


egy that can be employed across all tumor types. It might be used in mainly
indolent or incurable cancers. The argument for using this approach is for
long-term disease control by continued exposure to the drug. However, there
are important issues with this approach and several unanswered questions
remain.
First, this approach does not accurately isolate and quantify the clinical
benefit of maintenance therapy as compared with the standard approach of
fixed-duration induction followed by the second-line treatment at progres-
sion (see Figure 2.5). To address this, RCTs need to utilize an OS (or HRQoL)
endpoint; or, in settings where this is not feasible, utilize endpoints that
incorporate the effects of subsequent lines of therapy (e.g. time from ran-
domization to second progression or death). Toxicity and symptom infor-
mation over both the study treatment (maintenance) and the second-line
treatment should also be collected and reported. However, trials continue
to be designed with PFS as the primary endpoint in most cases. To reduce
the economic burden, a possibility is to cap the duration of continued treat-
ment to a fixed period to reduce cost (see, for example, pembrozulimab
(NICE, 2018d), nivolumab (NICE, 2017a), and atezolizumab for NSCLC
(NICE, 2018).

(iv) Assessment of PFS

Frequent radiological or other additional assessments may not reflect clini-


cal practice and may actually lead to increased health resource utilization.
For this reason, PFS and other surrogate endpoints can be prone to error and
bias because they are contingent on consistent timing of tumor assessment
in both control and intervention groups. That is, PFS is a design dependent
outcome. If one chooses to scan more frequently, a different median PFS
could be obtained. It could also artificially increase the total cost of treatment
(more or less frequent scans influence costs).

(v) Missing Data between PPS and Death

After PD, the potential for collecting post-progression data is limited by clin-
ical protocol requirements. For example, if the ‘end of a trial’ for a patient is
defined as when progression occurs, no further collection can be justified.
This will have obvious implications for collecting costs and consequences
over a lifetime horizon. Importantly, if no data are available post-progres-
sion, extrapolation between progression and death is even more uncertain.
Usually data on some patients after PD is required to entertain a plausible
model for extrapolation. Extrapolation methods are increasingly being
used to predict survival beyond the trial observation period to estimate the
expected future health benefits and costs.
Important Outcomes for Economic Evaluation in Cancer Studies 53

(vi) Combined Risk

Another concern relating to PFS during economic evaluation is that PFS


may not be suitable for capturing the combined risk–benefit profile. A short
observation period may mask the true incidence of serious or detrimental
side effects with limited knowledge about longer-term toxic effects of novel
treatments, which could make the new treatment less cost-effective (e.g. if
the trial stops after first evidence of PD). Delaying disease progression might
decrease a patient’s emotional distress but, on the other hand, it needs to
be balanced against understanding fully (longer-term) drug toxicity and
patient preference.

2.3 HTAs with Surrogate Endpoints


Some examples and case studies with surrogate endpoints are now discussed.

Example 2.2: Chronic Myeloid Leukemia


In chronic myeloid leukemia (CML) a number of different tyrosine
kinase inhibitors (TKIs) are approved based on surrogate endpoints of
complete cytogenetic response (CCyR) and major molecular response
(MMR). In 2012 NICE assessed three of these TKIs, dasatinib, nilotinib
(Ciani et al., 2013), and standard dose imatinib as first-line treatment of
CML. A systemic review and meta-analysis were undertaken to quan-
tify the association between CCyR and MMR at 12 months and OS. This
was acceptable to some agencies and a favorable cost-effectiveness con-
clusion was reached (Ciani et al., 2016).

Example 2.3: Increasing Use of Surrogate Outcomes


The trend over the past two decades suggests that tumor-centered end-
points are increasingly being used as a basis for oncology drug licens-
ing and approvals. The number of FDA approvals based on trials with
time-to-event (tumor-centered endpoints) as a primary endpoint has
increased while the number of approvals for cancer drugs using OS as
their primary endpoint has decreased. Del Paggio et al.’s review (2017) of
277 RCTs involving breast, non-small-cell lung cancer, colorectal cancer,
and pancreas cancer showed 35% of trials used OS as the primary end-
point, while 62% had a tumor-centered primary endpoint. The remain-
ing 3% of trials used HRQoL, toxicity, or another type of endpoint.
Figure 2.6 shows the number of trials for each type of primary end-
point used over the two decades 1990–1999 and 2000–2011. The chart
shows a clear increase in the number of trials that used tumor-centered
54 Economic Evaluation of Cancer Drugs

FIGURE 2.6
Changes in the use of primary endpoints for FDA drug approvals since 1990 split by decades.

Source: Adapted from Martell et al., 2013.

endpoints for successful FDA approval (Martell et al., 2013). For exam-
ple, between 1990 and 1999 there were 36 approvals and, by 2011, this
increased to 104. This increase does not take into account recent approv-
als (e.g. 2014 onward) where 16 out of 17 oncology drugs were approved
based on a surrogate endpoint (of these 17, 6 were based on PFS, 1 DFS, 8
ORR, and 1 using a complete remission with partial hematologic recov-
ery rate outcome).
Figure 2.6 shows the changes in the use of primary endpoints since
1990 split by decades (Martell et al., 2013).

Example 2.4: Types of Endpoints used for Approvals (License)


based on Common and Novel Cancer Endpoints
Table 2.3 shows endpoints used as the primary basis of drug approval
(for licensing). What is noticeable is that whereas cost-effectiveness of
cancer treatments is evaluated over a lifetime horizon (using OS as a pri-
mary outcome), several of the endpoints above demonstrate that clinical
benefit can be determined using outcomes other than OS.
Considerable uncertainty exists if extrapolation of survival is required.
In some cancers, survival may be very long and therefore trial follow-up
will be curtailed for practical and logistical reasons. Consequently, the
proportion of the extrapolated survival as a fraction of the total survival
(i.e. extrapolated time divided by the total survival time) might be very
large. This means that cost-effectiveness is based on modeled survival
patterns and not actual or overall survival for the most part. In some
tumor types, this is less of a problem because survival time is so short
that most of the survival time is observed during trial follow-up (in con-
trast to being predicted) (e.g. Lee et al., 2011).
Important Outcomes for Economic Evaluation in Cancer Studies 55

TABLE 2.3
Examples of Drug Approvals Based on Different Endpoints
Endpoint Drug Name/Year Indication/Tumor Type
OS Pemetrexed/2004 Non-small-cell lung cancer
PFS Sorafenib/2007 Advanced hepatocellular carcinoma
DFS Anastrazole/2010 Adjuvant postmenopausal estrogen receptor positive
breast cancer
TTP Gemcitabine/2004 Advanced breast cancer
ORR Atezolizumab/2016 Urothelial carcinoma/2016
DoR Fludarabine/2007 Chronic lymphocytic leukemia,
pCR Pertuzumab/2015 HER2 positive locally advanced, inflammatory or early
stage breast cancer

2.4 Emerging Tumor-Centered Endpoints


In addition to the traditional tumor-centered endpoints listed above, a num-
ber of novel endpoints have become available for cancer patients. These
include:

(a) Immune-related response criteria (irRC)


(b) Minimal residual disease (MRD)
(c) Pathological complete response (pCR)

These outcomes are related to how the body’s own immune system is used to
fight cancer. In some types of cancer such as acute or chronic leukemia and
early breast cancer, such novel endpoints based on disease burden have been
used selectively.

(a) Immune-Related Response Criteria (irRC)

Immune-related response criteria are appropriate for investigational medici-


nal products that require frequent repeated assessments and measurements
of tumor burden). One important consideration for the development of the
irRC is based on the observation that in some cancer patients, based on a
scan, the tumor may give the appearance of disease progression whereas the
tumor is in fact responding to treatment. Complete response (CR), partial
response (PR), and stable disease (SD) may only occur after an initial increase
in tumor burden. If conventional RECIST criteria were used, patients would
have been classified as having “progressive disease” (Wolchok et al., 2009).
For this reason, some have termed the appearance of initial progression as
‘pseudo-progression’. That is, even if tumor size is increasing, for certain
drugs (e.g. checkpoint inhibitors) the immune system may take some time
56 Economic Evaluation of Cancer Drugs

to ‘kick in,’ which will eventually lead to a decline of tumor burden in many
patients. Hodi et al. (Hodi et al., 2014) report that about 12% of patients (51 of
411 patients) with melanoma treated with pembrolizumab would have been
classified as having PD by RECIST but as SD or responding using iRECIST.
There appears to be a small percentage of patients who achieve responses
using irCR but not RECIST (Chiou & Burotto, 2015).
Although, widely used in cancer immunotherapy trials, at present, reg-
ulatory authorities have not approved any drug based solely on irRC, and
currently its role is limited to an exploratory assessment tool only. For cost-
effectiveness analysis, it may not fall directly into the calculus of the ICER,
but it could be considered as an endpoint with the potential to offer further
value.

(b) Minimal Residual Disease (MRD)

MRD identifies traces of cancer cells that may otherwise elude other test-
ing techniques before clinical symptoms and signs of cancer become appar-
ent. Sophisticated technology now enables the detection of the persistence
of blood cancers at lower thresholds than conventional methods, a level of
disease burden known as MRD (e.g. 1 leukemia cell in 1 million compared to
1 cell in 100). This endpoint applies to blood cancers, e.g. acute or chronic leu-
kemias and requires sophisticated technologies to detect traces of leukemia
cells. This is measured as a continuous outcome similar to laboratory-type
measures, and one objective is to measure effectively a reduction in cancer
cells below a pre-specified threshold (e.g. 1 leukemia cell in a population
of 1 million normal cells). Although MRD is increasingly used in clinical
practice, it is not currently accepted by regulatory agencies for registration
of new drugs, and its relevance for an economic evaluation at this time may
be limited.

(c) Pathological Complete Response (pCR)

Pathological complete response is limited to neoadjuvant (treatment given


before surgery) trials. pCR is a biological reflection of therapy’s ability to
eradicate micro-metastatic disease. Neoadjuvant therapy has permitted the
assessment of tumor response on this basis. Pathological complete response
can be assessed quickly (unlike progression or death, which can take longer)
and has been used as a surrogate for accelerated approval in patients with
operable breast cancer. The FDA accepted definition of pCR is “the absence
of residual invasive cancer on hematoxylin and eosin evaluation of the com-
plete resected specimen and all sampled regional lymph nodes following
completion of neoadjuvant systemic therapy” (FDA Guidance for Industry –
October 2014, accessed on 13 January 2019). Recent approval of pertuzumab
in combination with herceptin in early breast cancer is currently the only
Important Outcomes for Economic Evaluation in Cancer Studies 57

example of regulatory approval (for a license) based on pathological com-


plete response (pCR) (NICE, TA424 2016).
However, it may not be suitable for a cost-effectiveness analysis because
longer follow-up is required in order to confirm whether pCR is a reliable
surrogate for PFS or OS. As an example, in the cost-effectiveness evaluation
of pertuzumab (NICE, 2016, TA424) in early breast cancer, NICE acknowl-
edged that although there was some correlation between pCR and survival
outcomes (OS) it was, however, very weak (correlation coefficients of 0.03
and 0.24 for event-free survival and overall survival respectively). The expert
reviewers noted that the evidence for a beneficial treatment in terms of pCR
did not translate into an OS benefit or was not convincing:

there was considerable uncertainty about whether pathological complete


response could be viewed as a surrogate marker of long-term benefit…

2.5 Demonstrating Value from Other Cancer Endpoints


There is no single agreed methodology or definition of ‘value’ when it comes
to economic evaluation of cancer treatments across countries. Different coun-
tries and regions around the world use different methods to evaluate value
at the time of economic evaluation and/or reimbursement. For example,
Belgium, France, Germany, and Italy make reimbursement decisions based
on the clinical benefit of a product and take costs into consideration in a
subsequent separate pricing process. While Austria, the Netherlands, Spain,
Sweden, and the UK combine a clinical assessment with an economic evalu-
ation to make reimbursement recommendations. Where economic evalua-
tions are carried out, the most commonly used integrative measure of the
value of healthcare products is the QALY, which captures benefits in terms
of reduced HRQoL and improved survival. Values from other endpoints can
take shape in various forms such as those listed below:

• Relationship between tumor measurements and toxicity (e.g. dem-


onstrating tumor reduction does not lead to worsening toxicity)
• Relationship between toxicity and PFS, TTP, DoR, and other end-
points, with the objective to show better tolerability while benefit is
ongoing

Relationship between tumor endpoints and HRQoL. This is particularly


important because, after PD, HRQoL can deteriorate quite rapidly. This will
be the subject of the next chapter on using HRQoL in cancer for economic
evaluation.
58 Economic Evaluation of Cancer Drugs

2.6 Summary
In this chapter we presented the different endpoints that are commonly used
in cancer trials. We made a distinction between primary endpoints, such as
overall survival, and surrogate endpoints. We also discussed the relation-
ship between overall survival and progression-free survival and response
types. We introduced the notion for QALYs and presented some examples
of their use in HTA. Finally, we discussed some more recent tumor-centered
endpoints and their role in demonstrating ‘value.’

2.7 Exercises for Chapter 2


1. Discuss the relevance of each type of cancer outcome for an eco-
nomic evaluation.
2. Is overall survival always relevant for an economic evaluation?
When might it not be used and what other outcomes might demon-
strate value in a cost-effectiveness of a treatment for lung cancer?
3. What are the concerns around assessing disease progression?
4. Distinguish between irRC and RECIST. How can tumor response be
used to demonstrate value?
5. Discuss the challenges for cost-effectiveness of treating a cancer
until progression occurs.
3
Health-Related Quality of Life
for Cost-Effectiveness

3.1 Health-Related Quality of Life (HRQoL) in Cancer Patients


A universally acknowledged definition of ‘quality of life’ has not been
agreed upon. However, most researchers in healthcare adopt the definition
provided by the World Health Organization (WHO): “state of complete phys-
ical, mental and social well-being, and not merely the absence of disease
and infirmity” (WHO, 2014). Health-related quality of life (HRQoL), however,
is affected in a complex way by the person's physical health, psychological
state, the level of independence, social relationships, personal beliefs, and
their relationship to salient features of their environment.
HRQoL is measured through various methods – often questionnaires,
with specific questions about feelings, symptoms, ability, and preferences
among others, in relation to their state of health. HRQoL data are often
collected at several time points during a study (including clinical trials).
In cancer trials, an experimental intervention is often expected to yield
at least equivalent or better clinical benefit (efficacy), compared to a usual
(standard of care) treatment. However, the new treatment may also offer
improved HRQoL in addition to or despite a lack of improved clinical ben-
efit (e.g. the new treatment may be equivalent in terms of efficacy, but with
fewer side effects).

3.1.1 Limitations of Anti-Cancer Treatments


Cancer patients are concerned about their HRQoL during and after treat-
ment (Cykert et al., 2000). Anti-cancer treatments have sometimes resulted in
some harm without benefit as well as harm with modest benefit (Montazeri
et al., 2001), but at the cost of increased toxicity and long-term sequelae.
Consequently, HRQoL should be a key outcome measure when assessing
the cost-effectiveness of a new cancer intervention (Klein et al., 2009). The
implications for HRQoL during palliative therapy, for example, can be par-
ticularly acute because symptom palliation may contribute toward improved

59
60 Economic Evaluation of Cancer Drugs

quality of life, and in some cases impact survival as well (Temel et al., 2010).
Since no further treatments e.g. chemotherapy) are likely to be used during
the end stages of a patient’s life (End of Life, EoL), the HRQoL benefits for
patients and their carers from other forms of intervention (e.g. carer support)
may yield important HRQoL benefits. Some researchers have suggested that
a “treatment can be recommended … even without an improvement in sur-
vival if HRQoL is shown to improve” (Goodwin et al., 2003).
The importance of HRQoL in cancer trials is noted by the fact that it can
influence the choice of treatment. In about 8% of the RCTs in breast cancer,
for example, HRQoL influenced a treatment decision. In prostate cancer
studies involving chemotherapy and surgery, 25% and 60% of treatment
decisions were influenced by HRQoL, respectively (Blazeby et al., 2006).
Due to the increasing number of therapy lines, smaller treatment effect
sizes, and increasing costs of drugs, HRQoL plays an important role in
treatment, policy, rationing, and decision-making. This is likely to remain
an important factor in the short to mid term (Damm, Roeske, & Jacob,
2013).

3.1.2 Why Collect HRQoL Data?


There are several reasons why HRQoL data are collected:

(i) Researchers need to maximize the information about how anti-can-


cer treatments are working, so that informed decisions for treating
patients can be made.

It is essential to know (from both patients’ and clinicians’ perspectives) not


only what the side effects associated with treatments are, but also how these
side effects impact the patients’ HRQoL. It is now universally accepted that
HRQoL should be measured in cancer clinical trials, however, the debate has
been ongoing for some time as to what is the most reliable and practical way
to obtain, measure, and define clinical benefit from HRQoL data (Slevin et
al., 1988).

(ii) The risk–benefit relationship between competing treatments, espe-


cially when clinical effects are small, can be guided by HRQoL out-
comes (Montazeri et al., 2000).

The value of a new healthcare intervention may also have to be considered


through its benefit in terms of HRQoL and not only survival. Although some
cancers are curable (e.g. testicular cancer), many of them are still consid-
ered to be incurable at this time. Therefore, one of the objectives of cancer
patient management should be improving HRQoL, particularly toward the
end of life, when few further treatment options are available (Drummond &
O’Brien, 1993; Clauser, 2004; Gibbons, 2013).
Health-Related Quality of Life for Cost-Effectiveness 61

(iii) Pre-diagnosis (baseline) assessment of HRQoL can help in clinical


decision-making because baseline HRQoL may be directly related
to a patient’s survival time and can also be predictive of survival
benefits during chemotherapy (Montazeri et al., 2001). Therefore,
baseline HRQoL can be used to determine whether certain patients
are more or less likely to benefit from a given treatment. Baseline
HRQoL is also useful in controlling the considerable variations to
estimate treatment differences for effects and costs (as those patients
with better/poorer HRQoL at the start of the trial, may also be the
same patients with higher/lower post-baseline costs and effects).

3.1.3 Challenges with HRQoL in Cancer Studies


There are several features about measuring HRQoL in cancer patients that
are important. First, HRQoL assessments for the purposes of economic evalu-
ation are often omitted. There are several reasons for this omission. For some
countries economic evaluation is not important due to the specific healthcare
system. Measures such as QALYs may not be so important (e.g. in the US) for
decision-making. The issue becomes more complicated when a study con-
ducted in a region where QALYs are not relevant to decision-makers (who
pay for the treatment) is also submitted for licensing to, say, countries where
the QALY is important (e.g. some European countries). When this happens,
the payer perspective does become important. Countries that require HRQoL
for decision-making are likely to criticize or even refuse to pay for cancer
drugs, despite the drugs having a license for marketing authorization.
The second reason why HRQoL assessments for cost-effectiveness analyses
are omitted is because emphasis is placed on the clinical- or disease-specific
aspects of HRQoL. HRQoL measures for economic evaluation are often con-
sidered to lack sensitivity. A further reason for not collecting HRQoL for an
economic evaluation might simply be because two treatments are considered
to be equivalent, and therefore collecting HRQoL for an economic evaluation
may not be useful (e.g. as in the case of biosimilar or generic drugs).
One key feature of a disease such as cancer is that patients can deteriorate
rapidly thereby leading to an absence of data on both short-term and long-
term effectiveness. Economic evaluation is often determined over a lifetime
horizon, and without available HRQoL data performing a cost-effectiveness
analysis can become challenging. Some studies report at least 50% of the
data missing within 3 months of starting treatment (Temel et al., 2010) due to
disease progression, death, or loss to follow-up. This may be due to short sur-
vival time of patients and/or their rapidly deteriorating health. For example,
survival times for patients with NSCLC can be short (e.g. only 32% and 10%
alive 1 and 5 years after diagnosis, respectively) (Hollen et al., 1997).
Estimation of HRQoL within a study/trial and beyond protocol-defined
follow-up also plays a significant role in the economic evaluation of cancer
drugs. Improved methods are needed for estimating long-term HRQoL for
62 Economic Evaluation of Cancer Drugs

the cost-effectiveness of cancer drugs. The short survival time also restricts
the opportunity to collect HRQoL data when there is a limited time win-
dow. This is often despite inclusion/exclusion criteria in protocols specify-
ing a minimal life expectancy, because patients who progress quickly are
likely to die quickly, resulting in missing data. A further challenge is ensur-
ing the appropriate or optimal HRQoL is used. For example, the Functional
Assessment of Cancer Therapy (FACT) FACT-L (specific to lung cancer) and
Quality of Life Questionnaire (QLQ) QLQ-C30 (general cancer measure) can
both be used to measure HRQoL in cancer patients resulting in different
descriptions and measures of clinical benefit. HRQoL instruments used for
measuring cancer HRQoL are discussed in the next section.

3.2 Measuring Health-Related Quality of Life


Outcomes for Common Cancer Types
3.2.1 Condition-Specific Measures of HRQoL
Measuring HRQoL can be broadly classified into the two categories – con-
dition-specific measures (CSM), which measure specific HRQoL symptoms
(e.g. a cough, dyspnoea) and generic measures, which measure the broader
HRQoL areas (e.g. mobility). Figure 3.1 illustrates the relation between some
generic and condition-specific measures of HRQoL.

Examples
HRQoL Instruments Preference
QLQC-30
HAQ Based HRQoL:
KHQ EQ-5D
HUI
SF-6D
Examples
LC-14 Condion Non
FACT-O Specific Generic
Preference
(Very HRQoL HRQoL
Based
condion SF-36
specific)
Very
Condion
Specific
HRQoL

FIGURE 3.1
Relationship between generic and condition-specific HRQoL measures. (Note: LC-14: lung
cancer symptom-specific questionnaire; HAQ: health assessment questionnaire; KHQ: King’s
health questionnaire; HUI: health utilities index; SF-6D: short-form 6D; FACT-O: specific to
ovarian cancer.)
Health-Related Quality of Life for Cost-Effectiveness 63

In most cancer studies, HRQoL assessments, wherever collected and


reported, have been mainly restricted to condition-specific measures. A CSM
is an instrument that captures the specific quality-of-life issues in patients
who have a given disease. The QLQ-C30 and FACT-G are CSMs, but they
are generic for cancer as they are used across several tumors. Very specific
CSMs, often called subscales (e.g. European Organization for Research and
Treatment of Cancer (EORTC) QLQ-LC13 for lung cancer), attempt to cap-
ture information on specific symptoms of the particular cancer and are addi-
tional questionnaires to the generic CSM. The wide use of CSMs is due to at
least three reasons:

(a) First, CSMs were validated for estimating clinical effects and histori-
cally cost-effectiveness was not considered as part of their validation.
(b) Second, CSMs were considered more sensitive than other generic
measures for estimating the HRQoL, focusing on specific symptom
relief.
(c) Third, economic evaluation was not considered important. As bud-
gets for healthcare became constrained, while demand for health
resource use grew, the impetus for rationing healthcare resources
became essential. For cost-effectiveness, HRQoL assessments from
CSMs are not used unless responses can be converted into a generic
preference-based measure. A cancer-specific preference-based CSM
(e.g. QLQ-8D (Rowen et al., 2011)) could also be used for economic
evaluation, but this is not so common, and, in any case, clinicians
may be unlikely to use a short form when the full 30 questions could
be used.

As an example, 26 out of 43 (60%) NSCLC studies (including RCTs) that


assessed HRQoL included a symptom-specific measure such as the LC-13 (in
addition to a generic cancer-specific measure) and only 2 studies (5%) used
a generic measure. This suggests that using a generic HRQoL measure was
considered inadequate to detect the clinical difference of interest (Gibbons
et al., 2013).

3.2.2 Common General Condition-Specific Measures of HRQoL in Cancer


Table 3.1 shows a summary of the main features of some commonly used
HRQoL instruments in various cancers. The important thing to note is that
none of them are suitable for use in an economic evaluation for deriving
QALYs. This is not to say that improved clinical benefits in these measures
do not confer additional value. Certainly, if improvements in, for example,
physical function are observed, it would not be incorrect to conclude that a
treatment did not offer further value in HRQoL just because an alternative
generic measure of HRQoL could not demonstrate a QALY difference.
TABLE 3.1
64

Summary of Cancer-Specific HRQoL Instruments for Common Cancers.


HRQoL Common Analysis Useful for
Instrument Cancer Type Key Features Outcome Metric Reported QALYs
EORTC- Generic across most 15 subscales (5 symptom, 8 Each scale measured from 0 Mean difference No
QLQ-C30 cancers function, 1 global and 1 finance) to 100 Time to observe a
formed from 30 questions specified difference
Used in Europe Odd ratio
FACT-G Generic across most 4 subscales (physical social/family, 22/27 (80%) complete for a Mean difference No
cancers emotional and functional total score ranging from
well-being) from 27 items. 0–108 points.
Commonly used in US
Specific EORTC subscales
QLQ-LC13 Lung 13 additional symptom specific Each scale measured from 0 Mean difference No
questions to 100
QLQ-OV28 Ovarian 28 additional symptom specific Each scale measured from 0 Mean difference No
questions to 100
QLQ-MY20 Lymphoma/ 20 additional symptom specific Each scale measured from 0 Mean difference No
myeloma questions to 100
QLQ-BR23 Breast 23 additional symptom specific Each scale measured from 0 Mean difference No
questions to 100
QLQ-PR25 Prostate 25 additional symptom specific Each scale measured from 0 Mean difference No
questions to 100
Specific FACT subscales
FACT-L Lung Uses 4 scales of FACT-G plus the Trial Outcome Index (TOI) Mean difference No
lung cancer scale (LCS) – total across 4 scales of
FACT-G plus LCS
(Continued)
Economic Evaluation of Cancer Drugs
TABLE 3.1 (CONTINUED)
Summary of Cancer-Specific HRQoL Instruments for Common Cancers.
HRQoL Common Analysis Useful for
Instrument Cancer Type Key Features Outcome Metric Reported QALYs
FACT-O Ovarian FACT-G plus the ovarian cancer- TOI of FACT-G and OCS Mean difference No
specific subscale (OCS)
FACT-Lymp Lymphoma FACT-Lym and fifteen TOI of FACT-G and Mean difference No
additional disease-specific items FACT-Lymp
FACT-Lymp 15 additional items to
the FACT-G (42 in total)
FACT-B Breast 10 items addressing symptoms TOI: PWB+FWB+BCS Mean difference No
and issues specifically relevant to subscale
patients with breast cancer on a
4-point scale, from 0 (‘not at all’)
to 4 (‘very much’)
Prostate FACT-P – prostate cancer subscale TOI: PWB+FWB+EWB+
(PCS) plus including pain scale SWB + PCS + pain scale
subscale
FACT-Ntx Neurological 11 items subscale TOI of PWB+FWB+Ntx Mean difference
Health-Related Quality of Life for Cost-Effectiveness

toxicity
FACT: Functional Assessment of Cancer Therapy – General; P/F/E/S-WB: Physi​cal/F​uncti​onal/​Emoti​onal/​Socia​l/Wel​l-Bei​ng
65
66 Economic Evaluation of Cancer Drugs

Consider two of the most common HRQoL measures in cancer clinical


research: the EORTC QLQ-C30 and the FACT-G: Functional Assessment of
Cancer Therapy – General (Hollen et al., 1997; Gibbons et al., 2013; Damm
et al., 2013).

(i) The EORTC QLQ-C30 generic cancer instrument

The EORTC QLQ-C30 version 3 is a generic cancer instrument consisting of


30 questions, out of which 28 questions are measured on a 4-point scale (‘not
at all’ (1) to ‘very much’ (4)) and 2 questions are measured on a 7-point scale.
The 30 questions result in: 5 functional domains: Physical Functioning (PF),
Role Functioning (RF), Emotional Functioning (EF), Cognitive Functioning
(CF), and Social Functioning (SF); 8 symptom domains: Fatigue (FA), Nausea
& Vomiting (NV), Pain (PA), Dyspnoea (DY), Sleep Disturbance (SL), Appetite
Loss (AP), Constipation (CO), and Diarrhea (DI); and 2 further domains:
Financial Impact (FI) and Global Quality of Life (QL).
All raw responses are transformed to a scale of 0 to 100, where a higher
value represents better physical function for the function domains (includ-
ing global and financial scales) and the converse for the symptom domains
(a high value implies poorer symptoms).
The QLQ-C30 (Fayers, Aaronson, & Bjordal, 2001) has been well docu-
mented, has good psychometric properties, is validated, and is translated
into more than 48 different languages with a large number of possible
‘health states.’ A health state, in economic evaluation terms, means combi-
nations of different responses. For instance, one outcome for a particular
patient from the QLQ-C30 could be 11111 … 1 (i.e. 30 responses of a value
1). This combination of 1s represents a ‘health state. In this sense, there are
428+72 possible health states. Inferences across all the possible health states
are practically impossible. However, summary statistics are often computed
for each domain, in terms of the average (mean) scores for which the health
states are less relevant. Interested readers may refer to the EORTC’s website
for guidance on scoring (Fayers et al., 2001).

(ii) FACT-G

The FACT-G consists of 27 items: physical wellbeing (PW, 7 items), social


and family well-being (SW, 7 items), emotional well-being (EW, 6 items), and
functional well-being (FW, 7 items). Additional subscales can be added as in
Table 3.1 to derive the Trial Outcome Index (TOI), a key measure of improve-
ment in HRQoL with this instrument. This instrument also has a large num-
ber of possible health states (see Fayers et al., 2001).
The choice between utilizing FACT-G or QLQ-C30 is based on a subjective
clinical choice, rather than empirical evidence for the superiority of one over
the other. In fact, the empirical evidence presented for their relative superiority
can be considered minimal or nonexistent (Gibbons et al., 2013; Calman, 1984).
Health-Related Quality of Life for Cost-Effectiveness 67

A related issue is which instrument is more (or less) sensitive to detect-


ing a clinically relevant treatment benefit and what is the clinically and/
or economically relevant effect size from these measures (an economically
relevant effect in this context refers to a clinical effect size such that the
incremental net monetary benefit (INMB) has a high chance of being posi-
tive (see Khan, 2015) for sample sizes for cost-effectiveness). This aspect still
remains unknown and is not well understood. Maringwa et al. have sug-
gested important effect sizes of varying magnitudes (e.g. a difference of 10
points) (Maringwa et al., 2011). Comparing effect sizes between the varying
HRQoL measures has not been widely reported, particularly in NSCLC. In
contrast, generic measures of HRQoL have been criticized for the lack of sen-
sitivity to detect HRQoL benefits, something that has implications for later
cost-effectiveness (Ades, Lu, & Madan, 2013). A further issue is that a QALY,
a key outcome for estimating cost-effectiveness is not usually constructed
from CSMs, but from generic measures of HRQoL.

3.3 Measuring HRQoL for Economic Evaluation


Evaluating and measuring HRQoL benefit is an important aspect of economic
evaluation. Generic measures of HRQoL capture responses about health in
general and not the symptoms that might be associated with toxicity (result-
ing from chemotherapy, radiotherapy, and surgery). They are useful for com-
paring effects across a variety of diseases. Some generic measures reflect
a patient’s preference for certain health states, termed ‘preference-based’
measures (PBM) because they offer a way in which (subjective or perceived)
relative preferences (or values) for specific health states can be expressed by
individuals as ‘preference weights’ or ‘utilities.’ Utilities are based on subjec-
tive judgments by individuals presented with questions such as: “Do you
prefer an apple or banana?” and are derived from consumer choice theory in
economics. These utilities can subsequently be used to adjust observed clini-
cal effects for later cost-effectiveness analyses or used directly as a measure
of effect such as a QALY.
A utility value is often measured on a continuous scale and, depending
on which instrument is used, these values have different ranges (lowest
and highest values). The health utility index (HUI), for instance, generates
utilities on a scale from 0 to 1, where 0 represents ‘dead,’ which is the worst
health state possible and 1 represents ‘Full health.’ However, not all generic
measures of HRQoL generate utilities on a scale between 0 and 1, for exam-
ple the EQ-5D. Other preference-based generic HRQoL measures include:
SF-6D: Short Form 6D; HUI: Health Utilities Index (versions I, II and III); and
the more common EuroQol EQ-5D (EQ-5D-3L and 5L) (Carreon et al., 2013;
Herdman et al., 2011; Rabin et al., 2001).
68 Economic Evaluation of Cancer Drugs

Although responses from the cancer specific QLQ-C30 reflects how a


given patient might feel with regard to their symptoms, disease, or treatment
received, the responses do not necessarily reflect how payers (people who
ultimately pay for treatments through taxes) perceive the value of a given
patient’s health condition, even if the patient is suffering from a disease as
severe as breast cancer. It is possible that society (and not necessarily doc-
tors) might regard a specific patient’s health (state) as far worse and there-
fore believe any funds available to treat a patient’s illness should be spent
elsewhere (e.g. preference for a prostate cancer sufferer over a breast cancer
patient). CSMs do not incorporate a relative evaluation of the extent to which
a specific symptom or health state affects the overall perception of health.
For instance, severe nausea might be considered worse than severe pain for
some patients but not for others. The expression of such relative preferences
(utilities) is determined through preference-based HRQoL measures. The
QLQ-C30 and the FACT are not preference-based measures of HRQoL and,
therefore, cannot be directly used as such in an economic evaluation.
The most common preference-based measures used in the UK (and many
European countries) for economic evaluation are the EuroQol EQ-5D-3L and
the more recent EQ-5D-5L.

3.3.1 EuroQol EQ-5D-3L and 5L


The EQ-5D is a widely used generic measure, which is the shortest and per-
haps the least cognitively demanding instrument that appears to be at least
as responsive as the other community- (preference-) weighted instruments
(Brazier et al., 2007).
EQ-5D-3L consists of a descriptive health state classification system with
five questions (mobility, self-care, usual activities, pain/discomfort, and anx-
iety/depression), measured on three severity levels – ‘no problems,’ ‘some
problems,’ and ‘extreme problems.’ Combining one level from each question
defines 243 different health states, ranging from full health (value of 1) to
worst health or death (value of 0). A health state defined by the descriptive
system of EQ-5D can be described by a five-digit number. For instance, 12113
refers to a patient who has no problems with mobility (1), some problems
with self-care (2), no problems for usual activities (1) or pain/discomfort (1),
and extreme problems with anxiety/depression (3). Full health is indicated
by 11111 and the poorest health state by 33333. In practice, the raw responses
are converted into a utility scale from –0.594 to 1 (1 is full health and –0.594
represents the worst possible state – even worse than death), depending on
the method (called the tariff) of converting the responses into utilities. Some
tariffs result in utilities between –0.109 to 1 (Dolan, 1997; Shaw, Johnson, &
Coons, 2005). The Dolan UK tariff results in utilities in the –0.594 to 1.0 range.
The term ‘health state’ requires some elaboration. Given that the EQ-5D-3L
consists of a descriptive health state classification system with five domains
and 243 different health states ranging from full health (11111) to worst health
Health-Related Quality of Life for Cost-Effectiveness 69

(33333). Values such as 11111 or 21333 are not easily analyzed but converting
these responses to a utility value will allow analysis. Each of these health
states is therefore converted to a single number, called a utility value: a value
from –0.594 (worse than death) to 1 (full health). This is true if the conver-
sion from values such as 11111 is based on the ‘tariff’ provided by Dolan
(Dolan, 1997). An alternative tariff of Shaw et al. (Shaw et al., 2005) converts
the health states to a range between –0.109 to 1.0 (Lewis et al., 2010; Dunlop
et al., 2013). The word ‘tariff’ refers to the weights applied to health states to
generate the final EQ-5D value. Tariffs are country specific (if they exist) and
are estimated from general public surveys.
There are various reasons why the EQ-5D is emphasized, at least in some
parts of Europe. It is for example recommended for use in economic evalu-
ations in the UK, by NICE (Brazier et al., 2011; Brazier et al., 2016). It is not
uncommon that when NICE adopts a decision on the value of a cancer drug,
some (but not all) countries follow a similar decision (but not necessarily
using QALYs). Hence, the EQ-5D is also used in several countries as a part
of economic evaluation and health technology appraisal (HTA). The EQ-5D’s
properties are well documented (Brazier & Rowen, 2011) and it has been
shown to be a reliable and valid HRQoL measure (Hurst et al., 1997; Van Agt
et al., 2005).

3.3.2 EuroQol EQ-5D-5L
The EQ-5D-5L is a revision of the EQ-5D-3L. It consists of five questions,
identical to EQ-5D-3L (mobility, self-care, usual activities, pain/discomfort,
and anxiety/depression), but with an expanded 5-point scale and slightly
different descriptors for each of the levels compared to the 3-point scale
of the EQ-5D-3L (Carreon et al., 2013). These are: mobility, self-care, and
usual activities: 1: ‘no problems’; 2: ‘slight problems’; 3: ‘moderate problems’;
4: ‘severe problems’; and 5: ‘unable to’; for the pain/discomfort and anxiety/
depression scale, these are: 1: ‘no’; 2: ‘slight’; 3: ‘moderate’; 4: ‘severe’; and
5: ‘extreme.’ The scores are on a 5-point scale 1 to 5 (for each of the 5 domains).
A perfect health state is ‘11111’ and the worst possible health state would be
‘55555’. There are 3,125 health states that can be identified using EQ-5D-5L.
The corresponding minimum and maximal values are –0.281 for a health
state of 55555 and a value of 1 for the 11111 health state.
Predetermined scoring algorithms for EQ-5D have been developed in
order to yield community-based health utility estimates (i.e. relative prefer-
ences for health states not based on what the patients think but what the gen-
eral population believes) – specific to a given country. The derived utilities
are determined from a predetermined algorithm called a utility function.
The utilities from these instruments (such as the EQ-5D, HUI, and other pref-
erence-based measures) may subsequently be applied as a weight to clinical
measures, such as overall survival or progression-free survival (PFS) time in
order to derive a quality-adjusted life-year (QALY).
70 Economic Evaluation of Cancer Drugs

The primary difference between these two instruments (3L and 5L) is that
the latter has responses measured on a 5-point scale, with many more health
states (Oppe et al., 2014). EQ-5D-3L is reported to have limited discrimina-
tive ability (though it may have higher power to detect differences between
groups) compared to EQ-5D-5L (Dolan, 1997; Oppe et al., 2014; Van Hout et
al., 2012). Recently EQ-5D-5L tariffs have been developed for the UK and
several other countries (Zhao, Li, Liu, Zhang, & Chen, 2017).

3.4 Constructing Utilities
Utility elicitation methods may be classified as direct or indirect methods of
health utility elicitation (Figure 3.1) (Sacco et al., 2010). Most direct elicitation
methods include some sort of trade-off (standard gamble [SG], time trade-off
[TTO]), or a visual analog scale (VAS), but direct utility elicitation is rarely
performed in clinical trials. Furthermore, as cost-effectiveness studies are
intended to support health policy decisions, utility values from the general
population are preferred (Rowen et al., 2015; Batty et al., 2012), and healthcare
providers’ utilities are considered as valid. Terminally ill patients, children,
and dementia patients raise special problems where a proxy person (e.g. a
carer) may be used as a substitute for the patient, though as a second-best
option.
Direct elicitation, such as time trade-off involves asking people (patients
or members of the general public) to trade a given time (e.g. 10 years) in a
hypothetical or current health state worse than full health for a lower sur-
vival time in full health. The data collected from these choices (trade-offs)
are then analyzed to derive a utility score. An example of such elicitation
for chemotherapy in breast cancer patients can be found in Simes (Simes &
Coates, 2001).
The main indirect methods of utility measurement consist in the use of a
generic preference-based instrument (G-PBM), such as the EQ-5D, or a con-
dition-specific preference measure (CS-PBM), such as the QLQ-8D, with their
relevant health-profile associated utilities or utility-generating formula (Hao,
Wolfram, & Cook, 2016; Lorgelly et al., 2017). Another type of utility generat-
ing method is based on mapping utilities indirectly via a generic non-PBM
HRQoL questionnaire such as the QLQC30 or the FACT-G via a (published)
mapping algorithm that predicts (maps) the components of the HRQoL ques-
tionnaire to a PBM utility (Wailoo et al., 2017; Figure 3.2).
The benefit of direct elicitation alongside an RCT is that such valuation of
health states within an RCT framework has strong internal validity. In addi-
tion, for patients who survive beyond the median survival time, an accurate
reflection of the value of health states toward end of life might be possible.
However, using direct valuation methods from cancer patients alongside
Health-Related Quality of Life for Cost-Effectiveness 71

FIGURE 3.2
The EQ-5D utility function in terms of health states.

Source: Sacco et al., 2010.

a clinical trial is difficult in practice because of the poor prognosis, short


median survival times, and the complex logistics and ethical considerations
involved in clinical trial conduct. It may require extending the duration of the
trial perhaps by following up patients until death, which would be imprac-
tical in most cases. For example, in order to estimate longer-term HRQoL
effects (e.g. valuing health states toward the end of life), some patients would
need to be followed up in the trial much longer as part of the same proto-
col. In practice, after cancer progression, follow-up for many outcomes (other
than necessary longer-term safety) is often stopped. Trial governance may
also complicate the process of further assessments once a patient’s main
follow-up has been completed. Moreover, patients in some clinical trials are
often extremely ill and there is a serious debate about whether the patients
are capable of ascertaining their health status through TTO and SG methods.
The complex articulations can be daunting and time-consuming for some
cancer patients, especially when they are preoccupied with a variety of other
tests such as imaging scans, blood tests, and radiotherapy planning.
In health economics, the concepts of utility and utility functions are central
for appreciating why for example health states from EQ-5D assume values
between –0.594 and 1.0. A utility function is a mathematical representation of
determining utilities for preferences of given health states. Mathematically,
for each patient, i, the EQ-5D-3L utility Ui under the assumption of linear
additivity is equal to:

U i = 1 – (0.081 * K – a1 * Mi – a 2 * Si – a 3 * USi – a 4 * Pi – a 5 * Ai – a 6 )

Source: (Dolan, 1997)


where, α1 …α6 are weights derived from TTO utility elicitation, such that
for patient i: α1 = 0 if the mobility score Mj = 1, α1 = 0.069 if the mobility score
72 Economic Evaluation of Cancer Drugs

Mj = 2, and α1 = 0.314 if Mj = 3 (j = 1 to 3, referring to the 3-level response for


each question)
Similarly,

α2 = 0.104 if the self-care score Si = 2, and α2 = 0.214 if Si = 3,


α3 = 0.036 if the usual activities USi = 2, and α2 = 0.094 if USi = 3,
α4 = 0.386 if the pain score Pi = 2, and α2 = 0.123 if Pi = 3,
α5 = 0.071 if the anxiety score Ai = 2, and α2 = 0.236 if Ai = 3,
α6 = 0.269 if any of the Mi, Si, USi, Pi, or Ai is a score of 3, otherwise α6 = 0,

and finally, K is the indicator variable, which takes the value 1 if any health
state is dysfunctional (>1), otherwise, it is 0. The utility values are ordered
from lowest to highest and a numerical coding can be given to the ordered
health states (11111 = 1, 2 = 21112 = 0.878 … 3 = 33333 = –0.549). For example,
for a health state of 12123, Ui would be 1 – (0.081 + 0 + 0.104 + 0 + 0.123 + 0.236
+ 0.269) = 0.187.

3.5 Quality-Adjusted Life-Years (QALYs)


In economic evaluation, the use of HRQoL is particularly important for cost
utility analyses (CUA). CUA is a method of determining the cost-effective-
ness of a new health technology by combining HRQoL with clinical out-
comes (e.g. survival time) to derive a quality-adjusted life-year (QALY).
In Chapter 1, we briefly introduced the QALY in a rather simple way as a
combination of the quantity and HRQoL. Utility values determined from
the EQ-5D-3L (or 5L) were described in Sections 3.2 and 3.3. A commonly
described way of deriving the QALY is by computing the area under the
curve (AUC) as shown in Figure 3.3. The difference between the areas under
the solid line and the area under the dotted line in Figure 3.3 is the incre-
mental QALY.
In Figure 3.3, the HRQoL responses from the EQ-5D are converted into
utilities on a scale from either 0 to 1 or, as in the case of the EQ-5D-3L, from
–0.54 to 1.0. Utility measures were collected from baseline (time 0) until 24
months (when the follow-up is final). If continuous measures are available
from patients in each of the two treatment groups A and B, then we obtain
two curves as shown in Figure 3.3, one for each treatment group describing
the behavior of utility over time. In practice only a few HRQoL measure-
ments at fixed discrete time points are available (say every 3 months).
The next step is to calculate the AUC for each utility curve. This area
is the QALY. The area can be tricky to calculate sometimes, but it is usu-
ally straightforward. For example, if the utility score never changed from
Health-Related Quality of Life for Cost-Effectiveness 73

FIGURE 3.3
Theoretical QALY determination between treatment A and B with repeated measurements.

baseline and was always 0.80 every 3 months until 24 months, the area would
be the same as the area of a rectangle of 24 × 0.8 = 19.2 months = 1.6 QALY
years (i.e. 2 years × 0.80). The difference between the AUC for treatment A
and AUC for treatment B is the incremental QALY. When the utility profiles
are more complicated, the AUC is computed by dividing the area under the
curve into several trapeziums and using the trapezoidal rule to calculate the
area of each trapezium and add up all the areas. The area of a trapezium is
½(sum of the parallel sides) × perpendicular height: ½*(a + b)*t, where a and
b are the utilities on the Y-axis and t is the difference between adjacent time
points. A more complicated application in a cancer setting is demonstrated in
Example 3.1.

3.5.1 QALY Calculation in Cancer Trials


The description above for the calculation of the QALY is reasonably straight-
forward. There are some complications in that at the end of the follow-up,
some assumptions about the (future) unobserved utility need to be made.
For example, if the trial is completed after 24 months, and some patients are
still alive, what should we assume about the behavior of the utility beyond
24 months? One possibility is to assume it is the same as what was observed
at 24 months (last observation carried forward). However, if disease pro-
gression occurred at 24 months, we might expect the utility to get worse
and therefore the assumption of a constant utility is likely to bias the QALY
(upward).
A simple formula that weights the utility by the proportion alive (deter-
mined from the Kaplan Meier curve) can be used:

QALYAUC = S éë( Qi + Qi+1 ) / 2 ùû * éë( Si + Si+1 ) / 2 ùû * [ti+1 - ti ] (3.1)

where Qi are the mean utilities at time point t and Si are the survival propor-
tions (e.g. from the Kaplan-Meir curve) at the corresponding time point, t.
74 Economic Evaluation of Cancer Drugs

The subscript i refers to the time points (e.g. t0 = baseline, t1 is the first time
point at which utility are observed) and the summation (∑) is over the num-
ber of time points. A demonstration of the formula in equation (3.1) follows.

Example 3.1: Deriving the QALY from Survival and Utility Data
Using equation (3.1),

QALYAUC = ∑ (Qi + Qi +1 ) / 2 * (Si + Si +1 ) / 2 * [ti +1 − ti ]

we obtain

QALYAUC = (0.65 + 0.71) / 2 * (1.00 + 0.69) / 2 * [1 − 0 ]

+ (0.71 + 0.72) / 2 * (0.50 + 0.69) / 2 * [ 3 − 2]



+ (0.69 + 0.72) / 2 * (0.32 + 0.50 ) / 2 * [ 4 − 3 ]

+ (0.75 + 0.69) / 2 * (0.23 + 0.32) / 2 * [ 5 − 4 ] + etc.

Separate to the issue about what to assume about future non-observed


utility, is how the QALY is derived when taking into account the pre-
progression (progression-free survival) and the post-disease progres-
sion periods. A useful feature of the AUC approach is that the AUC is
computed by taking into account the corresponding utility during the
survival experience. In equation (3.1) the mean utilities are computed
for those patients who have and have not progressed. These are then
multiplied by the proportion alive. In cancer trials, partitioned survival
models (Chapter 7) separate the survival experience into a pre-progres-
sion survival (PFS) and a post-progression survival (PPS) period. Hence,
OS = PFS + PPS. The mean pre-progression utilities can be used to derive
a pre-progression QALY and similarly a post-progression QALY can be
derived. This will be discussed in Section 3.7.

3.6 Economic Evaluation in the Absence of Utility


Data: Mapping and Utility Studies
Damm (Damm et al., 2013) reports instances where a CSM, in this case the
QLQ-C30, was used yet no generic HRQoL was included. Moreover, an eco-
nomic evaluation of some form was performed using the same data at a later
point (e.g. the BR21 trial (Bradbury et al., 2010; Lewis et al., 2010)). In these tri-
als, generic HRQoL data for economic evaluation could have been collected
instead of relying on alternative sources (e.g. historically reported). There are
Health-Related Quality of Life for Cost-Effectiveness 75

several possible reasons why utility data may not be available or collected in
a trial, despite cost utility analyses being conducted later:

(i) Several examples in literature report the main results of clinical trials
(e.g. Bradbury et al., 2010) where utility data were not collected. One
reason is that in some countries the health system does not require
cost utility analyses. Therefore, cost-effectiveness was not part of
trial design (e.g. submission to the FDA in the US). However, the
same clinical data is used for licensing purposes in Europe, where
some countries consider QALYs important. Therefore, estimates of
patient-level utilities are not available but a CUA is required.
(ii) A second reason might be that EQ-5D are not considered sensitive
enough to detect treatment benefit, which may be a reason to avoid
them. Clinicians responsible for protocol development may not
focus on the ‘softer’ measures like HRQoL, and even less so on utili-
ties, and therefore do not include these outcomes in the protocols
and data collection.
(iii) Direct elicitation studies sometimes involve estimating utilities in
separate, smaller specific studies. These can yield biased or impre-
cise utilities, which affect the QALYs, giving a reason to avoid such
an approach (Dunlop et al., 2013). NICE recommends avoiding sepa-
rate utility studies (Brazier et al., 2011; NICE DSU 2016).
(iv) A further reason is that cost-effectiveness may not be considered
important. However, when the cost of cancer drugs is perceived to be
high, payers re-evaluate the value of cancer drugs, thus this reason
is unlikely to be sustainable. In addition, grant-awarding bodies (for
academic trials) request details of the cost-effectiveness of proposed
interventions that have potential to become the standard of care.

When utility data are not available, but a cost-utility analysis is required,
utilities can be determined by an indirect method called mapping or by
using published historical utility data. Mapping or ‘cross-walking’ can be
useful when patient-level utilities are not available in a clinical trial. A statis-
tical model sometimes termed a ‘mapping algorithm,’ is used to predict (esti-
mate) EQ-5D-3L utilities from a disease-specific measure such as QLQ-C30.
If patient level EQ-5D-3L cannot be obtained, then it becomes challenging to
conduct an economic evaluation with patient-level data, and reliance is often
placed on published aggregate utilities. Mapping is, therefore, another way
(and sometimes the only way) to estimate patient-level utilities for a cost-
effectiveness analysis. Details of mapping for cancer can be found elsewhere
(Crott & Briggs, 2010; Crott, Versteegh, & Uyl-De-Groot, 2013; Khan et al.,
2016; Brazier et al., 2010; Doble & Lorgelly, 2016). The alternative approach is
to use historical data or published utilities such as those reported in Nafees
et al. (2008) (see Section 3.8 for examples).
76 Economic Evaluation of Cancer Drugs

3.7 Sensitivity and Responsiveness of EQ-5D versus QLQ-C30


HRQoL for Detecting Improvement in Cancer Patients
Some generic instruments may not be adequate to demonstrate HRQoL ben-
efits, compared to condition-specific measures (CSM) (e.g. Lee et al., 2013;
De Vine et al., 2011; Malkin et al., 2013; Krahn et al., 2007). This is because a
brief and standardized HRQoL instrument across diseases will lack sensi-
tivity due to its nature (Malkin et al., 2013). Concerns about the sensitivity
and responsiveness of generic measures such as the EQ-5D-3L (Buchholz,
Thielker, Feng, Kupatz, & Kohlmann, 2015; Richardson, Khan, Iezzi, &
Maxwell, 2015) have been important enough to lead to the development of
the EQ-5D-5L, using a 5-point scale instead (EQ-5D-3L has a 3-point scale).
The issue becomes more acute and more relevant where a CSM appears to
offer an interpretation for a treatment benefit inconsistent with a generic one
(or vice versa).
It is also unclear, not only in cancer studies but also in many trials with
HRQoL endpoints (either as primary or secondary endpoints), what a rele-
vant clinical or economic treatment difference is (or the size of the minimum
clinical important difference (MCID)). Moreover, which among the common
HRQoL measures (in cancer) are more sensitive in detecting HRQoL benefits
remains highly uncertain (Buchholz et al., 2015; Richardson et al., 2015). This
is relevant whether the HRQoL is primary or secondary because HRQoL
effects are often considered to ‘add value’ to an intervention, especially when
the primary outcome result shows modest or borderline benefits.
For instance, with QLQ-C30, there are 15 possible different effect sizes but
only 1 for EQ-5D. The precise clinical or economic relevance of such effect
sizes still remains unknown or not well understood. Maringwa et al. (2011)
suggest important effect sizes of varying magnitudes (Maringwa et al., 2011),
although in practice these are rarely stated up-front. A suggested effect size of
10 points (it is unclear whether this is for a particular domain or any domain
of the QLQ-C30) is assumed to be an important difference yet it is unclear
why this should be the case. Moreover, this suggestion was made many years
ago when novel cancer treatments at the time were compared to an older class
of drugs. As the standard of care has improved, and because ­economic evalu-
ation often requires the ‘best’ standard of care for ­comparison, treatment ben-
efits are likely to be smaller and this definition, despite its wide used is likely
to be redundant (King, 1996; Osoba, 2007; Pickard et al., 2007).
Generic measures are considered to lack the sensitivity to detect HRQoL
benefits when compared to CSMs. However, if the treatment benefits are
similar between a CSM and a generic measure, then a simpler and shorter
preference-based generic measure (e.g. EQ-5D) could be used with a highly
condition-specific measure (e.g. such as the lung cancer symptom-specific
LC-14 questionnaire, as in Figure 3.1) without losing much information. With
preference-based measures, there may be a concern that preference weights
Health-Related Quality of Life for Cost-Effectiveness 77

from the general population may not reflect the similar relative importance
for certain health states that a cancer patient might portray. However, the
EQ-5D-3L is considered to lack sensitivity for measuring changes in health
states in a cancer setting (Bongers et al., 2011).
In a systematic review of 43 published articles in NSCLC (Damm et al.,
2013), for example, 28 of the studies used the QLQ-C30 with the objective
of detecting clinical improvements in HRQoL. Among these 28 studies, the
vast majority (>80%) did not report improvements with the QLQ-C30, either
between treatments or relative to baseline. Moreover, wherever an effect was
detected, the sample size was small. Khan et al. (2015) report that condition-
specific HRQoL treatment effect sizes in lung cancer trials are rarely large
(Khan, Bashir, & Forster, 2015).
Conclusions regarding HRQoL benefits are often provided in terms of
‘non-worsening HRQoL’ and although a few studies report small improve-
ments, most report no improvements in HRQoL. The conclusions are often
presented such that if patients did not deteriorate in their HRQoL, then this
is something worthy of comment or a favorable outcome (see Damm et al.,
2013). A more complicated situation is when a CSM suggests an HRQoL
improvement and a preference-based measure does not. One should recall
that utilities for the EQ-5D are based on societal preferences in general popu-
lations, whereas CSM measures are determined from cancer patients, which
may explain some of the discrepancies.
Small, but important differences in HRQoL should not be ignored (Khan
et al., 2015) but investigated further with a view to identifying the implica-
tions for an economic evaluation. For example, a small mean difference in
EQ-5D (e.g. 0.05 point improvement in physical function) on an odds ratio
scale might translate to 20% improvement in HRQoL (this can happen when
the data are heavily skewed). It would be misleading to conclude HRQoL
improvements do not exist when small mean differences are observed. This
approach may help to contextualize borderline QALY differences, particu-
larly where a generic measure lacks sensitivity.

3.8  Measuring Post-Progression (PP)


Utility: Some Approaches
Patients with cancer often have progressive disease during or after anti-can-
cer treatment. The consequence of this on utility estimation can be serious
because, as patients deteriorate, the availability of post-progression utility data
becomes problematic. In clinical trials, HRQoL is often planned to be collected
until disease progression and, in some instances, until death. PP survival can
vary depending on factors such as the underlying cancer, severity of the dis-
ease, and availability of other treatments. What happens in some clinical trials
78 Economic Evaluation of Cancer Drugs

is that some patients have available HRQoL after disease progression and oth-
ers do not. A further complication is that even where data are available after
progression, the follow-up times may vary from patient to patient.
Earlier, in Section 3.5, we noted mapping as a means to estimate utili-
ties. However, the development of mapping models depends on the avail-
ability of both generic measures and CSMs from the same patient sample.
Furthermore, many mapping models available in cancer do not specifically
offer algorithms for predicting PP utility data. When PD has occurred, both
generic measures and CSMs are not collected and the lack of utility data
after PD does have a consequence for cost-effectiveness. One approach might
therefore be to develop a model to extrapolate utilities after PD (Figure 3.4).
The difference between this approach and mapping is that the estimation
of utilities beyond disease progression does not need to depend on a CSM.
Mapping involves the use of a CSM to estimate utilities, whereas in this
approach, the utilities are extrapolated by modeling available data. In this
section, we therefore discuss an empirical examination of patterns of PP util-
ity data and consider the principles of some statistical models for estimating
PP utility data and their relevance to QALY estimation for cancer treatments.
It is not uncommon to model survival data in a similar way where long-term
survival patterns are estimated using complex models.

Why Is Estimation of Utility between Disease


Progression and Death Relevant?
There are several reasons why PP utility data are important:

(i) The QALYs computed in economic evaluations of cancer treatments


are often computed as a weighted measure of PFS and PPS, using
the partitioned survival model approach mentioned in Section
3.4. Therefore, how post-progression utilities are estimated, either
directly or in some other way, influences the overall QALY.

FIGURE 3.4
Post-progression linear decreasing utility function.
Health-Related Quality of Life for Cost-Effectiveness 79

(ii) Estimating utility beyond PD is important especially when treatment


is planned to be delivered beyond progression (e.g. as in the case of
more recent immunotherapy advances like Nivolumab (NICE, 2017;
Ulmeanu, Antohe, Anisie, & Antoniu, 2016). This will be important
to determine whether it is worthwhile for patients (and payers) to be
treated beyond PD.
(iii) For some anti-cancer treatments, late toxicities can occur (e.g.
from radiotherapy or surgery), which may have a marked impact
on HRQoL for years, or even lifelong, after the treatment has been
delivered.
(iv) If no HRQoL data are available after disease progression, then there
is likely to be greater uncertainty around the QALY (due to the
dependence of utility from external or historical data). In practice,
some patients are able to provide HRQoL data after PD as long as it
is not too difficult to collect. The EQ-5D is a short and simple HRQoL
measure, unlike the QLQ-C30, which facilitates collection of these
data. The availability of these data can have significant implications
for economic evaluation.

The Behavior of Utility in Cancer Patients


between Progression and Death?
One may reasonably expect, as patients’ health worsens, HRQoL deterio-
rates. Therefore, many patterns of post-progression HRQoL are possible:

(a) The utility between progression and death falls linearly at some con-
stant rate from the last observed time (i.e. last time when progres-
sion occurred) point it was measured (until death).
(b) The PP utility falls either in a concave or convex fashion (Figure 3.5).
(c) The PP utility oscillates between various health states over time and
eventually declines (e.g. due to multiple sequences of treatments).

The assumption that the PP utility falls at a constant rate between progres-
sion and death is unlikely to be realistic. A possible situation that might
entertain this assumption is in long-term survivors who are no longer at risk
for relapse and where the sequelae and the adaptation by the patient to the
sequalae has stabilized. However, this is still better than the even stronger
assumption that the utility is constant from the last observed utility, like
a type of last observation carried forward (see, for example, Dunlop et al.
(2012). This may be completely unrealistic in situations where the disease
worsens (or improves) over time.
In Figure 3.5, the region below zero reflects the state worse than death.
This implies that if a patient has a predicted utility of zero (death) at time t, it
is not possible to predict a utility at time t+1 because the patient is assumed
80 Economic Evaluation of Cancer Drugs

FIGURE 3.5
Post-progression non-linear decreasing utility functions.

to have reached an equivalent of a ‘death’ state. Using a linear decreasing


utility function (or the constant assumption for that matter) does not allow
estimates of utilities to weave between states worse than death (utilities < 0)
and death (utility = 0). Therefore, either a transformation has to be used to
map the utilities on the interval (0 to 1) or a more flexible utility function is
needed. A utility value of zero does not mean the patient is not alive (it is
representative of a health state equivalent to death).

3.8.1 Plausible Post-Progression Utility Behavior


Figure 3.6 shows the PP behavior from patients collected in an observational
study (Khan et al., 2016). The figure appears to show great heterogeneity in

FIGURE 3.6
Post-progression patient profiles.
Health-Related Quality of Life for Cost-Effectiveness 81

FIGURE 3.7
Mean observed utilities post-progression over time with superimposed possible models (using
data from Figure 3.5).

PP utility profiles. Most profiles tend to show a decline in utility after pro-
gression, even if there appear to be some spikes.
These profiles can in general be categorized in some key types of behavior
(Figure 3.7). Figure 3.8a shows rapidly deteriorating utility after progression,
with a ‘spike’ at around 5 months, followed again by a rapid decline. On
the other hand, Figure 3.8c shows an improvement followed by a steadier
decline. If a constant value or a linear function is used to estimate PP util-
ity, the estimated utility and QALY are likely to be misleading. Due to the
highly variable nature of the PP utility–time profile, a modeling approach
using the combined data (for those patients with PP data) who are alive or
dead is considered (because fitting a model to each patient’s PP utility data
profile is likely to be impracticable). Figure 3.7 shows the mean utility plot
post-progression with some possible models.
82 Economic Evaluation of Cancer Drugs

FIGURE 3.8
(a) to (f): Post-progression individual patient profile instances.

Source: Data from Study 3 in Khan et al., 2016.

3.8.2 Non-Linear Models
Several non-linear models are also plausible to model the PP utility: Two
graphical representations of models are shown as an example. The Y-axis in
each of the graphs in Figure 3.8 shows utilities and the X-axis is a time vari-
able showing the number of months PP:
Health-Related Quality of Life for Cost-Effectiveness 83

FIGURE 3.9
Bragg and Packer Model (1962) for post-progression utility.

(i) Using the Bragg and Packer (1962) 4-parameter equation (Ratkwosky,
1989)

EQ − 5D = α + β * exp{− γ (X − δ )2 }

An example of this equation depending upon the parameters α, β,


γ and δ shown in Figure 3.9)
(ii) Using a derivation of the Pareto distribution (Ratkwosky, 1989), the
equation:

EQ − 5D = 1 – 1 / X α

where α < –1
yields a convex shape that, although it does not allow for an
increase, indicates a slower rate of HRQoL/utility deterioration. This
function also allows for an estimation of utility at a time that would
equal zero (unlike the asymptote in (i) above) – (Figure 3.10).

FIGURE 3.10
Pareto-type model for post-progression utility.
84 Economic Evaluation of Cancer Drugs

Example 3.2: Fitting a Number of Models to PP


Utility Data to Estimate the PP QALY
In this example, data is used from Study 3 of Khan et al. (2016) to show
how PP utility can be used to estimate the PP QALY. In the economic
evaluation of cancer treatments, a partitioned survival model is often
used. The total survival experience is partitioned into progression-free
survival (PFS) and post-progression survival (PPS):

OS = PFS + PPS

Consequently, PFS is modeled along with OS and future (extrapolated)


survival proportions for each of PFS and OS are generated.

The steps in this approach are:

(i) Extrapolate PP utility.


(ii) Apply the extrapolated utilities to the PP survival data. Compute
the AUC (QALY) as the QALY for the PFS plus the QALY for the PPS
using: QALY as an area under the curve (AUC) using the formula of
equation (3.1):

QALYAUC = å éë(Qi + Qi + 1 ) / 2 ùû * éë( Si + Si + 1 ) / 2 ùû * [t i + 1 - t i ]

where, Si and Qi are the PP survival and PP utilities respectively


The details are provided in Khan et al. (2019) using data from a lung can-
cer population. Table 3.2 shows a summary of the PP utility models fitted to
extrapolate utilities as far as at least 24 months (the observed follow-up data
was 12 months). The five-parameter model had the best fit (smallest value of
a model fit statistic called the Akaike’s Information Criterion (AIC)).
The results in Tables 3.3 and Table 3.4 show:

(a) The best model to predict longer-term utility data using observed
patterns of utility was the five parameter model because the AIC
was the smallest (best model fit).

TABLE 3.2
Data for Example 3.2: Construction of the QALY
Month (t) Utility (mean) OS (% alive)
0 (baseline) 0.65 100
1 0.71 69
2 0.72 50
3 0.69 32
4 0.75 23
5 0.77 19
6 0.74 4
Health-Related Quality of Life for Cost-Effectiveness 85

TABLE 3.3
Models Fitted to Extrapolate PP Utility
Model Parameter Estimate p-value Equation AIC
Linear α 0.654 –<0.001 0.654 − 0.0219 * Time 115.7
β –0.0219
Exponential λ 0.1415 <0.001 Exp( −0.1415 * Time) 198.0
α
Bragg-Packer
β
–0.08323
0.693710
0.983
0.8617 {
α + β * exp − γ * (Time − δ )2 } 106.3

γ 0.005245 0.8990
δ 1.697112 0.6575
Pareto α 0.4073 <0.001 253.4
1 − 1 – 1 / Time0.4073 
Beta α 0.9595 0.0002 101.2
α Timeβ * (1 − Time)
γ
β 0.1270 0.1440
γ 1.3985 0.0059
Lorentz α –0.4832 0.968 106.2
α +β / 1 + γ * (Time − δ )2 
β 1.0939 0.927
γ 0.00532 0.948
δ 1.77192 0.784

Rational α –0.04831 0.5562 106.4


function β 0.5997 <.0001
(b + g * Time ) /
γ –0.01726 0.8670 (1 + a * Time + d * Time ) 2

δ 0.005538 0.7464

α
Five
parameter β
–0.01618
0.5985
0.7419
<.0001 (β + γ * Time+ε * Time 2
/
92.4

γ <.0001
δ
0.1000
0.003861 0.8397
1 + α * Time + δ * Time 2 )
ε –0.00148 0.7431

(b) Not all parameters were statistically significant, suggesting a model


with fewer parameters (and possibly easier interpretation) could be
used.
(c) The PP QALY was estimated to be lowest for the Pareto model and
the highest for the linear model (which assumes utilities are falling
at a constant rate over time).
(d) The model with the greatest uncertainty was also the Pareto because
the extrapolated QALY constituted more than 32% of the total QALY.
Uncertainty here is determined by how much of the QALY is deter-
mined through extrapolation relative to the total QALY.
TABLE 3.4
86

Extrapolated Mean Utilities Beyond 12 Months for Each Model


Month PP PPSa (%) Linear Exponential Bragg-P Pareto Beta Lorentz Rational 5-parameter

1 100 0.6323866 0.8680552 0.6087141 1.0000000 0.6035076 0.6077124 0.6082953 0.5783413


2 89 0.6104600 0.7535198 0.6101463 0.6699382 0.6193203 0.6097999 0.6104777 0.5606051
3 80 0.5885334 0.6540968 0.6043310 0.5299948 0.6109764 0.6042279 0.6053164 0.5550958
4 71 0.5666067 0.5677921 0.5914498 0.4488172 0.5919137 0.5912281 0.5924955 0.5720147
5 69 0.5446801 0.4928748 0.5719015 0.3945166 0.5667773 0.5713266 0.5722454 0.5519026
6 63 0.5227535 0.4278426 0.5462812 0.3550638 0.5378116 0.5452932 0.5453433 0.5255827
7 55 0.5008269 0.3713909 0.5153507 0.3248014 0.5063101 0.5140697 0.5130163 0.5140768
8 55 0.4789003 0.3223878 0.4800006 0.3006798 0.4731083 0.4786924 0.4767720 0.5421457
9 55 0.4569736 0.2798504 0.4412078 0.2808945 0.4387921 0.4402168 0.4382009 0.4000281
10 55 0.4350470 0.2429256 0.3999912 0.2643017 0.4038001 0.3996548 0.3987983 0.3997085
11 49 0.4131204 0.2108728 0.3573677 0.2501377 0.3684792 0.3579274 0.3598385 0.3285215
12 43 0.3911938 0.1830493 0.3143117 0.2378708 0.3331174 0.3158357 0.3223101 0.3172918
13* 36 0.3810000 0.1588969 0.2717202 0.2271183 0.2979837 0.2740473 0.2869069 0.2763705
14* 33 0.3600000 0.1379312 0.2303847 0.2175969 0.2632678 0.2330954 0.2540558 0.2368746
15* 32 0.3390000 0.1197319 0.1909714 0.2090917 0.2292018 0.1933870 0.2239648 0.1988992
16* 29 0.3180000 0.1039339 0.1540096 0.2014369 0.1959969 0.1552166 0.1966766 0.1627003
17* 29 0.2970000 0.0902204 0.1198883 0.1945018 0.1638710 0.1187826 0.1721177 0.1284338
18* 25 0.2760000 0.0783163 0.0888597 0.1881820 0.1330590 0.0842040 0.1501403 0.0961758
19* 23 0.2550000 0.0679828 0.0610497 0.1823931 0.1038273 0.0515365 0.1305537 0.0659408
20* 20 0.2340000 0.0590129 0.0364715 0.1770658 0.0764966 0.0207869 0.1131479 0.0376981
21* 18 0.2130000 0.0512264 0.0150440 0.1721430 0.0514822 –0.0080745 0.0977094 0.0113846
22* 15 0.1920000 0.0444674 −0.0033898 0.1675768 0.0293806 –0.0351042 0.0840311 –0.0130844
23* 13 0.1710000 0.0386001 −0.0190427 0.1633268 0.0112157 –0.0603764 0.0719187 –0.0358072
24* 11 0.1500000 0.0335070 −0.0321654 0.1593588 ~0 –0.0839771 0.0611935 –0.0568892
PP QALY# 4.648132 3.747115 4.160589 3.732875 4.272484 4.135935 4.304773 4.028435
%Extrapolated@ 28.25819 21.88337 18.89631 32.44121 21.13157 18.50418 21.69392 19.02141
a Note: Using a 3-knot Royston-Parmar model
* Extrapolated utilities using modeling; #using ∑ ( Qi+Qi + 1)/2  * ( Si +Si+1) /2  * [ ti+1- ti ]
   
@ percent of PP QALY extrapolated = (Total calculated QALY – QALY without extrapolated)/ Total calculated QALY*100.
Economic Evaluation of Cancer Drugs
Health-Related Quality of Life for Cost-Effectiveness 87

3.9 HRQoL Issues in Health Technology


Appraisals of Cancer Drugs
In HTAs of cancer drugs, there are several aspects of HRQoL issues that have
been identified during the reporting of cost-effectiveness analyses. These
are important to appreciate, especially regarding utility data. For example,
a summary of the issues from several HTA submissions for lung cancer are
presented in Table 3.5. For the moment, we focus on the issues relating to
utility (HRQoL) data.

TABLE 3.5
Data Source for Key HTA Submissions
Treatment Submissiona Utilities
Afatinib 920/13 1L (SMC, • Lack of details
2014) • Data collected from historical sources
TA310 1L (NICE, Health state utilities derived from LUX-Lung and
2014b) LUCEOR trials (Chouaid, 2012) in base case and
assumed to be the same across treatment arms
other sources used in sensitivity analysis (Doyle, 2008;
Lewis, 2010; Nafees, 2008)
Disutilities sourced from LUX-Lung 1, LUX-Lung 3, and
(Nafees, 2008)
Crizotinib TA296 2L (NICE, Utility collected in PROFILE 1007 using the EQ-5D
2013d) Calculated EQ-5D in PFS by weighting the value at each
time point by the number of patients at each time point
A weighted average utility at the end of treatment was
extrapolated to post-progression health states
No utility decrement was applied to AE occurrences
865/13 2L (SMC, Utility data was derived using EQ-5D from the clinical
2013) trials. Values for BSC were assigned using assumptions
pCODR, 2013 2L Utility data was derived using EQ-5D from the clinical
(pCODR, 2013b) trials
Erlotinib TA258 1L (NICE, Utilities were taken from (Nafees, 2008); a study
2012) commissioned for second line NSCLC, with 100
members of the general population, using SG and VAS
techniques
TA162 2L (NICE, No detail
2008)
749/11 1L (SMC, Primary study derived from a survey that used the
2012) standard gamble technique with 100 members of the
UK public
220/05 2L (SMC, Primary study from a sample of the UK general
2006) population using appropriate methods (not specified)
07-2013 1L (PBAC, Utilities for patients with stable disease and progressive
2013b) disease directly from (Nafees, 2008)
(Continued)
88 Economic Evaluation of Cancer Drugs

TABLE 3.5 (CONTINUED)


Data Source for Key HTA Submissions
Treatment Submissiona Utilities
Gefitinib TA192 1L (NICE, Most utilities were sourced from (Nafees, 2008)
2010b) Progression-free and therapy were sourced from an ERG
report (2006)
Disutility for anemia was sourced externally (2009)
615/10 1L (SMC, Sourced from literature (not specified)
2010b)
07-2013 1L (PBAC, Utilities were adapted from HRQoL data reported in the
2013c) trial
Pemetrexed TA181 1L (NICE, Utilities were taken from (Nafees 2008); a study
2009) commissioned for second line aNSCLC, with 100
members of the general population, using SG and VAS
techniques.
TA309 MTx (NICE, EQ-5D data from a mixed regression model of the
2014a) PARAMOUNT trial.
531/09 1L (SMC, Sourced from a utility valuation survey using SG in 100
2010a) members of the general population in the UK
342/07 2nd line Sourced from a study of 100 members of the general
(SMC, 2008) population using SG to value 18 states and adverse
events)
Key: AE: Adverse events; EQ-5D: EuroQoL five dimension; ERG: Evidence review group; NICE:
National Institute for Health and Care Excellence; NHS: National Health Service; NSCLC: Non-
small-cell lung cancer; OS: Overall survival; PBAC: Pharmaceutical Benefits Advisory
Committee; pCODR: pan-Canadian Oncology Drug Review; PFS: Progression-free survival;
PSSRU: Personal Social Services Research Unit; SG: Standard gamble; SMC: Scottish Medicines
Consortium; SPC: Summary of product characteristics; TA: technology appraisal; UK: United
Kingdom; VAS: Visual analogue scale.
a Note: References for the HTAs are provided in the bibliography.

The common features of HTA submissions, including HRQoL issues were:

(i) Utility data were collected from historical sources and limited
details were provided on the assumptions. Where assumptions were
provided, some of these were problematic (e.g. utilities the same
between the two arms of the treatment).
(ii) In some cases, utilities from later lines of treatment were used
(imputed) for earlier lines of treatment. In some cases it may also be the
case that utilities from earlier lines of treatments are used to impute for
later lines of treatments – this might happen when first-line therapy
has resulted in progression and utilities collected on the first-line treat-
ment are ‘carried forward’ while patients take later lines of treatment.
(iii) Utilities were used from separate utility studies (e.g. TTO or SG
methods). That is, utilities were not collected in the clinical trial.
Health-Related Quality of Life for Cost-Effectiveness 89

(iv) Utilities for patients in the SD and PD health states were determined
from an external study (Nafees et al., 2008).
(v) Lack of details about where health state utilities have been sourced
from, either directly from the trial or from external studies such as
Nafees et al. (2008) or through mapping (Scuffham, Whitty, Mitchell,
& Viney, 2008).

3.10 Summary
In this chapter we have discussed the reasons why HRQoL is important.
We have also discussed the differences between cancer-specific and generic
HRQoL measures. We showed an example of how utilities can be con-
structed from generic HRQoL measures for economic evaluation, focusing
on the EQ-5D. The computation of the QALY was outlined in a general and
a cancer-specific context. New methodology was introduced on modeling
HRQoL for economic evaluation in cancer by investigating the behavior of
post-progression utility – a common issue in many HTAs. We concluded the
chapter by reporting the issues around the use of HRQoL measures in cost-
effectiveness analyses of cancer drugs.

3.11  Exercises for Chapter 3


1. Generic HRQoL measures should be avoided at all cost. Discuss.
2. Compare and contrast the approaches to estimating HRQoL for eco-
nomic evaluation.
3. How would you go about designing a study to collect HRQoL data
for an economic evaluation in a cancer trial? What challenges would
you encounter?
4. Extrapolation of survival data is common and therefore we should
be allowed to extrapolate utility data. Discuss.
5. Do you think that treatment effects from CSMs and generic mea-
sures are comparable? Can effects from generic measures be under-
stood in the same way as CSMs?
4
Introductory Statistical Methods for
Economic Evaluation in Cancer

4.1 Introduction
In this chapter, we will introduce some essential statistical concepts that will
allow the reader to appreciate statistical methods used in economic evaluation.
Economic evaluation relies heavily on the use of statistical and clinical trial-
related methodology to quantify and present cost-effectiveness analyses. When
these methods are used in a cancer context, they may be more challenging.
Some methods presented are slightly more technical and advanced. However,
it is more important to understand the concepts rather than the finer technical
details for which statistical software can be used. References in the bibliography
can be consulted for interested readers. The statistical methods underlying eco-
nomic evaluation in cancer broadly consist of appreciating the distribution of
data, presenting data using summary measures, understanding survival anal-
yses methods, modeling, and simulation. Some of these will be covered in this
chapter. Statistical methods in economic evaluation do not really revolve around
complex hypothesis-testing problems and statistical inference. Although many
HTAs and cost-effectiveness analyses present p-values (the observed treatment
difference being due to chance) in some form or another, these are secondary
to the important objective of economic evaluation. Statistical methods in eco-
nomic evaluation are mainly used to quantify the uncertainty in the decision-
making process – whether to pay for a new cancer drug (or not). We therefore
start with the concepts of uncertainty and variability.

4.2 Uncertainty and Variability


Uncertainty is central to the decision-making process that determines whether
a new cancer treatment is cost-effective (or not). Uncertainty is sometimes
confused with words like ‘variability’ ‘and ‘probability.’ It is important to dis-
entangle some of these concepts so that we can be precise in our language.

91
92 Economic Evaluation of Cancer Drugs

Indeed, Kaplan (1997) noted that “50% of the problems in the world result
from people using the same words with different meanings” and “the other
50% come from people using different words with the same meaning.”

4.2.1 Uncertainty
Uncertainty in everyday language suggests we are “not sure” or “we do not
know” about some statement. Technically, uncertainty refers to an unknown
true value of some measure or quantity. For example, we might believe that
the proportion of deaths in the population is 30%. The statement: “the pro-
portion of deaths in the population is 30%” can be true or false. The quanti-
fication of this statement is made in terms of probability. That is, we may be
certain the statement is true (with a probability of 1) or is highly uncertain,
with a probability close to 0, or false if the probability equals 0. Similarly, we
might believe some risk factors are related to survival or increased costs. The
choice of selecting the risk factors (e.g. we choose age and gender, but we could
have chosen weight and ECOG) is also subject to uncertainty. Uncertainty can
arise from many factors (poor communication, subjective belief, imprecise
approximation, etc.) of which one might be statistical variation (variability),
which leads us to distinguishing between variability and uncertainty.

4.2.2 Variability
Variability is a feature of observed phenomena, or, for our purposes, data
collected from a clinical trial or some other study. Variability occurs where
multiple measures are observed. For example, if we measure the HRQoL of
10 ‘identical’ (or similar) cancer patients at 3 months, each value may be dif-
ferent. The dispersion of the values around some central point (e.g. the mean)
can be expressed as a numerical quantity – termed the ‘variance.’ The larger
this quantity is, the greater the variability (or dispersion). The question of
what the central measure (i.e. the mean) is allows us to make a statement such
as “The mean is 5.7.” The uncertainty of this statement is quantified through
the use of probability. If the variability is large (reflected in say a range of val-
ues between 0 to 10), we might be less certain compared to observing values
between 5.5 and 5.9 (more certainty about the mean).
In economic evaluation, whether for a cancer intervention or otherwise, we
are interested in expressing the uncertainty of a statement such as “The new
intervention is cost-effective” in terms of:

(i) The probability (a single value) of this statement being true.


(ii) The range of values (e.g. ICERs) for which the statement is plausible
with some level of probability (e.g. ICERs between £18,000 to £22,000
have between 85% to 90% chance of cost-effectiveness at a threshold
WTP of £30,000).
Introductory Statistical Methods for Economic Evaluation in Cancer 93

The statistical tools used to ultimately determine (i) and (ii) are described in
this chapter.

4.2.2.1 Hypothesis Testing
In classical (frequentist) statistics, the hypothesis-testing framework pres-
ents two scenarios: the null hypothesis and the alternative hypothesis. For
efficacy trials in cancer, with OS as a primary endpoint for two treatments
we could have:

H0 (null hypothesis): no difference in survival between treatments


H1 (alternative): there is a difference (a target effect size in mind)

After we conduct the trial, we collect the data and are then faced with a deci-
sion to choose (declare) one or the other hypothesis to be true/false, based on
the data (evidence). The p-value (the likelihood of rejecting H0, when in fact
it is true – that is akin to declaring a treatment as being efficacious, when in
fact it is not) is used as a way to judge the evidence in favor of one hypothesis
over the other (e.g. choose H0 or H1).
In a cost-effectiveness framework, we might have the hypothesis presented
in terms of incremental net monetary benefit (INMB, introduced in Chapter 1)

H0 (null hypothesis): INMB = 0 (not cost-effective, for some given WTP)


H1 (alternative): INMB not = 0 (cost-effective for some given WTP)

Rather than deciding in favor of one or the other hypothesis, a more infor-
mative question might be “What is the chance of H0” being true (or false). In
other words, we wish to quantify the uncertainty of a hypothesis rather than
simply choose one or the other. There could be several reasons why we may
not be comfortable in deciding whether H0 is true or not (for example, a small
sample size, some extreme values, excess variability, and so on). Alternative
inference paradigms are possible (Bayesian), which we will briefly discuss
later. The statistical measures used to determine uncertainty around cost-
effectiveness decisions will take the form of traditional statistical quantities
(e.g. mean, median, rates, proportions, variance, probabilities, and confi-
dence intervals) determined from simple to more complicated analyses.

4.3 Distributions: Cost, Utility, and Survival Data


The distributions of outcomes such as survival, health resource use, costs, and
utility data are important to appreciate before starting any statistical analy-
ses. Examples of some typical distribution of each are shown in Figure 4.1a–c.
94 Economic Evaluation of Cancer Drugs

FIGURE 4.1
Distribution of (a) survival times, (b) costs and (c) HRQoL.

Figure 4.1a shows the distribution of survival times typical of OS or PFS.


The distribution is skewed, with some patients having longer survival times.
These are consistent with a family of exponentially distributed outcomes
(see Collet, 2014; Crowther, 2016; Collet, 2017). The extremes of the tail might
also indicate patients still alive in the trial at the end of follow-up.
Figure 4.1b shows the distribution of costs and health resource. These are
positive (≥ 0) and like survival data also have some extreme values for dif-
ferent reasons (the two could be correlated because those who live longer
might also have more extreme costs). Some costs appear often (spike) at some
value(s) and are said to be ‘over-dispersed.’ When data exhibit skewness and
over-dispersion, that is, the presence of greater variability in a data set than
would be expected, special statistical modeling approaches are needed to
estimate the mean cost.
Figure 4.1c shows the distribution of HRQoL data (see also utility data,
Chapter 5). The distribution is also over-dispersed particularly at values
at 1. That is, there are lots of values near to 1 (ceiling effects) and this can
cause problems in presenting mean estimates, unless suitable statistical
approaches are used to handle excess variability. Here, over-dispersion may
also occur near to the extremes. This makes the distribution look like a ‘U’
Introductory Statistical Methods for Economic Evaluation in Cancer 95

shape and hence the term ‘U-shaped distribution.’ Special statistical models
can be used to analyze these types of data.
It is important to note that regardless of the shape of the distribution, we
will need to determine the mean value of the data for the purposes of economic
evaluation. It might be tempting to use the median or other statistic such as a
truncated or trimmed mean (the mean of some data with some extreme val-
ues omitted), but that would not be appropriate. At the current time, only the
arithmetic mean is considered suitable for an economic evaluation.

4.4 Important Measures Used in Cancer Trials


4.4.1 Time-to-Event Endpoints
The primary outcomes in confirmatory Phase III trials are often time-to-
event outcomes such as OS and PFS. The event is often death or disease pro-
gression and the time taken to reach the event is the outcome of interest.
Since some patients may live for a long time (long after the trial has been
completed), there are instances of some very extreme survival times that
result in a ‘skewed’ distribution. For this reason, the median is often used to
present the summary statistics. Figure 4.2 shows a Kaplan-Meier (KM) plot
of two treatments (A and B) from a cancer trial.

FIGURE 4.2
Kaplan-Meier plot of two treatments (A and B) from a cancer trial.
96 Economic Evaluation of Cancer Drugs

The y-axis shows the proportion (probability) of patients being alive at


given time points. The x-axis is the time since randomization presented in
years or months (or days, depending on how often the event occurs.) At time
0 (when the trial started), 100% of the patients are alive. It may happen that
some instantaneous deaths could occur after the patient has been random-
ized or on the day of randomization. Hence, it is not necessary that at time
0 exactly 100% of patients are alive. When reading numbers from survival
curves, we should be aware whether the time unit is in days, months, or
years. Over time, fewer patients are alive. The maximal follow up is around
6 months (the last time point on the KM curve) in this example.
The median survival is observed by drawing a horizontal line starting
from 50% (or 0.50) until it reaches the curves. By drawing a vertical line, we
can read off the values. In this example, the median survival times for treat-
ments A and B are each around 1 month. Hence, neither treatment appears
to improve median survival time. What is important to know is that the
proportions calculated at each time point are determined through calcula-
tions involving the number of deaths (events) occurring in the preceding
time interval based on those alive (called the risk set). The KM estimates of
the (cumulative) proportions alive at each time point are called nonparamet-
ric estimates because we do not need to fit a theoretical curve to the overall
survival times in order to estimate important measures of treatment benefit.
Figure 4.3 shows a KM curve for PFS. Here, there are more ‘steps’ in the
curve, due to fewer events. The steps get longer over time. These steps are
related to the frequency at which ‘progression’ or ‘death’ events accumulate.
The median is estimated in the same way as for the OS curve. The median
for Treatment A is about 3.25 months and for B is about 2.75 months. Hence
Treatment A improves survival by 0.5 months (about 2 weeks). The crosses
on the lines show the presence of censoring, or patients who have not had an
event during the observation time. Over time, as fewer patients live or have
not progressed, the curve takes a more discrete shape and is less informa-
tive (more uncertain) about the event rates, especially where there are fewer
patients and therefore fewer events.

4.4.2 Median Survival
In some KM estimates, the median may not exist (Figure 4.4). The reason for
this is because there are insufficient events, or that patients have not been
followed up for long enough. It is an important consideration in trial design
(Chapter 6) to ensure that patients can have adequate follow-up so that reli-
able cost and survival data can be determined.
The median is often reported with 95% confidence intervals. A median OS
of 5 months with a 95% CI of (3 to 7 months) tells us that the true median
lies somewhere between 3 to 7 months with 95% confidence. Note that the
median calculated from the survival curve is not the same as the simple raw
median when some data are censored (i.e. patients that have not died by the
Introductory Statistical Methods for Economic Evaluation in Cancer 97

FIGURE 4.3
Survival rates in the presence of censored data (fewer events).

FIGURE 4.4
Kaplan-Meier curve where median does not exist due to too few events.
98 Economic Evaluation of Cancer Drugs

end of follow-up). If there are no censored data, then the estimate of the raw
median and the median from the KM curve are the same.

4.4.3 Hazard Rate and Hazard Ratio


An important concept in analyses of time-to-event data is related to ‘hazard
rates.’ A ‘hazard’ in everyday language is synonymous with ‘risk.’ When
one says it is hazardous (and often illegal) to drive after drinking alcohol,
it suggests there is a risk of an event (e.g. a car crash or death). For patients
with cancer, unfortunately, there is a risk of death from their disease. That
risk might be immediate or it could happen over time. Sometimes, the
potential hazard for death changes. For example, immediately after com-
plicated surgery, the hazard for death is high; then, as the patient recovers,
the hazard falls. In other cases, the hazard might be increasing (e.g. as with
increasing age or severity of the disease). The hazard might also remain
constant (this does not mean patients do not die, but rather the hazard for
death during say months 2 to 3 is the same as it is during months 3 to 4). The
hazard rates are often depicted using graphs (called hazard functions) as
shown, for example, in Figure 4.5. A hazard rate is also referred to a ‘failure’
rate, where ‘failure’ might refer to a death event (thought of in simplistic
terms as the failure of a treatment to prolong survival or time until disease
progression).
An ‘incidence’ is a measure of the frequency with which a disease occurs
in a population over a specified time period and ‘incidence rate’ or ‘inci-
dence’ is numerically defined as the number of new cases of a disease within
a time period as a proportion of the number of people at risk for the disease.
So a failure rate is basically an incidence rate where the incidence is a nega-
tive event, namely a failure.

FIGURE 4.5
Examples of hazard functions.
Introductory Statistical Methods for Economic Evaluation in Cancer 99

A more technical description of a hazard rate is the chance of an event


occurring within a specified time interval, given or conditional on the patient
having survived to some time, t. If the interval is long (e.g. between 6 months
and 2 years), the rate might fall. If the interval is short (e.g. between 2 and 4
weeks), it might increase. The hazard rate, h(t) defined below is not a prob-
ability. Its value must be >0 and can be >1. A high value means that chances
of death, (death rates) will be high for a given time interval.

P(t < T ≤ t + ∆t|T > t)


h(t) = lim
∆t → 0 ∆t
For example, if the hazard rate of disease progression (for an individual)
at 3 months is 0.25, this means that, if the hazard stays at the value of 0.25
(assuming a constant hazard) over the next 2 months, one would expect dis-
ease progression 0.25 times. If the event is repeatable (e.g. second disease
progression), then the reciprocal of this value (1/0.25) = 4 means that we
can expect the next progression for that individual to take place in about 4
months’ time, on average. If the hazard was 1.5, then we would expect this
individual to have disease progression 1.5 times over the year (assuming
the hazard stays constant over that year); and the next progression would be
expected to occur in about 8 months’ time (1/1.5). This is important because,
in some cancers, very few events might be observed. This will involve longer
follow-up to get the events, the use of external (real-world data) or extrapola-
tion (Section 4.6.3), or even the abandonment of the trial (due to an inconclu-
sive result).
When one considers hazards, one could imagine that each person has his
or her own hazard or risk of an event. For example, as one sits at a com-
puter, the risk of death is small (but not zero). However as one gets into their
car to drive, the risk increases, and it will change again when they return
home. The true hazard function (the evolution of the hazard over time) will
depend on lots of factors and conditions. The combined hazard functions
in a group of patients in a cancer trial will include some patients with high
(short survival time) and some with relatively lower (longer survival time)
hazards, such that across lots of individuals, so that the general trend is
captured.
When we have hazard rates for two treatment groups, it is the relative
hazard rates that are important. This is termed as the ‘hazard rate ratio’ or
simply the ‘hazard ratio.’

4.4.4 Hazard Ratio
In its simplest terms, the hazard (rate) ratio (HR) is the ratio of chances of
events occurring in one group compared to the other. A more detailed defi-
nition is “the ratio of two hazard rates or of two hazard functions, either at a
particular point in time or averaged over a long period” (Day, 2002). The HR
100 Economic Evaluation of Cancer Drugs

determined from two patient groups can be interpreted as a reduction or


increase in the risk (the hazard) of an event in a similar way to the relative
risk or risk ratio when the time interval for an event is small. In this sense,
the HR describes the relative risk based on a comparison of events (Spruance,
2004). Time occurrence of the event is not of interest or not available (Saad et
al., 2018). The hazard ratio is frequently interpreted as a risk ratio (or relative
risk), but these are not technically the same (Stare, 2016).
In a comparison of two treatments for an outcome like death, an HR of 1
suggests no treatment benefit. An HR of 0.8 would be interpreted as the risk
of death being 20% lower in the group taking treatment A compared to B.
An HR of 1.20 suggests that the risk of death is 20% higher for A versus B.
In general, an HR < 1 suggests the experimental intervention is better when
compared to a control; and an HR > 1 implies the experimental intervention
is worse than the control for the outcome of interest. HRs are important for
interpreting clinical efficacy, but they are also important in economic evalu-
ation for simulating survival patterns.

Example 4.1
The following results are reported in NICE TA189 (May, 2010) for the treat-
ment of hepatocellular carcinoma with the drug sorafenib (Figure 4.6).
The HR of sorafenib versus placebo (S vs. P) was 0.69 with a 95% CI of
(0.55, 0.88): on average, the risk of mortality was reduced with sorafenib

FIGURE 4.6
OS in hepatocellular carcinoma (NICE, TA189).
Introductory Statistical Methods for Economic Evaluation in Cancer 101

by about 31% and the true risk reduction lies somewhere between 45%
and 12% with 95% confidence. The p-value of 0.00058 shows strong evi-
dence to reject the null hypothesis (of no difference between S and P). An
interim analysis was carried out using an adjusted p-value (p = 0.0077 >
0.00058) to take into account looking at the data early.

4.4.5 Survival Rates and Proportions


A survival rate is the estimated proportion of patients alive at a given time
point. This can be read from the survival curve. As noted, earlier estimates
are likely to be more precise than those at the latter part of the curve, where
fewer patients are alive. In Figure 4.2, the proportion alive at 2 months was
36%. At later time points (say 5 months), the survival rates are more uncer-
tain. This is likely to be reflected in wider confidence intervals at these and
later time points.
A common occurrence is when we might wish to estimate the proportion
of patients alive at a given time point (e.g. 12 months) if we are provided with
a median. For example, if the median is 6 months, then the proportion alive
is 50%. If we assume the KM is approximately an exponentially decaying
curve, we can use this information to estimate the survival proportion at 12
months. The survival proportion can be denoted as:

S ( t ) = exp ( - lt ) ,

where t is the time to an event of interest. The value of λ is important. It


represents the rate of death (event), also called a constant hazard (of death) –
i.e. the rate of death is constant over time. Hence, for a median of 6 months,
S ( t ) = 0.50 = exp ( -l * 6 ) at 6 months, the proportion alive is 50%. We can esti-
mate λ : −log (0.50 ) / 6 = λ ; λ = 0.1155. The proportion alive at 12 months is
estimated as S (12) = exp ( −0.1155 ∗ 12) = 0.25 or 25%.
We can convert a hazard rate into a probability and vice versa: The hazard
rate λ (or incidence rate of death, assumed constant) can be estimated by the
formula:

-Log ( 1 - p ) /t,

where t is the time point of interest, and π = the survival rate at time t.
When the hazard function is no longer constant but changes over time,
the survival curves may cross as in Figure 4.6a. In this HTA (TA179, 2009)
in gastrointestinal stromal tumors, the difference in median survival val-
ues was about 8 weeks. Differences in the proportion alive at 5 weeks are
larger. In Figure 4.7a, the survival curves cross at the median. Differences
at later time points are larger and using the median difference would be
misleading.
102 Economic Evaluation of Cancer Drugs

FIGURE 4.7
Nonproportional or changing hazards in general (a) and observed in a comparison of sunitinib
versus placebo (b) (NICE, HTA TA179, 2009).

4.4.6 Relationship between Hazard Rate and Survival Rate


In this section the relationship between transition probabilities and sur-
vival rates is briefly explored. This is important because in some situations
a health economic model (e.g. Markov model, Chapter 7) is used where sur-
vival rates are modeled. Determining transition probabilities from survival
rates becomes important for this purpose. In some cases, the primary end-
points for cancer trials are survival rates at fixed time points (e.g. two-year
survival rates compared between experimental and control treatments).
Therefore, the relationship between survival rates changing from one time
point to another (or a transition) is related to the death or hazard rate. It is
plausible that the hazard for death increase for some patients who progress
in their disease and therefore the rates of transition between PD and death
are high.
Earlier, we saw how an exponential model had a survival function of the
form: S (t ) = e − Φ06 Xt . In this expression λ is the hazard rate or the rate of an
event (such as death) per unit of time: e.g. 5 deaths per month (unit of time is
months). The probability of death can be expressed as a rate, where π is the
proportion alive at time t:

π= e − Φ06Xt and hence λ = −(log(π) / t)

For example, if after 2 years 50 patients out of 200 are alive, and assuming
the death rate is constant (the same number of deaths each year), the death
rate is: [ –log(1 - 0.25)][/5 = -log[0.75] / 5 = -0.287 / 5 = 0.0574 and the prob-
ability of death at year 3 would be: 1 – exp ( −0.0574 ∗ 3 ) = 0.158 (15.8% ).
To distinguish between a rate and a probability, as an example, 4 patients
are followed up of whom 3 die at various times (5, 3, 2, and 1 months).
Introductory Statistical Methods for Economic Evaluation in Cancer 103

The rate of death = 3/11 or 3 per 11 person-years or 0.27 persons per year.
The probability of death is 3 out of 4 = 0.75.

4.4.7 Transition Probability and Matrix


A transition probability is the probability of moving from a given health
state to the next health state in a Markovian process. In other words, the
transition probability is the chance of moving between health states over (dis-
crete) time. The Markovian process means that the chance of moving into a
future health state is determined only by the current health state (and not
other, previous, health states). A patient cannot move from a state of ‘death’
to ‘progressive disease,’ although the reverse is possible.
The source of the transition probability is also important – where was it
taken from, how reliable it is, whether it is a conditional or a joint probability.
For example, rates of adverse events are extracted from clinical trial reports.
Many adverse events summary tables report percentages using the denomi-
nator of the intent-to-treat (ITT) or safety population (i.e. the total sample
size). An example of a transition matrix for patients with health states mild,
moderate and death after 1 month of treatment with an experimental treat-
ment might be as shown in Table 4.1.
Prior to the computing the transition matrix in Table 4.2, patients would
have been randomized to one of two treatments: experimental or control.
Patients at the start of their treatment are considered to be in either a mild
or moderate state (e.g. based on ECOG or similar inclusion criteria). The pro-
portion of patients who started the trial at baseline in a mild health state and
then remained in the same state after treatment was 45%. Very few patients
(5%) who started the trial in a moderate state improved into a mild state post-
baseline. In fact, 60% of patients who presented with moderate disease did
not change states after starting treatment.
In its simplest form, it is assumed that the probability of moving from one
health state to another (namely the transition probability) is the same for
each future time point (i.e. is constant over time) although the probabilities
of being in each health state at each time point or cycle will be different. In
order to compute how the transition matrix will look like after 1 month, we

TABLE 4.1
Example Transition Matrix on Experimental Treatment after 1 Month
Post-Randomization
Post-Baseline (After Treatment)
Mild Moderate Death Total
Baseline Mild 0.45 0.35 0.20 1
Moderate 0.05 0.60 0.35 1
Death 0 0 1 1
104 Economic Evaluation of Cancer Drugs

TABLE 4.2
Example Survival Times for
Determining Restricted Mean
Survival Time
Patient (Months) Death
1 3 Yes
2 4 No
3 6 Yes
4 7 Yes
5 2 Yes
6 8 Yes
7 3 No
8 9 No

need to know the initial probabilities (or the probabilities at baseline). These
are assumed to be a0=[0.45, 0.35, 0.20] (the fact that these are the same as the
first row is coincidental).
After 1 month of treatment, therefore, the transition matrix is a0 multiplied by
the 3 × 3 transition matrix above: a1 = [0.45 × 0.45 + 0.35 × 0.05 + 0.20 × 0, 0.45 ×
0.35 + 0.35 × 0.60 + 0.30 × 0, 0.45 × 0.30 + 0.35 × 0.35 + 0.30 × 1] = [0.22, 0.37, 0.56]
The 1 × 3 matrix a1= [0.22, 0.37, 0.56] describes the probabilities in each of
the health states mild, moderate, and death; 1 month after treatment, this
1 × 3 matrix now becomes the initial matrix needed for further calculations.
After 2 months (cycle 2 of the process), this would be: a1 multiplied by the
3 × 3 transition matrix, and so on. In general, one can compute the proportion
of patients for the n + 1th step in any of the above 3 health states by simply
multiplying an by the given transition matrix, where an is the 1 × 3 matrix at
the current step:
a 1 = a 0∗P

a 2 = a 1∗P
a 3 = a 2∗P ……..a n = a n − 1∗P

4.4.8 Relation between Transition Probability and Survival Rates


The Kaplan-Meier method estimates probabilities of survival within given
discrete intervals. If the survival rate at 2 years = S(2) = 0.80, then the survival
rate in the previous year before (year 1) can be written as S(1) = 0.9. In general,
the survival during the previous year u, will be S(t – u), where the current
survival rate at time t is S(t).

More technical (can be left out without loss of generality)


If the survival rate is S(t), then the failure (death) rate is 1 – S(t). The prob-
ability of surviving during an interval of time is S ( t ) /S ( t - u ) , hence the
Introductory Statistical Methods for Economic Evaluation in Cancer 105

probability of death (failure) during an interval of time is 1 - éëS ( t ) /S ( t - u ) ùû .


The transition probability (TRp) for an event (e.g. death) is defined as
TR p = 1 − S (t )/S (t − u) .
In survival analysis, there is a key relationship between the survivor func-
tion, hazard function, and the probability density function (see Collett, 2014
for further details) is defined as:

f ( t ) = h ( t ) * S ( t ) and further, S ( t ) = exp {- H ( t )} ,

where f(t) is the probability density function, S(t) is the survival function,
and H(t) is the cumulative hazard function. Therefore, the transition prob-
ability, TRp is:

1 - éëS ( t ) / S ( t - u ) ùû = 1 - éëexp {- H ( t )} / exp {- H ( t - u )}ùû



= 1 - éëexp {H ( t - u ) – H ( t )}ùû

Example 4.2
For example, if the survival times follow an exponential distribution,
then the probability density function (PDF) is:

f ( t ) = lexp{-lt}

Here,

h ( t ) = l , S ( t ) = exp{lt} and H ( t ) = lt

Using,

TR p = 1 - éëS ( t ) /S ( t - u ) ùû = 1 - éexp{- H ( t )} / exp{- H ( t - u )}ù


ë û

= 1 - éexp{H ( t - u ) - H ( t )}ù ,
ë û

then

TR p = 1 – exp{l ( t - u ) – lt} = 1 - exp{-lu}

The above is the constant transition probability of moving from one


time point to another. Typically what is required in cancer trials is the
transition probability of moving between health states such as death,
progression, and being stable over time.
106 Economic Evaluation of Cancer Drugs

Example 4.3
For example, if t = 6 months and the proportion alive is 30%, the HR =
–log (1 – 0.30)/6 = 0.0594. Converting a rate into a constant probability is
determined from:

( )
π = 1 − exp − λ ∗t hence, for λ = 0.0594, at 6 months,

yielding a survival probability of 0.70, which agrees with the above.


We will revisit this in an example when discussing Markov models
(Chapter 7). At this junction, we simply need to appreciate that transition
probabilities, hazard rates, and survival rates are all related quantities.

4.4.9 Proportional Hazards
Many statistical analyses of survival data rely upon the validity of the pro-
portional hazards (PH) assumption mentioned earlier (strictly speaking, the
name PH model can be generalized to allow for non-PH). The PH assump-
tion means that the hazard functions for two different levels of a covariate
are proportional for all values of t. For example, if women aged 60, taking
treatment A have twice the risk of death compared to women at age 60 who
take treatment B, then it is assumed they will also have twice the risk of
death (on Treatment A compared to B) aged 70, or for that fact, any other
age. Violation of this assumption is often visible when survival curves cross
or are not parallel. When that happens, it suggests that the risk of death (or
the event of interest) is not constant over time. As another example, in a trial
comparing surgical intervention, there is a high risk of death initially after
surgery (see earlier Figure 4.5), which later falls as the patient recovers. This
is an example of non-constant hazards or changing hazards.

4.4.10 Mean Survival and Restricted Mean


In situations where there exist non PH, as noted from Figure 4.4, the median
difference may not be reliable and the HR too might be misleading or at
least need some special adjustment using more advanced statistical tests (e.g.
adjusted log-rank or Renyi statistics (Li et al., 2015)) that weight the treat-
ment benefit across the survival experience in different ways. An alterna-
tive approach suggested is to use the mean survival difference – termed the
restricted mean (Royston, 2011).
In economic evaluation, the mean is the choice of statistic despite the median
survival time often being cited as a measure of effect size. The mean survival
time is computed as the area under the survival curve. Mathematically this
is computed as an integral:



µˆ = Sˆ (t)dt. With no censoring, µˆ = t . (4.2)
0
Introductory Statistical Methods for Economic Evaluation in Cancer 107

Equation (4.2) is the area under the survival curve between the time 0 to
infinity. In practice, the survival times are restricted to some time point t
(since no one lives for infinity, at least not in this world). The raw mean and
the mean from the KM curve in the absence of censoring are equal.
The method of estimation of the mean is usually carried out using an
approximation rather than equation (4.2). Equation (4.2) could only be used
if knew the (true) equation of the survival curve (and we could actually use
that equation to model survival times). A KM-curve has no equation: it is
empirical and determined from observed data. In Section 4.6.2, where para-
metric survival curves are used, equations like those above may be useful.
When computing the mean survival time from a KM plot, some computer
packages issue a warning that if the largest survival time is censored, the
mean survival time and its precision (measured by the standard error) may be
underestimated. For this reason the mean is sometimes called the restricted
mean. The restricted mean cut-off (restricted by either using the maximum
overall survival time at which an event occurs, or using the maximum over-
all survival time regardless of an event, or some other cut-off point) could
result in different values of the restricted mean. There are several reasons
why the restricted mean might be preferable over a median estimate.

(a) It covers the whole survival curve (up to the restriction point T*).
(b) When used in cost-effectiveness analysis it is coherent with the use
of the difference in mean cost in the numerator of the ICER and the
definition of the effects.
(c) It is possible for the KM curves to cross at the median (no difference
in median survival) or near to where the median difference is small,
but the area under the curves (mean survival) might show a larger
difference (Dehbi, Royston, & Hackshaw, 2017).
(d) When the follow-up time is short, or a large fraction of observations
are censored earlier on, a large part of the (potential) survival curve
might be uncertain. The restricted mean may not be appropriate in
the presence of heavy censoring (Saad et al., 2018).

As an example, if the survival times were observed as in Table 4.2, the lon-
gest survival time is 9 months, where the patient was still alive. A restricted
mean could be based on using this value. If however, it was based on the
largest survival time with an event (i.e. 8 months), it would give a different
restricted mean (a smaller one in this example). In the latter case (8 months)
the mean survival time and corresponding standard error would be under-
estimated because the largest event time was censored.
As survival times are usually right-skewed the mean survival will gener-
ally be larger than the median. It is also more sensitive to outliers. An impor-
tant question is what restriction time point should be used. Several suitable
methods to define the restriction time (T*) have been proposed (see for
108 Economic Evaluation of Cancer Drugs

example Miller, 1981; Klein & Gerster (2008); Klein & MoeschBerger (2005).
The methods proposed in deriving the restricted mean suggest:

• If the last observation is censored, use this to compute the restricted


mean.

TABLE 4.3
Summary of Statistical Measures and Their Relevance for Economic Evaluation
Relevance to Economic
Measure When/How Used Evaluation
Median From the Kaplan-Meir or survival curve Limited, expect for
Describing clinical effects contextualizing clinical effect
Mean Area under the KM curve Used directly in the estimation of
Useful when non-PH as a clinical effectiveness
measure, but used in the ICER calculus
regardless of PH assumption
Hazard rate Used for describing the nature of event Useful for justifying choice of
patterns survival model for extrapolation
May also be used for deriving
transition probability
Hazard Describes the clinical effect Not used directly in the ICER,
ratio but may be used in modeling
survival data and transition
probability
Transition Describes the chance of patients moving Used in Markov modeling
probability from one health state to another
Restricted Area under the KM curve using a Used directly in the estimation of
mean specified cut-off survival time effectiveness
Useful when non-PH as a clinical
measure, but used in the ICER calculus
regardless of PH assumption
Log rank A statistical test to determine if the Not directly used except to
test survival curves are statistically different contextualize treatment
differences
p-Value Used as evidence for a decision to reject Not used for ICER or
the (null) hypothesis – often that the decision-making
experimental and control treatments do
not differ in terms of an outcome
Confidence Describes the plausible range of values Not used for ICER or decision-
interval for which the true difference (e.g. HR, making, but may be used to
mean difference) lies with some summarize results
specified degree of confidence (e.g. 95%).
Correlation Describes the relationship between Important to report and
several variables – e.g. costs and effects understand so that simulation
can use the correct correlation
matrix for sensitivity analyses
Quantiles Measures used to describe proportion of Not used for ICER or decision-
patients with the event of interest making, but may be used to
(usually, 25%, 50%, 75%) summarize results
Introductory Statistical Methods for Economic Evaluation in Cancer 109

• When there are two curves (two treatment groups), define T* as the
minimum of the largest overall survival times (with an event) in the
different trial arms or groups.
• Use the redistribution to the right (i.e. tail correction) algorithm for
censored distributions.

Other suggestions are of a more theoretical nature and have not been widely
used in practice like those of Andersen et al. (2004) or Susarla and Van Rizyn
(1980)
A summary of important statistical measures and their relevance for eco-
nomic evaluation are shown in Table 4.3.

4.5 Simulation: Bootstrapping and Monte-Carlo Simulation


An essential part of economic evaluation is using simulation to quantify the
uncertainty around cost-effectiveness decisions. The concept of a represen-
tative sample revolves around the idea that the data observed in a trial or
study is a representative sample from a population. Assume the data sup-
ported a conclusion of cost-effectiveness (ICER < £20,000 per QALY). This
conclusion is a single realization from numerous possibilities. If the trial
were to be conducted repeatedly with a similar (not necessarily identical)
set of patients, would it result with the same conclusion? If it were repeated
10,000 times how many of those conclusions would be consistent with our
observed finding of cost-effectiveness. If in 80% of these 10,000 simulations
we have the same conclusion as the observed data we might feel our decision
is more ‘credible’ or we might feel more ‘confident’ about it.
Usually, in an economic evaluation, for each patient, there are several
types of outcomes used in a cost-effectiveness analysis: costs, utilities, PFS
and OS, which are often correlated with each other and with costs). When
patient-level data are available, we can simulate individual patient-level
data by randomly selecting patients from the observed sample. When only
summary data are available and there are no patient-level data, we need to
make assumptions in order to generate data from a (probability) distribu-
tion. An example of simulation will be demonstrated using two well-known
approaches: bootstrapping and Monte-Carlo methods.

Example 4.3: Simulating Using Bootstrapping


Table 4.4 shows ten patients with cost and effectiveness measures, for
each of two treatment groups. The example is fictitious to demonstrate
the concept.
110 Economic Evaluation of Cancer Drugs

TABLE 4.4
Observed Data for Bootstrapping
Patient Treatment PFS OS QALY Drug Cost Toxicity Cost
1 A 3 5 0.8 2,000 1,300
2 A 5 7 0.7 2,500 1,000
3 A 6 9 0.8 2,800   800
4 A 2 4 0.6 2,600   700
5 A 7 8 0.5 2,400   550
6 B 4 5 1.2 4,000   230
7 B 6 8 0.9 3,800   220
8 B 7 9 1.1 4,120   420
9 B 4 8 0.8 3,800   330
10 B 3 7 0.9 3,200   190

TABLE 4.5
A Bootstrap Sample of Size 5 from Observed in Table 4.5
Drug Toxicity
Observation Patient Treatment PFS OS QALY Cost (£) Cost (£)
1 1 A 3 5 0.8 2,000 1,300
2 1 A 3 5 0.8 2,000 1,300
3 2 A 5 7 0.7 2,500 1,000
4 4 A 2 4 0.6 2,600   700
5 4 A 2 4 0.6 2,600   700
6 6 B 4 5 1.2 4,000   230
7 8 B 7 9 1.1 4,120   420
8 8 B 7 9 1.1 4,120   420
9 9 B 4 8 0.8 3,800   330
10 10 B 3 7 0.9 3,200   190

A bootstrap sample of ten patients (five in each group) is performed


separately for each treatment group. This involves taking a random
patient with replacement (as each patient is representative of the popula-
tion). Hence, in a bootstrap sample of the same original sample size, the
entire (row) set of data is sampled for that patient, ensuring that the cor-
relation between costs and effects remains intact.
Table 4.5 shows one bootstrap sample of size ten (five per group). Due
to the small sample size here, some patients have not been resampled.
In 10,000 bootstrap samples of a larger sample size (e.g. a sample size of
n=150 instead of n=10), we would expect a more representative sample
(if n=10, it is possible we could resample the same patients 50% of the
time). The data from each bootstrap sample is then used to construct
the mean (10,000 incremental means costs and QALYs). It is important
to note that the bootstrap is conditional on the sample. If the observed
sample is biased or of poor quality, the bootstrap sample is also likely to
share these characteristics.
Introductory Statistical Methods for Economic Evaluation in Cancer 111

4.5.1 Simulating Using Monte-Carlo Sampling


This type of simulation can be used when we need to use summary sta-
tistics to simulate the observed mean, or variance, or other parameter of
some observed data (patient-level data may not be available). This requires
assumptions to be made about the statistical distribution for each outcome.
For example, if the data were assumed to be normally distributed with a
mean of 3.4 and a standard deviation (STDev) of 1.4, we could simulate a
data set (or several data sets) such that when we compute the mean and
STDev of the simulated data set, it is close to the observed mean and vari-
ance values.
For example in Table 4.4, OS and PFS might be exponentially distributed
(see Figure 4.3), costs might be gamma distributed (due to skewed data), and
ensuring that the simulation holds, the correlation structure can be very
challenging. One difficulty encountered with Monte-Carlo simulation is that
sometime a survival (OS) time can be generated that is shorter than the PFS
(at the very least) such that OS < PFS; hence care needs to be taken with these
methods. Simulation of OS and PFS to generate PPS is commonly employed
to generate a distribution of the mean survival times for to derive the QALY.
Khan (2015) provides several examples of Monte-Carlo simulations of cor-
related data from any distribution.

4.6 Analyzing Data from Cancer Trials


The KM method does not involve modeling survival times. Other estimates
of survival rates are based on the methods such as the ‘life table’ approach
or the so-called Nelson-Aalen estimates. The KM is a descriptive method for
presentation of survival rates and has limited application when it comes to
describing the treatment effect over the entire overall survival curve.

4.6.1 Semi-Parametric Methods: The Cox PH Model


The Cox proportional hazards model (Cox PH) is a statistical method for
comparing survival times (events) between two or more groups while allow-
ing for other factors or covariates (e.g. age, gender). If we assume that there
are two treatment groups and we wish to determine whether treatment is
effective, the hazard rate for patients can be written as:

hi ( t ) = h0 ( t ) exp ( bXi ) ,

where Xi is an indicator variable that takes the value of 1 if the patient takes
the experimental treatment and 0 if they take control and h0(t) is the baseline
112 Economic Evaluation of Cancer Drugs

hazard (which can take any form). The baseline hazard corresponds to the
chance of an event (e.g. death) when all the explanatory variables assume a
value of 0. The baseline hazard function can be thought of as similar to the
intercept in linear regression. When the covariate assumes a value of 0 (e.g. if
the control arm was given a value of 0), the baseline rate could be interpreted
as the risk per unit time of death for an individual who does not take the new
treatment (i.e. takes the control). The Cox model differs from other models
because the covariates are used to predict the hazard function, and not a sur-
vival time (or failure time). Using the model is reasonably straightforward
with a computer program and the interpretation of treatment effects is also
straightforward as shown in Example 4.4.

4.6.1.1 Adjusting for Covariates with the Cox Model


The Cox PH model can be used to adjust for covariates when estimating the
HR. For example, where survival differences exist between treatments for
males and females (e.g. females live longer), an adjusted measures of treat-
ment effect (the HR) can be computed. An HR of 0.70 for treatment A versus
treatment B means that patients, on average, are 30% less likely to have the
event of interest (e.g. death) on treatment A compared to treatment B. An
analysis from a Cox PH model will not give estimates of median or mean
survival times. Cox PH models are used because, like KM methods, one
does not need to make assumptions (other than the PH assumption) about
survival times. Despite the widespread use of the Cox PH model, there are
nevertheless limitations. In practice the Cox PH model is used for estimat-
ing hazard ratios (or adjusted hazard ratios) and little else (Table 4.6).

Example 4.4: Interpreting Results from a Cox PH Model


Table 4.4 shows that prior to adjusting for treatment, the HR was 0.63
and statistically significant (p < 0.001). After adjusting for age, gender,
and ECOG, of which only ECOG appeared to be a statistical predictor of
survival, the risk of mortality on treatment A was now 27% lower (previ-
ously, ignoring ECOG, it was 37% lower).

TABLE 4.6
Example of Interpreting Adjusted Hazard Ratios
Effect HR 95% CI p-Value
Treatment (A versus B) 0.63 0.34, 0.89 <0.001
Treatment (A versus B) 0.73 0.51, 0.91 <0.001
Age 0.313
Gender 0.105
ECOG 0.002
Introductory Statistical Methods for Economic Evaluation in Cancer 113

4.6.1.2 
Using Hazard Ratios to Predict Survival Rates
Sometimes it is pertinent to use the HR to compute the percentage increase
in survival rates. An interesting example in NICE (TA189, 2010, sorafenib) for
hepatocellular carcinoma, is discussed. In the comparison of sorafenib, the
manufacturer stated:

The percentage increase in survival was calculated using the Hazard Ratio,
which takes into account the whole K-M survival curve by averaging the
treatment effect across the curves. Formula: HR = hazard of sorafenib/
hazard of placebo. Thus the relative improvement of sorafenib = 1/HR,
i.e. 1/0.6931 =1.44 (i.e. prolongation in survival by 44%). (Note: Under the
assumption of exponential survival distribution, the ratio of hazards is
the inverse of that of the medians. Comparing the medians directly is con-
sidered the most intuitive, but less reliable since it only takes one point of
the K-M curve).

The ERG commented on this:

The use of hazard ratio (HR) to calculate a % increase in survival time


is potentially misleading if the assumption of exponential survival dis-
tribution is not supported (see Spruance et al., 2004). HR informs on the
likelihood that a random patient from one group will reach an endpoint
before a patient selected randomly from the comparator group. When
the exponential assumption is not supported HR may inflate (or deflate)
the apparent survival benefit.
The ERG extracted individual patient data for the placebo group and
tested the exponential assumption. On the basis of this analysis the ERG
consider that the assumption is not supported and that a 44% increase
in survival benefit probably inflates the apparent benefit. A more reliable
indicator in this case is the % increase in median survival, which for
overall survival is 34.6%.

In Example 4.1, the HR was 0.69 (Figure 4.6). The relative improvement
was computed as 1/HR = 1/0.69 = 1.44 (a 44% improvement in OS). However,
before reporting this, it would have been prudent to check that the survival
data fitted an exponential model. This means checking the model for both
groups. In fact the data did not support an exponential model fit and the
conclusion was that the improvement in OS of 44% was an overestimate. The
median OS was 46.3 versus 34.4 weeks (a difference of 11.9 weeks). This cor-
responds to a 35% improvement. When the exponential fit cannot be satis-
fied, the improvement in OS (across the entire OS curve) can be determined
from the median values, or restricted mean, or from a parametric survival
model.
Spruance (2004) makes the point that the hazard ratio can be misleading
if used to assess the magnitude of treatment benefit. In some cases large
114 Economic Evaluation of Cancer Drugs

treatment effects (small HRs) can be reported along with small median dif-
ferences (KM curves cross near to the median but diverge significantly over
time); in other cases small treatment effects (larger HRs < 1) can result in
large median differences. One reason for this is due to the shape of the sur-
vival curves combined with non-constant hazards. A more reasonable esti-
mate of the treatment effect size when these situations arise is likely to lie
somewhere between these two extremes. The median could be a conserva-
tive assessment of OS improvement on the one hand, whereas the HR may
be overly optimistic. This leads us now to the issue of parametric survival
models.

4.6.2 Parametric Methods: Modeling Survival Data for Extrapolation


The Cox PH model is a useful model for estimating treatment effects in terms
of HRs as well as adjusting the effects for covariates. However, a key part of
economic evaluation lies in extrapolating survival rates beyond trial follow-
up. Prediction of survival rates beyond the last overall survival time point
(extrapolation) allows health benefits (and associated costs) to be computed
over the entire life time of patients for economic evaluation. Several para-
metric models are now discussed that may be used for predicting survival
rates. Details are also provided in the NICE TSD on survival models (Woods,
2017); Latimer, 2011).

(i) The exponential model

The exponential distribution is a simple parametric model with one param-


eter λ. It therefore assumes a constant hazard with a survival function;

S ( t ) = exp ( -l * t )

This is a PH model and treatment effects are estimated in terms of HRs,


interpreted in a similar to the Cox PH model. The model is easy to fit and
interpret.

(ii) The Weibull Model

The Cox PH model is often used when the probability distribution of the
sampled survival times is unknown, or it might be complicated to fit a model
to the data. Since we wish to predict survival rates at specific time points, the
survival function using a Weibull model can be used:

( )
S ( t ) = exp -lt a (4.3)
Introductory Statistical Methods for Economic Evaluation in Cancer 115

where t is the survival time. Equation (4.3) has two parameters, λ and α, which
need to be estimated (here λ is the hazard rate or scale parameter) and 03B1 is
the shape parameter. These are used to adjust the shape of the survival curve
and provide predictions of survival times at time t, once the shape and scale
parameters are estimated. Note that when α = 1, this becomes an exponential
model.
In practice one might fit this curve to an observed set of survival data,
estimate the parameters λ and α, then use equation (4.3) to predict future
survival data. The Weibull model is frequently used (sometimes inappropri-
ately) in HTAs. The reasons for its popularity is because it is relatively simple
to fit and it is also a PH model (which is helpful for interpreting coefficients
as treatment effects as it is similar to the Cox PH model).
The Weibull model has greater flexibility than the exponential model,
which assumes a constant hazard. Here the hazard may increase or
decrease, but cannot change direction. Where α = 1, the Weibull is the
same as an exponential. For α > 1 the hazard function increases and for α
< 1 the hazard function decreases monotonically. In order to fit a Weibull
distribution, it is important to check the nature of the hazard function
statistically and whether it has a sound clinical interpretation. Figure 4.8
also shows how the survival curve takes on different shapes for varying
values of α. Example 4.5 shows how an exponential and Weibull function
are used.

FIGURE 4.8
Changing shape of the survivor function for the Weibull model.
116 Economic Evaluation of Cancer Drugs

Example 4.5: Fitting an Exponential and Weibull Function to


Estimate Survival Rates: Cost-Effectiveness of Lenalidomide
BACKGROUND
The data for this analysis was kindly provided by Reck et al. (2014).
Despite lenalidomide and dexamethasone being highly effective, its
use in multiple myeloma (MM) is restricted due to a lack of evidence
of cost-effectiveness. In this example we performed a cost-effectiveness
analysis by using an alternate day dosing strategy where lenalidomide
commenced at 25 mg daily; Due to hematological toxicity, instead of
giving 15 mg daily, 25 mg was given every two days (alternate days).
Subsequent reductions of 5 mg were made due to potentially further tox-
icity (i.e. 15 mg then 10 mg on alternate days, instead of 10 mg then 5 mg
daily). The objective was therefore to compare the cost-effectiveness of
lenalidomide and dexamethasone alternate dosing (L+DAlternate) versus
lenalidomide plus dexamethasone standard (L+DStandard) dosing based
on the summary of product characteristics.

DESIGN, PATIENTS AND TREATMENT SCHEDULE


The data for this example comes from a real-world setting. It was a ret-
rospective review of patients with relapsed multiple myeloma treated
with lenalidomide and dexamethasone in a single UK center (University
College London Hospitals Foundation Trust (UCLH)). The main efficacy
endpoints were time-to-progression (TTP), progression-free survival
(PFS), and overall survival (OS).
Real-world patient-level data were available for a single cohort of
L+DAlternate patients (N = 39) who were given an alternate dosing regi-
men. Summarized data for lenalidomide alone, dexamethasone alone
and L+DSPC were taken from published data.
First a Kaplan-Meir curve was generated for TTP for L+DAlternate and
then a Weibull parametric function was used to predict survival proba-
bilities beyond 21 months. Figure 4.9 shows the KM curve with a Weibull
function for L+DAlternate fitted over the first 55 months. The value of the
scale and shape parameters were 0.817 and 0.0276 respectively.
For the comparison of L+DAlternate with dexamethasone, since no patient-
level data were available for dexamethasone alone, this data had to be
simulated using published statistics. The mean TTP for dexamethasone
alone was simulated using published data that compared L+DSPC versus
dexamethasone using a reported hazard ratio of 0.427. It was assumed
that the TTP followed an exponential model (see Table 4.7).
In Table 4.7, the mean OSs for L+DSPC (4.0 years) and dexamethasone
(2.3 years) were determined by simulating survival times from published
data. Recall, it is the mean survival times needed for cost-effectiveness
analyses. Mean survival times were not reported (only medians were
reported) and so had to be simulated. The L+DAlternate group was used
for the real-world data for which limited follow-up data were available.
Hence, TTP was modeled to compute the mean TTP for the L+DAlternate
group. In short, for each group, we required a mean OS, a mean TTP, and
a mean PPS, from which we could construct the QALY. The available
Introductory Statistical Methods for Economic Evaluation in Cancer 117

FIGURE 4.9
Estimated shape of the TTP L+dexamethasone and dexamethasone alone survivor function
estimated from a Weibull model.

TABLE 4.7
Summary of Estimates Used for Simulation
Dexamethasone
Parameter L+DSPC alone L+DAlternate
Efficacy Median OS 1 2.8 years 1.6 years 2.8 years
Mean OS2 4.0 years 2.3 years 4.0 years
Median TTP 0.87 years1 0.4 years 0.98 years3
Mean TTP 1.7 0.65 1.7 years4
Mean PPS 2.3 years 1.7 years 2.3 years
1 Notes: From ERG report (NICE, TA171, 2008; Stadtmauer, 2006).
2 Modeled assuming an exponential distribution (mean OS was not reported so had to be
simulated).
3 Observed in the data (also reported in the published studies).
4 Using a Weibull function for the observed data and assumed to be the same for the standard
regimen. The observed TTP was modeled using a Weibull model with a scale parameter of
0.817 and a shape parameter of 0.0276. The log likelihood test for model fit suggested the
model fit was appropriate.
5 Estimated using TTP/log(2) , assuming dexamethasone TTP is exponential (page 88, ERG
report, NICE TA171).
118 Economic Evaluation of Cancer Drugs

data (often medians) were used to construct (simulate) these values as


shown in Table 4.7.
From the above we were able to generate the effectiveness inputs for
both L+DAlternate and dexamethasone alone. Using the observed OS data
for L+DAlternate, assuming an exponential model to fit the observed KM
curve, we can extrapolate beyond the last time point to the maximum
time an ‘average’ human being could theoretically live – set to 100 years.
Consequently, the mean OS for L+DAlternate was 4 years. In order to esti-
mate the mean OS for dexamethasone alone, we used the fact that OS =
TTP/log(2) (also used by the evidence review group, NICE TA171, 2008).
For the dexamethasone group, therefore, the mean TTP was estimated to
be 2.3 years. Similar data for the L+DSPC was already reported (Table 4.7).
A strong and somewhat unverifiable assumption was that mean sur-
vival times between the alternative dosing arm versus the dexametha-
sone arm would yield similar effects to L+DSPC versus dexamethasone.
This could have been investigated using a well-controlled RCT; however,
it may not be in a manufacturer’s interest to conduct such a trial.
(iii) Other Models
The NICE technical support document (TSD) (Latimer, 2011) suggests
several other survival function models that can be used, including the
Gompertz, generalized gamma, log-normal, log-logistic models. Details
of how to fit these models can be found in several references (Collet, 2014;
Crowther, 2016; Klein, 2005). A further group of models called piecewise
and flexible parametric models are now discussed. For now, we limit our
discussion on the statistical aspects of these models.

4.6.3 Advanced Modeling Techniques for Survival Data


4.6.3.1 Flexible Parametric Survival Models
Standard parametric models (e.g. Weibull) can be used to model survival data,
but not all will retain the useful assumption of PH or fit the overall survival
pattern well. Flexible parametric modeling methodology (Beck & Jackman,
1998; Royston & Parmar, 2002) is based on using ‘flexible’ polynomial func-
tions that are fitted piecewise. This consists of several functions that are joined
together at ‘knots’ (which act as constraints) so that the overall fitted function
is smooth. The idea can be compared with linear spline fitting (basically join-
ing lines), but instead of linear splines (which are polynomial functions in the
first degree), ‘curves’ are joined together to fit the observed data.
The approach to flexible parametric modeling involves joining pairs of data
points using higher degrees of polynomial functions (e.g. quadratic for order
two and cubic for order three, or fractional polynomials). One useful property
of using splines is that the underlying functional form does not need to be
specified (Kruger, 2004). Splines can be used in the context of a Cox PH model
to smooth the hazard function (Sleeper & Harrington, 1990), however in this
section we present the use of splines for fully parametric models.
Although the Weibull model is a useful alternative to the Cox PH model,
especially when the assumptions around distributions are reasonable, a
Introductory Statistical Methods for Economic Evaluation in Cancer 119

more flexible approach (Royston & Lambert, 2011) directly modeling the
baseline hazard function h0(t) as a polynomial function has shown to be a
versatile approach to fitting smooth survival functions.

4.6.3.2 Applications in Cancer Surveillance


Cancer registries increasingly collect important prognostic factors of survival
useful for oncologists, policymakers, and others in order to make decisions
on expected survival rates for subgroups of patients. Calculating patient-
and strata-specific survival rates, although possible, is not straightforward
using Cox PH modeling. The main reason for using the Cox model is the PH
assumption for which the baseline hazard function is eliminated (through
special estimation methods called partial likelihoods). For a Cox model, the
important aspect is the hazard ratio and not the hazard function. The base-
line hazard can be thought of as nuisance parameter, which is eliminated.
For example, calculating the survival probability for an individual (or group
of individuals) who are female, aged 50 (on average), with Stage III breast can-
cer is a particularly useful statistic for policymakers and planners, especially
for future costing of cancer treatments. Latimer (2011) and Bagust and Beale
(2014) discuss approaches to extrapolating data in survival models, including
a selection process as a guide for modeling. Several issues merit consideration.
First, it is recognized that reimbursement decisions depend on having reli-
able evidence with respect to costs and effects accrued beyond what is often
presented, because trial follow-up is curtailed for practical purposes. Second,
methods for selecting or checking models/model fit and assumptions (such
as PH) should be assessed using appropriate plots and information criterion
methods (note model fitting is somewhat determined by the observed data).
Third, there is debate as to whether standard parametric functions should be
considered as the first choice (e.g. Weibull, log-normal), although it is agreed
that the data should determine the most appropriate method through a
systematic approach to modeling. In addition, a pre-specified cut-off point
should be identified for when the extrapolation should start. Often this is the
last observed time point or is dependent on the date of data cut-off, which in
itself is dictated by a regulatory constraint that requires a definition in the
protocol as to when the ‘end of trial’ occurs. Lastly, consideration should be
given to the physiological process. It is possible to fit a mathematical function
to survival data that shows an excellent fit, but the estimates of the param-
eters may not have sound clinical relevance.

Example 4.6: Modeling Survival Data Using a Flexible


Parametric Model in a Lung Cancer Trial
BACKGROUND
The data from Lee (2012) were used to fit a flexible parametric model.
Nearly all (98%) of the patients died at the time of analysis (658 deaths).
The objective was to fit a parametric survival curve to each treatment
120 Economic Evaluation of Cancer Drugs

FIGURE 4.10
(a) OS Erlonitib (b) OS Placebo of predicted survival rates from Weibull applied to data from
the TOPICAL trial.

group to predict the survival pattern and mean survival time. More
details are in Dewar and Khan (2015). Although in this example the
practical benefits of extrapolation are negligible, the exercise shows that
a flexible parametric model can show a better fit than standard models.
The Royston-Parmar (RP) model was fitted using SAS statistical soft-
ware. In this analysis the OS was used as the time to event variable. The
separate curves were joined using 1 ‘knot’ (or ‘2 separate curves’), hence
it was called an RP(1) model.

RESULTS
The empirical survival curve (Figures 4.10(a) and 4.10(b)) is also plotted
along with the Weibull model. The Kaplan-Meier (solid line) is approxi-
mated well by the RP (dotted line) with three knots. The Weibull (dashed
line) is a slightly worse fit (Table 4.8).
In this example, the mean survival times for erlotinib versus placebo
were 6.95 versus 6.53, 6.96 versus 6.47, and 7.05 versus 6.62 months for

TABLE 4.8
Estimates of Coefficients and Hazard Ratios from STATA and SAS
Cox PH Weibull* a RP(1)
HR(SE) 0.95 (0.08) 0.93 (0.09) 0.95 (0.08)
Lower 95% 0.82 0.78 0.82
Upper 95% 1.11 1.11 1.11
Predicted 6 month survivalb 40.1 vs. 38.4 34.1 vs. 32.4
AIC 7340 3818 2165
a Notes: RP(1): Flexible parametric using 1 knot.
b Observed 6 month survival rates for erlotinib vs. placebo were 39.1% vs. 36.6%.
* Using PROC LIFEREG in SAS.
Introductory Statistical Methods for Economic Evaluation in Cancer 121

the Kaplan-Meier, RP(1), and Weibull models respectively. The Weibull


model might therefore overestimate mean survival time and hence
QALYs (assuming quality of life is the same between treatment groups).

Example 4.7: Modeling Survival Data Using a Flexible


Parametric Model in the Presence of Censoring
In this trial, two treatments A and B were compared in terms of OS.
Patients were followed up in the trial for 12 months. Long-term cost-
effectiveness was required over a 5-year time horizon by modeling and
extrapolating survival time between randomization and death, after
examining the overall survival curves (Figure 4.7).
The follow-up period was only 12 months, and there were many
patients who were still alive by the end of 12 months. A flexible para-
metric model using cubic spline (Royston & Parmar, 2002) with a RP(3)
(3-knot) model was used. Extrapolation beyond 5 years was consid-
ered to be highly speculative and uncertain because most death events
occurred between randomization and end of follow-up. The fitted
Royston-Parmar model was used to predict the survival rates at each
time point (Figure 4.8). The survival rates at each of 3 monthly intervals
(3, 6, 9, 12 … 60 months) were used to determine the expected costs and
derive the QALYs (at an aggregate level).
After fitting, a Royston-Parmar 3-knot model (the best model based
on lowest AIC) using a 5-year time horizon yielded a mean survival
time of about 41.9 versus 33.3 months for A versus B, respectively.
From this, the mean overall survival (OS) for A versus B was 4.12 ver-
sus 4.07 months based on the observed KM estimates from the within-
trial data over 12 months. At 5 years (60 months) post-randomization,
67% of patients were expected to remain alive in group A compared
to 45% in group B (Figure 4.8a: KM survival plot). These survival rates
are lower than the national average as reported in mortality statistics
for the general population with a similar age distribution to this popu-
lation. These would be 95% at 5 years from a mean age of 63 years,
which logically are higher than observed here as these are patients
with cancer.
Plots of the log survival (Figure 4.11b and the hazard rate (Figure 4.11c)
suggest that A has a trend of decreasing hazards (lower risk of death
over time) while B appears to have an increasing hazard (higher risk of
death over time). This is mainly due to the spike in hazards after month
6 because of a later death event in group B. Both hazard functions sug-
gest risk of death is decreasing with a similar trend until 4 months, albeit
with a more erratic hazard for the group B.
After about 4 months, patients may die at a slightly faster rate (ignor-
ing the spike) in the group B. Due to a lack of further follow-up data,
future death patterns may or may not be similar, although the curve
trajectory for group B suggests the hazard curve may lie slightly above
that of group B. These plots partially support the extrapolated survival
curves, suggesting death rates in group A occur more slowly compared
with patients in group B, but survival rates over time may converge.
122 Economic Evaluation of Cancer Drugs

FIGURE 4.11
Observed and extrapolated OS over 5 years using RP (3) log survival (bottom left) and the
hazard function (bottom right).

4.7 Issues in Fitting Models


When a parametric model is fitted to survival data, two broad approaches
may be taken. One is to fit a single parametric model to the entire data (assum-
ing PH). The second is to fit an individual or piecewise parametric model
separately for each treatment group to each treatment arm. If an HR is used
(assume two groups) to fit a model for the entire data set, it should ideally be
fitted under the PH assumption – that is, the treatment effect is proportional
over time and the survival curves fitted to each treatment group have a simi-
lar shape. Proportional hazards models such as the exponential, Gompertz,
or Weibull could also be used. However, some models (e.g. log-logistic and
some accelerated failure time models) will not necessarily produce a single
hazard ratio (HR). With such models, the PH assumption does not hold.
Using separate parametric models fitted to each treatment arm requires
fewer assumptions (but requires the estimation of more parameters). Fitting
parametric models separately to each treatment arm is possible (e.g. a Weibull
for one treatment arm and a log-normal for the other) but will require strong
Introductory Statistical Methods for Economic Evaluation in Cancer 123

justification because different models allow very different shaped hazard


distributions. If the PH assumption does not hold, it may be reasonable to fit
separate parametric models. Where a PH model is used, justification will be
needed. Further details can be found in the NICE TSD report (Latimer, 2011).
In addition to the Weibull, other models such as the Gompertz model can
also be used. The approach to determining which model is the most useful
fit can be determined by the smallest value of the AIC. In general, where the
survival rates are to be estimated, an accelerated failure time (AFT) model
form would be most useful. In general, the PH models the hazard rate,
whereas the AFT form of the models is used for modeling the survival rates
over time. Klein and Moeschberger (2005) present a table of hazard functions
and survival functions for each model.

4.8 Handling Crossover, Treatment Switching,


and Subsequent Anti-Cancer Therapy
4.8.1 Introduction to Treatment Switching
Sometimes randomized clinical trials in oncology include the possibility for
patients in the standard care or comparator arm (i.e. the control arm) to be
able to switch to the experimental drug. This is due to ethical concerns that
when equipoise is lost, patients should not be forced to remain in a sup-
posedly lower-efficacy treatment. Earlier (in Chapter 1) we introduced the
issue of crossover (or treatment switching) in cancer trials, which can make
differences between treatment arms appear larger (or smaller) and there-
fore bias the estimate of the QALY. Jonsson et al. (2014) and Latimer et al.
(2014) provide an overview of the issues and implications of crossover for
economic evaluation. Crossover allows patients to experience the treatment
they did not get because the current treatment a patient has received did
not yield the desired effect (efficacy or safety). Crossover often occurs at the
point of unblinding (at the patient level) and results in a loss of information.
In a (double-blind) trial, at the point of progression (switching over), it would
not be known whether an experimental treatment is efficacious or not – that
would usually be known after the data analysis is complete or partially com-
plete (through interim analysis).
There are several issues that concern both efficacy and effectiveness relat-
ing to crossover. These are likely to be about the methods available for han-
dling crossover, the assumptions for their use, and how to interpret the
resulting treatment effects. From the efficacy perspective, we consider that
such methods can only be used as a form of sensitivity analysis around the
observed treatment effect. For cost-effectiveness, however, the issue may be
deeper. Whereas efficacy is essentially about an unbiased treatment effect,
124 Economic Evaluation of Cancer Drugs

effectiveness is often about effects in the real world in which crossover is


often unavoidable.
Designing a trial for potential crossover may be impractical or very chal-
lenging (i.e. you don’t always know if patients will switch over, or who
they are, and it is unlikely patients would want to be randomized again
to another ‘choice’ when they have just stopped taking the existing treat-
ment). Moreover, some patients can be treated with experimental treatments
(switch over treatment) by moving to a location where the treatment is avail-
able. For example, stereotactic whole brain radiation therapy offered to lung
cancer patients is only available at some particular hospitals. If some patients
decided they wished to receive this despite being randomized to the alter-
native treatment group, this too would have similar effects as crossing over
– biasing treatment effects.

4.8.2 Types of Switching
Figure 4.12a shows the simplest form of switching. Patients in the experimen-
tal arm are not allowed to switch to the control arm (arrows moving upward
or downward signify direction of switching). Usually switching is allowed
when or some time after progression has been detected. However, in practice
there may be other reasons for switching, such as the occurrence of a severe
adverse event, toxicity, or the treating physician’s decision. These are called
the (switching) trigger events or reasons (see Green Park Collaborative, 2016).
This case is called a unilateral switch as opposed to a bilateral switch where
both switches are allowed. Less usual is a bilateral switching process (Figure
4.12b) whereby patients in the experimental group switch to the control as
well as vice versa. Switching starts later in the experimental arm. This would
be the case if switching was allowed upon progression in the control arm
and progression is delayed in the experimental arm.
Other more complex switching patterns (Figure 4.9c) can be observed if
switching to other (e.g. second-line treatments) are allowed (including pal-
liative care in metastatic disease). In this case, both patients who received
experimental and control treatments, and to different 3rd line drugs H
or F. Figure 4.9d shows that both groups switch to treatment A, but some
or all control patients might switch to the experimental treatment first. It
is important to note that the assumption that patients switching from con-
trol to experimental treatment will not be subject to harm or deterioration
will be very strong but only hypothetical. Where efficacy is more impor-
tant than effectiveness, an OS free from confounding factors will be more
important.

4.8.3 Implications of Switching
The statistical issues around treatment switching also have similar implica-
tions for situations where patients take other concomitant medication, stop
Introductory Statistical Methods for Economic Evaluation in Cancer 125

FIGURE 4.12
Graphical demonstration of the types of switching/crossover: (a) Switching from control
to experimental; (b) bilateral switching; (c) patients in both groups switch to a different
treatment; (d) multiple switching patients on control switch to experimental (A), and then
subsequently take another treatment (F). Patients on experimental (A) take a subsequent
treatment (H).

taking treatment, take cocktails of treatment, and also take dose adjust-
ments (which are commonly captured through dose modifications in the
case report forms). The presence of significant crossover can have serious
implications for QALY estimates due to biased efficacy estimates. Hence, this
problem is not just a health economic evaluation question, but a more general
question on generating unbiased estimates of treatment effect in the pres-
ence of crossover in cancer trials. It is also related to a more recent theme
called ‘estimands,’ which is explained in the addendum to the ICH E9 guide-
line on statistical principles (ICH E9 Addendum, 2017). ‘Estimands’ attempt
to estimate the ‘true’ value of a new treatment by considering multiple sets
of analyses, where each analysis is relevant to a specific stakeholder. Hence,
efficacy and effectiveness will have their own corresponding ‘estimad’ or
measure of treatment effect. This ICH E9 addendum seeks to differentiate
the types of treatment effects that ensue according to the different ways in
which patients are handled for complex issues such as treatment switching
or missing data. The addendum may have been needed to clarify the dif-
ferent and sometimes conflicting requirements from reimbursement and
licensing agencies for determining efficacy and effectiveness under different
scenarios.
126 Economic Evaluation of Cancer Drugs

In general, a definition of a crossover treatment should be identified and


stated up-front. One should also determine whether, for a given reimburse-
ment agency, crossover is likely to be a concern, especially for long-term
treatment effects in the presence of crossover. One should identify and
distinguish between spontaneous versus treatment-related crossover (e.g.
patient decision rather than due to progression). It is always better to design
for switching, and where possible, to avoid crossover. It would also be use-
ful to have an idea of the rate of crossover expected based on experience
with previous trials. In addition, careful testing of assumptions and beliefs
about PPS after switching should be evaluated – this also extends to post-
progression HRQoL.

4.8.4 Handling Switching in Statistical Analyses


There are several approaches to handling crossover suggested in the pub-
lished literature:

(i) ITT analysis


(ii) Per protocol analysis
(iii) Including crossover treatment as a time-varying covariate in a Cox
model
(iv) Inverse probability-of-censoring weighting (IPCW)
(v) Rank-preserving structural failure time models (RPSFTMs)
(vi) Two-stage adjustment model
(vii) Other approaches

We will discuss somewhat approaches (iii) and (iv) in more detail since these
appear to be the currently recommended approaches (Jonsson et al., 2014;
Morden et al., 2011; Ishak, 2014).
No one particular method has been identified as the ‘best’ approach suit-
able for all situations. Interested readers may consult the references for spe-
cific details. However, each method aims to address particular questions
(or estimands). For example, the IPCW estimates the treatment effect as if
switching from control treatment to experimental treatment was absent, but
the estimate still includes the effects of subsequent therapies. Using RPSFT
on the other hand might allow for an estimation of the experimental treat-
ment benefit alone (disentangled from the subsequent treatments), although
in practice the estimate of treatment benefit is a combination of experimental
treatment and subsequent therapy. More complex statistical techniques have
been developed to adjust or correct for the switching problem.
Introductory Statistical Methods for Economic Evaluation in Cancer 127

4.8.4.1 Intent-to-Treat (ITT)
In the case of ITT, since patients are analyzed according the group they were
randomized to, effects measured after switching are considered as belong-
ing to the treatment group being switched from. For example, a patient is
randomized to the control group (C). The group’s median OS time (until pro-
gression) is 6 months but the PFS time was 4 months. Patients in the experi-
mental arm (E) have a median OS time of 8 months and a median PFS time
of 6 months. At 4 months this patient switched treatment to the experimental
arm. It would appear this patient lived for 7 months.
Using an ITT analysis, the patient would have an OS of 7 months attribut-
able to the control. If the median OS is 8 months on E, then the patient has
an OS of 1 month less than on average in E (Figure 4.13). The fact that the
extended survival might be due to post-switch experimental treatment is not
factored in. Consequently, the treatment effect is underestimated, as will be
the QALY. Conversely, if a switch from E to C happened (because E was too
toxic), the ITT analysis may lead to the wrong interpretation. The effects are
not necessarily biased (in the statistical sense), because data are treated as
belonging to a randomized experiment.
In a usual ITT analysis (where patients are grouped according to their initial
randomization therapy), even in the simple case of unilateral control switch-
ing, the overall effect is to dilute the difference in OS between the two arms
(∆E), so that the true drug efficacy is underestimated. In fact, any post-switch
subsequent endpoint is affected (e.g. PPS, OS, HRQoL, adverse effects, or
dropout). This can be important as in some trials up to 80% of the control arm
patients switch to the experimental drug some time after progression. One
should bear in mind that in practice, the transitions happen with some delay
after evidence of progression and switching is not necessarily instantaneous.

Example 4.8: Impact of Switching on Cost-Effectiveness


In a trial with a mean survival of 12 months for the experimental group
E and 6 months for the control group, switchers from C to E gain another

FIGURE 4.13
Relationship between switching and survival; C = censored; E = event.
128 Economic Evaluation of Cancer Drugs

3 months and 50% of the controls switch at progression (they increase


their PPS by 3 months). Had those in C stayed in the control arm, the
difference in mean survival would have been 12 – 6 = 6 months. With
switching, the mean survival of the controls is now equal to (6 x 0.50) +
(9 x 0.50) = 3 + 4.5 = 7.5, or a difference of 12 – 7.5 = 4.5 months in favor of E.
Now imagine also that the average experimental cost for a full treat-
ment is much higher than for the control drug cost, say respectively
€5,000 and €1,000. The same switching process also makes the cost
higher for C; instead of a cost difference of €4,000 without switching we
now face (1,000 x 0.50) + (5,000 x 0.50) = 500 + 2,500 = €3,000 (if all switch-
ers also receive a full treatment with E). These combined effects change
the ICER dramatically in favor of the new drug; ICER no switching =
(€4,000/6) ≈ €667 per life-month gained (LMG) compared to (€2,000/4.5)
= €444 per LMG.

4.8.4.2 Per Protocol Analysis


A per protocol (PP) analysis would censor patients from the date they
switched. This analysis can be subject to selection biases: patients who switch
may well be those with poorer prognosis. This has two main consequences:
first, to diminish the statistical power of the study because their observation
is cut short, reducing the number of observable events (deaths) in the control
arm. Second, unless switching is random, it will most probably introduce a
selection bias as the events (or reasons for it like progression) for switching
are likely to be correlated with the final endpoint (death for OS assessment),
and therefore distort the initial randomization in treatment groups.

4.8.4.3 IPCW
The IPCW approach (Robins & Finkelstein, 2000) involves calculating the
probability of censoring in relation to a set of confounders (e.g. age, perfor-
mance status, stage, etc.) using a logit model. We assume there are no hidden
or unmeasured confounders – an assumption difficult to verify. This is basi-
cally an adaptation of the marginal structural model (MSM) first developed
for observational studies.
The MSM inverse probability-of-censoring weighting (IPCW) model, fre-
quently used in epidemiology to correct for dropouts, can be used also for
correcting for switching. In this approach patients who switch are first cen-
sored at their time of switching. Then the bias introduced by the switching
is corrected by weighting each patient in the control arm by the inverse of
his predicted probability (estimated through logistic regression) of not being
censored. The predicted probability is estimated based on baseline patient
characteristics and further time-varying factors that could influence the
switching. Once these probabilities have been calculated they are used as
weights. This creates a (weighted) pseudo-population where control patients
who did not switch are weighted more heavily. Finally, a survival analysis
Introductory Statistical Methods for Economic Evaluation in Cancer 129

is performed on this pseudo-population, usually with a standard survival


method, such as Cox regression or a parametric model.
The whole method relies on the premise that the probability of switching
is correctly estimated from the observed covariates and that, conditional on
these, switching is independent of the outcome and its timing (the so-called
‘no unmeasured confounding’ assumption). Consequently, the trial must
collect enough information over time on the switching covariates. Trial sam-
ple sizes also have to be relatively large as generally the number of potential
covariates is large, and in smaller trials the logistic regression will not con-
verge or yield very high weights for some specific patients when the number
of non-switchers is low (Jonsson et al., 2014; Latimer et al., 2016).
The method is briefly described below.
For a given data set we identify the date of switching for each patient. If
a date is present a switch is considered to have occurred. The logit model is
then regressed against a set of covariates:

Response = intercept + covariates ( including time varying co


ovariates )

where response is a binary variable with 1 for a censored survival time and
0 if not censored. The approach to modeling is as follows.

Step 1: Determine covariates that may be predictive of switching.


Decide the rule to drop or keep covariate (e.g. if p-value >> 0.05, drop)
Step 2: Model the probability of switching (using a logistic regression)
and for each patient estimate the predicted probability of switching.
For those patients who have not switched calculate the inverse of
these predicted probabilities.
Step 3: For each patient that has not switched adjust the survival times
by the inverse of the probability weights.
Step 4: Fit a survival model to the data using the original randomiza-
tion allocation and censor patients who switch (as in a PP analysis)
on the date of switching.
Step 5: Compute mean survival and QALY.

4.8.4.4 RPFSTM
This method uses the accelerated failure time model (AFT) (see Table 4.9)
form of a survival model with the objective of presenting a measure of treat-
ment effect by adjusting for those who cross over.
The approach of the RPFSTM is to consider the total survival time Ti con-
sisting of two parts (using Jonsson’s 2014 notation): Ti before crossover on the
control arm, referred to as Tion and survival time after switching Tioff :

Ti = Ti on + Ti off [Overall Survival Time] (4.3)


130 Economic Evaluation of Cancer Drugs

TABLE 4.9
Summary of Survival Analysis Methods
Model PH Useful for Extrapolation Statistic AFT
Kaplan-Meier No No Median, Mean, No
Survival rate
Exponential Yes No HR Yes
Weibull Yes Yes HR, Survival rate Yes
Gompertz Yes Yes HR, Survival rate No
Log-normal No Yes HR, Survival rate Yes
Log-logistic No Yes HR, Survival rate Yes
Generalized gamma No Yes HR, Survival rate Yes

In addition we have something called the ‘unobserved’ survival time, Ui – or


the survival time that would have been observed on the control arm after pro-
gression if the patient had not switched plus a measure of effect, ψ (called the
acceleration factor, which is considered to be a causal factor; values of ψ < 0,
suggests a beneficial treatment effect).

U i = Ti off exp ( ψ ) * Ti on [Unobserved Survival Time] (4.4)

If exp(ψ) = 0.7, for example, this is like saying that 1 year taking the study
treatment (control arm) is equivalent to 0.7 years of not taking the treat-
ment. The value of ψ is iterated until a value is found for which a statistical
test (e.g. log-rank) yields the highest p-value. The process of estimation is
carried out through G-estimation – a flexible semi-parametric approach tor
estimating effects of exposure in non-randomized studies (Robins, 1986;
Faries et al., 2010). Assumptions for RPFSTM include assuming a common
treatment effect: i.e. the treatment effect from progression onward (until
death) in the control group is similar to the treatment effect from random-
ization until death in the experimental group. In addition Ui is consid-
ered to be independent of the randomized treatment group and there is
an explicit assumption that switchers and non-switchers are comparable
(or need to be comparable). Further details of this method can be found
in the references in the bibliography (e.g. Li et al., 2015; Faries et al., 2010;
Dukes, 2018).
There are several core assumptions in the model:

(a) The treatment effect is the same regardless of when experimental


treatment is initiated (common treatment effect).
(b) The randomized arms are properly balanced, this implies that the
average survival time in the two groups (control vs. experiment)
would have been equal if none had received the experimental treat-
ment (i.e. Scontrol1 = Scontrol2 = Scontrol).
Introductory Statistical Methods for Economic Evaluation in Cancer 131

(c) Patients have no other choice of being on or off treatment, so if multi-


ple treatment switches are possible, further assumptions about these
‘treatment effects’ are needed also.

The Ui values are used instead of the Ti values for the switching cases in
further survival analysis. So basically, this is a technique that adjusts or
shrinks the post-switching times of the crossover patients. But extra censor-
ing (referred to as ‘re-censoring’) is required to maintain the assumption of
independent random censoring (which results in a loss of information at the
end of the survival curve and thus reduces the precision of the estimates)
(Latimer, White et al., 2018).

4.8.4.5 Two-Stage Adjustment Model


A simplified two-stage method was developed by Latimer et al. (2016) for
situations in cancer where the switching occurs at or close to disease pro-
gression. For this, one must define a secondary baseline time to progression
(instead of randomization time as usual). The post-progression survival is
then estimated with an accelerated failure time (AFT) survival model for
non-switching control patients versus switchers at the time of the secondary
baseline, with prognostic covariates similar to those in the IPCW approach
but measured only at the time of progression.
Finally, the additional treatment effect associated with switching (based
on the estimated AFT factor) is ‘shrunk’ toward the survival times for simi-
lar non-switching control patients. This alleviates the need to collect time-
varying data throughout the trial period but still needs to respect the ‘no
unmeasured confounding’ assumption for the covariates. Compared to the
IPCW method this method does not, however, capture changes in covariates
occurring post-progression.
It is also possible to re-censor the survival times in order to correct for
the fact that when we shrink the survival time for switchers we also shrink
the administrative censoring time for those of who did not die during the
trial, but re-censoring then entails a loss of information. Generally, it is then
advised to present results with and without re-censoring (Walker et al., 2004;
White et al., 1999).

4.8.4.6 Other Approaches: Structural Nested Mean Models (SNNM)


When marginal structural models are not applicable, a structural nested
model (SNM) may be used (Yamaguchi & Ohashi, 2004; Greenland et al.,
2008). This has been proposed by Robins (1994) in the context of causal effect
estimation in the time-varying settings where both the treatment T (the
cause) and a set of (pre-treatment) moderator variables M vary over time
when assessing the effect of T on the outcome Y. In this context the modera-
tors may themselves be dependent or influenced by the outcome of earlier
132 Economic Evaluation of Cancer Drugs

treatment (Almirall, Ten Have, & Murphy, 2010). These have mostly been
studied in the presence of non-compliant, fully sequential treatments but
rarely for switching (Yamaguchi & Ohashi, 2004).
The underlying assumptions used for any analyses involving switching
will be difficult to demonstrate. An analysis that adjusts for crossover/treat-
ment switching will not salvage a trial that has shown to be negative prior
to adjustment. If the ITT population does not show treatment benefit, adjust-
ment is unlikely to be considered as strong evidence of treatment benefit
from the experimental drug. The regulatory questions as to what would
have been the treatment benefit without the presence of crossover cannot be
totally separated from a reimbursement question.
One last consideration for treatment switching relates to how probabilistic
sensitivity analysis (PSA) is conducted in the presence of switching. Where
switching is of concern, the PSA and ICERs should be presented by taking
into account the above methods. Estimates of ICERs and PSA from both the
standard (not taking into account switching) PSA and from those that result
from adjusting for switching should be reported. The issue also extends to
extrapolation of survival curves, since switching may continue well beyond
trial follow-up has completed.

4.9 Data Synthesis and Network Meta-Analyses


This section introduces an important area related to health technology
assessment covering two broad areas: mixed treatment comparisons (MTCs),
sometimes called network meta-analysis (NMA), and evidence synthesis,
which concern pooling together data from several randomized trials (or
other studies) and estimating a type of average treatment effect directly or
indirectly.

4.9.1 Mixed Treatment Comparisons


Randomized controlled trials (RCTs) that have two or more treatments are
an effective yet expensive and lengthy method of comparing treatment
effects. In some cases there is simply not enough time or enough resources
to perform additional trials to compare a new treatment with existing com-
parators. In such situations an alternative method is to carry out an MTC,
which uses existing data in a summarized form. The International Society of
Pharmacoeconomics and Outcomes Research (ISPOR) describes an MTC as
a network in which some of the pairwise comparisons have both direct and
indirect evidence (Hoaglin et al., 2011). This means that some pairwise com-
parisons can be carried out in a given trial (direct comparisons), but using
additional data can help to obtain more precise estimates. For example, in an
Introductory Statistical Methods for Economic Evaluation in Cancer 133

FIGURE 4.14
Diagram representing direct comparison for A versus B and A versus C (solid lines) and the
indirect comparison for B versus C (dashed line).

RCT comparing A versus B and another separate trial comparing A versus


C, the comparison of B versus C can be made indirectly.
There are two pieces of information for treatment A (A vs. B and A vs. C),
and a single piece of information for each of B and C (Figure 4.14). Treatment
B versus C is the indirect comparison.
Pharmaceutical companies may not be able to conduct a direct comparison
of a new treatment against the standard or ‘best’ treatments for practical or
commercial reasons. This may be because there is no standard treatment, or
because it is commercially risky or expensive. Therefore, an MTC can be the
only way to compare treatments for estimating effects that may otherwise
not be possible. MTCs can be described as an extension of the models for
pairwise meta-analyses. They are slightly more complex in that they can be
applied to connected networks of arbitrary size and complexity. Some defi-
nitions of interrelated concepts when conducting MTCs are now provided:

4.9.1.1 Direct Comparison
A direct comparison is described as a head-to-head randomized controlled
trial (RCT) of pairs of treatments under investigation; for example treatment
A versus treatment B, as in Figure 4.14.

4.9.1.2 Indirect Treatment Comparison (ITC)


An indirect comparison occurs when two or more treatments of interest are
compared using a common comparator. For example, in Figure 4.14, treat-
ment A is the comparator in common that facilitates an indirect compari-
son between treatments B and C. Indirect comparisons are also commonly
referred to as adjusted or ‘anchored’ indirect comparisons.
134 Economic Evaluation of Cancer Drugs

4.9.1.3 Meta-Analysis
A meta-analysis is carried out when there is more than one trial involving the
same pairwise comparisons. It combines the treatment effects (or other results)
from these trials and presents one overall or pooled measure of effect size.

4.9.1.4 Network of Evidence
A network of evidence is a description of all the trials that include treatments of
interest and/or trials including the comparator treatments. A network diagram
(Figure 4.14) gives a visual representation of all the direct comparisons that have
already been made between the treatments, and can make it easier to deter-
mine the potential for indirect comparisons. It can be as detailed or as simple
as required, i.e. it need not include all the treatments in the network but just the
ones that are of interest. A network meta-analysis (NMA) is a more general term
for describing MTCs and indirect comparisons, and can be defined as an analy-
sis where the results from two or more trials that have one treatment in common
can be compared. Jansen et al. (2011) provide a useful summary of these.

4.9.2 Assumptions for Carrying Out MTCs


There are several assumptions to take into consideration when carrying out
an MTC analysis. First, it is important to bear in mind that randomization
only holds within the original trial and not across the RCTs. Unlike meta-
analyses, where results might be pooled despite strong differences in patient
characteristics as well as differences in the circumstances in which outcomes
were collected, MTCs assume homogeneity across trials for (indirect) effects
to be plausible. Hence, the homogeneity assumption must be satisfied.
The similarity assumption (Jansen, 2011) states that if trials differ among
the direct comparisons (e.g. a trial comparing A vs. B differs from a trial
comparing A vs. C), and these differences are due to ‘modifiers’ (see exam-
ples listed below) of the relative treatment effects, then the estimate of the
indirect treatment effect is biased. To explain this, consider two trials where
one trial involves a comparison of treatments A versus B (Trial 1) and the
other compares A versus C (Trial 2). If Trial 1 includes patients within the age
range 20–30 years and Trial 2 includes patients within the age range 60–80
years, then age is an effect modifier. Hence, any indirect comparison will
produce a result where potential confounding can bias the indirect estimate
of B versus C. Thus, to avoid violations of the above two assumptions, trials
must be comparable on effect modifiers such as:

• Patient characteristics
• The way in which the outcomes are defined and/or measured
• Protocol requirements, such as disallowed concomitant medication
• The length of follow-up
Introductory Statistical Methods for Economic Evaluation in Cancer 135

In summary, with regard to NMA, it is important to check whether the trials


are sufficiently similar to yield meaningful results for the ITC and MTC. A
further assumption, the consistency assumption, must also hold. This means
that trials must be comparable in effect modifiers and there must be no dis-
crepancy between the direct estimates and the indirect estimates. Due to the
way it is defined, this assumption can only be, and need only be, verified
when there are direct and indirect comparison data available for particular
pairwise comparisons. If the consistency assumption does not hold, then the
indirect estimates may be biased. In MTC analysis, however, since there is
always a sampling error, a strict evaluation of consistency based on point
estimates would not be appropriate.
Lastly, an assumption of exchangeability should be satisfied.
Exchangeability means that the relative efficacy of a treatment would be
the same even if it were carried out under the conditions of any of the
other trials that are included in the indirect comparison. In practice, this
assumption cannot be verified conclusively, though verification of the
similarity assumption would give more reason for the exchangeability
assumption to hold.
All the above assumptions must be considered very carefully when select-
ing studies for inclusion in any sort of network meta-analysis because the
validity of indirect comparisons depends on both the internal validity and
the similarity of the trials conducted or reported.

Example 4.9: An MTC Comparing Gefitinib


with Erlotinib for NSCLC
A simple MTC was planned to compare erlotinib versus gefitinib.
Erlotinib versus placebo was compared in one trial (Lee, 2012) and gefi-
tinib versus placebo was compared in a separate trial (Thatcher, 2005).
Further details can be found in Khan (2015).
In order to determine whether the assumptions above were likely to
hold, attention was paid to ensuring that, where information was avail-
able, the trials were comparable on the effect modifiers mentioned above.
In particular: the male to female ratio was checked and the median age
was compared between the three treatment groups; all the primary out-
comes were measured in the same way; and outcomes for all studies
were measured by progression-free survival (PFS) and overall survival
(OS). Other factors such as whether the trials were double-blinded mul-
ticenter trials and patient recruitment per region were also checked.
However, there are often factors such as adherence to the research pro-
tocol, quality of staff conducting the study, and other contributing fac-
tors that might increase heterogeneity between studies. A summary of
results from seven trials is shown in Table 4.10.
Using the data from Tables 4.10 and 4.11, we now conduct an MTC to
estimate the treatment effect for gefitinib versus erlotinib. Three addi-
tional studies could also have been included (one Phase II study, one
retrospective multicenter study, and another single center retrospective
136 Economic Evaluation of Cancer Drugs

TABLE 4.10
Summary of Results from 7 Published Trials of Lung Cancer Treatments
Study Author (year) Treatment Comparator OS HR PFS HR
GP1 Thatcher (2005) Gefitinib (n = 1129) Pl. (n = 563) 0.89 0.82
GP2 Zhang (2012) Gefitinib (n = 148) Pl. (n = 148) 0.84 0.42
GP3 Gaafar (2011) Gefitinib (n = 86) Pl. (n = 87) 0.81 0.61
EP1 Shepherd (2005) Erlotinib (n = 488) Pl. (n = 243) 0.70 0.61
EP2 Capuzzo (2010) Erlotinib (n = 438) Pl. (n = 451) 0.81 0.71
EP3 Herbst (2005) Erlotinib (n = 526) Pl. (n = 533) 0.99 0.94
EP4 Lee (2012) Erlotinib (n = 332) Pl. (n = 332) 0.93 0.81
Notes: G: gefitinib; Pl.: placebo; OS HR: hazard ratio for overall survival; GP: gefitinib vs. placebo
comparison; EP: erlotinib vs. placebo comparison. Studies 1, 2, 3 compare gefitinib vs. placebo,
whereas 4, 5, 6, and 7 compare erlotinib vs. placebo. HR = hazard ratio.

TABLE 4.11
Lung Cancer Data for Example 4.11
Study Treatment OS HR [95% CI] PFS HR [95% CI]
1GP1 Gefitinib (n = 1129) 0.89 [0.77–1.02] 0.82 [0.73–0.92]
2GP2 Gefitinib (n = 148) 0.84 [0.62–1.14] 0.42 [0.33–0.55]
3GP3 Gefitinib (n = 86) 0.81 [0.59–1.12] 0.61 [0.45–0.83]
4EP1 Erlotinib (n = 488) 0.70 [0.58– 0.85] 0.61 [0.51–0.74]
5EP2 Erlotinib (n = 438) 0.81 [0.69–0.94] 0.71 [0.62–0.82]
6EP3 Erlotinib (n = 526) 0.99 [0.85–1.15] 0.94 [0.85-–1.03]
7EP4 Erlotinib (n = 332) 0.93 [0.93–1.10] 0.81 [0.68–0.95]
1 Notes: Thatcher et al. (2005).
2 Zhang et al. (2012).
3 Ghaffar et al. (2011).
4 Shepherd et al. (2005).
5 Cappuzzo et al. (2010).
6 Herbst et al. (2005).
7 Lee et al. (2012).

study could also have been included as RCTs). However, only the seven
Phase III RCTs in Table 4.11 are used.
A network diagram for the data in Table 4.11 is shown in Figure 4.15.
Rather than use just the pooled estimates as in a meta-analysis, the
treatment effects from each study are used in the MTC to avoid loss of
information.

Step 1: Identify the measure of effect. In this case it is the hazard ratio
and, therefore, we will work on the log hazards scale.
Step 2: Determine whether a Bayesian fixed effects or random effects
model is to be used. We will choose a random effects model.
Introductory Statistical Methods for Economic Evaluation in Cancer 137

FIGURE 4.15
Network meta-analysis diagram for Table 4.11.

Step 3: Identify the network structure. This also depends on whether


pooled estimates are used or the individual effect sizes from each
trial. For example, a meta-analysis (see Khan, 2015) generated pooled
effects of the direct pairwise treatment effects. These could be used,
but as noted earlier we use the ‘raw’ effects (treatment differences)
available from Table 4.12.
Step 4: Specify the model assumptions.
Let the log hazard ratio for a study be pooled from a distribution
that has a normal distribution with mean µi and a precision that is
determined by the standard errors (SEi) of the studies (subscript i is
for each study). Let yi be the log hazard ratio for study i, and ti the
precision of the distribution for the log hazard ratios, then the likeli-
hood for the model is as follows:

yi ~ N ( mI , ti )

where ti is: 1/[SEi * SEi]


There are 3 treatment effects: TE(1), TE(2), and TE(3) associated
with the treatments placebo, erlotinib, and gefitinib respectively,
each with the following identical non informative priors: Therefore:

• TE(1) ~ N(0, 0.00001)


• TE(2) ~ N(0, 0.00001)
• TE(3) ~ N(0, 0.00001)

Step 5: Identify the data to be used in the MTC.

These data include 7 hazard ratios (in Table 4.11) and the 7 standard errors. It
is important to ensure the direction of the comparison is correct. For example,
138 Economic Evaluation of Cancer Drugs

in Table 4.11 the HR for gefitinib versus placebo in study 1 is 0.89. The HR for
placebo versus gefitinib would be 1/0.89 = 1.12 and log (HR) = 0.11653, which
would be used in the analysis.

Step 6: Write the code (WINBUGS or SAS)

                WINBUGS CODE:
   # MODEL
   model {
    for (i in 1:N) { # indexes studies
    t[i] <- 1 / SE[i]*SE[i]
    y[i] ~ dnorm(mu[i], t[i]) # Likelihood function
    mu[i] <- (TE[t[i]] - TE[t2[i]])
    } # end i loop
   # OUTPUTS
    for (base in 1:(NT-1)) { # indexes treatments
    for (comp in (base+1):NT) { # indexes comparators
    theta[base,comp] <- exp(TE[base] - TE[comp])
    }} # end base, comp loops
   # PRIOR DISTRIBUTIONS
    for (k in 1:NT) {
   TE[k] ~ dnorm(0.0,0.00001)
    } # end k loop
    } # END MODEL
   #DATA
   list(N=7, NT=3, HR=c(0.1165338163, 0.1743533871,
0.2107210313,
   0.3566749439, 0.2107210313, 0.01005033585,
0.07257069283),
    SE=c(0.072, 0.155, 0.164, 0.098, 0.079, 0.077, 0.088),
    t=c(1, 1, 1, 1, 1, 1, 1), t2=c(3, 3, 3, 2, 2, 2, 2))

The indirect comparison of erlotinib versus gefitinib results in a very slight


advantage (likely to be clinically irrelevant) for erlotinib. However, the PFS
effect from gefitinib suggests it would offer a potentially more cost-effec-
tive treatment. One would also expect HRQoL during the progression-free
period to be better with gefitinib compared to erlotinib, which would influ-
ence the cost per QALY.

4.10 Summary
It is not possible to cover all the statistical methods used for modeling sur-
vival and other data for cost-effectiveness analyses in this book. We have not
discussed subjects such as, for example: hurdle and dispersion models (for
TABLE 4.12
Summary of Main Statistical Issues Relating to Economic Evaluation of Cancer Drugs
Issue Costs HRQoL Survival
Distribution Normal Gamma Any justifiable (normal, Normal Nonparametric (e.g. Exponential, Weibull or
beta, etc.) Beta (EQ-5D) Cox PH) suitable justifiable
Other justifiable
Other Normal Possible over-dispersion Possible over-dispersion Proportional hazards PH or non-PH
assumptions Gamma (PH)
Over-dispersion
Statistics of Mean costs Mean Mean Mean survival Mean
choice Median Hazard ratio (survival Truncated/restricted
Any justifiable rate) Mean
median
Missing data Multiple Multiple imputation, Multiple imputation, Right censoring or Right censoring or similar
imputation depending on mechanism depending on similar
methods of missingness mechanism of
assumption (e.g. MAR, missingness
MCAR, MNAR) assumption (e.g. MAR,
Worst case MCAR, MNAR)
Impute using placebo
Extrapolation Model -based None Model-based (e.g. post none Model-based (Weibull,
progression HRQoL) exponential or suitable)
Treatment Included in Not typically adjusted for Not typically adjusted Censoring at crossover Censoring at crossover
switching mean costs for IPCW IPCW
May assume that RPSFT RPSFT
HRQoL improves or Two-stage methods Two-stage methods
deteriorates depending As a sensitivity analysis See section above
Introductory Statistical Methods for Economic Evaluation in Cancer

on nature of switching since regulatory Important to harmonize


Mean statistic will be question is what estimands for efficacy
used regardless would have been the and effectiveness
effect on OS without wherever possible
139

switching
ITT: Intent to treat population.
140 Economic Evaluation of Cancer Drugs

TABLE 4.13
Results from WINBUGS
Comparison OS HR PFS HR
Erlotinib vs. placebo 0.855 0.777
Gefitinib vs. placebo 0.874 0.419
Erlotinib vs. gefitinib 0.996 0.544

cost data); joint modeling of costs and effects; landmark analyses; propensity
modeling (for real-world data) for non-randomized data; mixed modeling for
analyses of utilities; and multiple imputation. This would require an entire
book in itself. Nevertheless, we presented at the end of this chapter Table 4.13
that summarized the types of statistical methods that could be relevant for
modeling data for economic evaluation of data collected in cancer patients.

4.11 Exercises for Chapter 4


1. Distinguish between variability and uncertainty.
2. Explain the following terms in the context of survival analysis: rate,
hazard rate, risk, proportion, and probability.
3. What is a hazard ratio? interpret a hazard ratio of 0.80.
4. What distribution would you expect costs to follow and why?
5. What is a transition probability matrix in the context of healthcare?
6. How might parametric survival models be used in an economic
evaluation?
7. Distinguish between the different types of switching and their
implication for cost-effectiveness analyses.
5
Collecting and Analysis of
Costs from Cancer Studies

5.1 Types of Costs Typical of Cancer Trials


To estimate costs of healthcare in treating cancer, we need first to distin-
guish between unit price and health resource use (in physical units). Health
resource includes items such as number of visits to the hospital, number of
nights spent in hospital, number of milligrams of drug, and so forth. The
conversion of these healthcare resource items into costs requires a unit price
(P) associated with each resource. The quantity of the resource (Q) is then
multiplied by the price to derive a cost: C = P × Q. Since there are lots of dif-
ferent types of resources, we can reference each type by a subscript i, so that
the cost of each resource used is: Ci = Pi ´ Q i .
For each patient, we can compute the total cost of health resource consumed
(with an additional subscript j, for each patient): Cij = Pi ´ Q ij . The unit price of
a health resource is likely to be the same for each patient. The cost will change
depending on how much of the resource use is used. The ‘unit price’ is the
expense incurred by the purchaser/provider of the resource and will depend
on the decision-maker’s perspective of the analysis.
The individual patient-level costs are then added up to get a total cost per
patient over a given follow-up period. If the patient is followed up for 1 year,
the total costs over a 1-year period will be computed for that patient. Another
patient might have died after 6 months, and hence total costs will be com-
puted over 6 months. The above approach allows us to compute the average
(mean) cost per patient per time period (e.g. monthly, yearly, by cycle), the
average cumulative cost over time, and the overall average total cost, and
compare these between treatment groups. These costs could be plotted to get
an idea of the distribution of costs between treatment groups. Furthermore,
the total costs could also be categorized. It may be useful to know what the
breakdown of costs is, because some particular resource use might be used
more (e.g. hospitalization costs if there are severe toxicities). Costs could be
categorized as drug costs, surgery, radiotherapy, laboratory, imaging, post-
treatment surveillance, etc.

141
142 Economic Evaluation of Cancer Drugs

5.1.1 Categorization of Health Resource Use


In general, resources directly related to treatment or the consequence of
treatment may be categorized as shown in Table 5.1.
Some resource acquisition costs and resource use are published or easily
accessible. For other resource use, such as those involving complicated pro-
cedures (for example, surgery, radiotherapy, and imaging), primary research
may be needed to collect this data.
Typical health resource use in a breast cancer trial might include: first-line
chemotherapy, radiotherapy, additional (subsequent lines) chemotherapy,
treatments for adverse events, genetic testing (e.g. for the BRCA gene, whether
mutant positive or negative), surgical procedures (e.g. initial lumpectomy or
mastectomy, perhaps also reconstructive plastic surgery), duration of hos-
pital stay, and hospital outpatient appointments or day care. It is therefore
important to identify at the study design stage all the health resources that
will be monitored and where data will need to be collected during and pos-
sibly beyond the trial (for real-world data, see Chapter 8).

5.1.2 Resource Use Monitoring


Health resource use monitoring is particularly important in cancer trials
where a patient can undergo several lines of treatment between periods of
stable disease (SD) and progressive disease (PD). Consequently, one should
include health resource use from the whole patient pathway – from the start
of treatment until cure or death. That is, an economic evaluation over a life-
time horizon should be considered. This implies health resource use should
be collected even beyond protocol study follow-up. For example, for long-
term recurrence (sometimes several years after the end of the intervention)
of either the disease or adverse events, any costs of palliative care or longer-
term management, if required, need to be considered (and monitored).
A key consideration is to determine how in the future, downstream costs
associated with an experimental cancer treatment compare to those in the

TABLE 5.1
Categorization of Health Resource Use
Resources Related Directly to the Resources Related to the Consequences of
Drug Administering the Intervention
• Nurse time for administering an • Treatments for side effects, intensive care unit
infusion admission (e.g. due to a serious toxicity for
• Drug amounts (mg per tablet or example)
infusion) • Additional visits to the hospital, length of stay
• Materials and equipment in hospital, visits to the doctor
(single-use and also multiple-use • Social or personal services (carer support,
equipment) (compare for example physiotherapist, psychologist support), cost of
a throw-away plastic syringe and parking at the hospital, cost of not going to
the use of an infusion pump) work by either carer or family member
Collecting and Analysis of Costs from Cancer Studies 143

control arm. For example, if the type of palliative care and its duration is
expected to be the same in both groups (assuming a two-arm study) then
there is likely to be little benefit in monitoring these health resource costs for
an economic evaluation because we are usually interested knowing about
those health resource items where the difference in mean costs (incremental
costs) is likely to be larger.
It may be difficult to know beforehand which health resources items are
likely to be used more in one particular group than the other. It may be pos-
sible to postulate a hypothesis that a new treatment is associated with more
or less healthcare resource use at the trial design stage. For example, a new
chemotherapy might be expected to have a better safety profile, and so we
may expect costs from additional treatments for toxicities to be lower, mean-
ing fewer hospital or doctor’s appointments. In such cases we might focus on
specific health resource use at study design. Similarly, where there are higher
numbers of follow-up CT scans (i.e. additional CT scans after treatment has
been completed) between experimental and control arms, this not only biases
some outcomes such as PFS, but the costs may also be overestimated – the issue
here relates to inadequate trial design. Taking more scans on one arm com-
pared to the other offers greater opportunity to detect (and shorten) PFS, while
increasing the costs of scans (not to mention the risks of radiation exposure).

5.1.3 Baseline Characteristics and Health Resource Use


Baseline characteristics are sometimes relevant to clinical outcomes. The
typical approach is to adjust treatment effects for factors such as age, gen-
der, ECOG, and disease stage (to name a few). However, these (risk) factors
(see Chapter 1) may also be related to post-baseline costs. Patients who have
poorer ECOG, or are elderly, may well be the same ones with later post-base-
line health resource use. For statistical analyses of costs, not only are clinical
or demographic covariates used to adjust and control the variability of the
ICER, or the incremental costs, the baseline costs will also be important for
this adjustment. Baseline measures of medical resource use can be used to
reduce the variance in incremental cost estimates (Ramsey et al., 2005; 2015).
For example, some costs such as carer costs, concomitant medication, and
visits to hospitals will occur prior to randomization. These costs may or may
not be different between treatment groups (it is something we might check).
When computing the mean incremental costs, the baseline costs would be
included in any statistical model. Patients with high baseline costs often
have high post-baseline costs. The mean incremental costs will be based
on modeling the correlation between post-baseline costs and baseline costs
(along with other factors). This usually provides shorter confidence intervals
for the incremental costs (depending on the type of analyses).
Baseline costs might be presented following categories as in Table 5.2 over-
all and by baseline characteristics to show the relationship between baseline
costs, post-baseline costs, and baseline characteristics. Note that in Table 5.2,
144 Economic Evaluation of Cancer Drugs

TABLE 5.2
Example of Mean Costs Collected at Baseline and Post-Baseline
Treatment A (Mean) Treatment B (Mean)
Baseline (£) Post-Baseline (£) Baseline (£) Post-Baseline (£)
Gender
Male 4,000 7,000 2,000 4,000
Female 2,000 4,000 1,000 1,500
ECOG
0 2,500 2,400 2,450 1,890
1 2,000 2,100 1,980 1,400
2 4,000 3,800 3,800 2,200
Stage
I-II 3,000 6,000 3,000 3,000
II-IV 1,500 5,000 1,450 2,500

age is not included. Age would need to be categorized for presentation (how-
ever, it can be incorporated as a continuous covariate in statistical analyses
without categorization).
Table 5.2 shows that mean baseline costs for ECOG and stage are similar
between groups. Treatment B appears to have lower post-baseline mean costs
compared to treatment A. There seem to be differences in costs at baseline
for males and females between treatments: males in particular have lower
baseline costs (£2,000 vs. £4,000) for treatment B, reflected in post-baseline
costs. Here baseline adjustment would result in a different mean incremental
cost compared to when baseline is ignored (raw mean difference).

5.1.4 Costs Determined by a Study Protocol


An important factor that can be overlooked is the fact that an economic eval-
uation does not seek to estimate the costs of a trial or study. Rather, it is the
value of the intervention that needs quantifying. Such costs are derived from
health resource identified in a clinical or study protocol. Since controlled
clinical trials do not reflect real-world practice (as they are experimental set-
tings), it is important to capture those costs related to the intervention dur-
ing the experiment and beyond it. A study protocol may restrict the use of
a number of resources that might otherwise be used in a real-world setting
(e.g. restrict the use of some concomitant drugs or subsequent treatments).
On the other hand, a protocol might increase the use of other resources (like
imaging or genotyping procedures and laboratory tests) – perhaps to cap-
ture PFS better. Several issues require consideration.

(i) Differences in hospital sites from which patients are recruited.


Most clinical trials recruit a selected sample of patients (through
application of inclusion/exclusion criteria). The trials are conducted
Collecting and Analysis of Costs from Cancer Studies 145

in a nonrandom sample of hospital sites or other health settings


(often larger or academic hospitals).
(ii) Identifying unit prices and costs of health resource use from different
hospitals.
This can be challenging because one would need to have access
to the billing data located in the finance departments of hospitals,
or information from insurance claims, or one may need to perform
primary research to find out these unit costs. An expert panel could
be used to elicit expected unit prices. However, opinions may differ
widely.
(iii) Changes to the trial.
Where a substantial protocol amendment is carried out such that
the trial design is altered in some way, or additional assessments
are collected, this will impact health resource collection. Prior to the
amendment, different costs may have been collected compared to
post-amendment resulting in health resource data from two differ-
ent types of experimental conditions.
(iv) Using early phase data. In Phase II trials, an early ICER might be
derived to inform Phase III trial design and future economic evalu-
ation. However, a Phase II trial may have more exhaustive protocol-
defined assessments, such as more scans to determine the optimal
time when disease progression occurs, more laboratory tests and
HRQoL data assessments. These are likely to be optimized in a
Phase III trial and hence an ICER from a Phase II trial will need to be
treated with some degree of caution when informing trial design for
cost-effectiveness for a later Phase III trial.

5.2 Perspective of Analysis and Costs Collection


It is the study perspective that will define which costs will be included and
which unit cost (price) will be used to value the resources used. Two extreme
situations can exist: one where all the costs are borne by the patient (full out-
of-pocket expenses) or by private health insurance in situations/countries
where a public healthcare system is not available; and, at the other end, full
coverage of all healthcare expenses by a central government, or other agency,
or administration.
Most developed countries have a healthcare financing system that falls
somewhere between these two extremes. For example, although the UK has
a fairly centralized National Health Service (NHS) with widespread respon-
sibilities over the whole healthcare spectrum, there are some interventions
or drugs that are not covered by the NHS, or not recommended by NICE.
146 Economic Evaluation of Cancer Drugs

Moreover, in many countries, for some interventions or treatments such as


dental treatments or (certain) drugs, a patient contribution (co-payment) may
also be expected (Burau, 2015; Wendt et al., 2009).
Most countries have a mix of partially/fully covered or reimbursed inter-
ventions depending on the status of the patient, their income, disease, type of
intervention, and/or perceived efficacy of the intervention or drug. Such sys-
tems of healthcare can result in complex approaches to payment for treatment
for patients and can often result in patients paying out-of-pocket expenses.
In such cases there may be an argument for cost-effectiveness analyses to
be conducted from a societal perspective. In the majority of cases, the main
perspective will be that of the public payer for the healthcare in a restricted
budgetary perspective. In the UK, for example, cost-effectiveness analyses
are conducted mainly from an NHS (and sometimes Personal Social Services
(PSS)) perspective.
In several countries there are special budgets for drugs with their own
coverage and reimbursement rules. This results in the so-called ‘silo’ effect
where, historically, decision-makers sitting on drug committees did not feel
that non-drug expenses were important when considering the financial
impacts of new drugs. For the same reason costs borne by patients or orga-
nizations outside the health budget (societal costs) will often be overlooked.
Many national economic evaluation guidelines have an explicit statement
regarding their perspective choice. Therefore, many studies use only the
country official fees or tariffs as unit cost (see Section 5.5).

5.3 Collecting Health Resource Use Across


the Treatment Pathway
Table 5.3 shows how collecting health resource use information can be sepa-
rated by periods of time. Each time period has a specific emphasis. Some
costs may be irrelevant (e.g. the pre-staging process) whereas others are
clearly important.
Pre-staging: The health resource use required for assessing asymptomatic
disease, screening tests, or symptomatic visits to a GP or specialists before
cancer definitive diagnosis is related to costs. These costs cover the pretrial
period generally and are often either unknown or not relevant for the trial
objectives. Such costs are not ‘treatment emergent’ and therefore can be
ignored for an economic evaluation in the context of a clinical trial.
Staging: Cancer suspicion is high, and a number of tests, imaging, and
surgical procedures (biopsies) are undertaken to get a final diagnosis and
cancer stage. Although health resource use takes place, these costs should
not be confused with the costs of the intervention. Sometimes the costs of
a performing a clinical trial can get tangled up with the costs related to the
TABLE 5.3
Health Resource Use Categorized by Time Periods for a Clinical Trial
Pre-staging Staging Start of Treatment Post-treatment Post-trial Palliative Care
Definitive Imaging Chemotherapy Maintenance Additional End-of-life care
diagnosis of Biomarker tests Radiotherapy therapy medical care Equipment and
condition Resource use for confirming Surgery Additional scans Hospital, doctor, aids for self-care
No health diagnosis not relevant for Concomitant medication Subsequent nurse visits Palliative
resource use economic evaluation Laboratory testing treatments Social care (carer radiotherapy
relevant as not Only tests that are considered Pathology support) Specialist nurses
‘treatment predictive of treatment (e.g. Hospitalization for adverse Family support (e.g. Macmillan or
emergent’ EGFR testing) events Treating long-term Marie Curie
Collect any baseline costs Administration costs of or lifelong nurse)
– such as care, support, family delivering intervention toxicities
help at baseline as this could (e.g. infusions)
increase or decrease Scans/imaging
post-treatment
Collecting and Analysis of Costs from Cancer Studies
147
148 Economic Evaluation of Cancer Drugs

consequences of delivering the treatment. Since patients entering a clinical


trial already have a diagnosis of cancer, any health resource use related to
establishing their diagnosis, eligibility for the trial or part of inclusion/exclu-
sion criteria are unlikely to be relevant for an economic evaluation. Some
health resource use that occurs just prior to randomization or baseline might
be useful and can help to manage the variability in post-baseline costs (after
adjusting for baseline).
Start of treatment: Health resource use after randomization is usually
the most important, often covering the entire spectrum of treatment options
including chemotherapy, concurrent chemotherapy with radiotherapy, sur-
gical procedures, and any laboratory and/or pathology testing, as well as any
other costs likely to be incurred when the treatments are taken (or adminis-
tered) in a ‘real’ or ‘natural’ setting beyond protocol follow-up time. In some
trials there is an open label (or extended follow-up) period where the ‘real-
world effects’ (see Chapter 8) can be observed, which will also include costs.
Post-treatment period: After treatment has concluded, follow-up contin-
ues (e.g. 6 cycles of chemotherapy have been taken but follow-up continues
for 2 more years) and an observation period is scheduled where the response
status (i.e. any progressive disease in terms of tumor growth, metastases or
additional malignancies, still alive or dead) of the patient is assessed and
monitored. In case of relapse/recurrence generally, a new treatment line will
be started depending on whether the patient is able to sustain it. This may
depend on the previous treatment and, in some cases, the patient’s genetic
profile where targeted therapies are concerned.
Post-trial period: For patients who are alive or even cured of their cancer
at the end of the trial, further medical care could still be needed. This can
be either lifelong treatment such as when definitive sequelae (e.g. remain-
ing symptoms related to the cancer, but not the cancer) are present (such as
lymphoedema in breast cancer, permanent stoma in bowel cancer, etc.). The
associated costs could therefore be higher or lower on a given arm, depend-
ing on the initial trial therapy choice (arm).
Palliative care: This is sometimes referred to as the ‘end-of-life’ stage and
is associated with failure of all previous treatments or if the patient has dete-
riorated to an advanced metastasis, where the cancer has spread at multiple
sites in the body. Palliative care may also include chemo- or radiotherapy,
without intention of cure. Apart from the costs of chemo- or radiotherapy,
the costs of the carer and specialist palliative nurse, as well as additional
societal costs play a role here.

5.3.1 Time Horizon
An important consideration for collecting health resource use data follows
on from the above in terms of the time horizon. In cancer trials, at the end
of the maximum follow-up period (it would be expensive to conduct a trial
that follows all patients until death, if some patients lived for a long time),
Collecting and Analysis of Costs from Cancer Studies 149

some patients may still be alive. For these patients, their costs are censored at
the last time point known alive. Hence, future costs are unknown for those
patients still alive and would need to be estimated. In Section 5.8, an example
is provided on how to adjust for censored costs.

Example 5.1
In a trial in newly diagnosed glioblastoma patients comparing two treat-
ments (experimental E and control C), after 3 years of follow-up, 35%
of patients are alive on E and 25% on C. The mean costs for E over the
3-year period were $21,000 and $35,000, respectively.
To estimate future costs (ignore discounting for now), we first need to
predict the proportion of patients alive after 3 years for each group. We
can do this by using a special type of statistical model called a paramet-
ric survival model (introduced in Chapter 4). Assume that the survival
model predicts no patient to be alive by year 8 and beyond. Once this is
done, we need to make one of several possible assumptions about the
behavior of future costs.
The simplest method is to assume that the average per patient costs
over the previous 3 years or over the last year would be constant over the
next 5 years. This is known as last observation carried forward (LOCF).
This assumption of constant costs will need to be evaluated separately.
Table 5.4 shows the computations for future costs assuming an average
(mean) constant cost of $3,000. If we assume 1,000 patients were enrolled
and the average cost per patient for E in year 3 is equal to $3,000, then if
we also assume costs are constant from year 4 and beyond, we can com-
pute future expected costs.
The assumption of constant mean future costs however may not be
realistic. There is no reason to believe that once further away from the
start of treatment, a patient would have the same level of care and there-
fore costs for several reasons:

• If a patient is still in remission, then patient monitoring will be


less intensive over time and therefore less costly
• Conversely, if disease progression has occurred, then treatment
costs would increase significantly because the patients may
even be removed to an intensive monitoring unit or moved to
palliative care.
• Cancer is associated with deteriorating health and disease pro-
gression (and related health states), hence from a physiological
perspective it is unlikely that the assumption of constant costs
over time is tenable. In the same way that we identified the
problem of extrapolation of utilities in Chapter 3, it is possibly
better to extrapolate the behavior of future costs using appro-
priate modeling techniques, rather than assume constant costs.
Markov or multistate survival models identified earlier could
be used for this purpose (Chapter 4 and Chapter 7).
• Patients that relapse later may be treated differently to those
who relapsed earlier in the trial because medical practice and
150 Economic Evaluation of Cancer Drugs

TABLE 5.4
Example of Future Cost Estimation
% Alive % Alive (Predicted)
Year 3 Year 4 Year 5 Year 6 7 8 Total (Y4–Y8)
E (% alive) 35% 25% 10% 5% 2% 0%
C (% alive) 25% 15% 12% 3% 1% 0%
E (costs) 3,000 750 300 150 60 0 1,260,000a
C (costs) 3,000 450 360 90 30 0 930,000
a Note:  Calculated as $3,000 × 0.25 + $3,000 × 0.1 + $3,000 × 0.05 + $3,000 × 0.02 = $1,260 × 1,000
patients.

technology change over time – the so-called ‘technological


drift.’ The nature of the ‘shift’ is, however, unknown since all
estimated costs are calculated using current technology,

Actual and future healthcare costs unrelated to the disease under


study are normally ignored in the cost analysis although there has
been some discussion about this in the literature (Versteegh et al., 2016;
Hoefman et al., 2013; Rapange et al., 2008; Van Baal et al., 2017, Van Baal
& Wang, 2011)

5.4 Costing Methods: Micro versus Macro Approach


There are two main approaches to calculating costs: a macro approach and
a micro approach. The macro or gross-costing approach uses a ‘top-down’
approach for breaking down the costs borne in a traditional accounting way,
resulting in estimations such as, for example, the average (mean) cost per
case, or per diagnostic-related group (DRG) or average cost per hospital day.
The micro approach uses a bottom-up methodology by tracking down the
individual resources and activities performed during an intervention or treat-
ment and aggregates the intermediate costs up to a total cost. The advantage
is its precision and level of detail. The disadvantage is that the level of detail
with which costs are collected is time-consuming, and sometimes may not
be worthwhile because differences between groups in terms of some of these
cost items may be negligible, although they might be collected for interest.
In practice, a mixture of both (macro and micro) approaches might be used
by collecting drug costs in more detail (e.g. drug name, drug dose, number
of days or cycles given, dose reductions, or amount of wastage), whereas
the cost of chemotherapy administration as an outpatient in a given cancer
hospital would be determined from the average cost per visit, either at that
hospital or, eventually, at a national level.
Collecting and Analysis of Costs from Cancer Studies 151

5.4.1 Average versus Marginal and Incremental Cost


The average total cost (ATC) is the sum of average variable costs and average
fixed costs; thus, the ATC is equal to the total cost divided by the number of
units produced. At the patient level, this is equivalent to computing, for each
patient, the total cost across all subcomponents (drug cost, cost of treating
adverse events, cost of hospital visits, etc.) incurred for their care. The aver-
age or mean total cost is subsequently calculated for all patients in a given
treatment group.
Marginal cost is the change in the total cost that arises when the quan-
tity produced is incremented by one unit, that is, it is the cost of produc-
ing one more unit of a good. In general terms, marginal cost at each level of
production includes any additional costs required to produce the next unit.
Therefore, the marginal cost will tend to decrease most when production
is increased in cases where fixed costs are large compared to the variable
costs. An example is a proton therapy machine or robotic surgery that needs
high-cost equipment, and possibly investment in buildings that need to be
renewed/upgraded over a relatively short time period (8 to 20 years)

Example 5.2
In this (simple) example, we will show that when there are high fixed
costs, such as expensive equipment (e.g. imaging equipment, biomarker/
DNA extraction machines), the average cost of using the equipment will
decrease if they benefit a large number of patients.
Assume a family practitioner with a secretary where both are paid
on a per patient visit basis. Assume a fee for service of €25 per visit for
the practitioner and €10 for the secretary, and no other costs for the time
being. On day 1, 30 patients are seen and on Day 2 31 are seen. We now
can calculate the cost for the service for these 2 days

Day 1 Day 2
Number of patients 30 31
Practitioner 25 * 30 = 750 25 * 31 = 775
Secretary 10 * 30 = 300 10 * 31 = 310
Total cost 1050 1085
Average cost 1050/30 = €35 1085/31 = €35
Marginal cost 1085 – 1050 = €35

We see in this case that the average cost is constant and that the mar-
ginal cost equals the average cost. Let us now assume the practitioner
bought some IT equipment for both of them to use for a total of €3,000
and that it will have to be replaced in 3 years’ time (without deprecia-
tion). Assuming 200 working days per year at 30 patients per day, we can
now recalculate the average and marginal cost.
152 Economic Evaluation of Cancer Drugs

Day 1 Day 2
Number of patients 30 * 200 = 6000 6001
Practitioner 25 * 6000 = 150,000 25 * 6001 = 150,025
Secretary 10 * 6000 = 60,000 10 * 6001 = 60,010
Equipment €3000/3 = 1000 €3,000/3 = 1000
Total cost €211,000 €211,035
Average cost €35.16666 €35.16663
Marginal cost –€0.0002

We now see that the average cost is only slightly higher, but the mar-
ginal cost is negligible given the large number of patients seen. This
is because fixed costs are still low compared to variable costs and are
spread out over a large number of patients. Had the practitioner invested
much more in equipment, say €180,000, then the average cost would be
much higher because the influence of fixed costs becomes greater. It also
means that the average cost for the same procedure with similar technol-
ogy may vary widely between institutions according to their operating
level at full or less than full capacity. We leave it to the diligent reader to
calculate the different costs for this case.
The above implies that when two alternatives are compared they
should be compared at the same capacity level. In general, at the overall
hospital level about 70% of costs are labor costs but this proportion var-
ies widely between departments and procedures. Compare for example
radiotherapy with psychiatry or a visiting nurse service.

5.4.2 Inflation
Some clinical trials take several years to complete. Health resource use con-
sumed by patients earlier in the trial, say in 2008, may have a different price
by the end of the trial, which might be completed in, say, 2013. The unit
prices, or rather their value due to inflation, may have changed. Dvortsin et
al. (Dvortsin, Gout-Zwart, Eijssen, Van Brussel, & Postma, 2016) noted that
drugs such as cetuximab, bortezomib, and bosutinib had different prices:
“Treatments in the late stage were found to be more expensive per QALY by a
factor ranging from 1.5 to 12” (this may be explained by the fact that patients'
health changes over time, e.g. deteriorates from Stage I to Stage IV cancer,
and therefore costs of care later on are likely to be higher). Hence, price dif-
ferentials will need to be adjusted. Dvortsin et al. reported that late stage
treatment was more expensive by a factor 10, and could lift the ICER over the
threshold, potentially rendering a new treatment as not cost-effective.
We would need to inflate or deflate the prices by bringing them to a
standard year and either report the mean costs at 2013 prices (usually the
reporting takes place using later prices at the end of the trial or at the time
of analysis). In some countries a national statistics office or some other public
agency publishes a price index for healthcare services. When using these
price indices attention should be paid to check whether the indices published
Collecting and Analysis of Costs from Cancer Studies 153

relate to a healthcare industry, or other retail goods. If no healthcare price


index can be found, then we would need to rely on the more general cost-of-
living or macroeconomic GDP price deflator. The drawback is that healthcare
prices grow more rapidly than in other sectors of the economy.

Example 5.3
A clinical trial was conducted between 2008 and 2013. The unit prices
for hospital stays are reported as £400 per night using 2008 prices. The
final analyses will be performed using 2013 prices. First, we need to find
an inflation index (which is published somewhere). We need one for the
year 2008 and one for the year 2013. Assume the 2008 index = 105 and the
2013 index = 125.
The formula for inflating prices is:

price index current year (2013 ) - price index base year (2008 )
∗ 100
price index base year (2008 )
= (125 - 105) / 105 = 20 / 105 = 0.19 × 100 = 19% (a 19% increase)

The cost of £400 per night in 2008 is now (400 × 1.19) = £476 in 2013 prices.
A caveat here is that this applies only when the healthcare resource is
fixed and does not change over time in the construction of the index (a
so-called chained price index). This could become particularly problem-
atic when using a disease-specific price index with rapid technological
change (Dunn et al., 2018; Hall & Highfill, 2013).

5.4.3 Time Preference and Discounting


Time preference refers to the idea that people prefer to receive benefits
sooner and pay costs later. Some people may exercise today in the hope that
they live longer later; others may or prefer to enjoy their leisure time through
smoking or watching TV now. Individuals are considered to have a positive
time preference in the sense that they might have a preference for health ben-
efits earlier rather than in some distant future even when the inflation rate
is zero (which may or may not always be the case for a specific individual).
The current practice is therefore to discount both future costs and future
benefits (e.g. QALYs or life-years gained) by a discounting factor after the
first year to estimate the present value. Generally, a constant discount factor
(between 1.5% and 5%) is used and fixed by the national guidelines for eco-
nomic evaluation. In some countries, a different discount factor for costs and
outcomes is used (e.g. 3.5% per annum in the UK).
The net present value (NPV) is the sum over a number of years (n years) of
the present value of the cost incurred in each year NPV = ∑ FV/ (1+r ) where
n

n is the year and r is the discount factor. Reporting using undiscounted and
discounted results is useful because the discount rate can have an important
154 Economic Evaluation of Cancer Drugs

impact on the conclusions of an analysis. In cancer trials, the proportion of


patients who remain alive at the end of the trial is not often zero. Hence the
future survival rates for each treatment group is a future benefit from which
future QALYs are generated. It is these future benefits that are subject to
discounting. Discounting is not applied when the cost-effectiveness analyses
take place over a time horizon of less than 1 year (for a general discussion on
discounting see Claxton et al. (2011) and Drummond et al. (2015).

Example 5.4
The total costs for a patient in each of years 1, 2, and 3 are £3,000, £7,000,
and £5,000 respectively (total of £15,000). Discounting starts at year 2,
hence, we will discount at 3.5% in each of years 2 and 3. After discount-
ing, the value of £15,000 received by year 3 is £14,045

Year 1 Year 2 (2016) Year 3 (2017)


(2015) (£) (£) (£) Total (£)
Cost 3,000 7,000 5,000 15,000
Discounted 3,000 6,5351 4,510 14,045

1£7,000 × 1/(1 + 0.035)2 = £6,535.

5.5 Charges
Unit costs should not be confused with charges. Charges are the amounts
paid by insurers, or national/local health systems, or similar public agen-
cies, or private insurers to individual care professionals or organizational
healthcare providers, including hospitals. These can be defined at the diag-
nostic-related group (DRG) hospital episode-level for hospitals, or in fee-for-
service reimbursement systems, down to individual care procedures, drugs,
and laboratory tests, and so on. The reimbursement in this case is based on
official positive reimbursement lists such as the ‘Red Book’ in the US for
drugs, the NHS drug tariff list in the UK, and other similar sources for other
countries (Truven, 2018).
In some countries (e.g. Canada, Spain, Italy), tariffs are decentralized, to a
greater or lesser extent, by region or province. Many countries, especially for
hospital reimbursement, use a combination of fixed daily fees, DRG or epi-
sode-based fees, and for-service fees, making cost estimations quite complex.
However, in these cases the diffusion of electronic billing systems, whether
hospital-based or those of (national) insurers, makes it, in principle at least,
easier to retrieve billing data either for groups of patients or individually
tagged ones. Access to such data is, however, governed by national privacy
laws and access can be restricted for external parties.
Collecting and Analysis of Costs from Cancer Studies 155

5.5.1 Cost-to-Charge Ratios
Charges rarely reflect the actual costs of the goods or services provided. In
the US, through its Medicare Cost Reports, the Center for Medicare Services
(CMS) publishes cost-to-charge ratios that represents the total amount of
money required to operate the hospital, divided by the sum of the revenues
received for patient care and other operating revenues. These are hospital
averages, however, and are difficult to apply to specific bundles of proce-
dures therefore (Brent, 2002)
For clinical procedures, the Center for Medicare and Medicaid Services
(CMS) has also developed the resource-based relative value system (RBRVS)
approach that is used to calculate reimbursement fees through the Medicare
Physician Fee Schedule (MPFS). Each current procedural terminology (CPT)
code in the MPFS is assigned a relative value unit, which is then multiplied
by the annual conversion factor (a dollar amount) to yield the national aver-
age fee. These are further adjusted according to geographic indices based on
provider locality. Payers other than Medicare, such as private insurers, may
also adopt these relative values and apply their own conversion factor(s).

5.5.2 Other Non-Medical Costs (e.g. Societal Costs)


When taking a societal perspective in a welfarist approach, productivity
losses and related costs, which measure the cost of the ‘time of ill health’ of
the patients (and possibly their informal caregivers) should also be assessed
because unwell individuals are no longer partially or completely ‘produc-
tive’ in a broad sense (see Drummond et al., 2015; Drummond & McGuire,
2002).
The traditional approach called the human capital approach (HCA) is
to value productivity losses by the patients using the average gross wage
(including the employer’s tax share and overheads). This has the advantage
of being an easily accessible information source – the only information to
be collected is time out of ’productive work.’ However, for non-employed
individuals (e.g. students, retirees, stay-at-home parents) this amount may
be different but is certainly not equal to zero. For example, the average net
income replacement ratio for retired individuals is generally in the range of
40% to 60% of the average salary in developed countries.

5.6 Distribution of Costs
In this section, we discuss several issues in the statistical modeling of
patient-level cost data. We will first assume non-zero and complete cost data (no
censoring or ‘missingness’) for each patient. These assumptions will then be
156 Economic Evaluation of Cancer Drugs

relaxed and discussed separately to allow situations where missing data is


possible.
It is important to note that the key objective of a cost estimation model in
an economic analysis is to provide estimates of the mean cost, and the differ-
ence in mean costs between treatments is used for cost-effectiveness analysis
(the mean incremental cost), regardless of whether the distribution is skewed
or not. What this implies in practice is that a suitable statistical model needs
to be determined that will allow an estimate of the mean costs for each group
and then calculate the mean incremental cost. There are several features of
cost data that are worth noting:

(i) In general, cost data are strictly positive (C > 0) . It would be dif-
ficult to justify the costs of patients in a clinical trial for a cancer
treatment as ‘true’ zeros. This is because they are likely to have used
some health resource after having been randomized to treatment.
In some non-cancer trials, it may be possible that patients are cured
(e.g. pain relief, reflux) and there may be zero future costs. However,
this could only happen after treatment (which incurs costs).
(ii) Cost data are most likely to be right-skewed (positive skew) and
have no theoretical fixed upper limit. They are termed a ‘leptokur-
tic’ distribution with excess kurtosis (with a tall peak). In some cases
the distribution of costs may even be multimodal (have more than
one peak). The reason for this is because, inevitably, some patients
are more ill than others and therefore usually have high costs due to
higher comorbidities or lengthier hospital stays.
(iii) In clinical trials, there are varying time periods of observing costs,
so that we have repeated measures. However, in general, it is the
total mean cost cumulated over all periods and resource items that
is primarily of interest.
(iv) The mean costs may depend on a number of covariates such as age,
gender, comorbities, cancer stage, baseline health status, etc., which
therefore need to be taken into account when calculating mean incre-
mental costs. For example, if a clinical trial is stratified by ECOG
status, those with poorer performance status (ECOG > 2) may have
different (higher) mean costs compared to those with a better prog-
nosis. These differences could be reflected in the ICER.
(v) There will inevitably exist costs that are missing. This is not the same
as the zero costs discussed earlier. Missing health resource use can
happen due to patient withdrawal from the study because of disease
progression, death, or for other reasons. It is worthwhile taking into
account the burden one is placing on patients for the collection of
health resource as this can contribute toward the amount of missing
data. A commonly used collection tool in the UK is the client ser-
vices receipt inventory (CSRI), which collects health resources over
Collecting and Analysis of Costs from Cancer Studies 157

many aspects of care (e.g. hospital stays, therapists, social services


costs). However, these can be excessive and time-consuming to col-
lect, especially when the incremental differences are likely to be neg-
ligible. Hence, careful thought needs to be given before subjecting
cancer patients to this level of data completion. Excessive amounts
of time and energy expended on cleaning this type of data (as well
as finding the associated unit costs), which is unlikely to have an
impact on the ICER, should be avoided. Medications is a good exam-
ple of this. In a reported database for a non-cancer trial in demen-
tia (e.g. the dementia and physical activity (DAPA) trial; Khan et al.,
2018) the medications ran into over 5,000 individual records, which
took a very long time to clean, yet showed little difference between
treatments in terms of costs and impact on the ICER.

5.6.1 Transforming Cost Data


Often costs are not normally distributed showing a combination of skew-
ness and over-dispersion (e.g. where lots of patients have zero costs or many
patients have a specific high cost), and, therefore, a transformation might be
considered appropriate. One common transformation is logarithmic (natu-
ral logs), when data are positively skewed in order to make the distribution
more normal. Figure 5.1 shows costs that were skewed and transformed
resulting in greater normality. Note in Figure 5.1 how the numbers (scale)
change with a quite marked impact on the high costs. Some special statisti-
cal tests exists to check the degree of non-normality in the data. Some com-
monly used tests are the Kolmogorov-Smirnov, the Shapiro-Wilks (SW), or
Shapiro-Francia (SF) tests. For the data in Figure 5.1a, normality is rejected
(the data are not normal) prior to transformation; after the transformation in
Figure 5.1b, especially the square root transformation, the data are now more
normal.
An important consideration in transformation is when costs are zero. Zero
costs cannot be transformed using a logarithmic function. An arbitrary small
constant (e.g. 0.01) is sometimes added to whenever a zero cost occurs, as this
allows a log transformation. However, this could be misleading when com-
puting mean costs because the logarithm of very small numbers is negative.
For example, log(0.001) = –6.91 and hence causes the mean costs to be lower.
A second important consideration is that after logarithmic transforma-
tion, the mean after logging values is called the ‘geometric mean.’ It is well
known that this value is in fact an estimate of the median and not the arith-
metic mean. The variance too, after log transformation, is not an unbiased
estimate of the variance of the original cost data prior to transformation.
This is important for interpreting incremental costs on the original scale.
Hence, for this reason, statistical models for costs analyses might be prefer-
able to the use of a transformation for estimating the mean incremental cost.
Some cumbersome corrections can be used to derive the unbiased estimates
158 Economic Evaluation of Cancer Drugs

FIGURE 5.1
Example of transformation of a gamma distribution before and after square root and logarith-
mic transformation.

of mean costs (after transformation). A so-called ‘smearing factor’ was pro-


posed by Duan (1983) for when the treatment groups have a common vari-
ance; Khan (2015) gives details of other transformations.

5.7 Handling Censored and Missing Costs


We distinguish between two types of missing data:

(i) Missing in the sense that the patient missed the visit, or a page from
the case report form (CRF) was lost or the health resource was used,
but the quantity was unknown (for example, the date of hospital
admission was recorded in the CRF, but the duration of stay was
unknown). Another example might be where a health resource use
page from the CRF is lost, but the data lost are similar between treat-
ment groups. An important issue for this type of missing data is that
missing is not the same as zero cost; and a resource use set to zero is
not the same as saying the resource use is missing
Collecting and Analysis of Costs from Cancer Studies 159

(ii) An incomplete record of health resource use because the patient was
either lost to follow-up or the study was ‘complete’ before the pri-
mary outcome was observed. This type of ‘missing’ is known as cen-
sored data – censored at the last date of contact. In this case, the total
costs for that patient represent a minimum (because, if followed up,
the costs might turn out to be larger if the reason why they dropped
out was because they were more ill). Consequently, calculating a
simple mean cost would be biased. It has been suggested (Willan &
Briggs, 2006) that patients who are censored are likely to have lower
costs. This will impact the mean costs across patients.

One reason for missing resource use (cost) data is when patients withdraw
from the trial early, due to lack of effect, adverse events, early death (due
to a competing event), or when the study has ended before the outcome of
interest can be observed. Missing data are problematic in any clinical trial
when this occurs, whether the outcome is resource use or any other clinical
endpoint, and it is more likely to occur in clinical trials where the primary
outcome of interest can occur several years after randomization (e.g. survival
endpoints). There is often little or no plan to collect any additional data after
study withdrawal or adverse events.
Missing data of type (i) can be handled through well-established analysis
methods such as multiple imputation and other complicated approaches (e.g.
shared parameter models) involved in testing assumptions about the mecha-
nism of missingness, such as: missing completely at random (MCAR), missing
at random (MAR) and missing not at random (MNAR). A detailed discussion
of these would require a separate volume, and so they are not elaborated on
further here. References in the bibliography can be consulted for interested
readers (e.g. Carpenter & Kenward, 2013; Farclough, 2010; Van Buuren, S. (2018).
F). Most literature on missing data in economic evaluation tends to focus on
censored costs. The focus here is less about estimating missing health resource
(e.g. number of GP visits for a particular patient), but on the monetary value
itself of the missing resource. Therefore, attention will be paid to missing data
of type (ii) for the purposes of deriving the incremental costs.
In many clinical trials the ITT population is often the primary popula-
tion for analysis. This means that regardless of any violations, compliance,
or early withdrawal from the trial, if the patient was randomized, their data
should be analyzed. If data are missing at some point after randomization, or
midway through treatment, strategies are needed to deal with how to handle
such missing data. What makes this issue particularly relevant to the analy-
sis of costs is that patients who have missing data might well be the same
patients who also have higher costs (because the missing values might be
associated with problems with side effects and ultimately the treatment).
Patients who drop out from the trial due to toxicity from the experimental
medicine may go on to receive additional medication or treatment for their
adverse events. If the patient has been lost to follow-up then the costs are likely
160 Economic Evaluation of Cancer Drugs

to be underestimated. Although it is difficult for patients to recall their pain


response over the last 7 days, the patient is less likely to forget they went to
visit a GP or saw a nurse. Therefore, if the CRF was ticked as ‘Yes’ to a question
regarding whether a GP visit had been carried out, but the date of the visit was
missing, this would not necessarily be considered a missing data problem.

5.7.1 Strategies for Avoiding Missing Resource Data


There are several approaches to preventing missing or censored cost data:

• Design clear and well understood CRFs, with clear instructions,


when collecting resource use.
• Think carefully whether assessment time points are adequate.
Assessments that are too far apart, for example 1 month and 6
months, may result in costs being missed between these months;
the recall period may also be too long. For example, a question at
6 months “Did you have any other medication for your pain since
your last assessment?” requires the patient to remember everything
that happened between the first and sixth month.
• Continue to collect all or some data after the patient has discontin-
ued from the study or has discontinued from the trial (if possible).
• Select outcomes that are easily ascertainable.
• Use those clinical sites that have good experience with following up
(not just the ability to recruit).
• Train trial staff on how to communicate with the site staff so that
follow-up data can be obtained. It is not helpful when a trial coordi-
nator or clinical research associate (CRA) demands (possibly rudely)
that site staff obtain the missing data. There would not be much moti-
vation to obtain follow-up data when communication is a problem.
• Pay site investigators for the quality of follow-up and the complete-
ness of CRF data or use other types of incentives.
• Inform patients why collecting resource use data is important:
because it will help determine whether the new experimental treat-
ment offers value for future patients (this may not go down too well
with all patients!)

5.7.2 Strategies for Analyzing Cost Data When


Data Are Missing or Censored
Several analysis strategies can be suggested if data are missing or censored.

(a) Complete case analysis


(b) Imputation methods
Collecting and Analysis of Costs from Cancer Studies 161

(c) Model-based methods


(d) Complete case analysis

When there are complete cost data, any method can be used to analyse the
costs. When patients who do not have complete cost data are excluded from
the analysis, not only would this violate the ITT principle, but would lead
to biased estimates of mean costs (Huang, 2009; Wijeysundera et al., 2012).
Although data from patients who are censored are omitted, leading to loss
of information, the associated loss of power for statistical inference is less of
a practical concern because differences in mean costs are not powered for
statistical significance. What is more important is the resulting bias in the
estimate of the mean and standard errors of the incremental cost.

5.7.3 Imputation Methods
Single imputation methods such as last observation carried forward (LOCF)
should generally be avoided. Just as in clinical endpoints, when the disease
severity deteriorates over time, the LOCF is not realistic; similarly, with cost
data, as the disease progresses the costs might increase over time. Other
imputations, such as using the mean costs, worst case, or baseline carried
forward, sometimes used for clinical endpoints (and suggested in some
clinical regulatory guidelines, EMA, 2012) also result in biased estimates of
mean costs and are not appropriate for costs. For example, the worst case for
a clinical endpoint involving pain on a scale of 0 to 10, might be 10; but the
‘worst’ or ‘maximum’ cost could be anything; similarly, baseline carried for-
ward might be a way to determine a conservative treatment effect, but using
baseline costs carried forward can result in lower mean costs, because as the
disease progresses, costs are likely to increase.
Multiple imputation methods on the other hand might be suitable for han-
dling missing data of type (i) above (Section 5.7) to improve estimates of the
mean cost. Whereas the single imputation approaches are effectively guesses
for the data that are missing, multiple imputation is a slightly more compli-
cated method that results in a 'better' estimate of the missing data compared
with the above methods (e.g. such as LOCF). These methods are discussed
extensively elsewhere (Rubin, 2004; Carpenter & Kenward, 2013). An impor-
tant point concerning multiple imputation is that it is important to state what
is being imputed from where. For example, it makes no sense to impute miss-
ing costs from the control arm for the experimental arm. In addition, patients
who dropped out are likely to be different in some way to those who remain
on the trial and have data. Hence imputing costs from a group who did not
drop out for those who did (even if on the same arm) is likely to lead to
biased estimates of mean costs. To generate the mean incremental cost when
using this approach, an imputation model is assumed and the missing costs
are predicted several times (meaning several complete data sets are gener-
ated). Consequently, the mean cost is determined for each treatment group
162 Economic Evaluation of Cancer Drugs

for each of the data sets. Finally, algorithms are used to derive the final point
estimates of mean costs for each treatment group and the mean incremental
cost is derived.

5.8 Handling Future Costs


Future costs related to the intervention are always to be included in a cost-
effectiveness analysis. An example of a future-related cost might be antici-
pated longer-term costs of side effects from radiotherapy and the consequent
costs of treating them. Depending on the perspective one takes, such costs
may include medical or healthcare system costs and even wider societal
costs (e.g. direct out-of-pocket costs sustained by the patient, informal care,
productivity loss). Unrelated medical and non-medical costs can be ignored
as these can be assumed to be similar between treatment groups.
In cancer trials where overall survival is often the primary outcome, and
patients survive longer in the experimental arm, future costs may also accrue
for a longer period since an increased mean life span for a number of indi-
viduals over and above that of the control arm is likely to be a direct conse-
quence of experimental intervention, and such future costs should therefore
be included. For example, if patients incur non-cancer-related medical costs
after 33 months (say for arthritis) one should also take these into account
because these are directly linked to the patients living longer. Some others
(Weinstein & Fineberg, 1980; Weinstein & Manning, 1997; Garber and Phelps,
1997) take an opposing view. This is a complex decision-making problem
because it seems contradictory: on the one hand, a new groundbreaking can-
cer treatment is efficacious and offers a longer survival benefit, while on the
other hand, for patients with comorbidities, the associated costs of treating
the comorbidities are increased resulting in more expensive treatment that
may possibly render the new treatment to lack cost-effectiveness, because it
is ‘so good.’
To date, there seems to be no clear-cut answer or consensus on how to
handle this problem, and often the choice is left to the health economist.
One (simple) way to incorporate these costs is to add gender-age average
overall population-wide medical costs (eventually corrected for cancer costs)
from national health expenditure data. In fact, this aspect would perhaps be
more important for budget impact predictions (a budget impact relates to the
affordability of new treatment by a health system) than for trial-based cost-
effectiveness analysis (for a discussion see Drummond & McGuire, 2001;
Drummond & Sculpher, 2015).
A further complexity is the definition of cancer-related and non-cancer-
related (future) costs. Cancer treatments have numerous, sometimes long-
lasting side effects that impact other organs (e.g. central and peripheral
Collecting and Analysis of Costs from Cancer Studies 163

neuropathologies, cardiac toxicity, dermatology) where the causality cannot


always be clearly established, even during the trial observation period, let
alone beyond the trial.

Example 5.6: Simple Extrapolation of Costs


A common situation occurs where at the end of the trial there are
patients still alive. In this case one would need to extrapolate the long-
term survival after the trial ends, together with the corresponding costs,
until the last surviving patient dies. For example, assume a trial with a
follow-up duration of 2 years and a maximum survival of 3 years in the
control arm, and 5 years in the experimental arm (after extrapolation
of the survival curves). During the trial the cancer treatment costs are
collected (see Table 5.5); the non-cancer medical expenses are however
considered irrelevant and are therefore set to zero. The new treatment
is more expensive partly due to drug costs and partly due to increased
toxicity, but it yields improved survival for some patients.
At the end of the trial (post-trial), the average (estimated) annual medi-
cal cancer care cost per surviving patient is equal to €50 in the control
arm but somewhat higher (€60) in the experimental arm because of long-
term treatment adverse events. This cost is a mix of care and follow-up
for patients with no sequelae, for patients with sequelae needing more
intensive follow-up, and for patients who die in that interval (year) and
therefore receive possibly a further treatment line and palliative care.
In the year following the end of the trial, non-cancer costs are still lim-
ited in both arms. Since the experimental arm patients survive longer
and are therefore aging further, their care costs for other comorbidities
increase as well. Hence, the total lifetime costs are due to the complex
interplay between the proportion of patients surviving over time, until
all are deceased. The time profile of their medical care costs follows a
typical bathtub (U-shaped) profile (i.e. high cost during active therapy
at the start, decreasing after the end of the treatment, increasing slowly
with age, and finally increasing rapidly in the last year of life.

TABLE 5.5
Example of Costs During and After the Trial
Period Year Cancer-Related Costs (€) Non-Cancer-Related Costs (€)
Control Experimental Control Experimental
During trial Year 1 100 120 0 (NC) 0 (NC)
During trial Year 2 80 100 0 (NC) 0 (NC)
Post-trial Year 3 (50 ) (60) (20) (20)
Post-trial Year 4 0 (60) 0 (30)
Post-trial Year 5 0 (60) 0 (40)
Post-trial Year 6 0 0 0 0
Notes:  ( ) = estimated amount; NC = not collected.
164 Economic Evaluation of Cancer Drugs

In this simplified example, we see that the per patient cumulative trial
cancer treatment costs are respectively equal to €180 and €200 but over
the lifetime can be substantially higher (€250 for control vs. €490 for
experimental) because patients who survive longer incur both medical
costs for their cancer and for their other comorbidities, and this would
possibly change the ICER (Faria, 2014; Clement, 2009). This was observed
in the recent study (Olchanski et al., 2015) who reported that by includ-
ing all medical costs, that is, cancer-related medical costs plus cancer-
unrelated medical costs, the ICER drastically increases (i.e. worsens) and
may reverse the cost-effectiveness decision. Therefore, inclusion of unre-
lated medical costs during added years of life may “implicitly penalize
therapies that add expensive life years.”

5.9 Case Report Forms and Health Resource Use


An important aspect of cost analysis is data collection and how we collect
data for health resource use. In clinical trials the case report form (CRF)
is a critical document that is reviewed by several professionals to ensure
that clinical data can be collected accurately. This is no less true of CRFs
designed for health resource use. In this section we give several examples of
CRFs that can be used in clinical trials. These CRF designs are examples and
modifications can be made to accommodate specific trial needs. One impor-
tant consideration when collecting resource use data in a clinical trial are
unscheduled visits, such as additional GP visits. Not all unscheduled visits
are direct treatment costs. For example, a repeat visit for a laboratory test is
not necessarily a direct treatment cost. Therefore, a distinction needs to be
made between collecting the resource use associated with treating patients
and the costs of running the clinical trial. Where adverse events data are col-
lected for reporting incidence rates, the treatment emergent adverse events
(TEAE) are more important because these again are associated directly with
costs of treatment (these adverse events emerge after treatment has been
taken). Figures 5.2 through 5.5.

FIGURE 5.2
Example CRF from a lung cancer trial.
Collecting and Analysis of Costs from Cancer Studies 165

FIGURE 5.3
Example CRF for a general cancer trial.

5.10 Statistical Analyses of Costs


Statistical analyses of costs data can be determined using a variety of meth-
ods, from simple univariate (i.e. just calculating the total mean costs for each
group using complete cases), to complex model-based methods. In cancer
trials, it is important to take into account the full trial design features so
that estimates of mean incremental costs take into account features such as
heterogeneity, over-dispersion, censoring, and other features (e.g. repeated
measures or clustering, where each patient has costs at several time points).
In general, the types of statistical analyses of patient-level costs may take the
shape of the following statistical models:

(i) Linear regression (ordinary least squares, OLS)


(ii) Generalized linear models (GLMs) that allow one to specify the dis-
tributions of costs as:
a. Gamma distributed for skewed data (or log-normal)
166 Economic Evaluation of Cancer Drugs

FIGURE 5.4
Example CRF from a sarcoma cancer trial.

b. Poisson or negative binomially distributed where there is over-


dispersion (for count data such as number of hospital visits)
(iii) Using models (i) and (ii) above, but also including a random subject
effect, where the difference between each patient’s individual cost
(effect) and some average cost (e.g. average of all patients at one site)
is taken into account in the analyses (often known as mixed effects
models)
(iv) Two-part models (such as hurdle models) that first model the chance
of zero resource use, and then use a second model that computes the
expected (mean) intensity of the resource use.
Collecting and Analysis of Costs from Cancer Studies 167

A. PATIENT ACCOMODATION
1. Usual place of residence Owner occupied house/flat
during the last three months?
Privately rented house/flat
House/flat rented from housing associated/local authority
Sheltered housing/warden control
Extra care housing
Care home providing nursing care
Care home providing care
Dual Registered home (providing both personal and nursing care)
Acute psychiatric ward
Rehabilitaon ward
General medical ward
Other:
2a. Has the parcipant lived anywhere else in the last three months? Yes (Go to Q2b)

No (Go to Q3a)

2b. Please state the approximate No. of nights


number of nights spent in this
Owner occupied house/flat
accommodaon in the last
three months: Privately rented house/flat
House/flat rented from housing associated/local authority
Sheltered housing/warden control
Extra care housing
Care home providing nursing care
Care home providing personal care
Dual Registered home (providing both personal and nursing care)
General medical ward
Rehabilitaon ward
Acute psychiatric ward
Other:
3a. Has the individual been in receipt of direct payments during the last three months? Yes
No
3b. If yes please state the total
weekly value £ .

4a. Has the individual been in receipt of individual budgets during the last three months? Yes
No

FIGURE 5.5
Extract of CSRI inventory of broader societal health resource use.

(v) Models used to estimate the mean health resource use (and not the
costs). Once the mean health resource use is estimated by using, for
example, two-part hurdle models, the unit costs can be applied to
estimate the costs. This is different from modeling derived costs.
168 Economic Evaluation of Cancer Drugs

Example 5.5: OLS Model after a Square Root Transformation


In this example, a simple regression model of the form:

Cij = m + b * Treatment j + eij

where Cij is the cost for patient i on treatment j, μ is the common intercept
(the overall mean ignoring treatment) and εij is the error term (the dif-
ference between each patient’s cost and the overall mean cost for a given
treatment); and β is the rate of increase (or decrease) in costs.
For the purposes of this example, an OLS model is used after a square
root transformation. The square root transformation involves taking the
square root of costs first and then analyzing the data. Since costs are
positively skewed, then this transformation can work well. A log trans-
formation would have problems with costs equal to zero whereas the
square root transformation does not (the square root of zero is zero).
Table 5.6 gives the mean cost difference between the two groups equal
to 1.02 (4.00 vs. 2.97) after a square root transformation. After a back
transformation (by squaring), the mean costs are 16.01 and 8.88 respec-
tively or a mean incremental cost of 7.13 (17.37 vs. 10.05) between the two
groups. On the raw, non-transformed scale, this mean difference was
7.32, which is quite close to 7.13.

Example 5.6: Modeling Costs with and without


Covariates (Normally Distributed)
The following example of 48 patients with patient-level total costs is used
to generate the mean incremental costs before and after adjusting for age
and country effects. In this example only costs are modeled (ignoring
effects). The INMB is estimated using a suitable value of λ, the willing-
ness to pay, in order to generate the base case estimate of the INMB. If the
correlation between costs and effects is also included (modeled), a more
complicated bivariate model (two response variables) will be required
(see Table 5.7 and 5.8).
TABLE 5.6
Results of Mean Costs after Transformation (Square Root)
Mean estimation Number of obs = 2,000

0: group = 0
1: group = 1

Over Mean Std. Err. [95% Conf. Interval]

gamma_all
0 10.05558 .2259303 9.612495 10.49866
1 17.37788 .3107348 16.76848 17.98728

sqrtgamma
0 2.979869 .0343094 2.912583 3.047155
1 4.001552 .0369706 3.929047 4.074057
TABLE 5.7
Data for Example 5.6
Patient Treatment Country Age Cost (£) Patient Treatment Country Age Cost (£)
1 A UK 34 5,268 25 B Germany 63 8,126
2 A UK 26 1,535 26 B Germany 34 6,535
3 B UK 58 8,261 27 A Germany 30 12,111
4 A UK 64 526 28 A Germany 59 4,236
5 A UK 44 3,126 29 B Germany 54 7,126
6 A UK 32 2,671 30 B Germany 71 7,671
7 B UK 66 4,319 31 A Italy 66 4,151
8 A UK 42 2,111 32 A Italy 34 4,213
9 B UK 34 8,881 33 B Italy 31 5,481
10 B UK 68 9,123 34 B Italy 54 3,923
11 A France 74 18,146 35 A Italy 78 22,146
12 A France 59 5,111 36 A Italy 34 25,111
13 B France 34 5,998 37 A Italy 57 8,199
14 B France 62 4,129 38 B Italy 34 12,129
15 A France 48 3,112 39 A Italy 32 13,199
Collecting and Analysis of Costs from Cancer Studies

16 A France 61 2,189 40 B Italy 34 23,489


17 B France 34 3,777 41 A Spain 57 13,797
18 B France 34 6,453 42 B Spain 58 16,111
19 A France 29 7,268 43 A Spain 33 7,658
20 A France 44 9,121 29 B Spain 44 9,311
21 B Germany 34 4,321 45 B Spain 79 1,321
22 B Germany 61 4,139 46 B Spain 35 1,590
23 A Germany 34 5,912 47 B Spain 69 12,917
24 A Germany 34 3,998 48 B Spain 34 9,654
169
170 Economic Evaluation of Cancer Drugs

TABLE 5.8
Summary Output for Example 5.6
Model LS Mean Incremental Cost 95% CI p-Value
Without Covariates A 7,705 6a (–3356, 3367) 0.997
B 7,699
With Covariates LS Meanb Incremental Cost 95% CI p-Value
A 7,844 180a (–3090, 3450) 0.912
B 7,664
Age 0.596
Country 0.029
Country*Treatment 0.549
a Notes:  Difference between A versus B: treatment A has higher mean costs.
b After adjusting for covariates (age p-Value = 0.596; country p-Value = 0.029).

TABLE 5.9
Summary Treatment by Country Mean Costs
Country Treatment A Cost (£) Treatment B Cost (£)
France 7,491 5,089
Germany 6,564 6,320
Italy 12,837 11,256
Spain 10,728 8,484
UK 2,540 7,646

Interpretation:

• The mean incremental cost is £6 without accounting for covari-


ates and increases substantially to £180 when accounting for
age and country.
• Age is not associated with higher (or lower) mean costs, but
country is associated with costs (i.e. there are significant differ-
ences in mean costs between countries). The treatment*country
interaction p-value is 0.549, suggesting that on the whole (across
all countries) the incremental cost does not differ significantly.
• We should be careful about the interaction terms, because tri-
als are often not powered to detect interactions (for clinical
effects, let alone costs), especially for a sample size as small as
this. Table 5.9 shows the country-specific mean costs for each
treatment. There is large variability in incremental costs, which
partly explains the lack of significance; in addition, in the UK
the costs of treatment B are much higher than those for treat-
ment A. Local payers (e.g. NICE) may be more interested in this
aspect of incremental cost than the overall much smaller incre-
mental cost. The mean differences in costs between treatment
in Germany are smaller (£244).
Collecting and Analysis of Costs from Cancer Studies 171

Example 5.7: Missing Costs due to Censoring


Method of Lin et al. (1997)
When patients drop out of the trial due to disease progression (or other
reasons), the full costs are unknown because the patients’ survival (or
follow-up) times are censored. For patients who have died, future costs
are sometimes set to zero, which would bias estimates of mean incre-
mental costs. Several methods are available to take into account censor-
ing for estimating costs. One method is Lin’s method (Lin et al., 1997).
The method shown in this example provides an estimate of the mean
cost by taking into account patients who are followed up to a particu-
lar time point and then are lost to follow-up (censored). The estimate
of mean costs uses the survivor function (Kaplan-Meier) to generate
weights that are multiplied by the mean costs for specified intervals. For
endpoints that do not have a mortality endpoint, the event of interest
might be time-to-dropout. In a 12-week study where mortality is not the
primary endpoint of study, almost all patients are likely to remain in the
study and most patients would be censored.
The calculations for mean costs can be broken down into the following
steps:

(i) Divide the follow-up period into smaller (not necessarily equal)
time intervals. For example a follow-up of 1 year could be split
into 12 equal intervals (1 month apart).
(ii) Calculate the mean costs within each interval (i.e. for each of
the 1-month intervals) only for those patients alive at the start
of the interval. For example if 100 patients are alive at the start
of month 1, then the mean 1-month cost will be determined for
the 100 patients.
(iii) Compute the survival rates at each interval (every month in this
example) using survival methods (typically a Kaplan-Meier
plot). For example, the OS rates at 1 month might be 90% (after
1 month, 10% have died).
(iv) Calculate the expected monthly cost by multiplying the
monthly survival rates by the monthly costs.
(v) Add up the mean costs for each of the 12 months (i.e. month 1 +
month 2 + … month 12) to get the total mean costs (total of the
means).
(vi) Repeat for each treatment group.

One limitation of this method is that it does not adjust for covariates
when estimating the mean cost. However, Lin (Lin, 2000) provides an
extension of this that can adjust for covariates.
Table 5.10 shows a table of costs and mean costs calculated for each
treatment over a 12-month follow-up period using the Lin (1997)
approach. Firstly, we split the 12 months into 2-monthly intervals and
derive the costs (treatment costs, nurse visits, etc.). One might think of
each interval as a cycle of 8 weeks (roughly 2 months) of treatment for
a particular cancer corresponding to months [0–2], [2–4], [4–6], [6–8],
[8–10], and [10–12].
172 Economic Evaluation of Cancer Drugs

TABLE 5.10
Adjusting for Censored Costs
Interval
Patient Treatment 1 [0–2] 2 [2–4] 3 [4–6] 4 [6–8] 5 [8–10] 6 [10–12]
1a A 5,000 3,000 4,000 7,000 6,000 5,000
2 A 7,000 6,000 2,000 300 0 0
3 Etc. A Etc. Etc. Etc. Etc. Etc. Etc.
51b B 7,000 2,000 0 0 0 0
52 B 5,000 3,000 4,000 8,000 4,000 0
53 Etc. B Etc. Etc. Etc. Etc. Etc. Etc.
Mean B 5,300 3,900 1,200 4,650 6,170 3,195
S(t)A A 90% 85% 80% 40% 20% 10%
S(t)B B 90% 75% 65% 20% 5% 2%
a Patient censored.
b Notes: Patients 2 and 51 died during interval 4 and 2 respectively.

In Table 5.10, for each interval, the mean costs are computed taking into
account whether patients have complete cost data in the interval. Cost
calculations are based on patients alive at the beginning of the interval.
The survival rates are obtained by plotting the survival times (assum-
ing the endpoint is mortality) and using the observed product limit
estimates (i.e. KM estimates). For each group, the cumulative (i.e. total)
mean cost for treatment A is computed as (£6,300 × 0.9) + (£4,900 × 0.85) +
(£4,200 × 0.8) + (£3,650 × 0.4) + (£4,520 × 0.2) + (£5,450 × 0.1) = £12,744. The
same is then performed for treatment B; and then the incremental cost
can be calculated. Note that this method is based on patients alive at the
beginning of the interval. An alternative method is also available (see
Lin, 1997, method 2), which computes the mean costs of those patients
who die within an interval and then multiplies these costs with the prob-
ability of death within the interval (from the KM survival estimates).
Adjustment for covariates when estimating mean costs in the interval is
also possible (Lin, 2000). Whatever method is used, care should be taken
to check the assumption that censoring within the interval is not some-
how systematically related to treatment groups.

5.11 Summary
Chapter 5 described the different costs that need to be tracked and how
the analysis perspective drives their choice. We also described the type
of costs in relation to the treatment pathway and showed some examples.
Collecting and Analysis of Costs from Cancer Studies 173

Micro-costing (bottom-up) and macro-costing (top-down) cost collection


methods were discussed and compared as well as the difference between
cost and charges. A number of statistical features of cost data were described
such as distributions, transformations, and the handling of censored and
missing cost data. We also discussed some analysis methods and strategies
to minimize missingness. The handling of future costs and their extrapola-
tion beyond the trial horizon were introduced, and, finally, we presented
examples of economic case report forms (CRFs) that can be used for collect-
ing data on health resource use in cancer trials.

5.12 Exercises for Chapter 5


1. Distinguish between the types of costs one might expect in a cancer
trial. How different might they be compared to a trial in epilepsy?
2. What are the distinct stages of collecting health resource use in a
cancer trial?
3. Explain the difference between inflation and discounting.
4. Design a case report form for collecting health resource use data in
a lymphoma trial.
5. In a cancer trial, health resource use data were observed to be heav-
ily skewed with censored and missing data. Distinguish between
censored and missing data; and discuss how you might analyse
such data.
6
Designing Cost-Effectiveness
into Cancer Trials

6.1 Introduction and Reasons for Collecting


Economic Data in a Clinical Trial
There are several reasons for designing the cost-effectiveness of a clinical trial
prospectively. First, it is often cheaper and easier to collect health resource
use data alongside a clinical trial rather than having to design separate ret-
rospective studies of costs and effects (Drummond 1993). In some cancer tri-
als where the follow-up period can last for many years, it may be costly to
follow patients extensively (e.g. until death) to collect relevant data. Delaying
publication of important clinical findings on the efficacy of a new treatment
in order to collect additional ‘real-world evidence’ for cost-effectiveness may
also not be considered ethical.
Second, several reimbursement agencies require economic data to be col-
lected alongside clinical trials to demonstrate value of new treatments. With
several new treatments given market authorization showing ‘small’ or ‘mod-
est’ treatment effects, the burden of proof is on the claimant to demonstrate
why any healthcare system should finance one treatment over the other. In the
UK, NICE makes specific reference to clinical trial data being used in assess-
ing cost-effectiveness. When government agencies make specific requests,
especially where it concerns payments for the provision of new drugs, such
requests are not to be taken lightly – which might alone justify why designing
the economic component carefully in a clinical trial is important.
In Chapter 1, it was shown how an early decision to reimburse a new treat-
ment can influence future revenue. Reimbursement authorities may request
economic evaluation of new cancer treatments closer to (and sometimes
before) a decision is made for licensing. Some reimbursement agencies may
prefer to have an early review of the value argument (submission dossier) put
forward. This may be in the interests of both the sponsor and the patients.
In the UK, the early access to medicines scheme (EAMS) gives a promis-
ing innovative medicine (PIM) the possibility to have an accelerated route
to licensing (see Medicines and Healthcare products Regulatory Agency

175
176 Economic Evaluation of Cancer Drugs

(MHRA) website for details). Similar schemes may exist in other European
countries. Any delay in a recommendation from reimbursement authorities
can unnecessarily restrict access to treatments by patients and healthcare
professionals – despite receiving market authorization. This is not entirely
the same situation in all countries. For example, in Germany, a period of
free pricing is offered where the manufacturer/pharmaceutical company is
offered an agreed price for the new treatment, while value is assessed.
The economic value of a new treatment, however important, is unlikely to
trump the clinical reasons for patient access to it – either privately or through
the local health system. Many HTAs are conducted after marketing authori-
zation has been provided (although pharmaceutical companies may generate
evidence for both efficacy and effectiveness in tandem). It is possible that the
future relationship between the regulatory and reimbursement procedures
may be altered to take into account the overlap and (sometimes unnecessary)
duplication that takes place. In several HTAs there have been concerns from
manufacturers that questions raised by payers belong to the domain of licens-
ing authorities (there is no reason why this should be case). Some of these ques-
tions had no apparent consequences on licensing during the initial assessment
only to be raised later by reimbursement agencies For example, in the HTA
review of ofatumumbab for chronic lymphocytic leukemia by NICE (NICE,
HTA TA202, 2010) it appeared that evidence review experts were critical as to
why interim OS results were not made available, not appreciating that report-
ing unplanned interim analyses has huge implications for bias and future
trial conduct. The potential for contradictory advice from two agencies whose
objectives are different is of concern. If, for example, NICE offer advice on trial
design that might compromise licensing, this is clearly a cause for concern and
government agencies need to align more closely to avoid such situations.
In academic clinical trials, where licensing is not relevant, reasons for collect-
ing economic data are associated with evaluating the impact of the proposed
health technology from the National Health Service (NHS) and government
policy perspective. In some grant award application forms, there is a desire to
know from a funder perspective (e.g. National Institute for Health Research
(NIHR), Cancer Research UK (CRUK) whether the planned trial will impact
future national healthcare resource use. The new health technology proposed
need not be a new drug. For example, in a grant application, a new treatment
might propose to compare patients followed up intensively for tumor pro-
gression (e.g. >4-monthly scans) compared to patients with a less intensive
follow-up schedule (3-monthly scans). The main objective might be to deter-
mine whether more intensive follow-up results in the earlier detection of dis-
ease progression. More intensive follow-up may lead to more efficient health
resource use in the long term compared to less intensive follow-up. Treatments
are not compared in this type of trial (both take the same treatment): the effects
of differences in PFS between the follow-up schedules is compared.
A further reason for collecting economic (health resource) data in a clini-
cal trial is because clinical trials provide strong internal validity of estimates
Designing Cost-Effectiveness into Cancer Trials 177

of average costs and effects, although they may lack external validity.
Additional evidence is sometimes needed to make informed decisions about
the cost-effectiveness of new treatments, particularly in a real-world setting
(Sculpher, 2006). If a decision cannot be reached regarding the cost-effec-
tiveness of a new treatment based on data from one or more clinical trials,
the impetus for collecting economic data loses some force. If the ‘weight of
evidence’ from clinical trial data is considered too ‘weak’ to offer a robust
conclusion for cost-effectiveness, then it becomes debatable whether the eco-
nomic data collected alongside a clinical trial adds to a demonstration of
the value of a new treatment. Gheorghe et al (2015) suggest operationaliz-
ing the idea of generalizability and incorporate it into trial design – such
as trials designed for the real-world setting – with a view to getting market
authorization. It may be argued that despite the lack of external validity in
RCTs, they remain the optimal recognized framework to derive unbiased, or
at least less biased, estimates of treatment effects.
Evidence from nonrandomized trials (single arm Phase II trials) has
been acceptable in certain settings such as unmet needs or rare tumors (the
licensing of gefitinib is an example in NSCLC). Therefore, it could be pos-
sible to use nonrandomized evidence as a basis for a cost-effectiveness deci-
sion. This might involve combining data from the trial with external data.
The question of mixing data from controlled and noncontrolled sources to
provide unbiased estimates of treatment effects is a challenging one. Cost-
effectiveness analysis is not necessarily an inferential problem like that of
efficacy analyses in a regulatory setting, where there is a need to provide
an unbiased estimate of effect; it is a decision problem with an objective of
reducing uncertainty with more information (external and internal) around
measures of effectiveness and value.
For example, if we wish to carry out an economic evaluation comparing a
new treatment with a standard, it is not straightforward to extract costs and
HRQoL effects from published sources and combine this data with clinical
trial data (efficacy data) as inputs into an economic model. Costs and effects
from randomized and nonrandomized trials will be handled (combined)
separately by trial type; the nonrandomized evidence may be pooled with
the randomized evidence for sensitivity analyses in some situations where
there is limited data, using perhaps some special meta-analyses methods
(e.g. network meta-analyses).
Some of the above issues relate to the magnitude of the differences between
clinical trial and nonclinical trial conclusions. (Kunz et al., 2007) conclude:

On average, non-randomized trials and randomized trials with inade-


quate concealment of allocation tend to result in larger estimates of effect
than randomized trials with adequately concealed allocation. However,
it is not generally possible to predict the magnitude, or even the direc-
tion, of possible selection biases and consequent distortions of treatment
effects.
178 Economic Evaluation of Cancer Drugs

There are differences in the way costs (resource use) are collected in a study
that is nonrandomized compared to a clinical trial (Hltaky, 2002). For example,
side effects on a placebo arm in a randomized trial may not translate into real
costs in practice. It is not unusual to collect data on resource use that are of
highest monetary value (bias), or that are likely to show differences between
treatment groups. Not every item of health resource use in a trial is likely to
be captured and hence data collection forms (case report forms (CRFs)) are
often designed to collect selected treatment-related health resource use data.
One important reason for collecting economic data in a clinical trial is
because it is considered unethical to conduct a trial purely for the purposes
of demonstrating value for money (although arguably, it is also unethical to
waste resource where it could be put to better use elsewhere). If bias is to be
minimized, then the RCT framework may still be the only framework that
accommodates both efficacy and effectiveness. The idea of a balance between
internal and external validity was proposed earlier, (Drummond, 1993).
More than twenty years later the argument has somewhat shifted from the
idea that a pragmatic clinical trial with minimal inclusion/exclusion might be
acceptable for showing cost-effectiveness, to a situation where a single clini-
cal trial may no longer be admissible for demonstrating the value argument
(see for example Sculpher, 2006). We shall see in Examples 6.1 and 6.2, that
evidence from smaller trials can be used to demonstrate value arguments.
Combining clinical trial data with ‘other’ evidence to demonstrate value
using complex methods (with not always realistic assumptions) appears to
be one way of showing the value of new treatments through combining evi-
dence and cross-trial comparisons. If researchers are led to believe that the
RCT framework does not have ‘enough’ external validity for a conclusive
reimbursement decision, then the impetus for collecting economic data may
be lost in the trial. This puts control of the value argument into the hands of
the payers and not the manufacturer. A further question here is how internal
or external validity can be quantified. When is a lack of external validity so
serious that a cost-effectiveness decision no longer becomes valid?
It remains important to design a clinical trial for cost-effectiveness in addi-
tion to efficacy, noting the lack of external validity from RCTs. We discuss
some of these trial design aspects below.

6.2 Clinical Trial Designs for Cancer Studies


6.2.1 Clinical Trial Designs

(i) Parallel Groups and Crossover Designs

Where an economic evaluation has been or is required for a clinical trial, some
knowledge of trial design is useful. In general, cancer trials are designed to
Designing Cost-Effectiveness into Cancer Trials 179

compare the effects of treatments in different groups of patients with the


same condition. In a randomized parallel group design, two (or more) inde-
pendent groups of patients receive experimental treatment or a control treat-
ment (e.g. the current standard of care). The responses from these patients
are compared between the groups. These designs are typical of cancer trials.
In some situations, a patient may receive both drugs, the experimental and
the control. This is usually a crossover design. Trials with respiratory drugs
for example may involve giving each patient both drugs in two separate
treatment periods. Here, the patient acts as his/her own control (crossover
trial). Treatment effects are determined within patients. Crossover designs
(not to be confused with switching) are very unusual and unsuitable for can-
cer trials for several reasons (e.g. if the outcome is death, some patients can-
not be treated in the second period).
Figure 6.1 shows a parallel and a crossover design. The arguments for and
against crossover trials can be found in Jones (2014).

(ii) Single Arm Phase II Designs

A single arm trial is designed such that a single cohort of patients receive
the same treatment. There is no randomization. The trial is designed with a
comparison against a historical (control) response or survival rate. These are
typically conducted in Phase II trial designs, where preliminary evidence of
efficacy is required, in contrast to Phase III, where confirmatory evidence of
treatment benefit is determined. Variations of this design are:

(i) Fleming’s (single stage) design (Khan, 2012). Here, for example,
20 patients could be treated and a criterion is derived such that if
10 patients or more out of 20 (≥50%) achieve a complete or partial
response (i.e. the tumor has been reduced), then the new treatment is
considered worthy of further investigation. When there are no other
treatments available and the cancer is very rare, it may be possible
to get a license based only on a surrogate outcome such as PFS or
tumor response. If follow-up is too short for OS, economic evalua-
tion will involve the extrapolation of survival data.
(ii) A variation of the single stage design in (i) is a two-stage design. An
example of this is Simon’s two-stage design, which is further classi-
fied as either minimax or optimal design (Khan, 2012). The decision
to either investigate efficacy further (or declare efficacy) is conducted
in two stages. As an example, a sample size could be required to
demonstrate a difference in PFS rates between an experimental drug
and a historical control, let us assume n = 50 for the total trial. The
criteria to proceed are based on two steps:
(a) If after the first n = 20, we observe 10 or more patients (50%
response) who are alive and progression-free at 6 months (since
starting treatment), then recruit a further n = 30.
180 Economic Evaluation of Cancer Drugs

FIGURE 6.1
Parallel and crossover designs.

(b) After observing the full 50 patients, if 25 out of 50 are alive and
progression-free at 6 months, then the treatment is a good candi-
date for further evaluation or possibly even registration.

Clearly, if only 3 patients are alive and progression-free at 6 months out of


the first 20 (15%), the likelihood (probability) of 22 being alive without pro-
gression in the next 30 (73% PFS rate) will be very low. Thus, a low num-
ber that are alive without progression can lead to an early decision to stop
the trial. As far as economic evaluation is concerned, one should still plan
for collecting health resource use data assuming the trial will continue to
n = 50 (unless evidence suggests otherwise) in the second stage. All patients
will be valuable for a cost-effectiveness analysis and not just those in Stage
2. Further variations of these designs include single and two stage designs
Designing Cost-Effectiveness into Cancer Trials 181

for dual outcomes (safety and efficacy) such as Bryant and Day (1995) and
Yap (2013).
Recently, several trials using data from a single patient cohort have been
used to provide evidence for efficacy. Moreover, an economic evaluation has
also been performed using data from the experimental observed trial and
compared against a historical (rather than a concurrent) control. We present
below two examples for the same indication, with different drugs resulting
in two different decisions (one recommended and one rejected for reimburse-
ment). In Section 6.6, we will present a more detailed exposition of the design
implications for HTA assessments and point out in practical terms how these
need to be considered. For now, the point is that Phase II evidence alone may
be used for economic evaluation, but it must be robust and substantial.

Example 6.1: Single Arm Trial in Patients with


Ofatumumab for Chronic Lymphocytic Leukemia
BACKGROUND
Ofatumumab (Arzerra, GlaxoSmithKline) is a fully human, high-affin-
ity, monoclonal antibody that is targeted against the CD20 cell surface
antigen of B-lymphocytes. Ofatumumab received marketing authoriza-
tion for the treatment of chronic lymphocytic leukemia in patients who
are refractory to fludarabine and alemtuzumab. It is delivered by intra-
venous infusion. Further details are to be found in HTA TA202 (NICE,
2010) This was an uncontrolled (nonrandomized) trial in n = 154 patients.
The primary outcome was tumor response rate; secondary outcomes
included time to onset of response, duration of response, progression-
free survival, time to next therapy for chronic lymphocytic leukemia,
overall survival, reduction in tumor size, safety, and pharmacokinetic
endpoints.

EFFICACY
After an interim analysis from 59 patients, an observed response rate of
58% (34/59) [99%CI: 40–74%, p < 0.0001) was compared to a planned tar-
get of 15%. All responses were partial remissions (i.e. partial response).
The ERG (NICE Economic Review Group) comments from an economic
evaluation perspective can be summarized as:

(i) Absence of robust evidence from randomized controlled trials.


It was not possible to accurately assess the impact of bias on the
outcomes, including adverse events.
(ii) Limited data because of small sample size (n = 59) and an
absence of recent data.
(iii) No HRQoL.
(iv) Differences in baseline characteristics existed between groups
who responded and did not respond (e.g. the patients who
responded might have been younger compared to the ones who
did not respond: possibly older and with worse prognosis).
182 Economic Evaluation of Cancer Drugs

(v) The drug was not considered cost-effective despite European


Marketing Authorization approval for a license.

One issue following point (v) is that efficacy is determined on the tumor
response rate but the economic evaluation was based on the OS. This
difference in approach between two agencies is related to the different
decision-making problems – one of efficacy and one of cost-effectiveness.
The relationship between response and OS should have been examined
further. Indeed, the assessment report states:
The Committee concluded that, based on expert evidence, it was
plausible that ofatumumab may offer clinical benefits to patients,
but that that it was not possible to determine the magnitude of the
effect from the evidence presented.
The Committee recognized the difficulty of conducting random-
ized controlled trials in small populations of patients with limited
life expectancy, as highlighted by the clinical specialists. However,
the Committee concluded that such difficulties could have been
addressed more effectively than they had been in the manufac-
turer's submission.
The Committee further discussed the potential for using data from
historical controls, for example from retrospective observational
Ofatumumab data for the treatment of chronic lymphocytic leukemia
refractory to fludarabine and alemtuzumab.
The Committee also noted that the manufacturer had not provided
more recent data from the Hx-CD20-406 study (the interim analysis
was from May 2008, with no further data expected before 2011). The
Committee heard from the manufacturer that this interim analysis was
planned in the study protocol and that an unplanned analysis would not
be possible, in accordance with best statistical practice in clinical trials,
which discourages unplanned interim analyses.
This appears an unusual statement in that the magnitude of the over-
all response rate was 58% – 15% = 43%. Unless there was no correla-
tion between ORR and survival, it is hard to see why a plausible benefit
should not exist. This drug was not approved for reimbursement.

Example 6.2: Single Arm Trial in Patients Treated with


Venetoclax for Chronic Lymphocytic Leukemia
Venetoclax was approved for the treatment of chronic lymphocytic leu-
kemia (CLL). Prior to marketing authorization, venetoclax was desig-
nated a promising innovative medicine and was available to patients in
the NHS through the EAMS (a scheme whereby patients may have the
drug available at a given price because it has shown promising effects
and is innovative). Details can be found in the HTA TA 487 (NICE, 2017).
The evidence of efficacy came from a single Phase I (n = 67) and two
single arm Phase II trials (n = 158 and n = 64). The primary outcome was
overall objective response rates (ORR). Secondary outcomes included
OS. The ERG concluded:
Designing Cost-Effectiveness into Cancer Trials 183

The committee was concerned that the single-arm design of the trials
made it difficult to assess the efficacy of venetoclax (that is, there was no
comparator arm of patients having best supportive care) …
The committee concluded that interpreting the results from the veneto-
clax trials was challenging without a direct comparator, and that this was
compounded by the small patient numbers in the trials.
Nevertheless, it was recommended for reimbursement. We now pres-
ent a side-by-side summary of the evaluation of evidence from two dif-
ferent ERG committees for a drug for the same indication with opposite
decisions.
A summary of Table 6.1 showing two reimbursement decisions from
Phase II trial data indicates that the evidence from the venetoclax is con-
sidered to be:

(a) More substantial in quantity (i.e. more data).


(b) Appears more effective.
(c) However, the evidence was criticized for several reasons such
as:

(i) Whether rituximab was the best choice of comparator,


or BSC, or a drug called idealalisib in combination with
rituximab.
(ii) The choice of utility for progression-free survival (0.71 vs.
0.853).
(iii) Lack of comparative data (no randomized trials).
(iv) Whether the end-of-life (EoL) criteria were met; there is a
possibility to increase the cost-effectiveness threshold to
£50,000/QALY if EoL criteria apply.
(v) Most patients are diagnosed at age >65 and there are conse-
quences for affordability, intolerance to treatment, and side
effects.
(vi) Improvements in HRQoL should affect the whole family
and not only the patient.

(iii) Adaptive Designs

(a) Group Sequential

Where a decision to stop a trial early due to efficacy is considered, an


adaptive trial may be used. There are various forms of adaptive design.
The Phase II designs mentioned earlier (single and two stage designs in
Section 6.3) are also a type of adaptive design (for single arm trials). In the
two-stage design earlier, the decision to stop or continue after Stage one is
based on availability and strength of efficacy data in the first n = 20 (if >10
out of 20 are alive and progression-free at 6 months, one may continue and
recruit more subjects). An example of a group sequential design is shown
in Figure 6.2.
184 Economic Evaluation of Cancer Drugs

TABLE 6.1
Two Reimbursement Decisions
Venetoclax [TA487] Ofatumumbab [TA202]
Year November 2017 October 2010
Recommendation (NICE) YES NO
Recommendation YES YES
(License)
Drug company AbbVie GSK
Price £4,789.47/28 days £182.00 per 100 mg vial
Dosing is 300 mg for 1st infusion
plus 2,000 mg for 8 weekly
infusions

Duration of treatment Treat until PD (£58,752.23/ 8 weeks


year)
Number of patients N = 67 (Phase 1)a 154
planned N = 158 (Phase II)b
N = 64 (Phase II)c
Number of patients for 283 59
evidence
Interim analyses Yes (n = xxxTBD) Yes (n = 59)
Trial design One Phase I and two Phase IIa Single arm
Primary efficacy outcome ORR (RECIST) ORR (RECIST)
Target effect (primary) 60% (Phase II single arm) 15% ORR
Observed effect (primary) 77% (Phase II single arm) 58%
Median (mean) OS Not reported/available 13.7 months
Median (mean) PFS 27.2 to 41.4 months 5.7 months
Median (mean) PPS Not reported
Grade 3–5 toxicity Not reported/ available 64%
Time horizon 10 years 10 years
Comparator BSC (ritiximab/ BSC (inappropriate/not optimal)
inappropriate)
ERG used idelasilib (study
116)
HRQoL (EQ-5D) Collected in trials (EQ-5D-5L) None – externally used
Utility 0.853 company, 0.71 (ERG) 0.65 for PFS and 0.47 for PD
and agreed at 0.748
Incremental QALY Not reported/available 0.353
Incremental cost Not reported/available £13,565
ICER £47,370 £38,421/QALY
ERG revised ICER £57,476 to £77,779 £50,300 to >£81,500/ QALY
a Notes: One Phase II was a two-arm nonrandomized trial, and the other Phase II trial was a
single-arm design.
b Study M12-175 (n = 67, Phase I dose escalation).
c Study M13-982 : two-arm nonrandomized (n = 158).
d Study M14-032 (n = 64 single arm) plus (n = 41 open label safety extension).
Designing Cost-Effectiveness into Cancer Trials 185

FIGURE 6.2
An adaptive group sequential trial with two interim analyses.

Figure 6.2 shows the decision procedure for stopping a trial early for effi-
cacy or futility. The Y-axis is the observed hazard ratio related to the observed
treatment effect and the X-axis is the number of events of interest. The two
boundaries (lines) relate to stopping early for efficacy (top line) or futility
(bottom line). There are 3 points, one at 15 events, another at about 29 events,
and the final at around 49 events. These have been computed mathematically
and relate to the 2 interim analysis time points (1 interim analysis occurs
when a total of 15 events have been observed and the other at around 29
events). The final analysis occurs at around 49 events. If, after 15 events, the
observed HR is around 6.5 (this compares control vs. experimental, so the
HR for experimental vs. control would be 1/6.5 = 0.15, a very strong efficacy
signal), there is an option to declare early efficacy and stop. If the decision
is to continue, then after 29 events have occurred and the HR is around 2.5
(or 0.4), another opportunity to stop the trial early for efficacy is possible. In
contrast, if after 15 or 29 events the observed HR is 1, the trial may be stopped
for futility.
Figure 6.2 relates to a trial with two groups that was designed to detect
an HR of about 1.78 (or 0.56 for experimental vs. control), using 80% power
and an overall 2-sided significance level of 5%; a total of 120 patients (60 per
group) and about 30 death events in total were needed. Two interim analyses
were planned: the first at 25% of the total events (15 out of 60 events) and the
second at about 50% (29 out of 60 events). A stopping rule for futility was
considered (i.e. stopping the trial if evidence appeared there was unlikely to
be benefit for future patients). The top line in Figure 6.2 is a boundary for the
effect size for stopping early and the bottom line is for stopping for futility. If
186 Economic Evaluation of Cancer Drugs

the observed HR crosses either of these boundaries, the trial may be stopped
for either efficacy or futility. A rule called the O’Brien and Fleming rule was
used to take into account the multiple interim analyses (number of times
analyses are conducted). If we wish to stop early and declare the treatment
to be efficacious, this is very difficult earlier on because the HR ratio would
have to be around 6.5 or 1/6.5 = 0.15 (depending on the direction of compari-
son), which is a very large clinical effect.
If the trial does happen to stop early because the drug shows promising
benefit, evidence for effectiveness may nonetheless be limited. Adaptive
designs are designed to stop based on efficacy endpoints, but cost-effective-
ness calculations do not appear in the derivations of the stopping rules. These
can be incorporated, but is beyond the scope of this book. For a more practi-
cal demonstration see Bartha et al. (2013) and Chabot et al. (2010). Bartha et
al. (2013) report an instance where payers were receptive to the idea of using
immature survival data for their decision.
It is very important that statistical principles are followed and the integ-
rity of the trial is not compromised by requests for additional data to assess
effectiveness. In Example 6.1, the ERG noted an absence of interim data
and the manufacturer was criticized for not providing sufficient OS data,
meaning that a cost-effective judgment could not be made. The wording of
the response from the ERG to the manufacturer appears to suggest a criti-
cism for not performing an unplanned analysis. However, such unplanned
interim analyses can lead to bias and can compromise the entire trial. Hence,
requests for data for the purposes of cost-effectiveness needs careful consid-
eration before it is provided.

b) Multi-Arm, Multi-Stage (MAMS)

The so-called MAMs design (Royston, Parmar, & Qian, 2003) allows mul-
tiple arms to be compared against one or more controls in a single trial. The
nature of the adaptation allows comparisons to be made between multiple
treatments and a control at multiple time points (i.e. at several interim analy-
ses). Treatments that do not show sufficient promise in terms of an interme-
diate outcome (such as PFS or ORR) may be discontinued. Recruitment to the
control and remaining experimental arms continues to determine evidence
for efficacy. Currently, only efficacy outcomes (whether mean, proportion,
or time-to-event) are considered in the decision rules for assessing benefit.
What is likely to be important is whether specific arms are likely to be cost-
effective, not just efficacious. This implies that the methodology of MAMs
designs may need to be extended to several outcomes that include costs and
effects. Decisions on whether arms should be dropped or future patients
(who were initially randomized to treatments that were not efficacious)
should be randomized to more promising arms should also take account of
information on whether in the real-world they are likely to offer value. In
other words, continuing the trial on the basis of efficacy alone may not be
Designing Cost-Effectiveness into Cancer Trials 187

FIGURE 6.3
Example of the STAMPEDE MAMs design. (Sydes et al., 2009.)

sufficient guarantee patients will have access to these treatments if they are
later found to lack cost-effectiveness (Figure 6.3).
In summary, novel designs such as adaptive, response adaptive, and group
sequential designs for demonstrating early benefit, may limit the possibility
of observing medium- to longer-term costs and benefits if the trial stops for
early efficacy. The treatment benefit in such trials would need to be large to
stop early. Such designs can rely on statistical significance (p-values) or poste-
rior probabilities for decisions on efficacy, rather than the clinical effect size,
which is important for economic evaluation. The driver for cost-effectiveness
is related to the clinical effect size and not the p-values. For example, a trial
can be stopped early for a modest clinical effect with a smaller than expected
variability, but that may not be cost-effective.
Longer-term measurements for economic evaluations may not be a prior-
ity in a sequential/adaptive framework and therefore another separate trial
might be needed to demonstrate longer-term benefits/effects, which not only
raises ethical issues (e.g. why should a trial be run for economic consider-
ations when there is no longer clinical equipoise), but also risks invalidating
the conclusions of the first trial (e.g. ensuring additional follow-up measures),
especially if it was positive (although one would expect consistency in the
results between trials for the same population, endpoint, drug, conditions,
etc.). Where trials are stopped early, some additional evidence may be sought
from observational (real-world evidence) studies or retrospective evaluation
of databases (like the Hospital Episode Statistics (HES) data or audit data
such as the National Lung Cancer Audit (NCLA)).
188 Economic Evaluation of Cancer Drugs

6.2.2 Interim Analyses and Data Monitoring Committees (DMC)


A related issue is that of independent data monitoring committees (IDMC),
on which health economists are increasingly playing a more active role,
particularly in some academic trials in the UK. The reasons for this are
essentially to evaluate how the opportunity costs for continuing a trial are
reflected in the decision-making.
A small treatment benefit that is likely to result in a nonclinical yet statis-
tically significant difference may not result in a positive cost-effectiveness
conclusion. In order to ensure public expenditure is used in an optimal way,
funds could be directed to other trials that do show promising benefit. While
one may argue that such a decision could be made purely on clinical grounds
without using cost-effectiveness arguments, the two are likely to be inextri-
cably linked. The challenge for health economists here is that while arguing
that cost-effectiveness from a lifetime perspective needs to be evaluated to
reduce uncertainty (as is often stated in ERG reports), the interim data do not
provide for such decision-making.
Several important considerations need to be taken into account with interim
analyses, especially where a cost-effectiveness component is included.

(i) All relevant major health resource use data are adequately col-
lected (e.g. to ensure the IDMC has sufficient information to make a
decision).
(ii) Data collected does not compromise the trial design. For example,
where PFS is used as a surrogate, the desire to also look at OS should
not constitute an unplanned interim analysis that renders both the
final efficacy and cost-effectiveness analyses to be biased (or even
invalid).
(iii) The ongoing conduct of the trial is not compromised. Where the trial
is analyzed blind (even the IDMC will not know treatment group
allocation), health resource use data collected does not provide the
opportunity to make a good guess as to which arm patients are allo-
cated to.
(iv) Data are not shared, or information is not shared between people
appointed as members of the IDMC because they are colleagues.
Many academic trials work with in groups of collaborators who
are often familiar with each other’s work (they might meet at con-
ferences or be joint investigators); information from one arm of a
trial could be shared to influence the conduct of a separate trial
where there is a common comparator. Whereas there may be legal
consequences in industry trials where financial interest exists if
confidentiality is compromised, such penalties are not known in
academic trials (Sartor & Halabi, 2015). We believe that if such a
practice can happen in an industry-sponsored trial, the impetus
Designing Cost-Effectiveness into Cancer Trials 189

and pressure to secure grant funding may be equally as great,


hence a more robust approach is needed. Clinical trial units that
conduct academic trials need to be much more robust to ensure
such practice is removed, to avoid the credibility of published
results being open to question.

(v) Open Label Extensions

Open label extensions (OLE) occur where, at the end of the randomized
phase, patients can be followed up outside the randomized protocol-defined
conditions. An open label extension after an initial double-blind phase can
be helpful for understanding real-world effects. During the open label exten-
sion, longer term costs (e.g. for later adverse events, effects of maintenance
of new treatment, etc.) can be evaluated. Sometimes after the double-blind
phase of a trial, all patients are switched over to the experimental treat-
ment (despite the benefits of the new experimental treatment remaining
unproven). In that case, longer-term effects and costs can become available
for the experimental arm only, and not for the comparator.
The importance of an open label extension should not be underestimated
or glossed over, in terms of both design and statistical analysis. The empha-
sis is often placed on the double-blind part of the trial. The open label part
can often be used more efficiently to maximize the value argument because
this part of the trial reflects real clinical practice better than the controlled
phase. For example, patients might take concomitant medication, additional
patients might be entered who were not previously allowed, or longer-term
effects can be assessed. This would suit an economic evaluation that is usu-
ally performed after the results of the clinical effects are made available
(although the planning of economic models would be much earlier). An
OLE may be conducted in a separate protocol (one protocol for the double
blind and one for the open label), or as a single protocol. A clear definition
of the starting and stopping between the two stages needs to be carefully
determined.

(vi) Observational Studies

Not all clinical evaluations can be performed through RCTs. For example, we
could not (ethically) randomize one group to tobacco and other to placebo to
determine if they developed lung cancer. Similarly, it will be challenging to
randomize a group of children to the MMR vaccine and another group to no
vaccine to see if they developed autism (following the Wakefield controversy
on MMR related to autism, see Maisonneuve & Floret, 2012).
An observational study provides data on the natural history of a condition
where data are collected in a representative population from the disease of
interest. Clinical trials may not provide the data needed for a decision due to
limitations in the selected population (e.g. due to inclusion criteria), choice of
190 Economic Evaluation of Cancer Drugs

comparator (new treatments licensed after the main trial results submitted),
limited length of follow-up (e.g. with OS, may be able to have longer follow-
up to determine rare outcomes), limited sample size, and absence of HRQoL
data. This limits the generalizability of the trial results.
Observational studies can therefore provide an important source of data
and may be the only source of evidence for collecting cost-effectiveness data
in some cases. In observational studies, patients are not randomized, and
the timing of measurements and other procedures is not controlled. These
are often conducted in real-world (naturalistic) settings. One major issue is
the presence of selection bias. For example, if we select patients in one group
who have the risk of an expected outcome of interest (e.g. PD) and in the
other group, patients are those with the outcome, this type of comparison or
‘case mix’ is likely to be problematic. Ways to account for the selection bias
will be discussed in Chapter 8 (RWD) through the use of propensity scores
and other adjustment models, Guo and Fraser (2014).
In an observational study, the estimate of the treatment effect (or ‘drift’)
may be subject to considerable selection bias; it will be important to control
the data quality with complete histories and patient characteristics. Baseline
characteristics data will be needed such that adjusting treatment effects for
potential selection bias can be adequately determined. Observation studies
are particularly useful for evaluating the often-needed longer-term conse-
quences of treatments not observed during clinical trials. This also includes
data on HRQoL, utilities, and health resource use.

(vii) Retrospective Case Control Studies

Case control studies are used to retrospectively (data collected on past


events) determine if there is an association between an exposure and a spe-
cific health outcome. Patients are selected in a case control study based on
their known outcome status (e.g. died, progressed). For example, we might
collect data from hospital notes about incidence of disease (e.g. a particular
cancer) after being exposed to a number of variables (explanatory variables,
e.g. smoking, age, gender, disease history, prognostic markers). We wish to
investigate whether these variables explain or are related to the outcome of
the disease. Consequently, explanatory variables are subject to variability
and outcomes are known. This type of data is easier (and faster) to collect
and usually cheaper than an observational study or conducting an RCT.
A case control study might be useful where the disease (and/or outcome)
is rare because investigators can intentionally search for the cases. A cohort
study in a rare disease requires a large number of patients with the outcome
of interest (exposed) to get an adequate number of cases. ‘Cases’ and ‘con-
trols’ should be clearly defined prior to commencing the data collection. Data
on health resource use, too, needs to be determined a priori to avoid biased
conclusions.
Designing Cost-Effectiveness into Cancer Trials 191

6.3 Planning a Health Economic Evaluation in a Clinical Trial


6.3.1 Important Considerations When Designing a
Cancer Study for Economic Evaluation
One key aspect of planning a health economic evaluation is to identify:

(i) What data will be collected for economic evaluation?


(ii) How will the data be collected?
(iii) When will the data be collected?

The answer to these three questions is closely related to the overall objectives
and the design of the trial, in particular whether the economic evaluation is
prospective or retrospective. Whereas in almost all cases clinical trial data
are collected prospectively, cost and quality of life data are not always col-
lected prospectively. Some possibilities are summarized in Table 6.2 below
which outlines the type of economic evaluation that could be undertaken
(last column).

(a) Costs/Resource Use and Effects Collected Prospectively

In the context of a clinical trial, costs and resource data are collected at the
patient level prospectively. For each patient there are several measures of
effects and several measures of resource use (such as hospital visits, consul-
tations, scans, number of doses, etc.). The trial is typically designed for a clin-
ical endpoint, but the economic component is accommodated or added on.
In some cases, both clinical and economic endpoints are formally designed
(including sample size calculations) into the trial. For example, sample size is
likely to be based on the clinical endpoint, as will timing of the main primary
and secondary endpoints. However, the data collection forms (also called
case report forms (CRFs)) might be designed to capture resource use and
specific adverse events in detail, especially those associated with important

TABLE 6.2
Collecting Data in a Trial for an Economic Evaluation
Clinical Endpoint Other Outcomes Possible Economic
Costs collected (Primary) (e.g. utility) Evaluation Method
Prospectively Prospectively Prospectively Stochastic (patient level)
Retrospectively Prospectively Prospectively Stochastic/decision tree/
Markov
Retrospectively Prospectively Retrospectively Decision tree/ Markov
Retrospectively Retrospectively Retrospectively Decision tree/ Markov
192 Economic Evaluation of Cancer Drugs

costs. Extended patient follow-up time may allow for collection of data in
the form of an open label extension with relaxed inclusion criteria (whereas
in a trial without an economic component, the follow-up period might be
restricted).
An economic evaluation is usually performed after the results of the clini-
cal effects are made available (although the planning of economic models
will be much earlier). There is unlikely to be any benefit in carrying out an
economic evaluation if there is no clinical benefit (unless value is shown in
some subgroups). The economic evaluation is usually carried out using sto-
chastic methods, that is, using patient-level observations for the statistical
analysis of costs and effects. Another approach to analysis is to summarize
the individual cost and efficacy data (e.g. means and/or frequencies) and use
these as ‘inputs’ into a health economic simulation model to estimate the
ICER.

(b) Clinical Effects are Collected Prospectively but Some Health Resource
and Quality of Life Data is Collected Retrospectively

The clinical trial in this scenario is designed only for the clinical component,
and costs and utility (HRQoL) are extracted from external sources or pub-
lished literature. For example, in an oncology trial the clinical effects might
be a measure such as progression-free survival (PFS) time – calculated from
the time from randomization until disease progression. The health resources
involved with the new treatment, along with HRQoL data, are extracted
from the clinical trial database (from compliance, exposure rates and HRQoL
modules of the CRF), but concomitant medication use might be taken from
published sources. Collecting concomitant data in a CRF can be an ardu-
ous task in a clinical trial because the data often needs to be queried for
items such as missing dates, details of mode of administration, converting
names from active ingredient to either brand or generic name (brands are
more expensive), and so forth. In practice, the percentage of patients who
take a specific form of concomitant medication might be determined from
literature rather than using data collected in the trial.
Patient-level safety data will also be collected (e.g. specific adverse event
grades for each patient), where the average cost of treating each adverse
event can be extracted from external published sources using reported dura-
tion of adverse events. The duration of adverse events in the trial give an
idea of actual resource use to treat adverse events in real clinical practice.
For example, rash is a well-known side effect of the drug erlotinib (Lee et al.,
2012) that is treated with some topical ointment; the incidence of rash can
be collected but details of the treatment for rash can include information
such as frequency, dose, name, and route of administration, which may not
be collected in the CRF. If the time to rash disappearance could be deter-
mined from the CRF, this might provide a useful estimate of the duration
Designing Cost-Effectiveness into Cancer Trials 193

of concomitant medication use. However, unless the duration of an adverse


event is a critical clinical or cost outcome, one generally restricts computation
of duration for the most important adverse events (usually most costly) to
minimize database and programming time. To compute duration, the start
and stop times of adverse events will be necessary to compute and these are
not always available. Lewis et al. (2010) estimate the costs associated with the
duration of an adverse event by using concomitant treatment from published
data. Data managers and clinical programmers would need to ensure that
important data such as the dates of onset and termination were available for
this type of calculation.
In some situations, clinical effects may need to be extrapolated from the
clinical trial, even where effects are measured prospectively. For example, 5
years of follow-up time may not be long enough to monitor PFS (progression
might occur at 6 or 7 years after the start of treatment) in some cancer trials.
Estimates of PFS times in the specific patient population could be made at
year 6 and beyond based on literature data. Alternatively, Dukhovny et al.
(2012) modeling and simulation might be used to estimate PFS effects beyond
the fifth year of follow-up; The follow-up period could also be extended
(more costly) during the design stage of the trial so that extrapolation and
therefore uncertainty can be minimized. Having observed data (outcomes)
available is always less uncertain compared to extrapolating data, no matter
the complexity or ingenuity of the models used.

(c) Clinical effects and outcomes are collected retrospectively

If no clinical trial data are available, data are likely to be extracted from pub-
lished sources. An important feature of the data here is that cost-effective-
ness analyses are usually determined from summary data (e.g. mean costs
or effects). These (inputs) are often used for simulation in a cost-effectiveness
model. The key difference compared to prospectively collected data is that
since data collection is retrospective the inputs for an economic model are
based on whatever data has been collected previously. The quality of the
economic evaluation, therefore, depends on where the data were extracted
from, their quality, and their consistency with the population under study.
In many cases the retrospective data are extracted from published clinical
trials such as in Dukhovny et al., 2012 or O’Connor et al. (2012). Dukhovny et
al. (2012) described their economic evaluation as being ‘retrospective,’ how-
ever, the trial was designed prospectively for an economic evaluation. In this
book, when we consider retrospective analyses, we take this to mean that
the clinical trial did not plan for an economic evaluation or collect economic
data prospectively – not that the analysis was carried out at some much later
time. If the former were true, all analysis would be retrospective (economic
evaluation and statistical analysis) because analyses are almost always done
after the trial has been completed (except perhaps in some sequential (or
194 Economic Evaluation of Cancer Drugs

Bayesian) type of trial designs, where analysis is conducted while the trial
continues).
In some situations, prospectively collecting data may be too risky commer-
cially or may just not be the most efficient way (e.g. comparing head-to-head
with a competitor drug, such as nivolumab vs. pembrozulimab, see NICE,
2017 on nivolumab). In Examples 6.1 and 6.2, the company withheld the OS
rates from public view during the review of its economic model by NICE.
The details of cost or utility data may also be withheld for commercial sen-
sitivity. Consequently, in such cases the use of retrospective data may be the
only option available.
One common scenario is to compare a new treatment A with the current
standard of care S. In a three arm trial of A, S, and best supportive care
(BSC), reimbursement assessors may argue for including S as the third arm
to address value for money in terms of superiority (A vs. S). A pharmaceuti-
cal company may have its own commercial (or regulatory) reasons to avoid
a direct comparison of A with S (e.g. to minimize impact on market share).
One possibility is to carry out a comparison of A versus S in some indirect
way (Section 4.9) (Ades et al., 2011):

(a) Treatment A (the new treatment).


(b) BSC (needed to demonstrate efficacy).
(c) Treatment S (needed to demonstrate efficiency and effectiveness).

Suppose treatment S has nonetheless been compared with BSC in a separate


trial and a comparison with treatment A results in A being on average worse
than S (A < S). It may therefore be commercially risky for the manufacturer
of A to compare A with S.
Notwithstanding, an indirect comparison would allow an estimate of
treatment benefit between A and S that would otherwise not be made – or
even known. The so-called mixed treatment comparison allows for the pos-
sibility of avoiding direct comparisons, albeit in a noncontrolled setting. A
mixed treatment comparison would use published data from a trial that
compares treatment S with treatment BSC and A versus BSC as a way to
compare the efficiency of A versus S. Figure 6.4 shows an indirect treatment
comparison between A versus S through a common arm BSC.

6.3.2 Integrating Economic Evaluation in a Clinical Trial: Considerations


Integrating a health economic component into a clinical trial has been dis-
cussed previously (Haycox, 1997; Drummond, 2005; Glick et al., 2011, 2014;
O’Sullivan et al., 2005; Petrou & Gray, 2011; Ramsey, 2015). We now expand
further on the major aspects of trial design for economic evaluation below
and summarize these in Table 6.3 (Figure 6.4). More details can be found in
Khan (2015).
Designing Cost-Effectiveness into Cancer Trials 195

FIGURE 6.4
Diagram representing direct comparison for A versus B and A versus C (solid lines) and the
indirect comparison for B versus C (dashed line).

TABLE 6.3
Summary of Key Clinical Trial Design Issues for a Health Economic Evaluation
Key Issue Key Considerations
Endpoints and outcomes • Identify which endpoints are truly clinical, which are
economic, and which may be both
Choice of comparator • Standard of care or another comparator
• Direct vs. indirect comparison
Timing of measurements • Open label extensions
• Duration of follow-up
• Capture later and early costs
Trial design • 3-arm trial or 2 separate trials (efficacy and CE)
• Adaptive designs may have limited follow-up
CRF design • Identify resources to collect
• Focus on big cost items
Power and sample size • Number of patients required to demonstrate CE
• Power-specific subgroups where CE most likely
• Value of information
Treatment pathway • Identify costs and benefits in the treatment pathway
Generic entry/ patent expiry date • Identify dates for project management and speed of
preparing reimbursement argument
Compliance • Compliance may be related to side effects and hence
more costs
Subgroups/heterogeneity • Identify subgroups a priori defined where treatment
benefit is more likely and greatest
Early ICER/INMB • An early ICER helps to work out where the new
treatment is in the CE plane
Multicenter/multinational trial • Country-specific CEAC or ICERev
• Multilevel modeling techniques
196 Economic Evaluation of Cancer Drugs

6.3.3 Endpoints and Outcomes


Those endpoints that are clearly clinical, clearly economic, and that have
potential overlap should be identified as early as possible. The primary end-
point is often thought to be unambiguously clinical, whereas HRQoL may
be considered ‘somewhere in the middle’ between an economic and clinical
endpoint (Figure 6.5).
Compliance endpoints may be relevant to both clinical (e.g. for per proto-
col populations) and economic evaluations (cost of actual drug use). The type
of HRQoL measure is also an important consideration: should the HRQoL
measure be disease-specific, like the QLQC-30 for cancer, or should it be a
generic one such as the EQ-5D? Careful thought needs to be given as to how
important secondary endpoints can be used to add to the value to a new
treatment. Demonstrating value of a new treatment should take into account
more than just one endpoint alone. If the endpoint is rare such as chondroma
of the mandibular condyle (a benign tumor in the head and neck) then per-
haps a clinical trial is not the best design.
The QALY discussed earlier is not an endpoint but a composite of 2 end-
points (HRQoL and survival, but not necessarily limited to OS). This metric
is considered to be a preferred choice in some countries to demonstrate the
value argument, although not accepted universally (e.g. Germany and some
other countries do not consider the QALY as the primary basis for demon-
strating value). The value argument should not be dependent on a single
outcome, even if it is a key measure. In situations where a primary endpoint
is positive but all secondary endpoints are negative (worse effects), a decision

FIGURE 6.5
Relationship between clinical and economic endpoints in a clinical trial.
Designing Cost-Effectiveness into Cancer Trials 197

in support of cost-effectiveness may be difficult compared to the case where


the secondary endpoints are, on average, not worse (no difference).
Some generic HRQoL measures lack the sensitivity to detect treatment
benefit, such as the EQ-5D in lung cancer patients (Khan, 2015). A cancer-
specific HRQoL measure, such as the QLQ-C30, can have greater sensitivity
to detect treatment effects. It can be incorrect to conclude an absence of cost-
effectiveness, when the problem itself lies in the measures used to assess
cost-effectiveness. Other outcome measures, such as Quality of Time Spent
Without Symptoms of Disease and Toxicity (QTWiST), a special type of
QALY (Gelber et al., 1996) that subtracts from the overall survival the period
of time during which either treatment or disease reduces HRQoL, may be
considered as an alternative to the QALY in some cases. Disability-adjusted
life-years (DALYs) and other measures may be considered as outcomes.
The failure of an economic evaluation to demonstrate a cost-effective
QALY difference is not sufficient evidence to conclude an absence of an over-
all value argument. Indeed, in Germany, IQWiG does not consider QALYs as
a prominent measure (or the only measure) that demonstrates the value of
the new technology, but any measure that captures a combination of mortal-
ity, morbidity, and QoL. A translation of the text from IQWiG is shown:

As the benefit of an intervention should be related to the patient, this


assessment is based on the results of studies that have investigated the
effects of an intervention on patient-relevant outcomes. In this context,
‘patient-relevant’ refers to how a patient feels, functions or survives. In
this context, consideration is given to both the intentional and uninten-
tional effects of the intervention that allow an assessment of the impact
on the following patient-relevant outcomes, in order to determine the
changes related to disease and treatment:

1. Mortality
2. Morbidity (complaints and complications)
3. Health-related QoL

(IQWiG, version 3.0, 27/05/2008)

A comparison between a new treatment and the standard of care is often


needed in an economic evaluation. If a potential comparator has not been
approved for licensing because it is undergoing a review by the EMA, this
does not mean that a new experimental treatment cannot be compared with
this potential (future) comparator. For example, at the time of submission
of lenalidomide for licensing (NICE TA171, 2008), bortezomib was under-
going review by the EMA. Later bortezamib became the standard of care
while lenalidomide was under review. The sponsor argued that a compari-
son for economic evaluation with bortezamib was not possible because at
the time bortezamib was not the standard of care. The assessors (for reim-
bursement) argued by noting that although this may be true for licensing,
198 Economic Evaluation of Cancer Drugs

such an argument did not apply to economic evaluations and the sponsor
should have considered a comparison with bortezamib (see NICE TA171,
ERG Report). The method of indirect or mixed treatment comparisons may
be used in such situations.

6.3.3.1 Timing of Measurements
The primary efficacy endpoint is usually the main focus of a trial with col-
lection occurring at some ‘optimal’ set of time points (e.g. for PFS, scans
taken at some ideal time points). For other (secondary) endpoints, such as
HRQoL, the assessment times of collecting data are important. Ensuring
enough pre- and post-disease progression HRQoL measurements are avail-
able will allow more precise QALY estimates. If the median PFS is 6 months,
for example, then HRQoL might be collected every month. However, if
median PFS is only 3 months, more frequent HRQoL assessments might be
made prior to 3 months. For other endpoints, extended follow-up for moni-
toring compliance, end-of-life care, long-term efficacy, and safety might also
be needed.
In many trials HRQoL is often collected until disease progression (e.g.
cancer trials), although in theory it could be collected until death or to the
onset of palliative care. However, it might be felt that collecting HRQoL after
disease progression is very difficult because patients will deteriorate rap-
idly. If some patients continue to take some treatment even after he/she has
progressed, time to second progression might be a useful endpoint. In such
cases, having information about HRQoL between the first and second pro-
gression will help inform decision-makers about the value of treatment over
a longer period of time (and justify treatment with the new drug beyond first
progression). The period between first and second progression (or death)
is essential to estimate the value of a new treatment over the lifetimes of
patients. Moreover, during the post-progression period, patients tend to take
additional concomitant/anti-cancer treatments, which may have a noticeable
impact on HRQoL, because many of these are third- or fourth-line treat-
ments with higher toxicity burdens.
In some cancer trials, all patients may not be followed until death, so costs
are censored for some patients, whereas complete costs are available for oth-
ers. Adequate follow-up is needed to capture all the later as well as the earlier
costs. Follow-up time should not be so long that demonstrating cost-effec-
tiveness and reimbursement is delayed or becomes too imprecise.

6.3.3.2 Trial Design
Trial design was discussed earlier in Section 6.1. In this context, trial design
refers to the experimental design of the trial and not the broader aspects of
trial design such as blinding, choice of comparator, timing of measurements,
etc. Parallel group designs are the most common in cancer clinical trials.
Designing Cost-Effectiveness into Cancer Trials 199

6.3.3.3 CRF Design
In clinical trials, data is recorded on a case report form (CRF) or a data col-
lection form (DCF). Until relatively recently, CRFs were designed without
considering resource collection. Resource use was estimated from surveys
or based on external publications. In a clinical trial designed prospectively
for economic evaluation, resource data, especially for ambulatory care, is
generally captured through interviewing patients using nurses or trial staff
during scheduled clinic visits (note: this applies only to community outpa-
tient care; for in-hospital care, whether for a standard stay or otherwise, data
are tracked by extracting data from the hospital’s patient file either manu-
ally or electronically; in-hospital costs in a fee-for-service health system can
be extracted from the hospital billing system). Questions such as: “When
was the last time you visited the GP since your last visit to the clinic?” or
“Was an outpatient visit to the hospital made?” are included in the CRF. The
responses to such questions are then used to calculate the costs per patient.
Resource use is entered into the CRF in physical units and not monetary
value. The unit prices of each health resource are obtained from elsewhere
(see Chapter 4).
Resource use cannot always be planned, unlike scheduled clinic visits.
Often a patient might attend a protocol scheduled clinic visit and the ques-
tion in the CRF might be “Since your last visit, did you see a GP?” in order
to capture costs associated with GP visits. These unplanned visits could be
captured in a patient diary. Recording data in a diary may be more suitable
for health resource use (costs) because usually the total cost per patient can
be derived in their personal real-world setting. Diary data however have a
certain notoriety for being incomplete (missing data), unless electronic dia-
ries are used, and this could result in unreliable health resource use being
recorded. Compliance of medication might be captured through a diary, but
the simplest option is to count the returned tablets. For intravenously (IV)
administered infusions or radiotherapy given at hospitals, the CRF would
be more appropriate.
Recording data in the CRF (or electronic CRF (eCRF)) involves patients
being asked directly about resource use that has taken place. Sensible
judgment is needed to determine whether a diary is needed or whether
CRFs can be used to record resource use. It might be useful to run a test
or pilot of the CRFs, especially if electronic data capture, such as eCRFs,
are used. Chapter 5 provided some examples of CRF designs for health
resource use.

6.3.3.4 Sample Size Methods for Cost-Effectiveness


Traditionally, sample size calculations are carried out to design clinical trials
to detect clinically relevant differences between two treatments. In clinical
trials designed for economic evaluation, it is usual to collect patient-level
200 Economic Evaluation of Cancer Drugs

data on resource use (costs) in addition to efficacy and safety. While there
may be arguments to power a clinical trial for demonstrating clinical benefit,
the arguments for powering for cost-effectiveness have not been fully appre-
ciated. There are several reasons why calculating a sample size for cost-effec-
tiveness may not be considered relevant.
First, it is argued that without showing a clinical effect there is unlikely
to be any cost-effectiveness argument – therefore efforts should be directed
toward the clinical endpoint. Second, although the magnitude of a clinical
effect can be justified, the size of a measure of ‘effectiveness’ may be less
clear. For example, in trials where the endpoint is a combination of quality
of life and quantity of life, the composite ‘economically relevant’ QALY dif-
ference is usually unknown. Third, ethics committees rarely query a trial
protocol that has a cost-effectiveness objective but no sample size justifica-
tion, whereas they would almost certainly query a protocol with no sample
size justification for the primary clinical outcome. Finally, it may not be con-
sidered ethical to power a trial purely for demonstrating cost-effectiveness,
because it is construed as an economic argument and not a clinical argument
for the trial.
Despite the above reasons, the risks for a new experimental treatment that
‘misses’ the opportunity to demonstrate value either for reimbursement pur-
poses or for national health policy reasons are real and should not be under-
estimated (many cancer drugs get rejected for reimbursement). This is akin
to a ‘false negative’ where the likelihood of showing cost-effectiveness is real,
but the trial may not have been designed appropriately to demonstrate i.e. it
lacks power. It is perhaps for this reason that funding bodies such as CRUK
or the NIHR have sections on sample sizes for cost-effectiveness objectives
in their funding application forms.
In the UK, NICE offers an advisory service that provides advice to opti-
mize designs of clinical trials aimed at demonstrating cost-effectiveness.
The ability to power for cost-effectiveness, therefore, allows researchers,
policymakers, funders, and drug companies to plan for another level of
uncertainty toward the path of licensing and reimbursement or chang-
ing the standard of care in an economically viable way. A detailed expo-
sition of sample size methods for cost-effectiveness in a frequentist and
Bayesian framework is found in Khan (2015, Chapter 8) and Willan and
Briggs (2006).
In the past, sample sizes for cost-effectiveness were calculated in different
ways. One method was to calculate a sample size for clinical effects and costs
separately, and then choose the maximum of the two. However, it is unclear
what an important difference in costs between treatment groups is sup-
posed to be. Moreover, this approach treated costs and effects as indepen-
dent. In practice, patients with smaller treatment effects could have larger
costs because they might be treated with other more expensive treatments,
or might have stopped taking the treatment (e.g. for lack of effect) or side
effects, which might also result in increased costs.
Designing Cost-Effectiveness into Cancer Trials 201

In clinical trials of efficacy, the sample size depends on several variables,


including the hypothesized clinically relevant difference (e.g. measured
using a hazard ratio) and the variability associated with the clinical effect.
A sample size formula might look like the one below, used for continuous
measures (as in a two-sample t-test):

nE = ( zα + zβ )
2 (σ 2
E1 + σ E2 2 ) (6.1)
∆ 2
ER

In this formula, n is the sample size for each treatment group, σ E12
and σ E2
2
are
expressions of the variability of the response in each of the two groups, ∆ ER 2

is the clinically relevant difference, and the values of Zα and Zβ are special
values taken from published tables (or a software) that express the risk of a
false positive (often 5%) or a false negative (often 20%).
A sample size calculation for a cancer trial is calculated in a different way
in that it is generally driven by the number of events (death or progression
events) and the treatment effect (clinically relevant difference) is the hazard
ratio (HR).
Sample Size Formula for Cost-Effectiveness

 2 2
n1 = ( zα + zβ ) 
( 2 2 2
) ( 2 
2 r k σ E1 + σ C 1 − 2kρ1σ E1σ C 1 + k σ E 2 + σ C 2 − 2kρ2σ E 2σ C 2  )
r [ k ∆ ER − ∆ CR ]
2

(6.2)

n2 = ( zα + zβ )
2  ( ) ( )
 r k 2σ E2 1 + σC2 1 − 2kρ1σ E1σC1 + k 2σ E2 2 + σC2 2 − 2kρ2σ E 2σC 2 
.
[ k ∆ ER − ∆ CR ]2
For example, for a drug that is expected to improve OS by 20% (i.e. HR =
0.80) would be:

(1.96 + 1.28)2 / 0.25∗ log (0.80)2 = 845 death events needed (90% pow
wer )

The sample size here is driven by the number of events. If we assume the
probability of an event is 0.75 (this is usually calculated in some complicated
way) over the year (e.g. late stage lung cancer patients), the sample size is
1,127 or about 564 patients per group.

6.3.3.5 Sample Size Formulae for Cost-Effectiveness: Examples


In the above formula and calculations, there is no mention of costs for any of
the treatment groups or a (co-)relationship between costs and clinical effects,
nor is there detail of a cost-effectiveness threshold, nor anything about a
cost-effectiveness ratio or benefit. When cost-effectiveness is also considered
202 Economic Evaluation of Cancer Drugs

in the same trial (cost-effectiveness trial), the sample size formula needs to
include additional factors:

(a) The way the effectiveness element is evaluated – i.e. using the ICER
approach or the INB approach
(b) Costs in each group
(c) Variability of costs in each group
(d) Correlation between costs and effects in each group
(e) The cost-effectiveness threshold

Taking into account the above factors (a)–(e) makes the sample size formula more
complicated. In some situations (such as using Bayesian methods), a formula
(even if it is complex) might not be available. The sample size in these situations
is calculated using simulation methods. Although, the sample size problem can
be approached through simulation (see Khan. 2015), it is better (easier) to deter-
mine sample sizes by using a sample size formula rather than having to doing
lots of programming. In a clinical trial for comparing two groups, where costs
and effects are collected prospectively, we propose using the following sample
size formula when patient-level data are available (see Khan, 2015).
In equation (6.2), the denominator of the expression k∆ER – ∆CR expresses
the cost-effectiveness argument in terms of the desired incremental net ben-
efit (INB) (see Chapter 1). We use λ= K to represent the cost-effectiveness
threshold for the remainder of this chapter. Therefore, in reference to point
(a) above, the measure of effectiveness is a single numerical quantity that
combines separate measures of clinical efficacy and costs to form a measure
of economic value worthwhile pursuing.
For example, if an existing treatment compared with the current standard
of care offers an INB of £5,000 (measured in money terms), then we might
want to ensure the new treatment has a sample size that can show INB values
>£5,000. Sometimes, the measure of effectiveness might also consist of two
separate measures: HRQoL and efficacy, to form a QALY. Therefore, the terms
∆ER and ∆CR in K∆ER – ∆CR are the mean difference in QALYs and the mean
difference in costs between treatments, respectively. Therefore, the objec-
tive is to estimate the sample size that yields a specific INMB based on mean
effects and costs. Table 6.4 summarizes the inputs for the sample size formula.

Example 6.3
In a two-group trial (treatment A vs. B) in a 1:1 allocation (r = 1), the fol-
lowing parameters are considered in order to calculate the sample size
for showing an INB of at least £3,000.
We assume that the mean costs of the new experimental treatment aver-
ages to £20,000 per year (treatment A). The mean costs for treatment B, the
current standard of care are £10,000, substantially cheaper. The new treat-
ment will need to show a positive INMB, given that it is more expensive.
Designing Cost-Effectiveness into Cancer Trials 203

TABLE 6.4
Summary and Explanation of Terms for Equation (6.2)
Term Definition
Population variance for QALY measure in treatment group 1
σ Ej
2

Population variance for QALY measure in treatment group 2


σ Ej
2

Population variance for cost in treatment group 1


σCj
2

Population variance for cost in treatment group 2


σCj
2

n Sample size per group (multiply by 2 for the total sample size in a 2-group trial)
ρj Correlation coefficient between efficacy (QALY) and cost for the treatment group 1

ρj Correlation coefficient between efficacy (QALY) and cost for the treatment group 2
K=λ The cost-effectiveness threshold unit cost the healthcare provider is prepared to pay
to obtain a unit increase in effectiveness (in the UK, this is £20,000 to £30,000)
∆ ER Mean difference between group 1 and 2 in terms of (QALY) effects

∆ CR Mean difference between group 1 and 2 in terms of costs


r Patient allocation ratio between two treatment groups (e.g. in 1:1 allocation takes
value 1)
zα Value associated with false positive rate (typically 1.96 for 5% level)
zβ Value associated with false negative rate (typically 1.28 for 20% (80% power) level)

The clinical effects in this (hypothetical) example are based on mean


estimates of QALYs and survival rates from earlier trials. In the experi-
mental arm, the HRQoL based on EQ-5D is estimated to be a mean of
about 0.60 in each group. The mean survival for the new treatment was
17 months; for the standard treatment group it is expected to be around
14 months. The QALY is therefore about 10.2 (0.60 × 17) for treatment A
and 9.4 (0.60 ×14) for treatment B. If the cost-effective threshold is £30,000,
the number of patients required to show a positive INB (e.g. £5,000) on
average assuming no correlation between costs and effects (ρ1 = ρ2 = 0)
although in practice a positive correlation would be the rule.
We assume the variance of costs in the new and standard treatments
are £10,000 and £6,000 respectively and for QALYs are 10.2 and 8.4 for
treatments A and B respectively. The WTP value of λ = K = £30,000, gives
a value for the denominator in equation (6.2), (λ*∆E – ∆C)2 of : £30,000 ×
(10.2 – 9.4) – (£20,000 – £10,000) = £24,000 – £10,000 = (£14,000)2.
The value of £14,000 tells us that when the WTP = £30,000, the net ben-
efit of the new treatment is at least £14,000. The sample size for each treat-
ment group for a WTP of £30,000 in a 1:1 allocation, assuming a mean
QALY difference of 0.8 and a mean cost difference of 10,000 using equa-
tion (6.2), : is computed as:

( )( )
n = (1.96+1.28 ) * 1 * £30,000 2 * 3.5+£10,000 + £30,000 2 * 2.9 + £6,000 
2
 
 1 * £30, 000 × (10.2 − 9.4 ) − (£20, 000 − £10, 000 )
2

n = 605
204 Economic Evaluation of Cancer Drugs

Therefore, a sample size of 605 per group (1,210 in total, allocated in a


ratio of 1:1) is likely (80% power, 5% significance) to:

(i) Show an INB > 0.


(ii) Reject H0: that the new treatment would offer no value to the
(tax)payer – i.e. INB < 0.
(iii) Be unlikely to have sufficient power to show INB values <
£14,000. From a payer perspective, the larger the INB, the better.

6.3.4 Treatment Pathways
In the same way clinical pharmacologists attempt to understand the mecha-
nism of action of a drug, health economists seek to understand the mecha-
nism of cost structures. This requires an understanding of the full treatment
pathway. Without appreciating the pathway, key component costs could be
lost. An example of a treatment pathway for diffuse large B-cell lymphoma
is given.
Diffuse large B-cell lymphoma (DLBCL) is a fast-growing, aggressive
form of lymphoma. It is fatal if left untreated, but with timely and appro-
priate treatment, approximately two-thirds of all patients can be cured.
The standard treatment of advanced DLBCL is combination chemother-
apy plus immunotherapy. The most common chemotherapy regimen for
advanced DLBCL is called R-CHOP. R-CHOP includes rituximab, cyclo-
phosphamide, doxorubicin, vincristine, and prednisone. The first four
drugs are injected into a vein intravenously over the course of 1 day, while
prednisone is taken by mouth for 5 days. Patients are treated with up to 8
cycles of this combination. Approximately one-third of the patients who
fail to respond to this treatment regimen, receive more aggressive treat-
ment, such as bone marrow transplant, if they are fit and young, or pallia-
tive treatment (Figure 6.6).

6.3.5 Time of Generic/Competition Entry


An entry of a generic drug onto the market has huge implications for
demonstrating cost-effectiveness. If the reimbursement strategy is not
planned adequately, revenue could be lost. Even with 6 months of patent
exclusivity for an agreed price of €50 for a 28-day regimen, an estimated
market of 20 million patients in Europe can yield in excess of €6 billion
sales. For a drug receiving market authorization on 1 January 2010 and
a reimbursement decision made on 1 December 2010 (because activities
relating to health economics were delayed until after approval) with a
generic entry date of 1 June 2011, about 1 year of potential sales revenue is
lost. If the cost of the trial is £50 million and revenue in 1 year is £200 mil-
lion, the impact of late preparation for reimbursement/ cost-effectiveness
activities is not small.
Designing Cost-Effectiveness into Cancer Trials 205

FIGURE 6.6
Example treatment pathway for diffuse large B-cell lymphoma (DLBCL).

6.3.6 Treatment Compliance
Treatment compliance (or lack of) is related to efficacy, safety, and efficiency.
Lack of compliance may result in lack of effect, or even safety issues, par-
ticularly where the therapeutic window is narrow. Patients who take less
medication or drop out of the trial are likely to be the same ones who have
had more side effects.
Noncompliance in these patients is likely to result in greater costs. Patients
who drop out also tend to be those who are more ill or where there is a lack
of effect. Compliance during open label extensions is particularly important
for monitoring (Hughes et al., 2001) as it may reflect what is likely to hap-
pen in real practice. Clinical trials often report trial results using per proto-
col population analyses that take into account both protocol deviations and
treatment compliance. Only actual dosing needs be considered rather than
planned treatment – which can contrast sharply with the ITT definition. In
some ITT definitions, efficacy can be based on randomized patients (regard-
less of whether study medication was taken). Effectiveness should be based
on those randomized patients who took study medication and therefore dif-
fer from usual clinical practice. Treatment compliance is likely to be higher
in clinical trials because of the ‘Hawthorne effect’ as compared to real-life
practice.
In some instances, not all the drug is administered and so some is sub-
sequently wasted (sunk cost). For example, some intravenous infusions are
very expensive (e.g. the cost of rituximab is $500 per infusion). If only 70% of
206 Economic Evaluation of Cancer Drugs

the dose is used in the infusion, then the actual cost per mg used is $350 on
the condition that the remaining 30% are used in another patient.

6.3.7 Identify Subgroups/Heterogeneity
Heterogeneity has been the subject of much interest (Claxton, 2011). It is often
found that some (pre-specified) subgroups of patients in a clinical trial dem-
onstrate greater benefit than the whole sample studied. If a large treatment
effect is observed in a given subgroup of patients (e.g. those with the pres-
ence or absence of a biomarker), the argument for cost-effectiveness can be
more forceful in this subgroup. For example, Lee et al. (2012) showed greater
OS improvements with erlotinib compared to the standard of care in those
lung cancer patients who showed evidence of rash within the first 28 days.
There was no improvement in OS and only a modest improvement in PFS in
the whole sample. An economic evaluation (Khan et al., 2015) demonstrated
a stronger cost-effectiveness/value argument in this subgroup of patients
with rash. However, rash is not a pre-randomization stratification variable
and care should be taken with such subgroups for selection bias.
Reimbursement agencies (and regulatory agencies) may specifically look
for effects/lack of effects in subgroups of patients. For example, in the com-
parison of lenalidomide with dexamethasone, several subgroups were con-
sidered; ICERs were presented for each of these subgroups (NICE HTA 171;
ERG Report 2008). It is not uncommon to see a submission to reimbursement
agencies where various subgroup analyses have been presented. In Examples
6.1 and 6.2, treatment benefit was highlighted for the chromosome 17p dele-
tion biomarker for treatment with venetoclax (tested for prior-to-treatment
initiation) for patients with lymphocytic leukemia (NICE HTA TA487, 2017).
Addressing subgroup heterogeneity of treatment benefit is therefore impor-
tant, not only for reimbursement requirements – but also to identify those
subsets of patients who have a higher chance of receiving benefit (and show-
ing cost-effectiveness) with a view to hopefully obtaining a premium price
for these groups of patients.
The sources of heterogeneity might present through diagnostic testing, pre-
specified genetic subgroups, or even differences in trial results. However, the
INMB is unlikely to be >0 for every patient within a subgroup. The health
benefits and effects are presented as averages (means), therefore, the INMB
will be either higher or lower than the mean for some patients in that sub-
group. At best we can say that the chance of an INMB > 0 is higher in the
subgroup of patients compared to patients not in the subgroup. Subgroups
should be predefined to avoid data dredging resulting in false positives.

6.3.8 Early ICER/INMB
There may be interest in generating an early estimate of the ICER/INMB
to determine where the new treatment might fall in the cost-effectiveness
Designing Cost-Effectiveness into Cancer Trials 207

plane, possibly using Phase II trial data. However not many Phase II tri-
als collect health resource use data for cost-effectiveness purposes, although
critical safety data and secondary endpoint data could be used from Phase
II trial data to provide an initial estimate of the ICER. Costs and resource
use in a Phase III trial may be different to those of Phase II trials however.
For example, dosing may change, some endpoints may be dropped, or the
target population to treat may change. The open label extension of a Phase II
trial could, however, provide some insight into how patients respond to the
new treatment in real-world situations, especially if the target populations
between Phase II and III are the same. In Phase II trials, the selected dose
may also be the same dose used in a larger Phase III trial; in some cases, it
is hard to differentiate a Phase II trial and a Phase III trial, other than the
sample size.

6.3.9 Multicenter Trials
One of the realities of clinical trials, particularly large Phase III trials, is
that all patients are very rarely found in a single center (and sometimes a
single country). For Phase I and small Phase II trials this might be true, but
it is not the case for a large Phase III trial or a trial where rare tumor types
are treated. Local reimbursement agencies may ask whether patients from
their country were represented in the trial; and may raise an eyebrow if in a
10,000-patient multinational Phase III trial, patients from their country were
absent. However, for some European countries, it is assumed that treatment
effects are generalizable (Willke, Glick, Polsky, & Schulman, 1998).
Although generalizability might be true for clinical effects because the biol-
ogy and mechanism of action of a treatment should be the same in a homog-
enous group of patients whether from Italy, Spain, or the UK, this may not
be true for costs and other outcomes in an economic evaluation. Differences
between costs of GP visits, for example, may just reflect underlying policy
and societal differences between the countries. What is of particular interest
is whether cost differences between treatments differ between centers (treat-
ment by center interaction). Such interactions are not statistically powered
(i.e. sample sizes are not large enough to determine whether cost differences
between treatments depend on the country) and there is some concern that
cost differences between centers are not being addressed properly in multi-
center trials (Grieve, Nixon, Thompson, & Cairns, 2007; Thompson, Nixon, &
Grieve, 2006) (Manca, Rice, Sculpher, & Briggs, 2005).
The specific question of an economic analysis in a multicenter trial is how
the INMB varies between centers and how best to capture and then interpret
this variability. One approach is to address this concern through statistical
techniques such as multilevel modeling. The benefit of using a multilevel
(or random effects) model is that an estimate of INMB can be generated
for each center or country. While this may appear attractive and useful in
some cases, several considerations should be noted before a multilevel or
208 Economic Evaluation of Cancer Drugs

random effects approach is considered for multicenter analysis in an eco-


nomic evaluation:

(i) First, it is assumed that centers are sampled at random and are
representative of a population of centers. In practice, centers are
selected based on competitive recruitment and other logistical rea-
sons, therefore centers in a clinical trial are unlikely to be random.
In some disease areas, we can be almost certain the centers were not
selected randomly (e.g. only a handful of specialist centers may offer
stereotactic whole brain radiotherapy).
(ii) Second, although modeling does allow for an estimate of the INMB
across all centers and addresses the question of whether effects are
generalizable across centers, the practical benefits of presenting cost-
effectiveness results for each center for reimbursement purposes can
be limited (as well as restricting use if only one subgroup is shown to
benefit). For example, the UK BNF reports pricing for the UK in gen-
eral and not by location. However, reporting the CEAC in a multina-
tional trial by taking into account country-specific costs and effects
may have some benefit for local reimbursement decision-making.
Careful design considerations such as the number of patients within
each country and demographic characteristics of each country
should be examined so that reasonable values of the INMB and its
precision can be estimated.
(iii) Third, a multilevel model or random effects model may not always
be necessary. A fixed effects model with a treatment by center inter-
action will provide a simple yet robust conclusion as to whether the
INMB differs between centers, especially when these were not cho-
sen at random. Given the issues raised in (i), generalizability may
be less of a concern because the centers in themselves are seldom
randomly selected in a clinical trial. Although there is a loss of (sta-
tistical) degree of freedom if there are many centers, a loss of power
to determine the statistical significance of an interaction may not be
of great concern because clinical trials rarely power for cost-effec-
tiveness, let alone interactions for showing cost-effectiveness.

The validity of any assumptions of multilevel models (MLM) needs to be


critically evaluated before one can be sure about the conclusions (Grieve et
al., 2007). For example, it is assumed that the parameters (e.g. mean costs)
are exchangeable (Spiegelhalter & Best, 2003), that is, it is assumed there are
no a priori reasons why mean costs in one center are higher than in another
center, and further it is assumed that the variability of costs is constant
between centers. Reasons for differences in INMB could be due to differ-
ences in demographics between centers or countries. For example, in a clini-
cal trial where there are more patients with a disease diagnosis in one or
Designing Cost-Effectiveness into Cancer Trials 209

two centers compared to the others, then the INMB is likely to be different
between centers.

(iv) Multilevel models (MLM) might be valid where the data is clus-
tered or correlated within a center such that the usual OLS esti-
mates are biased and inefficient (Manca et al., 2005). But this can
also be remedied using approaches such as weighted least squares
or other approaches for the OLS estimates to remain valid so long as
the other conditions of the Gauss Markov theorem hold. It has also
been suggested that MLMs are more appropriate for determining
why resource use (e.g. costs) vary between centers (Grieve, Nixon,
Thompson, & Normand, 2005).

Figure 6.7 shows an example of clustered data where the INMB (Y-axis)
shows considerable heterogeneity between centers (X-axis) and in particular
varies for males and females across the 30 centers. One might expect the
INMB across centers (the hospitals where the patients are recruited) to be
similar (since there may be no real reason why a new treatment is more or
less cost-effective in one center compared to another). In addition, for some
sites, the INMB was markedly different between males and females – this
might be expected, or again, have occurred by chance. A statistical test for
differences in INMB between sites and also a center by gender interaction
might (if there is sufficiently large sample size) help identify such differences
in the statistical sense. One objective of MLM might therefore be to adjust
observed differences in net monetary benefit (NMB) between treatments for
the differences due to centers and gender.

FIGURE 6.7
Clustered data within each center.
210 Economic Evaluation of Cancer Drugs

6.4 Case Study of Economic Evaluation of Cancer Trials


Table 6.5 compares design issues relevant to cost-effectiveness analyses from
two recent cancer trials reported in an HTA (NICE HTA TA516, 2018). In
Chapter 9, we will interpret in detail the results from both of these evalua-
tions. For now, we briefly summarize the trial design features.

6.4.1 TA516 Cabozanitib + Vandetanib


Two Phase III RCTs of cabozanitib versus Best Supportive Care and vande-
tanib versus placebo (Table 6.5) were conducted in what appeared to be of
good quality trials.
The trial endpoints were well defined (PFS and OS), and assessment time
points for measures such as HRQoL were defined. However, HRQoL data
were not collected in the trial and external sources were relied on (it is not
mandatory for licensing to collect EQ-5D). A complicating feature of this trial
was that four different data sets were defined, each varying on the labeling
use and/or the specific subpopulation. The comparator was considered to be
BSC for the licensing authorities, however for economic evaluation, vande-
tanib and cabozanitib should have been compared against each other. This
had to be done using an indirect comparison, which was inadequate because
of differences in the trial populations.
A further, important problem was that the OS was confounded because
patients took vandetanib upon progression. This is likely to dampen the size
of the effect between vandetanib and BSC for OS. The economic evaluation
was based on the list price and not the purchasing discounted price. Hence
the reason in the recommendation for discounted prices.
Results show that it is clear that price played a fundamental role in the
calculation of the ICER. Even if the costs accruing from adverse events were
lower, the ICER is unlikely to fall.

6.5 Summary
In this chapter, we have outlined the reasons for collecting economic data in
a clinical trial, highlighting the importance for internal validity of RCTs. We
discussed a number of trial designs including the challenges innovative trial
design present for cost-effectiveness analysis, particularly during interim
analysis. We presented the key trial design components important for cost-
effectiveness and then summarized with a case study in the HTA literature.
The next step is to discuss how these fit in the context of economic models.
Designing Cost-Effectiveness into Cancer Trials 211

TABLE 6.5
Comparison of Cost-Effectiveness Considerations from 2 Licensed Drugs
Detail of HTA Cabozanitib + vandetanib TA516
Disease indication Medullary thyroid cancer
Year 2017
NICE recommendation Recommended only if the company provides cabozantinib with the
discount agreed in the patient-access scheme
Population Rare cancer (90 cases in 2014); Cabozanitib: progressive,
unresectable locally advanced or metastatic medullary thyroid
cancer
Vandetanib: aggressive and symptomatic medullary thyroid
cancer unresectable locally advanced or metastatic medullary
thyroid cancer
Price Cabozanitib: £4,800 (84 x 20 mg pack)
Vandetanib: £5,000 (30 x 300 mg pack)
Duration of treatment Until PD or no further clinical benefit
Comparator Vandetanib vs. cabozanitib or BSC (including radiotherapy)
Design Two Phase III trials (parallel group): double-blind RCT
Cabozanitib vs. BSC (n = 330)
Vandetanib vs. placebo (n = 331)
Sample size N = 330 (EXAM Trial); 80% power
N = 331 (ZETA Trial)
Time horizon 20 years
Multicenter Yes
Multi-country Yes (European)
Comparator in trial BSC
Primary outcome PFS
Secondary outcomes OS, ORR, DoR, disease control at 24 weeks, RET mutation status,
time to worsening pain
Observed PFS effect C vs. P: 11.2 vs. 4.0 mo (HR = 0.28) [n = 330]
V vs. P: 30.5 vs. 19.3 mo (HR = 0.46) [n = 331]
Target PFS effect C vs. P: HR = 0.50
V vs. P: TBD/not reported
Observed OS effect C vs. P: 26.6 vs. 21.1 mo (HR = 0.85) [n = 330]
V vs. P: 28.0 vs. 16.4 mo (HR = 0.99)
Subgroups Yes, Biomarker RET +ve showed improvements in PFS and OS
Target OS effect C vs. P: HR = 0.67
V vs. P: NR
PPS Not identified
Utility [ERG utility] FACT-G mapped to EQ-5D, no utility recorded in trial
Pre-progression: 0.84 [0.8]
Post-progression: 0.64 [0.54]
Decrements of –0.11 for adverse events
Incremental cost C vs. P: £72,734
V vs. P: £79,745
Incremental QALY C vs. P: 0.48
V vs. P: 0.23
(Continued)
212 Economic Evaluation of Cancer Drugs

TABLE 6.5 (CONTINUED)


Comparison of Cost-Effectiveness Considerations from 2 Licensed Drugs
Detail of HTA Cabozanitib + vandetanib TA516
HRQoL No overall improvement in HRQoL based on a condition-specific
measure (FACT-G)
ICERa C vs. P: £150,874
V vs. P: £352,508
Other ICER Range from £66,779 to £195,593, depending on data set
Mean survival time The mean survival time and its standard error were
underestimated because the largest observation was censored
and the estimation was restricted to the largest event time
ERG main concerns
Subsequent therapy Confounding of OS due to subsequent treatments occurred;
confounding/ vandetanib was not compared to placebo but some other
crossover treatment including vandetanib itself
Patients in the placebo arm who discontinued randomized
treatment were unblinded and given the option to take open
label vandetanib
Rank-preserving structural failure time (RPSFT) approach
reported to be unsuccessful in disentangling crossover effect
Modeling Partitioned survival approach based on a partitioned survival
structure implemented using the discretely integrated condition
event (DICE) approach
Extrapolation it was not possible to fit a parametric regression model to the
observed KM data; due to relatively sparse data in the restricted
population as producing KM curves with long steps would lead
to inaccurate estimates of the median survival function when
extrapolated for the economic model
Evidence synthesis Currently, there is one Phase III trial supporting the use of
vandetanib according to the EU label. Therefore, it is not possible
to undertake a meta-analysis at this time. No direct comparison
of V vs. C
NICE has identified cabozantinib as a comparator in this
appraisal, as part of this submission, however Sanofi Genzyme
have not undertaken any indirect or mixed treatment
comparisons for this treatment for two main reasons.
Mixed treatment comparison uncertain due to differences in
populations
Overall No improvement in OS
Improvement in PFS
No improvement in HRQoL
Time-to-pain worsening improved
OS confounded by subsequent therapy
Cost-effective
a Notes:  ICER based on modeling; V: vandetanib; C: cabozantinib.
Designing Cost-Effectiveness into Cancer Trials 213

6.6 Exercises for Chapter 6


1. Compare and contrast the different types of adaptive trial designs
and their implications for cost-effectiveness analyses.
2. Design a clinical trial for cost-effectiveness in any cancer trial. What
important issues should you consider?
3. What is the difference between a retrospective and prospective eco-
nomic evaluation?
4. Distinguish between a multicenter trial and a trial conducted in sev-
eral countries. What are the challenges one might encounter in an
economic evaluation from such trials?
7
Models for Economic Evaluation of Cancer

7.1 Types of Health Economic Models


Economic evaluation of cancer drugs is often conducted using special mod-
eling techniques that are of two kinds: mathematical simulation or statistical
(or stochastic, which refers to analyzing patient-level data). A health eco-
nomic model can take on several forms. Table 7.1 shows the common mod-
els that have been used for HTAs of in the UK. We shall introduce some of
these models in brief and discuss the more ubiquitous ones at some length.
What is noticeable from Table 7.1 is that a type of statistical model called a
‘partitioned survival model’ is more common for the economic evaluation
of cancer drugs. This chapter is more about understanding the key concepts
around these models rather than expecting the reader to ‘build’ models,
although it should provide some direction on how to approach the problem.
Interested readers on building such models may consult references such as
Drummond et al. (2015), Khan (2015), and Gray (2011) others.

7.2 Decision Tree Models


Decision trees are adequate for a decision at one specific point in time and
are mathematical constructs. Although less useful for economic evaluation
of cancer treatments for deriving cost per QALY from survival outcomes,
they have been used for management of cancer; for example, to determine
whether the use of 2-fluoro-2-deoxyglucose positron emission tomography
(FDG-PET) in addition (Kenneth et al., 2001) to computed (axial) tomography
(CT) is helpful in managing recurrent colorectal cancer (CRC). A decision
tree set-up is described in Figure 7.1.
The objective of the decision tree model in Figure 7.1 was to compare the
cost-effectiveness of FDG-PET (experimental) with CT alone (standard of
care) in the management of patients with recurrent CRC. The squares are
decision nodes; circles are chance nodes; and triangles terminal nodes.

215
TABLE 7.1
216

Health Economic Models used in Non-Small-Cell Lung Cancer


Disease
Progression Time
HTA body Indication Status Comparator Analysis Type Model Design No. of States Criteria Horizon Cycle Survival Analysis Drug

pCODR 2013 1L EGFRm+ Accepted Pemetrexed/ CEM (QALY and Partitioned 3 states: PFS; RECIST 10 years Not Kaplan-Meier data Afatinib
(pCODR, cisplatin life-years) survival PD; death reported used up to month
2013a) Gefitinib model 16
Erlotinib Extrapolation of
the data not
detailed

TA310 1L Accepted Gefitinib CUA (ERG have Partitioned 3 states: PFS; RECIST Lifetime 1 month Kaplan-Meier data Afatinib
(NICE, EGFRm+ Erlotinib later survival PD; death (10 (half used up to month
2014b) (TKI-naïve) recommended model years) cycle) 16
CMA) Extrapolated using
standard
parametric
survival models
920/13 1L EGFRm+ Accepted Erlotinib CMA – – RECIST 1 year Per day – Afatinib
(SMC, 2014) (TKI-naïve)
07-2013 1L EGFRm+ Accepted Gefitinib CMA and CUA Partitioned 3 states: PFS; RECIST 5 years – Kaplan-Meier data Afatinib
(PBAC, Erlotinib survival PD; death for 22 months
2013) Cisplatin/ model extrapolated to 10
gemcitabine years
TA296 2L Rejected BSC Docetaxel CUA Semi-Markov 3 state: RECIST Life time 30 days OS and PFS Crizotinib
(NICE, ALK+ (pooled (using AUC) PFS, PD, death (15 (half extrapolated from
2013) chemotherapy) years) cycle) the clinical trials
using standard
parametric
survival analysis
and using hazard
ratios
(Continued)
Economic Evaluation of Cancer Drugs
TABLE 7.1 (CONTINUED)
Health Economic Models used in Non-Small-Cell Lung Cancer
Disease
Progression Time
HTA body Indication Status Comparator Analysis Type Model Design No. of States Criteria Horizon Cycle Survival Analysis Drug

865/13 2L Accepted BSC CUA Markov 3 state: RECIST Life time – Standard Crizotinib
(SMC, 2013) ALK+ Docetaxel PFS, PD, death (15 parametric
(pooled years) survival analysis
chemotherapy) used to
extrapolate
beyond trial data
(including one
single arm trial)
pCODR 2013 2L Accepted Crizotinib/ CEM (QALY and – – RECIST Lifetime – Standard Crizotinib
(pCODR, ALK+ (resubmission) pemetrexed/ life-years) (5 years) parametric
2013b) docetaxel vs. survival analysis
gemcitabine/ used to
platinum extrapolate
agent/ beyond trial data
pemetrexed/
Models for Economic Evaluation of Cancer

erlotinib
TA258 1L EGFRm+ Accepted Gefitinib CUA Semi-Markov 3 state: PFS, RECIST 10 years Per month Kaplan-Meier data Erlotinib
(NICE, model PD, death used up to month
2012) (partitioned 16. Standard
survival parametric
model [PFS]) survival analysis
used to
extrapolate
beyond trial data
TA162 2L Accepted with Docetaxel CUA Markov 3 state: PFS, RECIST 2 years Per month No extrapolation Erlotinib
(NICE, restriction (Partitioned PD, death
2008) (revision of survival
TA162 + TA175 model)
is under
development
[ID620])
(Continued)
217
TABLE 7.1 (CONTINUED)
218

Health Economic Models used in Non-Small-Cell Lung Cancer


Disease
Progression Time
HTA body Indication Status Comparator Analysis Type Model Design No. of States Criteria Horizon Cycle Survival Analysis Drug

TA227 MTx (platinum- Rejected BSC (ITT; stable CUA Partitioned 3 state: PFS; RECIST Lifetime 1 month Standard Erlotinib
(NICE, based first-line disease) survival PD; death (5 years) (half parametric
2011) chemotherapy) Pemetrexed model Subgroups cycle) survival analysis
(non-squamous) defined by used to
response extrapolate
beyond trial data
749/11 1L Accepted Pemetrexed/ CUA Semi-Markov 3 state: PFS, – – – Standard Erlotinib
(SMC, 2012) EGFRm+ cisplatin model PD, death parametric
survival analysis
used to
extrapolate
beyond trial data
220/05 2L Rejected Docetaxel CUA – – – – – – Erlotinib
(SMC, 2006) 2L Accepted Docetaxel CUA – – – – – – Erlotinib
(resubmission
2006)
S0037 2L, EGFRm+ or Accepted Docetaxel CEM (cost per – – – – – – Erlotinib
(CADTH, unknown life-year)
2005)
1066/2005 EGFR+ Accepted Docetaxel – – – – – – – Erlotinib
(TLV, 2008)
07-2013 1L Accepted Cisplatin and CUA Markov 3 state: PFS, RECIST 5 years Month Constant hazard Erlotinib
(PBAC, EGFRm+ (resubmission) gemcitabine PD, death from months 1–3
2013) Gefitinib and then from
month 3 onward
TA192 1L Accepted Gemcitabine/ CUA Markov 4 states: TR, SD, RECIST Lifetime 21 days Standard Gefitinib
(NICE, EGFRm+ carboplatin (partitioned PD, death (5 years) parametric
2010b) Paclitaxel/ survival survival analysis
carboplatin model) was used to
Vinorelbine/ extrapolate
cisplatin beyond trial data
Gemcitabine/
cisplatin
Economic Evaluation of Cancer Drugs

(Continued)
TABLE 7.1 (CONTINUED)
Health Economic Models used in Non-Small-Cell Lung Cancer
Disease
Progression Time
HTA body Indication Status Comparator Analysis Type Model Design No. of States Criteria Horizon Cycle Survival Analysis Drug

615/10 1L Rejected Gemcitabine/ CUA Markov (using 4 states: TR, SD, – 5 years 21 days Standard Gefitinib
(SMC, carboplatin survival PD, death parametric
2010b) Gemcitabine/ models) survival analysis
cisplatin was used to
Paclitaxel/ extrapolate
carboplatin beyond trial data
Vinorelbine/
cisplatin
Pemetrexed/
cisplatin
07-2013 1L Accepted Gemcitabine/ CUA − − − 5 years – Extrapolated (17 Gefitinib
(PBAC, carboplatin months median
2013) in trial)
S0003 2L Accepted Gemcitabine/ − − − − − − − Gefitinib
(CADTH, cisplatin
Models for Economic Evaluation of Cancer

2004) Paclitaxel/
carboplatin
TA181 1L Accepted Gemcitabine/ CUA Markov 4 states: RECIST Lifetime 21 days 30-month trial data Pemetrexed
(NICE, cisplatin (treatment response, (6 years) (half converted to
2009) sequencing) stable, PD, cycle) constant
death probabilities
TA124 2L Rejected BSC CUA Markov 4 states (plus AE – Lifetime 21 days Constant transition Pemetrexed
(NICE, (treatment sub-states): (3 years) probabilities
2007) sequencing) response;
stable; PD;
death
(Continued)
219
TABLE 7.1 (CONTINUED)
220

Health Economic Models used in Non-Small-Cell Lung Cancer


Disease
Progression Time
HTA body Indication Status Comparator Analysis Type Model Design No. of States Criteria Horizon Cycle Survival Analysis Drug

TA190 MTx (following Accepted BSC CUA Trial based + 4 states: RECIST Lifetime 21 days Empirical data for Pemetrexed
(NICE, other treatment) survival pre- (6 years) (half 29 months;
2010a) model (OS progression cycle) extrapolated
AUC) (based on using exponential
treatment and Weibull
cycles); PD; remainder 43
terminal; months
death (OS)
TA309 MTx (following Rejected BSC CUA Markov (based 3 states: RECIST Lifetime 21 days A survival hazard Pemetrexed
(NICE, the same on PFS and pre- (15.99 (half model to
2014a) treatment) OS AUC) progression; years) cycle) extrapolate
post- beyond the trial
progression; (exponential,
death Weibull,
log-logistic,
log-normal,
Gompertz and
gamma)
531/09 (SMC 1L Rejected Gemcitabine/ CUA Markov 3 states: TR, SD, – Lifetime – – Pemetrexed
2010a) cisplatin PD (6 years)
1L Rejected Gemcitabine/ CUA Markov 3 states: TR, SD, – Lifetime – – Pemetrexed
(resubmission cisplatin PD
2009) Gemcitabine/
carboplatin
Docetaxel/
cisplatin
1L Accepted Gemcitabine/ CUA – 3 states: TR, SD, RECIST Lifetime – A survival hazard Pemetrexed
(resubmission carboplatin PD (6 years) model to
2010) Gemcitabine/ extrapolate
cisplatin. beyond the
29-month trial
(Continued)
Economic Evaluation of Cancer Drugs
TABLE 7.1 (CONTINUED)
Health Economic Models used in Non-Small-Cell Lung Cancer
Disease
Progression Time
HTA body Indication Status Comparator Analysis Type Model Design No. of States Criteria Horizon Cycle Survival Analysis Drug

342/07 (SMC 2L Rejected Docetaxel CUA Markov – – 3 years 21 days – Pemetrexed


2008) 2L Rejected BSC CUA Markov – – – – – Pemetrexed
(resubmission
2008)
2L Accepted Docetaxel CUA Markov 4 states: TR, SD, – 3 years 21 days – Pemetrexed
(resubmission PD, death
2008)
PBAC 2010 1L Rejected Gemcitabine/ CUA – – – – – – Pemetrexed
03-2010 cisplatin
(PBAC,
2010)
pCODR 2013 2L Accepted BSC or placebo CUA – – – 9 years 21 days – Pemetrexed
(pCODR,
2013c)
Models for Economic Evaluation of Cancer

Notes: AUC: area under the curve; BSC: best supportive care, CEM: cost-effectiveness analysis, CMA: cost-minimization analysis, CUA: cost-utility
analysis;
DSA: deterministic sensitivity analysis; SA: sensitivity analysis; PSA: probabilistic sensitivity analysis;
NICE: National Institute for Health and Care Excellence; SMC: Scottish Medicines Consortium; PBAC: Pharmaceutical Benefits Advisory Committee;
pCODR: pan-Canadian Oncology Drug Review; TLV: Dental and Pharmaceutical Benefits Agency; CADTH: Canadian Agency for Drugs and Technologies
in Health;
MTx: maintenance treatment; OS: overall survival; PD: progressive disease; QALY: Quality-adjusted life-year; RECIST: Response Evaluation Criteria in
Solid Tumors; SD: stable disease; TR: treatment response;
221
222 Economic Evaluation of Cancer Drugs

+ CT + biopsy other + biopsy liver


other
- biopsy other
+ CT - biopsy liver
liver Surgery (liver only)
- CT + biopsy liver Surgery (extrahepac
other meta found
CT alone
strategy Recycle through
- biopsy liver tree once
stop
+ CT + biopsy other Recycle through
-CT other tree once
liver - biopsy other
stop
- CT Recycle through
other tree once
stop
+ biopsy other
+ CT
Follow-up other + biopsy liver
posive - biopsy other
+ CT
- biopsy liver
liver
-CT + PET other + PET liver
other - PET liver
+ PET liver
- PET other
CT-plus-PET - PET liver
strategy
+ CT + biopsy other Recycle through
other tree once
- biopsy other
stop

-CT + PET liver


liver + PET other
- PET liver
-CT
other
+ PET liver
- PET other
-PET liver

FIGURE 7.1
Example of a decision tree.
Source: Kenneth et al., 2001.

At each branch, the chance of patients in each subsequent ‘group’ (‘recurrent


in liver,’ ‘other part of the body,’ and ‘no recurrence’) is determined from
literature. Costs and effects (life expectancy, surgery) are also determined
from literature. These are then used to determine the incremental costs and
effects of FDG-PET versus CT alone. In this example, all inputs were from
literature. However, one may also build a decision tree model using data
from a clinical trial as inputs, to determine the expected costs and effects
(determined by multiplying the probabilities at each branch with the costs
Models for Economic Evaluation of Cancer 223

associated with each branch). The expected life expectancy and the expected
cost of care associated with each strategy are determined by multiplying the
probabilities of certain events (e.g. recurrence) by the life expectancy and
costs. For example, if the mean cost of surgery is £3,000 and the probability
of recurrence is 0.20, then the expected cost is £600.

Example 7.1: Costs and Effects of Treating


Side Effects of Chemotherapy
Take as another example the need to treat a patient for a single infectious
episode with a given antibiotic. If one is concerned about the possibility
of, say, permanent hearing loss (a rare side effect of chemotherapy), this
simple decision can be decomposed into its constituent parts:

• The treatment alternatives (aminoglycoside vs. another


antibiotic)
• The consequences (permanent hearing loss or no hearing loss)
• The probability of hearing loss

The percentage of patients with permanent hearing loss as a side effect is


estimated to be between 20% and 60% with an average of 40% (Table 7.2).
There are only two possible outcomes (health states): it is either present
(hearing loss is valued as 1) or not (no hearing loss valued as 0). Based
on this simple example we will introduce some further basic concepts.
All patients are in the same initial health state (no hearing loss) and
can move (transit) only to permanent hearing loss or stay in their ini-
tial health state. The average possibility of a transition is expressed as a
probability (between 0 and 1). Each outcome has been assigned a value
(a pay-off). To formally choose between the two alternatives we then cal-
culate the expectation (in its mathematical sense) of each outcome. That
is the value of the outcome in question multiplied by its probability of
occurrence (its incidence in this case) and we then sum these for all out-
comes. So the expected pay-off of aminoglycoside treatment is equal to
(1 × 0.40) + (0 × 0.60) = 0.40 and otherwise equals (1 × 0.10) + (0 × 0.90)
= 0.10. We then choose the option with the minimum expected loss as a
basis for deciding the antibiotic.

TABLE 7.2
Decision Table for Example 7.1
Expected
Outcome Probability Outcome Valuation Pay-Off
Decision Permanent No hearing Permanent No hearing
hearing loss loss hearing loss loss
Aminoglycoside 0.40 0.60 1 0 0.40
Alternative 0.10 0.90 1 0 0.10
antibiotic
224 Economic Evaluation of Cancer Drugs

7.2.1 Further Possible Improvements to the Decision Model


• Valuation of the Outcomes

Instead of using a 0 or 1 (absence/presence) we could have given a score for the


hearing loss state say (0 and 100) (the higher the score the worse the hearing
loss). We could also give the hearing loss a subjective HRQoL value either as
a score or as a utility loss, in this case by interviewing patients with existing
hearing loss or those who lost their hearing after the treatment. Assume that
no hearing loss is anchored at 0 and hearing loss is evaluated at a mean of 0.50
utilities (on a utility scale of 0 to 1). If we had chosen to value the outcomes as
utility gains we could simply change the sign of the utility score (0 gain if no
loss, –0.50 if loss) and choose the option with the highest expected outcome; or
also give a score of 1 to no loss and a score of (1 – 0.50) = 0.50 to hearing loss.
This is what is commonly done in cost-effectiveness analysis where patients are
in some health state at the time of the decision with some (average) utility value
attached to it (either 1 or some lower value depending on their starting health
state). For example, cancer patients who have been diagnosed shortly before
starting treatment would have a utility score below that of the general popu-
lation of the same age and gender because of the psychological impact of the
diagnosis; for some other patients it may be due to the presence of symptoms.

• Introducing More Health States

We now assume that hearing loss can be either complete or only partial, so
we have three health states: ‘partial loss,’ ‘complete loss,’ ‘no loss.’ For each
of these we will have corresponding probabilities. To even further refine the
model, we could also introduce a difference between temporary hearing loss
and permanent hearing loss. By now we are realizing that by adding more
and more states, the decision tree will start to yield more branches and get
more complex. If we were now to introduce another adverse side effect of the
treatment like diarrhea (none, mild, severe) we would end up with 6 × 3 = 18
outcome states in each arm. This is sometimes called the ‘outcomes curse’ as
it rapidly leads to an exponential explosion of health states to account for and
is typical of all ‘state transition’ models.

• Probability Distributions

Instead of using a single fixed (average) probability of some event, one could
use values from a distribution of probabilities. For example, if the hearing loss
probability is between 0.20 and 0.60 (depending on the published literature)
we could use a uniform distribution (a distribution that assumes an equal
probability across possible values, as for example in the roll of a dice where
the chance is 1/6 for each possible outcome) of all possible values between
0.20 and 0.60 instead. The mean of a uniform distribution for some range (a, b)
Models for Economic Evaluation of Cancer 225

is (a + b)/2, which in this case is 0.40 (0.6 + 0.2/2). Alternatively, one could
simulate from some given values (e.g. simulate from a distribution with a
probability of 0.40). Inevitably, for each simulated probability, we would need
to compute our expected costs and effects repeatedly. However, this has now
replaced a fixed value with a random element and introduced uncertainty
(rather than assuming a fixed value). It would be rare to believe the true
probability of hearing loss can only be 0.40. There are likely to be several
ways to introduce uncertainty by using different assumptions around the
probability of distributions (see also Chapter 9 on uncertainty). The same
is true about other estimates (mean costs, mean life-years). A caveat is that
care should be taken when simulating or using probabilities such that their
values sum to 1.

• Extending the Time Horizon of the Consequences (Outcomes)

In a decision tree, events unfold over time in some implicit way, from the
initial decision node to the outcomes of interest. The period (of time) over
which events happen and the way consequences are assessed are not explic-
itly defined in such models. For example, in the above scenario, we could
use a 7-day antibiotic therapy period (the model used just states antibiotic
use, but usually such drugs are taken as a course of treatment). Similarly, the
outcome duration is undefined, i.e. hearing loss is permanent over a patient’s
remaining lifetime. It is up to the analyst to specify the time frame. For
example, extending the duration of antibiotic therapy to 10 days instead of 7
would likely impact the cost of therapy, but also the adverse event probabili-
ties and possibly their severity. If we were to define the outcome in QALYs
then we would also need to specify the time horizon over which these are
calculated (for the next 5 years, 10 years, lifelong).

• Time-Dependent Parameters

Once we consider longer time periods, i.e. extend the time horizon, most
clinical and HRQoL parameters and costs will vary over time. For example,
the incidence of post-surgery complications is highest in the first few days,
or first weeks, after surgery, and then wanes over time. The same can be said
about other treatments such as radiotherapy or chemotherapy. A decision
tree approach accumulates these incidences over its implicit time horizon.
For example, if we observe a scar infection incidence or wound dehiscence
of 5% in the first month and this falls to 2% in month 2, and 1% in month 3,
we could use a 3-month time horizon and consider using an adverse event
probability of 8%. However, we would need to adjust all parameters and
rerun the model for different periods if we change the time horizon. Hence,
it would be better to construct a model that takes into account the time
component.
226 Economic Evaluation of Cancer Drugs

• Repeating Treatments/Events

Many clinical situations involve several repeating episodes like those typi-
cally observed in migraine attacks, asthma, and other conditions. These are
treated in a sequential (step-up or step-down) fashion with a mix of differ-
ent interventions. This is typically the case in oncology where patients are
usually treated many times (therapy cycles) with the same treatment combi-
nation and go through different combinations of treatments (surgery, radio-
therapy, chemo- or immunotherapy, and combinations of these) over several
different lines of therapy (generally from 1 to 7 and sometimes more) and
over time. This therefore calls for a modeling approach where time is explic-
itly modeled.

7.3 Markov Models
A Markov model is an alternative approach to using decision tree models
in tackling some of the above problems (more states, time dependency, etc.).
It can be thought of as a recurrent number of decision trees over time. In
Markov models, time is divided into separate, fixed, discrete time periods
(weeks, months, years). In each time period, patients can ‘move’ or transit
from varying health states. There can be a large number of health states. In
oncology, there are usually three health states: alive and progression-free,
progressive disease, and death. Patients enter the model in the alive and
progression-free health state. Figure 7.2 shows such a model. Once death has
been reached (an absorption state), the patients remain there. Patients may
‘transit’ between the other two states.
All patients are considered to start in a similar health state (alive and pro-
gression-free). Patients can move between health states over time, at discrete

Alive and
Progression Death
Free

Progressive
Disease

FIGURE 7.2
A 3 state Markov model for cancer.
Models for Economic Evaluation of Cancer 227

time points (e.g. by week, month, year). The transition matrix is essentially
a table of probabilities describing the chance of moving from one health
state to another at a given time point. An elementary example of a transi-
tion matrix (see Chapter 4) would in fact be a survival curve where only two
transitions are allowed: from ‘alive to alive’ and from ‘alive to death’ – two
health states. In cancer trials, movements between health states is described
by a transition matrix similar to that shown in Table 7.3.
In Table 7.4, all patients start in the same health state and are considered
similar (homogeneity assumption) in time period zero (t0). In the next period
t1 they could have progressed (e.g. after month-1 follow-up) or have died with-
out progression. Each time period, ti, corresponds to a new state of the system,
i.e. a new distribution of the patients across the different health states.
At the first time point 1 (t1), we see that 25 patients have progressed and
5 died (t1 could be days, months, years, or any sensible discrete division of
time). Using days for time increments would require more calculations for
patients living years; using time increments of years for patients who live
only a few months would be not be sensible either. All the deaths originated
from the alive state in that period. The patients at risk in time 1 number

TABLE 7.3
Example of Probability Transition Matrix
From/to Alive PF Progression Death
Alive PF 0.70 0.25 0.05
Progression 0 0.85 0.15
Death 0 0 1

TABLE 7.4
Example of Markov Chain Calculation for a Given Treatment Group (e.g. Control
Arm)
Total
Time (t i) Alive Stable Progression Death Deaths Total Probability
0.00 100.00 0.00 0.00 0.00 100.00
1.00 70.00 25.00 5.00 5.00 100.00
2.00 49.00 38.75 7.25 12.25 100.00
3.00 34.30 45.19 8.26 20.51 100.00
4.00 24.01 46.98 8.49 29.01 100.00
5.00 16.81 45.94 8.25 37.25 100.00
6.00 11.76 43.25 7.73 44.99 100.00
7.00 8.24 39.70 7.08 52.06 100.00
8.00 5.76 35.81 6.37 58.43 100.00
9.00 4.04 31.88 5.66 64.09 100.00
10.00 2.82 28.10 4.98 69.07 100.00
228 Economic Evaluation of Cancer Drugs

70 alive with stable disease and 25 in progression. At the start of period 2,


from these 70, 49 (70 × 0.7) were still in stable condition, but 17.5 (70 × 0.25)
had progressed and 3.5 (70 × 0.05) had died. Of those in progression 21.25
(25*0.85) were still in progression and 3.75 (25*0.15) had died after progres-
sion. We therefore end up in that period with 49 stable, 38.75 (21.25 + 17.5) in
progression and 7.25 (3.5 + 3.75) dead patients, to which we have to add the
previous deaths (5) to account for all the 100 patients, and so on.

• Different Starting Health States

To improve the above, where we assume all patients start in the same health
state, we could assume that some patients enter the model in differing health
states (any other state, except death), so long as we assume that their tran-
sitions are similar. We could therefore start with a mix of stable and pro-
gressed patients for example, with the progressed patients following their
own transition trajectory from time 0 onward. We leave this as an exercise
for the diligent reader (Table 7.5).

• Time Varying Transition Probabilities

The above are examples of Markov chains because:

(a) The transition probabilities do not vary over time but are fixed (con-
stant) for all periods.
(b) The distribution between the different states at time period t depends
only on the number present at the previous time period, t–1. So, the
process has no memory.

TABLE 7.5
Markov Chain for Patients with Different Starting States
Alive
Time (t i) Stable Progression Death Total Deaths Control All
0.00 80.00 20.00 0.00 0.00 100.00
1.00 56.00 37.00 7.00 7.00 100.00
2.00 39.20 45.45 8.35 15.35 100.00
3.00 27.44 48.43 8.78 24.13 100.00
4.00 19.21 48.03 8.64 32.76 100.00
5.00 13.45 45.63 8.16 40.93 100.00
6.00 9.41 42.14 7.52 48.45 100.00
7.00 6.59 38.17 6.79 55.24 100.00
8.00 4.61 34.10 6.06 61.29 100.00
9.00 3.23 30.13 5.34 66.64 100.00
10.00 2.26 26.42 4.68 71.32 100.00
Models for Economic Evaluation of Cancer 229

One can allow probabilities to vary with time either by creating a set of tran-
sition matrices for different periods or by introducing a time-related equa-
tion for all or some probabilities, linking probabilities and the period. Care
should be taken again to satisfy the summation of the probabilities to 1.

• Relaxing the ‘No Memory’ Assumption

Often in cancer treatments there appears to be a relation to past events. For


example, an occurrence of febrile neutropenia in earlier cycles is likely to
result in occurrence(s) at later cycles. The same can be said for many other
treatment-related adverse events (AE). Resistance or progression observed in
previous chemotherapy treatments may decrease the efficacy of subsequent
treatment lines, including the same drug or drug family. Cross-resistance of
drug A that induces resistance to drug B should also be acknowledged. A
well-known effect is also the increased probability of AEs in relation to the
number of chemotherapy cycles or increasing cumulative radiotherapy dose.
This means that most toxicity-related probabilities will not only increase
with average dosage, but also increase over time (or over cycles of treatment).
These therapeutic aspects can be dealt with, at least partially, by mak-
ing the probabilities dependent on, for example, the total previous number
of time periods or treatment cycles; or, as an alternative, dependent on the
time spent in a given state following the cumulative dose received, by track-
ing these during the model simulation process and adjusting the transition
probabilities accordingly. These models are sometimes called semi-Markov
models.

• Reducing the Time Interval and Using Different Time Intervals in


the Same Model

In reality, time is continuous; the shorter the discrete time intervals, the more
closely they resemble continuous time. To reduce the approximation error
with large intervals, one needs to apply a half-cycle correction (HCCor) or
another type of continuity correction in state transition models. The stan-
dard HCCor consists of adding half a cycle of the state membership at the
beginning of the first cycle and subtracting half a cycle of the membership
at the end of the last cycle. There is some debate about the accuracy of the
HCCor, leading to the recommendation to use the cycle-tree HCCor, which
calculates state membership as the average of state membership at the begin-
ning and end of each cycle (for further details see Mahony (2015); Elbasha
(2016). It is also possible to construct a Markov chain with different time
intervals, for example, a short one for the treatment phase (say monthly or
weekly ) and a longer one post-therapy (Chattwal, 2016). In the next section
we can observe the impact of discrete cycle times on the expected survival
time. More on the different simulation modeling approaches can be found in
Petrou and Gray (2011a).
230 Economic Evaluation of Cancer Drugs

7.4 Continuous Time Markov Models


Reducing the time interval in a discrete Markov chain leads to a continuous
Markov chain model. That is, the time interval t, t+1 (e.g. month 1, month
2) can become smaller (week 1, week 2, or day 1, day 2). As the interval
gets smaller, the discrete Markov model with the time to a transition (e.g.
from progressive disease to death) follows a (continuous) exponential dis-
tribution with rate λ(t). This approach might be used for modeling disease
transmission through infections because the health state of another person
might change at any time due to infection. In cancer, however, health states
are likely to be reported at discrete intervals. Although death can occur at
any continuous time, the transition from progression until death will not
be continuous because progressive disease is determined through scans at
discrete time intervals (e.g. every three months). Time, therefore, progresses
at fixed intervals (e.g. yearly or monthly cycles) whereas in continuous mod-
els, transitions do not have to occur at say the beginning or end of a cycle.
Cost-effectiveness is therefore not influenced by the cycle length (and so
applying corrections, such as the half-cycle correction as noted above, is not
required). We can demonstrate the difference between a continuous and dis-
crete Markov model in Example 7.2

Example 7.2: Discrete and Continuous Markov Models


Let us assume there are only two health states, alive and dead. Alive is
the starting position of the Markov chain (obviously!). The transition to
death is assumed to occur at a constant rate λ. This means that the time
to the event of death follows an exponential distribution exp(–λt). The
average time to death is 1/λ.
Let λ = 0.1, t =1 year, and the cycle length varies according to the form
1/2k (k takes on discrete values from 0 to 6). Hence, the transition prob-
ability (transition from alive to dead) is: P10 = 1 – exp(–0.1 × 1) = 0.0952
(here 1 is the cycle length, or k = 0 in the expression 1/2k). The expected
mean time to death based on the continuous distribution is 1/λ = 1/0.1 =
10. Based on a discrete form, the expected time to death is determined as
1/P10 × (1/2k). Hence for k = 1, this is 1/0.0952 = 10.51. The expected mean
time to death is biased upward. As the cycle length becomes smaller, say
k = 6 (the larger the value of k, the smaller the cycle length and the more
it appears to be continuous), such that 1/2k = 0.015625, P10 =1 – exp(–0.1 ×
0.015625) = 0.9984. Hence, the expected time to death in a much smaller
cycle length is: 1/(1 – 0.9984)*0.015625, which approximates to 10 (in fact
10.01), and has a much smaller bias.
Few continuous semi-Markov models have been applied in cancer
treatment, although some have been applied in cancer-screening applica-
tions (Graham et al., 2014; Hettle et al., 2015; Rivera et al., 2017; Anderson,
D.F., & Kurtz, T. 2015). For this reason, we turn to the most common mod-
els used in economic evaluation, specifically, using Markov modeling
with individual patient data from clinical trials.
Models for Economic Evaluation of Cancer 231

7.5 The Partitioned Survival Model


A common question is what would be the use of building a mathematical
simulation model (like those above) if we have already conducted a trial and
have access to the individual patient level data? This situation is the most
common, where an RCT has been conducted, and a cost-effectiveness analy-
sis is required using the available patient-level data. In contrast to using data
from a reported trial where summary statistics (mean costs, mean effects,
or proportions), available patient-level data eliminates the need to simulate.
However, simulation for assessment of uncertainty is still required.
Using patient-level (Markov-type) micro-simulation also allows for greater
patient heterogeneity, reflecting the original trial data better (having sta-
tistical summaries such as mean values does not tell us the relationship
between these statistics and factors such as age, gender, etc.). This could lead
to a better understanding of the uncertainty in specific subgroups (e.g. older
patients might have larger effects, or where there are only a few patients in
this subgroup).
Despite the advantages of using patient-level data, evidence from a single
trial is seldom enough (except in rare or ‘ultra-orphan’ indications). Hence,
for health policy purposes it is usually preferable (and requested by reim-
bursement agencies) to include all available evidence beyond a single trial.
Here a ‘de novo’ model construction would probably be warranted where
patient-level trial data and published reported data might be synthesized to
provide inputs for the economic model.

7.5.1 Developing an Economic Model Using Patient-Level


Data Using a Partitioned Survival Model Approach
An economic model using data from a cancer trials consist of two parts:

(a) Modeling the efficacy data (survival data)


(b) Modeling the cost and utility data

7.5.1.1 Modeling the efficacy data (survival data)


The total survival experience is divided into progression-free survival (PFS)
and post-progression survival (PPS). PFS is computed from the time from
randomization (or first dose received) until progression or death (whichever
occurs first). Overall survival (OS) is computed from the time from random-
ization (or first dose of treatment) until death. In clinical trials, patients are
not always followed until death (it might be impractical or too expensive to
do so). Consequently, the PFS is modeled along with OS and future (extrapo-
lated) survival proportions for each of PFS and OS are estimated. Survival
proportions over time are typically represented as a Kaplan-Meier curve that
232 Economic Evaluation of Cancer Drugs

plots the proportion of patients alive, died, or progressed (depending on the


outcome) over time. In general, the mean survival time is represented as:

OS = PFS + PPS (7.1)

In general, the approach to modeling is as follows:

(i) Plot the KM curve for each treatment group for each of OS and PFS.
(ii) Extrapolate these survival times to generate mean OS and PFS.
Hence, derive PPS by subtraction (OS minus PFS). This requires
modeling survival beyond the trial follow-up. That is, ‘estimating’
the survival trajectory once the trial follow-up period is complete.
This will require choosing a parametric survival model (Chapter 4).
If all patients have died in the trial, extrapolation is not required. A
judgment should be made whether extrapolating in the case of just
one or two patients who are censored (alive at the end of the trial) is
required (or useful).
(iii) If covariates are included that might influence the predicted sur-
vival times, then these could be included as part of the model.

Modeling the Cost and Utility Data

(i) Generate the mean utility for each treatment group.


(ii) Generate the mean costs for each treatment group, taking into
account any censoring. For both costs and utility, model-based esti-
mates using covariates should be used. For estimation of future costs
(for patients left alive), these could be estimated by multiplying the
predicted survival by the predicted cost (or using methods such as
those of Lin (2861997; 2872000) mentioned in Chapter 5).
(iii) Derive the mean QALY for the PFS and PPS, that is, generate a mean
PFS QALY and a mean PPS QALY for each treatment group.
(iv) Compute the incremental (mean) cost and QALY and derive the
ICER

We now present a step-by-step example of a partitioned survival model


introduced earlier in Chapter 3 applied in the context of an economic evalu-
ation using a case study from a UK NICE HTA.

7.5.2 Case Study of an Economic Model Using Patient-


Level Data: A Partitioned Survival Model
We use the example of the multiple technology appraisal, NICE TA516
case study: cabozantinib and vandetanib for treating unresectable locally
Models for Economic Evaluation of Cancer 233

advanced or metastatic medullary thyroid cancer. Full details are found on


the NICE website (NICE HTA516, 2018).

(i) The indication: medullary thyroid cancer. This is a rare cancer (<1%
cases in the UK). About 71% of patients survive 10 years if they have
Stage III and 21% survive 10 years if they are diagnosed as Stage IV.
Table 7.6 provides a summary of the trial design and results.
(ii) Cabozanitib and vandetanib (CV) are the only disease-modifying
drugs licensed for this indication.
(iii) Comparators and existing treatments: surgery is the preferred
(curative) treatment if the disease is locally resectable. Otherwise,
depending on the prognosis either of:
(a) Vandetanib + best supportive care (BSC).
(b) Cabozantinib + BSC.
(c) Vandetanib (300 mg once daily (QD)) or cabozantinib (140 mg
QD) + BSC (treatment continued until disease progression).
(d) BSC (palliative care – symptom relief).
(iv) Clinical trials: in this evaluation, data from one Phase III trial was sub-
mitted (ZETA trial) for economic evaluation. A further Phase III trial
(EXAM trial) was included for comparing the efficacy but cost-effective-
ness analyses were not provided. The two trials were not considered to
be directly comparable. The differences in the trial population, impact
of prior and post-progression therapies, and presence of crossover in
the ZETA trial (not allowed in the EXAM trial) made it inappropriate
to formally compare the two treatments, in the absence of head-to-head
trials comparing vandetanib with cabozantinib. The results presented
here are for the restricted population of patients (those with symptom-
atic and progressive medullary thyroid cancer plus a biomarker status).
(a) EXAM trial: a Phase III double blind of cabozantinib versus
placebo (BSC), sample size of 330 (219 on cabozantinib, 111 on
placebo).
(b) ZETA trial: a Phase III double blind of vandetanib versus pla-
cebo (BSC).
(v) Sample size of 331 (231 on vandetanib, 100 on placebo).
(vi) A summary of the ZETA trial and cost-effectiveness parameters are
shown in Table 7.6.

Summary of the Methods, Results and Conclusions

(i) Trial Results

Both vandetanib and cabozanitib showed statistically improved PFS (primary


endpoint). Vandetanib does not appear to show improvement in OS after 50%
of the death events (HR = 0.99). However, the lack of statistical difference in
234 Economic Evaluation of Cancer Drugs

TABLE 7.6
Trial Design and Cost-Effectiveness Features of the ZETA Trial
Design EXAM ZETA

Trial Design Phase III double blind Phase III double blind

Sample size 330 (219 vs. 111) 331 (231 vs. 100)
80% power, HR = 0.67 for 80% power, HR < 0.5
OS
Treatment Cabozantinib vs. placebo Vandetanib vs. placebo until PD
until PD
Trial follow-up Median 13.9 months Median 24 months
Primary PFS PFS
endpoint
Secondary OS, response, DoR, safety OS, response, DoR, HRQoL, safety
endpoints
Trial results PFSa 11.2 mo vs. 4.0 mo (median) 30.5 mo vs. 19.3 mo (median)
HR = 0.28 [CI: 0.19, 0.40; p HR = 0.46 [CI: 0.31, 0.69; p <
< 0.001] 0.0001]
OS 26.6 vs. 21.1 mo (median) Not reported/not reachedb
HR = 0.85 [CI: 0.64, 1.12; p = HR = 0.99 [CI: 0.72, 1.38; p = 0.975]
0.241] death events about 50% (50% still
Death events: about 30% alive at end of trial, hence
died (70% still alive by the extrapolation of OS will be
end of the trial, hence needed)
extrapolation of OS will be
needed)
Crossover Yes – crossover from BSC to Yes – crossover from BSC to
present cabozanitib; hence vandetanib; hence adjustment for
adjustment for treatment treatment switching needed
switching needed
Health Horizon 20-year horizon (life time)
economics
Discounting Yes, 3.5% for costs and QALYs
Cycle length Monthly
Perspective UK NHS
Comparator Placebo/BSC
Costs Drug costs £5,000 per 30 × 300 mg and £2,500
per 30 × 100 mg tabletsf
Adverse event Varied depending on the type of
costs AE
BSC costs £788 if in PFS state, £7,195 if in PDc
Palliative care £189.75d per day
Palliative £827
chemotherapy
Monitoring costsf £400 in year 1 (per patient) and
about £200/year in subsequent
years
Utility EQ-5D None collected. Utility determined
from FACT-G using Dobrez et al.
algorithm (see below for details)
PFS utility 0.84
PD utility: 0.64
Efficacy PFS Modeled using Weibull, log-
normal, log-logistic
(Continued)
Models for Economic Evaluation of Cancer 235

TABLE 7.6 (CONTINUED)


Trial Design and Cost-Effectiveness Features of the ZETA Trial
Design EXAM ZETA

Trial Design Phase III double blind Phase III double blind

OS Modeled using Weibull, log-


normal, log-logisticg
Crossover Crossover to cabozantinib Crossover at any time
handling not allowed, but other Extensive crossover could not be
treatments allowed adjusted for statistically (see
(including vandetanib) below)
10% of those on vandetanib
crossed over and 28%k
Uncertainty Not provided PSA using CEAC
Deterministic by varying
parametersi
Incremental QALY
Scenario analysesj
Results ICER Incremental QALY: 1.34
Incremental costs: £42,215
ICER: £31,546h
Probability of CE 88% at a WTP of £50,000/QALY
a Notes: Both central review- and investigator-based assessment of progression were reported.
We only report the independent assessments for simplicity. Other results could be included as
part of sensitivity analyses.
b For the ZETA trial, for the vandetanib group, the median could not be estimated, although for
placebo it was estimable. However, reporting a median for one group and not the other is of
little comparative benefit.
c These were estimated by multiplying the unit costs for items such as scans (£148/scan) by the
frequency in either a PFS state or a PD disease state. mo: months; CI: 95% confidence
intervals.
d These were estimated as the mean of different types of palliative care such as inpatient, spe-
cialist, or outpatient.
e Prior to applying discounts.
f Once a patient starts treatment, they are monitored (e.g. for ECG, biochemistry, etc.) – for
vandetanib this was 8 times during the first year.
g This included extrapolated survival.
h This is based on the manufacturer’s model. The ICER based on the NICE assessor’s analysis
was £66,000/QALY.
i Parameters varied included cost of monitoring, cost of care, AE costs, utilities in PFS and PPS
states, and crossover rates (i.e. the proportion that crossed over).
j An example of one scenario was setting discount rates at 0% and 5%, removing post-progres-
sion costs, alternative methods of utility adjustment, setting the time horizon to 5 and 10 years
and assuming a crossover vandetanib dose of 300 mg (patients could have taken either 100 mg
or 300 g, but in this scenario only 300 mg is assumed).
k What this means is that some patients on both arms received alternative treatments. Since
progression is determined by central review (experts who look at scans to determine progres-
sion), some patients took alternative treatment before the final date of progression was known
(it takes time to get all the information across to determine PD status). Hence, the PFS was also
confounded (that is some of the PFS could have been due to alternative therapy). One
approach is to censor the patient at the date the crossover treatment was taken.
236 Economic Evaluation of Cancer Drugs

OS does not imply that a positive cost-effectiveness conclusion is not possible.


OS was a secondary endpoint and the benefit is likely to be driven by the PFS
endpoint. In addition, part of the reason for a lack of OS benefit is largely due
to patients on the BSC switching over to vandetanib, which makes the treat-
ments appear more similar. This is discussed further below.

(ii) Cost-Effectiveness Model

A partitioned survival model was used by the HTA assessor group using
three health states: progression-free, post-progression (i.e. PD), and dead.
The available patient-level data were used to estimate transitions (survival
rates). Since there was censoring, parametric survival models were used
including covariates for the presence of symptomatic disease and biomarker
status. The empirical Kaplan-Meier for both PFS and OS were generated and
then Weibull functions were selected to model both OS and PFS, assuming
independent (non-proportional) hazards between treatment groups. Once a
model was fitted, for uncertainty (PSA) the parameter estimates from these
models were simulated to generate thousands of QALYs (and compute incre-
mental QALYs). The mean survival times were computed, but it was noted
that these could be underestimated because the longest survival time was
censored. An example of a model fit is shown in Figure 7.3.

7.5.3 Crossover
An attempt was made to adjust the treatment effect for crossover. Methods intro-
duced earlier such as rank-preserving structural failure time (RPSFT) models

FIGURE 7.3
Source page 118 of HTA TA516: empirical PFS with modeled PFS beyond trial follow-up using
a parametric survival model.
Models for Economic Evaluation of Cancer 237

and inverse probability-of-censoring weights (IPCW) method were used. These


methods failed to provide an estimate of treatment effect unconfounded by the
crossover (from placebo to vandetanib). This resulted in an analysis that restricted
the label to patients who were progressive and with symptomatic medullary
thyroid cancer with rapid biomarker doubling (hence the consequences of cross-
over can be severe). RPSFT has been attempted in several previous NICE HTAs.
Since adjustment for crossover could not properly be carried out, the treat-
ment benefit of vandetanib over placebo is likely to be underestimated. If
vandetanib is indeed effective and prolongs survival, crossover is likely to
result in rapid attenuation of any differences in OS. Hence, a traditional ITT
analysis, will underestimate vandetanib’s effect on OS. This also relates to
the theme of estimands raised in Chapter 4.

(iii) Utilities

No EQ-5D utilities were collected during the trial. HRQoL data using a
condition-specific FACT-G were collected. An algorithm to convert FACT-G
responses to time trade-off (TTO) utilities was published by Dobrez et al.
(Dobrez, 2004). The algorithm is based on directly elicited TTO utilities pro-
vided by a large sample of patients (n = 1,433) with cancer for their health state
at the time as well as the patients’ responses to the FACT-G. The algorithm
gave an equation for utility –interested readers can consult the HTA report.
In addition, utility in the progressed state was determined from a few
patients who completed FACT-G while in the PD state. These were not con-
sidered representative and a utility decrement was applied to the utility
measured for the progression-free state (0.84). This decrement was obtained
from an external study Beusterien et al. (2009).

(iv) Costs

Several cost items are shown in Table 7.6 and described in the footnotes of the
table. The model structure states: “At each apportioning, values (in terms of
quality of life and costs) are applied to each state and the resulting accruals
are accumulated as outputs. This happens cyclically until the end of the time
horizon.” In other words, the proportions from each of the PFS and PD states
are multiplied and summed over the time horizon. Little was mentioned by
way of missing data, although its presence was noted.

(v) Results, Conclusion and Issues

The conclusions of the results were:

• Vandetanib was considered to be cost-effective with an ICER of


around £66,000. This was almost twice that of the manufacturer’s
ICER of around £32,000.
238 Economic Evaluation of Cancer Drugs

• Some reasons for the discrepancy are due to:


• The manufacturer included a single (first) progression event
that corresponds to the partition between the PFS and PD health
states. Patients receiving vandetanib after this in either arm are
assumed to continue to do so until death, whereas in practice
these patients are likely to have a second progression before
death. This would likely mean that vandetanib would be discon-
tinued. The manufacturer/company model failed to reflect this.
Although adjustment for crossover was unsuccessful, including
these costs of the drug in both groups would usually be reason-
able, but assuming that all treatment post-progression continues
until death will overestimate the costs of vandetanib in both
groups. This bias strongly favors the intervention group because
a higher proportion of patients receive vandetanib post-progres-
sion within the BSC group (proportion of patients on treatment
post-progression). Therefore, removing the costs of vandetanib
received post-progression in both groups will increase the ICER
from around £32,000 per QALY gained to around £60,000 per
QALY gained.
• Failure to adjust for treatment switching resulting in estimates of
treatment effect that are potentially confounded by the use of open
label vandetanib. The impact of open label vandetanib could not be
addressed. The company’s model included the estimated costs of
post-progression vandetanib used within both the intervention and
comparator treatment groups. The economic comparison made by
the company’s model is therefore vandetanib including continued
use in some patients post-progression versus BSC with vandetanib
use in most patients post-progression. The assessment group noted
that this may not have been useful for decision-making. The manu-
facturer used a mapping model to estimate the EQ-5D utilities from
the FACT-G whereas the assessor used estimates using a published
algorithm.
• The duration over which QALY losses due to AEs are applied and
differences in the inputs relating to the proportion of patients who
discontinued vandetanib prior to PD.

The above example shows the complexity and detail with which HTAs are
undertaken, and the care needed to evaluate all possible uncertainties. In the
above case the drug was recommended, despite a high estimate of the ICER.
However in other cases, drugs are not recommended after the biases have
been addressed.
Models for Economic Evaluation of Cancer 239

7.6 Summary of Cost-Effectiveness Models for


Cancer Used in HTA Submissions
Table 7.1 presented the common economic evaluation models reported in the
economic evaluation of (lung) cancer drugs from a UK NICE HTA perspec-
tive. Many of these models used individual patient-level data from clinical
trials and then simulation to describe the uncertainty around the ICER. Table
7.7 shows a summary of health economic models in cancer. The sources for
the model inputs – health resource use and utilities (not always from trial
data) are shown. For example, for afatinib (NICE TA310, 2014), patient-level
data were taken from two different trials (the LUX-Lung 3 and LUX-Lung
6 trials). Several parametric survival models were fitted to estimate future
(extrapolated) survival rates (exponential, Weibull and Gompertz models);
treatment costs were obtained from BNF 2011; and NHS reference costs were
obtained (NHS reference costs for a given year are on the website, seehttps://
improvement.nhs.uk). Utilities were taken from the clinical trials and all
these data were fed into a model to derive the ICERs. Note that while com-
bining trial data and historical data might be more informative, it could also
introduce greater uncertainty because populations and experimental condi-
tions from historical data may be different.
In Table 7.6, we have highlighted where survival models were used, often
for extrapolation for estimating QALYs. Most submissions extrapolated the
time-to-event survival data to cover a lifetime horizon, either through sur-
vival model fitting or by using Markov assumptions, using observed hazard
ratios for the comparators. In addition, the key cost and utility inputs are
provided for the model. The information in Table 7.7 can be summarized as:

• NICE in the UK have, to a large extent, recommended cancer drugs


for first-line treatment and generally recommended against drugs
for second-line and maintenance treatment. The SMC have also
rejected, or approved with limitations, a number of drugs for first-
and second-line treatment. Outside the UK, there has been a higher
rate of success in terms of approval for use, with only two treatments
(crizotinib first-line in Canada and afatanib second-line in Australia)
being rejected.
• The main sources of efficacy data for the models were from the
clinical trials: LUX-Lung 3, LUX-Lung 6, INTEREST, BR21, EURTAC,
OPTIMAL, IPASS, PARAMOUNT, JMDB, PROFILE 1005, PROFILE
1007 (see references to HTA).
• Survival data were often modeled to generate QALYs using parti-
tioned survival models.
• Costs were determined from literature and established sources
such as National Health Service (NHS) reference costs, British
TABLE 7.7
240

Cost-Effectiveness Models Used in Lung Cancer


Treatment Submission Clinical Data Resource Use/Costs Utilities

Afatinib TA310 1L Estimates of OS and PFS based on data from Treatment costs from BNF 2011 NHS reference costs Health state utilities derived from
(NICE, 2014b) LUX-Lung 3 (28% maturity) and LUX-Lung 6 (2010–11) and HRG data used for hospitalization (currency LUX-Lung and LUCEOR trials
(43% maturity), corresponding parametric codes are provided); and PFS 2011 used for outpatient (Chouaid, 2012) in base case and
survival models visits assumed to be the same across
Exponential, Weibull and Gompertz explored as Other costs sourced from literature reviews such as treatment arms (not between 1L
proportional hazard models; log-logistic and (Billingham, 2002; Lees, 2002) and 2L); other sources used in
log-normal included for sensitivity analysis Costs of AEs only applied in first year (no separate health sensitivity analysis (Doyle, 2008;
(goodness-of-fit provided) states) – frequencies and duration from MTC, LUX-Lung 1, Lewis, 2010; Nafees, 2008)
Second-line (docetaxel) PFS data from INTEREST and LUX-Lung 3 Disutilities sourced from LUX-Lung
trial EGFR mutation test from SMC 2012 (only in the first cycle) 1, LUX-Lung 3, and (Nafees, 2008)
Relative efficacy data retrieved from MTC
07-2013 1L Non-inferiority against gefitinib and erlotinib Utility from LUX-Lung 3 and
(PBAC, 2013a) from MTC – submitted a cost minimization LUCEOR 2 trials. Utility
analysis (CMA). Outcomes were extrapolated decrements from studies conducted
from data at maximum follow-up of 22 months in patients receiving second- or
observed in LUX-Lung 3 third-line therapy (LUX-Lung 1
and Tabberer et al.)
Crizotinib TA296 2L Extrapolation of OS based on the PROFILE 1005 Administration and routine medical management from NHS Utility collected in PROFILE 1007
(NICE, 2013) trial, and PFS based on the PROFILE 1007 trial. reference costs and PSSRU, acquisition cost from BNF 63 using the EQ-5D
Trial 1005 was single arm but more mature. Trial (2012), and resource use from clinical experts. Treatment Calculated EQ-5D in PFS by
1007 was assumed to have the same trends but cost until disease progression weighting the value at each time
included comparators. Both proportional hazard ALK testing costs were applied to the crizotinib arm of the point by the number of patients at
models (Weibull, exponential, and Gompertz) model – assumed as the cost of one test multiplied by the each time point
and accelerated failure time models (log-logistic number of patients needed to be tested to identify one A weighted average utility at the
and log-normal). ALK+ patient (all patients identified eventually) end of treatment was extrapolated
Crossover accounted for using RPSFT, IPTCW Treatment for AEs related to crizotinib and chemotherapy. to post-progression health states
(base case) Cost of AEs occurred the first out of the 30 treatment days No utility decrement was applied to
(only included neutropenia) AE occurrences
865/13 2L Taken from clinical trial, including Phase II (single Costs included cost of medicine and administration, cost of Utility data was derived using
(SMC, 2013) arm) study to increase sample size and ALK-testing (FISH test), routine care before and after EQ-5D from the clinical trials.
extrapolate the data progression Values for BSC were assigned
Price with PAS was proposed by SMC using assumptions
pCODR 2013 2L Extrapolation of the PROFILE 1007 clinical trial All costs relevant for the publicly funded healthcare. Utility data was derived using
(pCODR, using information from Phase II trials EQ-5D from the clinical trials.
Economic Evaluation of Cancer Drugs

2013b)
(Continued)
TABLE 7.7 (CONTINUED)
Cost-Effectiveness Models Used in Lung Cancer
Treatment Submission Clinical Data Resource Use/Costs Utilities
Erlotinib TA258 1L EURTAC clinical trial Cost data sourced from previous NICE technology Utilities were taken from (Nafees,
(NICE, 2012) appraisals in a NSCLC, specifically: TA227, TA181, and 2008); a study commissioned for
TA192 second-line NSCLC, with 100
members of the general population,
using SG and VAS techniques
TA162 2L BR21 clinical trial for erlotinib (Holmes, 2004) for Cost data sourced primarily from published sources
(NICE, 2008) docetaxel Docetaxel drug administration based on data from expert
panel
749/11 1L EURTAC clinical trial Cost data sourced from NICE appraisal of 1L pemetrexed/ Primary study derived from a
(SMC, 2012) cisplatin survey that used the standard
gamble technique with 100
members of the UK public
220/05 2L Indirect comparison from 2 trials (erlotinib vs. Cost data sourced primarily from published sources Primary study from a sample of the
(SMC, 2006) placebo; docetaxel vs. pemetrexed Resource use and AEs were estimated from an expert panel UK general population using
appropriate methods (not
specified)
Models for Economic Evaluation of Cancer

07-2013 1L Main clinical trial was EURTAC, supplemented by Utilities for patients with stable
(PBAC, 2013b) OPTIMAL trial compared to chemotherapy. Also disease and progressive disease
used IPASS, NEJGSG, Study 0054, and directly from (Nafees, 2008)
WJTOG3405 to compare to gefitinib.
Gefitinib TA192 1L IPASS study (gefitinib vs. paclitaxel/carboplatin) Various sources including: BNF, NHS reference costs, Most utilities were sourced from
(NICE, 2010b) Odds ratios for treatment response for the indirect (Nafees, 2008)
comparators were sourced from an MTC Progression-free and therapy were
sourced from an ERG report (2006)
Disutility for anemia was sourced
from Eli Lilly (2009)
615/10 1L Effectiveness of gefitinib and paclitaxel/ Adverse events costs: from industry submissions to NICE Sourced from literature (not
(SMC, 2010b) carboplatin: from the pivotal gefitinib trial results specified)
Effectiveness of the other double chemotherapy:
from an MTC, within which paclitaxel/
carboplatin provided the link to the results of the
pivotal gefitinib trial
07-2013 1L IPASS study (gefitinib vs. paclitaxel/carboplatin) Utilities were adapted from HRQoL
(PBAC, 2013c) data reported in the trial
241

(Continued)
TABLE 7.7 (CONTINUED)
242

Cost-Effectiveness Models Used in Lung Cancer


Treatment Submission Clinical Data Resource Use/Costs Utilities
Pemetrexed TA181 1L Transition probabilities calculated based on Acquisition costs and administration doses taken from the Utilities were taken from (Nafees,
(NICE, 2009) response rates and PFS from JMDB trial data trial and the treatments’ SPCs 2008); a study commissioned for
(pemetrexed/cisplatin and gemcitabine/ UK unit cost sourced from the BNF second-line NSCLC, with 100
cisplatin) by estimations of the median time. Administration costs estimated using national HRGs members of the general population,
Death derived from median time of OS and PFS, Resource use associated with adverse events informed by a using SG and VAS techniques
converted to transition probability. This assumes survey of clinician experts commissioned by the
an exponential form. manufacturer (Lilly)
No direct comparison of gemcitabine/carboplatin Unit costs were sourced from the UK NHS reference costs
or docetaxel/cisplatin was available. 2008
Indirect analysis conducted to adjust the data to
the JMDB population
TA309 MTx Curve fitting and extrapolation of OS and PFS KM NHS reference costs and HRG used for hospitalization and EQ-5D data from a mixed regression
(NICE, 2014a) curves were undertaken (maturity up to 80%) monitoring (currency codes provided) model of the PARAMOUNT trial.
using exponential, Weibull, log-logistic, Treatment costs from BNF 2012 No significant different in AEs
log-normal, Gompertz and gamma (goodness-of- Resource uses (including AEs) taken from the between maintenance treatments in
fit, curves, and values are presented) PARAMOUNT trial trial
Monitoring use from (Beckett, 2012)
531/09 1L Data was sourced from the 29-month pivotal Acquisition costs of pemetrexed/cisplatin and gemcitabine/ Sourced from a utility valuation
(SMC, 2010a) Phase III trial of pemetrexed/cisplatin vs. cisplatin were based on doses used in the trial survey using SG in 100 members of
gemcitabine/cisplatin and extrapolated using The dose for gemcitabine/carboplatin was based on that the general population in the UK
standard survival model fitting applied in the NICE appraisal of pemetrexed for first -line
treatment of NSCLC
Other resources and costs were taken from literature,
guidelines, or using expert opinion
342/07 2st line Median survival rates were taken from a Most resource use associated with adverse events was based Sourced from a study of 100
(SMC, 2008) retrospective subgroup analysis of the results of on clinical expert opinion members of the general population
the clinical study of pemetrexed and docetaxel NHS reference costs were used using SG to value 18 states and
(randomized, open label Phase III study) Data for febrile neutropenia was taken from literature adverse events)

Notes:  AE: adverse events; ALK: anaplastic lymphoma kinase; BNF: British National Formulary; BSC: best supportive care; CI: confidence interval; CMA:
cost-minimization analysis; EGFR: epidermal growth factor receptor; EQ-5D: EuroQoL five dimension; ERG: evidence review group; FISH: fluorescence
in situ hybridization; HRG: healthcare Resource Group; HRQoL: Health-related quality of life; IPTCW: Inverse probability of treatment and censoring
weighted; KM: Kaplan Meier; MTC: Mixed-treatment comparison; MTx: maintenance treatment; NICE: National Institute for Health and Care Excellence;
Economic Evaluation of Cancer Drugs

NHS: National Health Services; NSCLC: non-small-cell lung cancer; OS: overall survival; PAS: patient-access schemes; PBAC: Pharmaceutical Benefits
Advisory Committee; pCODR: pan-Canadian Oncology Drug Review; PFS: progression-free survival; PSSRU: Personal Social Services Research Unit;
RPSFT: rank-preserved structural failure time; SG: standard gamble; SMC: Scottish Medicines Consortium; SPC: summary of product characteristics; TA:
technology appraisal; VAS: visual analogue scale.
Models for Economic Evaluation of Cancer 243

National Formulary (BNF), and Personal Social Services Research


Unit (PSSRU). Some resource use has also been sourced from clini-
cal experts and systematic reviews of the literature, especially for
adverse events.
• Health state utilities have commonly been sourced from a study by
Nafees et al., 2008 and where available from the clinical trial.
• MTC has been used frequently but some submissions use direct
comparisons.

Criticisms of Health Economic Models: Examples from Lung Cancer


Criticisms of some cost-effectiveness models used in lung cancer are shown
in Table 7.8. Many of these models used patient-level data. For example, in
the case of afatinib, there were issues with model fit, mix of the patient popu-
lation (EGFR + ve and EGFR –ve), generalizability, and inadequate survival
projection.

7.7 Summary
We have discussed at some length the types of cost-effectiveness models
used for cancer. A common approach is the partitioned survival model. We
have also seen the common criticisms from HTA assessors in the context of
lung cancer, but these criticisms can be generalized. We have not discussed
some other types of models such as discrete event simulation (DES) models.
A DES model consists of entities (patients) that enter the system, get serviced
(by a resource unit) one or more times, and then exit, or make a delay and
then exit the system. Resource units can be physical agents (doctors, nurses,
other hospital personnel) and/or equipment or materials, or a mix of these, as
is usual in healthcare. DES has not yet gained wide acceptance in the cancer
field. A search in PubMed with search terms ‘discrete event’ AND ‘model’
OR ‘simulation’ AND ‘cancer’ yielded only 34 hits (as of June 2018). Most of
those before 2011 mostly concerned cancer-screening policies. A complete
introduction with practical applications of DES models in healthcare can be
found in Caro et al. (2015).

7.8 Exercises for Chapter 7


1. Distinguish between a decision tree, Markov model, and patient-
level model in the context of cancer.
244 Economic Evaluation of Cancer Drugs

TABLE 7.8
Key criticisms of HTA Submissions: Lung Cancer
Treatment Area Critiques Submission

Afatinib Clinical data The results of an MTC carried out using the currently available NICE, 2014b (TA310) 1L
trial evidence comparing erlotinib and gefitinib were unreliable, SMC, 2014 (920/13) 1L
especially as they included both EGFR-positive and EGFR- PBAC, 2013a (07-2013)
unknown/mixed patient populations in the networks 1L
Eliminating these studies results in predominantly studies with
Asian patients that may not be generalizable to all patients
The MTC relied on proportional hazards, but the ERG suggested NICE, 2014 (TA310) 1L
that the assumptions supporting this were not valid
Despite a statistically significant improvement in PFS, there did not NICE, 2014 (TA310) 1L
appear to be a corresponding gain in OS for TKIs vs.
chemotherapy, based on similar information from previous
erlotinib and gefitinib submissions, after adjusting for crossover
Survival The modeled PFS survival projection for afatinib did not reflect the NICE, 2014 (TA310) 1L
analysis LUX-Lung 3 trial afatinib data from which it was derived
The OS data was not yet mature at the time of submission NICE, 2014 (TA310) 1L
SMC, 2014 (920/13) 1L
PBAC, 2013 1L
Recommended to use RPSFT to account for crossover data. The SMC, 2013 (865/13) 2L
SMC tested this and it gave a higher ICER than in the original
model
Comparator Erlotinib and gefitinib were appropriate comparators in first-line, NICE, 2014 (TA310) 1L
but data were derived from an MTC that had methodological SMC, 2014 (920/13) 1L
limitations
The data in the MTC was only in first-line use. As such, relative
benefits in the second-line setting are unclear
Chemotherapies from LUX-Lung 3 and LUX-Lung 6 were not NICE, 2014 (TA310) 1L
appropriate comparators for the stated decision problem
Proposed comparators should be (a) first-line that includes EGFR PBAC, 2013 1L
testing compared to TKI for positive tests and chemotherapy for
negative tests, and (b) compared to chemotherapy treatment
without EGFR testing
Utilities The generated QALYs were not supported by clinical data PBAC, 2013 1L
The utility decrement between stable and progressive disease was
not supported
Costs Maintenance treatment with pemetrexed before disease progression PBAC, 2013 1L
would only incur costs and no additional efficacy
Others The submission did not present clinical- or cost-effectiveness NICE, 2014 (TA310) 1L
evidence to support the use of afatinib for TKI-naïve patients in a
second- or third-line setting
Since the clinical data was not appropriate to calculate reliable NICE, 2014 (TA310) 1L
ICERs, the ERG proposed a CMA exercise SMC, 2014 (920/13) 1L
PBAC, 2013 1L
The assumption that all patients receiving first-line chemotherapy PBAC, 2013 1L
who progress will receive a second-line treatment was
inconsistent with clinical practice
Assumption that EGFR mutation negative patients in the PBAC, 2013 1L
comparator arm will receive erlotinib in second-line may not be
reasonable
A significant limitation is that the model failed to account for false PBAC, 2013 1L
positives in the EGFR testing, as this would probably not be 100%
and therefore lower the efficacy of afatinib
(Continued)
Models for Economic Evaluation of Cancer 245

TABLE 7.8 (CONTINUED)


Key criticisms of HTA Submissions: Lung Cancer
Treatment Area Critiques Submission

Crizotinib Clinical data Limitations of the MTC include the small number of studies SMC, 2013 (865/13) 2L
included NICE, 2013 (TA296) 2L
Probabilities for progression and mortality were updated in this pCODR, 2013 2L
resubmission from the PROFILE 1001 study in the first
submissions as these were inappropriate
Survival No significant difference in OS data as it was immature and subject SMC, 2013 (865/13) 2L
analysis to high crossover. Extrapolation of the trial data using single arm NICE, 2013 (TA296) 2L
studies may introduce bias
The survival data did not focus on ALK+ patients NICE, 2013 (TA296) 2L
The extra trials used to estimate the survival (PROFILE 1005, GFPC NICE, 2013 (TA296) 2L
05-06, JMEI, and TAX 317) were not comparable to the main
clinical trial (PROFILE 1007)
Comparator The primary relevant comparator is docetaxel NICE, 2013 (TA296) 2L
Utilities The utilities from PROFILE 1007 were overestimated – particularly pCODR, 2013 2L
the assumption that treatment effect was maintained, to some NICE, 2013 (TA296) 2L
extent, post-progression
Costs Testing for ALK mutation was included but assumed the same in pCODR, 2013 2L
both arms – the ERG raised concerns that this underestimated the
costs
Removing the cost of ALK-testing reduced ICER by £1,000 – this SMC, 2013 (865/13) 2L
scenario may be likely in the future
Treatment duration was assumed to last until progression but in SMC, 2013 (865/13) 2L
some instances within the trials treatment continued longer. NICE, 2013 (TA296) 2L
Using trial duration increased the QALY
Discontinuation rule for crizotinib was inappropriate
Other The screening of ALK patients contained three issues: validity of NICE, 2013 (TA296) 2L
the approach where the model did not use the same test that was
used by NHS), cost of the test, and prevalence rate of ALK+
The ERG proposed a 2-year time horizon as opposed to 5 years pCODR, 2013 2L
The base case ICER was estimated to be overly optimistic toward NICE, 2013 (TA296) 2L
crizotinib for several reasons (as mentioned above)
Erlotinib Clinical data Questioned the strength of the indirect comparison/mixed NICE, 2L
treatment comparison due to the comparability of the studies SMC, 2011 (749/11) 1L
included SMC, 2006 (220/05) 2L
Survival Extrapolation method utilized did not appropriately fit the trial NICE, 2006 (TA162) 2L
analysis data for both the intervention and comparator; the method
utilized overestimated benefit for the intervention and
underestimated benefit for the comparator
The model solely relied on PFS data since no significant difference NICE, 2012 (TA258) 1L
was identified for OS, and assumed that PFS gains automatically
convert to OS gains, which was unreliable
Comparator Comparators included in the model were not comprehensive of NICE, 2012 (TA258) 1L
clinical practice. Efficacy of pemetrexed as a first-line treatment
for patients with EGFRm+ did not exist and therefore was
inappropriate as comparator
Pemetrexed should not be included as maintenance only in the PBAC, 2013 1L
comparator arm

(Continued)
246 Economic Evaluation of Cancer Drugs

TABLE 7.8 (CONTINUED)


Key criticisms of HTA Submissions: Lung Cancer
Treatment Area Critiques Submission

Utilities Assumption that utilities are independent of time is not reflective NICE, 2006 (TA162) 2L
of real-world circumstances
Utility estimates were not based on the full EQ-5D questionnaire NICE, 2006 (TA162) 2L
All utilities should preferably have been derived from trial rather PBAC, 2013 1L
than the use of vignettes
Different utility values for first-line progression-free treatment and PBAC, 2013 1L
second-line progression-free treatment were not supported by the
evidence
Costs Assumption that costs are independent of time is not reflective of NICE, 2006 (TA162) 2L
clinical practice
Assumption that there is no drug wastage is not reflective of NICE, 2006 (TA162) 2L
clinical practice
Assumption that there is no vial sharing is not reflective of clinical NICE, 2006 (TA162) 2L
practice
Cost and resource use data extracted from expert opinion was not NICE, 2006 (TA162) 2L
validated against any observational datasets
The duration of therapy was uncertain, especially if treatment PBAC, 2013 1L
would continue beyond disease progression in real-world practice
The model was not comprehensive of the costs included in clinical NICE, 2006 (TA162) 2L
practice. Specifically, transportation costs associated with
treatment delivery were omitted
Others The detection of all EGFR mutations using a standard test was PBAC, 2013 1L
considered too optimistic and that false positives should have
lower efficacy. PBAC had identified a number of studies showing
inferior results of TKI in patients with no EGFR mutation
(TORCH, DELTA, TAILOR, TITAN, IPASS) – lowering the
prevalence of EGFR increased the ICER
Assumption that patients cannot suffer from multiple adverse NICE, 2006 (TA162) 2L
events is not reflective of clinical practice
Gefitinib Clinical data The trial data (IPASS) did not focus on EGFRm+ and was not NICE, 2010 (TA192) 1L
powered to evaluate this

The trial data (IPASS) may not have been generalizable to England NICE, 2010 (TA192) 1L
and Wales

ERG believed that the first SIGNAL trial should have been
included in the submission

Survival Concern was expressed over whether using the Cox proportionate NICE, 2010 (TA192) 1L
analysis hazards method to calculate hazards ratios was appropriate (only
valid if the hazard ratio for the two groups remains constant over
time)
Concern was expressed about the immaturity of the survival data NICE, 2010 (TA192) 1L
as relatively few deaths had occurred
Concern was expressed about the poor fit of the parametric model NICE, 2010 (TA192) 1L
to the data SMC, 2010 (615/10) 1L
(Continued)
Models for Economic Evaluation of Cancer 247

TABLE 7.8 (CONTINUED)


Key criticisms of HTA Submissions: Lung Cancer
Treatment Area Critiques Submission

Comparator Comparators included in the model were not a comprehensive NICE, 2010 (TA192) 1L
reflection of clinical practice
The handling of second-line treatments may be inappropriate SMC, 2010 (615/10) 1L
Costs Comparator costs included in the model were inaccurate NICE, 2010 (TA192) 1L
Others Concern that resource utilization was not reflective of clinical NICE, 2010 (TA192) 1L
guidelines
Cost for the management of adverse events was considered PBAC, 2013 1L
inaccurate
The model assumes 100% specificity of the EGFR test, which likely PBAC, 2013 1L
overestimates the benefit of the treatment (since treatment effect NICE, 2010 (TA192) 1L
on false positives would be lower than on true positives)
Other cost data were considered to be inaccurate PBAC, 2013 1L
Uncertain prevalence of EGFR, which affects the ICER SMC, 2010 (615/10) 1L
Time horizon (5 years) was not considered to be reflective of the NICE, 2010 (TA192) 1L
approximated length of life for the patient group, where the
longest possible time horizon should have been used (6 years)
Pemetrexed Clinical data Clinical trial population differed from clinical practice NICE, 2013 (TA310) MTx
Unlimited maintenance treatment cycles not reflective of clinical NICE, 2013 (TA310) MTx
practice
The ERG believed the approach to evidence synthesis (pooling of NICE, 2007 (TA124) 2L
absolute median) adopted by the company was not meaningful;
as a consequence, indirect comparisons provided in the
submission were not considered to be appropriate
Re-analysis of clinical trial data indicates that there is no additional NICE, 2013 (TA310) MTx
benefit provided to patients by pemetrexed once disease NICE, 2007 (TA124) 2L
progression is confirmed
Survival The ERG questioned whether the estimated survival from the NICE, 2009 (TA181) 1L
analysis economic model was consistent with the mean and median SMC, 2010 (342/07) 2L
clinical outcomes in the trial
The survival estimates are inaccurate in the long term NICE, 2009 (TA181) 1L
Comparator Comparators included in the model were not comprehensive of NICE, 2009 (TA181) 1L
clinical practice PBAC, (2010) 1L
The validity of the indirect comparisons was questioned NICE, 2009 (TA181) 1L
Utilities The QALY weights used were considered inappropriate as they PBAC, 2010 1L
were not treatment-specific
Some of the utility values were not justified PBAC, 2010 1L
Others The chosen model design was not obviously suitable for modeling NICE, 2009 (TA181) 1L
the disease and treatments
The ERG questioned whether the model structure accurately NICE, 2009 (TA181) 1L
replicates the trial data
The ERG highlighted several flaws in the economic model making NICE, 2009 (TA181) 1L
it impossible to estimate robust ICERs
The ERGs corrected errors in economic model and it resulted in NICE, 2013 (TA310) MTx
substantially less favorable results than in the submission

Notes: ALK: anaplastic lymphoma kinase; BSC: best supportive care; CMA: cost-minimization
analysis; EGFR: epidermal growth factor receptor; ERG: evidence review group; EQ-5D: EuroQoL
five dimension; ICER: incremental cost-effectiveness ratio; MTC: mixed treatment comparison;
MTx: maintenance treatment; NICE: National Institute for Health and Care Excellence; NSCLC:
non-small-cell lung cancer; OS: overall survival; PBAC: Pharmaceutical Benefits Advisory
Committee; pCODR: pan-Canadian Oncology Drug Review; PFS: progression-free survival;
QALY: quality-adjusted life-year; RPSFT: rank-preserved structural failure time; SMC: Scottish
Medicines Consortium; TA: technology appraisal; TKI: tyrosine kinase inhibitors.
248 Economic Evaluation of Cancer Drugs

2. Discuss the main criticisms of cost-effectiveness models submitted to


reimbursement authorities. How can these criticisms be addressed?
3. Conduct and appraise an HTA submission (find a published one
from a relevant website). Provide a summary of your findings and
compare them with those of a colleague. Do you agree?
4. Explain in detail how a partition survival model works including
how uncertainty (PSA and deterministic) could be undertaken.
8
Real-World Data in Cost-Effectiveness
Studies on Cancer

8.1 Introduction to Real-World Data


Real-world data (RWD) is a relatively recent term that refers to data gener-
ated outside an experimental setting (often a clinical trial). It is the fact that
these data are collected outside experimental conditions, that is, under real
clinical practice conditions, that properly defines it as ‘real-world data’. The
conclusions derived from the use of RWD are sometimes termed as ‘real-
world evidence’ (RWE). The strength of RWE must be subject to many of the
principles of statistical inference and methodological rigor.
RWE offers a means of evaluating the natural history of disease devel-
opment and understanding important factors that influence clinical out-
comes outside of controlled settings. Generalizability of treatment effects,
regional equity, under studied populations and access to cancer care, can-
not be properly addressed using data collected in a multicenter RCT of say
5 or 20 centers all located in a confined geographical area. Some questions
about treatment and outcomes are also more appropriately addressed in
observational or other types of trials and not RCTs. For example, whether
some NSCLC patients live shorter lives because of lack of access to treatment
due to age, ethnicity, or social demographics cannot be adequately (or eas-
ily) answered from data generated in a clinical trial. Moreover, in clinical
trials, patients may receive treatment at specialist center; however, the target
population may actually be treated in a community setting or general hospi-
tals where the quality of care given can be quite different. Hence, treatment
effects from patients treated at specialist centers may be different compared
to those treated in real-world community settings.
Although the terms RWD and RWE are new, collecting RWD is not so
new. Its scope, however, has increased in recent years. In many clinical tri-
als, the use of adverse events (adverse events monitoring well beyond the
trial is completed) continues through the use of pharmacovigilance data-
bases. This practice has been ongoing for many years but is often subject
to underreporting. Their use has often been limited to adverse event signal

249
250 Economic Evaluation of Cancer Drugs

detection for public safety reasons. RWD is now seen in some ways as an
enhancement and extension of such data collection to include outcomes
beyond safety.
RWD are generated through designing studies, collecting data from elec-
tronic health records (EHR) such as pharmacy claims data sets, disease reg-
istries, and other data sources for which:

(a) The medicinal product is prescribed in the usual way in line with
marketing authorization approval; and, in some cases, through off-
label use.
(b) The assignment to treatment is not determined on the basis of a pre-
specified protocol but is given according to current treatment guide-
lines or practices.
(c) No further additional diagnostic or monitoring procedures are used
to gather data, outside routine practice.

Table 8.1 shows the differences in objectives between an RCT and a study
using RWD.
For the purposes of economic evaluation, RWD can provide valuable infor-
mation on actual health resource use involved in delivering a cancer inter-
vention, rather than protocol-defined healthcare. These aspects have been
discussed in Chapters 2–5.

TABLE 8.1
Key Features that Differentiate RWD and Clinical Trials
Key Feature RCT RWD
Objective Efficacy Effectiveness and safety
Where does treatment fit in the current
pathway
Setting Usually small sample sizes Larger, possibly millions of records
Design prospective Retrospective or prospective
Time to get the Longer Relatively quick, especially retrospective
results designs
Costs High Generally higha for prospective
(observational type), mid for
retrospective case control type studies
and low for registry data studies
Statistical Usually less complex More complex
methodology
Economic ICER based on trial ICER based on external factors, model
evaluation follow-up; relatively simpler uses complex simulation to quantify
approaches uncertainty
Implications Drug registration Clinical, economic, policy evaluation
a Note:  The costs are lower in relation to comparable RCTs.
Real-World Data in Cost-Effectiveness Studies on Cancer 251

8.2 Using RWD to Support Cost-Effectiveness Analysis


In Chapter 6 we introduced observational studies as one of several methods
for collecting data when an RCT is not feasible. Such data might provide
insights on, for example, real-world drug utilization (e.g. compliance or pref-
erence) and treatment regimens (e.g. which doses are more common, dose per
body weight in the wider community), patient characteristics, longer-term
safety clinical outcomes, additional treatments, or, indeed, additional care.
The longer-term side-effect data of oncology drugs are often limited in clini-
cal trial databases. Moreover, once the drug is prescribed in a wider commu-
nity setting, the treatment and management of the consequences of cancer
treatments becomes apparent in hospitals or visits to GPs. For example, for a
new drug with a novel mechanism of action such as palbociclib (Kish, 2018),
an observational study was set up to examine the real-world occurrence of
neutropenia and how providers monitored and managed these events (e.g.
through dose modifications or other interventions or care). In a clinical trial,
if neutropenia is severe enough, patients come off treatment and exit the trial
if dose modifications fail. No further data is often collected when the patient
leaves the trial. During the first year post-licensing, such types of data might
be particularly important for economic evaluation.

The main reasons for generating RWE are:

(i) To enhance the external validity and generalizability of clinical trial


evidence. Clinical trials may involve highly selected patients with
limited follow-up.
(ii) It is sometimes impractical to conduct research in wider populations
(not used in the main clinical trials because of heterogeneity introduced
in the data and also the costs of running the trial). The recruitment
challenges and costs associated with RCTs can be significant. Access
to readily available data can be a much cheaper option. In any case,
some form of data collection in trials might be considered burdensome
(patients already have many other assessments) and intrusive.
(iii) To bridge the gap between the experimental and real-world setting by
adding in the knowledge that RCTs are unable provide. In addition,
longer-term prediction of event rates or outcomes can be made beyond
the (limited) RCT follow-up. This is especially important for populat-
ing transition probability matrices used for Markov modeling.
(iv) Technology and computing power have increased substantially so
that the generation of RWE is more feasible (large amounts of data
on patients are readily available, yet nothing is being done with it to
improve patient healthcare). The opportunity cost for not using the
data is greater than for using it.
252 Economic Evaluation of Cancer Drugs

(v) To support claims or challenge claims such as: “response rates for
drug A appear to be higher than drug B.” The hope is that such
claims (whether factual or embellished) will influence the prescrib-
ers or other groups, depending on the strength of evidence. A mar-
ket access department has an objective to increase market share and
the use of RWD is one means to this objective. The methodology for
such claims should be rigorous.
(vi) Some licensing authorities (e.g. FDA) endorse gathering further data
to support clinical trials when, for example, survival data are not
mature (i.e. many patients are censored at the end of the study).
Further follow-up might be needed to ensure survival rates (i.e. the
primary efficacy endpoint) are sustained over a longer period. This
might be part of a conditional approval process – where, so long as
the manufacturer provides additional data, a license or reimburse-
ment may be awarded.
(vii) To document the interconnection between the different sequential
lines of therapy in oncology patients, which are often poorly docu-
mented or not documented at all in cancer RCTs.

In single arm trials, where there is limited evidence of efficacy, but a high
unmet need or rare tumor population, follow-up data from patient regis-
tries could be used to complement clinical trial evidence (see Examples 6.1
and 6.2); one way might be to compare outcomes from a group of similar
(matched) patients using RWD to those from the single arm trial. This use
of real-world data might provide useful comparative evidence of safety
and effectiveness for decision-makers. Another useful context might be
in the case of ‘promising’ cancer medicines through early access schemes
(such as EAMs). Here, a license may be provided ‘quickly’ under a special
EAMS protocol (a document that states the conditions under which the
treatment may be given). Data collection from such patients under routine
clinical care could be generated for understanding the economic value of
the treatment.

(viii) Where enhanced anti-tumor activity is observed in patients with


some specific genetic (bio)marker, the number of patients might be
limited in such subgroups. The proportion with or without a bio-
marker depends on the prevalence of the biomarker in the target
population The potential to provide more evidence when the drug
is used in the wider community can be realized by collecting such
data from patient registries. However, this does require each patient
having recorded in the registry the biomarker (or test) outcome (e.g.
EGFR +ve or –ve) along with clinical and demographic data; or, at the
very least, the ability to link the biomarker data with registry data
that holds other clinical data merged (linked) through a common
Real-World Data in Cost-Effectiveness Studies on Cancer 253

unique identifier (e.g. NHS number or health insurance number), or


in some cases through probabilistic matching.
(ix) For generating evidence for cost-effectiveness and hence the value
of the new treatment in real-world clinical practice. Real-world data
sources can contain a variety of data not collected or not possible in
clinical trials – such as elective and non-elective surgery, outpatient
visits, GP visits – all of which might be related to the cost-effec-
tiveness of the drug. Even where this is collected in an RCT using
health resource CRFs, the data is limited to the trial follow-up and,
as we noted earlier in Chapter 5, the data depends on retrospective
recall.

Table 8.2 shows possible ways RWD could be used during pharmaceutical
development.

8.3 Strengths and Limitations of Using RWD to


Support Cost-Effectiveness Analysis
The strengths and limitations will depend on where the RWD are generated
from (e.g. case control study, observational study, registry, administrative
data). Camm and Fox (2018) provide a useful summary of the strengths and
limitations of using RWD from various sources. The main strengths of RWD
are: the wider populations studied; the ability to evaluate real-world effec-
tiveness; a limited need, if any, for scheduled assessments; more treatments
able to be compared, rather than just the comparators in the trial; and data
captured at the point of care (or in real time). In particular, some larger data-
bases have massive amounts of data that allow detailed interrogation of the
relationship between multiple factors like treatment, outcomes, compliance,
dosing, ethnicity, geography, social class, and so forth. Such large databases
allow for the investigation of treatment benefit in some rare populations. The
access to data can be relatively cheap and quick (in contrast to large expen-
sive RCTs). Where healthcare resource use is available, the unit costs are also
readily available (e.g. NHS reference costs in the UK, or from claims data-
bases) and complement this, so that comprehensive economic evaluations
can be performed.
RWE is also increasingly accepted by regulators and other bodies to sup-
port disease management and decision-making. It has been argued (Khoizin
et al., 2017) that current FDA regulations support the use of evidence from
emerging sources as evidence for making regulatory decisions. In 2008, the
FDA launched the Sentinel Initiative in response to the FDA Amendments
Act (FDAAA) calling for monitoring the safety of approved drugs and
TABLE 8.2
254

Possible Real-World Data Use During Drug Development


Phase I Phase II Phase IIb/III Launch Phase IV
Epidemiology and Incidence of myeloma in Source data sources and Compare Phase III observed data with
treatment patterns population registries that expected in the general
Incidence of biomarker population
mutation status
Trial design Scoping of secondary source Feasibility of Feasibility of inclusion/ Real-world findings that have
data for the target product inclusion/ exclusion implications for altering or
profile exclusion modifying the trial design in future/
expanded indications
Health economics Identify appropriate HRQoL Burden to health system?
measures Cost-efficiency and budget
HRQoL features that impact impact of new drug
symptom and function
Post-marketing How is the treatment used How is the treatment used compared
clinical evidence compared to alternatives? to alternatives? Real-world survival?
(therapeutic Optimal duration of therapy?
utilization, Subgroups
effectiveness)
Post-marketing drug Incidence of specific SAEs in usual
safety care compared to RCT
Comparative OS? e.g. at 1 year
Comparative Real-world benefit in a different
effectiveness vs. tumor type; market size of new
usual care indication; potential long-term safety
concerns to be evaluated using
long-term studies or real-world data
Economic Evaluation of Cancer Drugs
Real-World Data in Cost-Effectiveness Studies on Cancer 255

medical products. In collaboration with insurers, universities, and hospitals,


several surveillance reports have been generated using data contained in
Sentinel’s (FDA, 2017) network of claims and electronic health record- (EHR)
related content.
In oncology, the Information Exchange and Data Transformation
(INFORMED) initiative (FDA, 2018) is a multidisciplinary group focusing on
building technical and organizational infrastructure in several key areas of
‘big data’ analytics to investigate how data from EHRs and digital health
solutions can be used in making regulatory decisions. The 21st Century
Cures Act in 2016 in the US also allowed for the potential use of RWD to
support new product indications and post-approval requirements. In other
words, whereas a post-approval clinical trial might have been conducted
previously to gather evidence of longer-term efficacy and safety post-mar-
keting, EHRs may be used as an alternative. However, a caveat in using RWE
of this type is that it must still subscribe to scientific rigor in terms of study
design and data collection (e.g. minimizing bias). Before discussing design-
ing studies for gathering RWE, it is useful to determine where RWD will be
collected from.
In Europe, the European Medicines Agency (EMA) does not dismiss the
use of RWD out of hand. While it is acknowledged that such data cannot
be used primarily as a basis for providing evidence of efficacy, they may be
used to contextualize the evidence provided in a clinical trial. For example,
if a single arm trial is conducted and reference to historical data is made, it
may not be acceptable to compare a historical control arm using RWD of,
say, several thousand patients with an experimental arm of 150 patients.
For cost-effectiveness, however, because the decision-making framework is
not always inferential (p-values do not matter), providing the biases were
addressed as best as possible, expected net benefits could be generated to
provide a direction of cost-effectiveness. As a caveat, just because the prob-
lem is not inferential, this does not mean that biased estimates of treatment
effects should be used to justify cost-effectiveness.

8.3.1 Limitations
The main limitations of RWD are issues such as recall bias, loss to follow-
up (censoring and missing data), selection, and other biases due to lack of
randomization (for example, there may be limited data on some treatment
due to clinical preference for one treatment over another). There can also
be restrictions on the amount of data that may be extracted, and it can cost
more, for example, to access 20 million records compared to 5,000. There is
also a lot of additional programming time that may be needed, because data
are not often collected at fixed time intervals, but as and when care is needed,
and may also need complex extraction and linkage procedures.
A key issue in the use of EHRs is the quality of the data. This relates to the
systematic and non-systematic bias (see Section 8.4) in the way the data have
256 Economic Evaluation of Cancer Drugs

been collected (e.g. outcomes on only those who might attend a hospital).
Registry data often have, for example, incoherent dates (e.g. date of death
before a date of progression) and incoherent data collected – often left to
be collected by untrained staff – whose main reason for collection is part of
their administrative role, rather than with a research objective in mind. The
coding of the data may also be erroneous (see 8.4.4) and therefore unreliable
for inferential purposes.
In clinical trial data, rigorous data management and monitoring efforts are
used to be able to verify the data source. Where EHRs are used, the funda-
mental principles of Good Clinical Practice – to be able to reconstruct the trial
results may not be possible. The governance issues in ‘source data verification’
(i.e. making sure that the data in the records corresponds with patients’/doc-
tors’ notes) might require access to patients’ personal records. In Europe, given
the new GDPR directive, this will be difficult. Acceptance of data that does not
conform to the basic principles of GCP may not be acceptable to regulatory
agencies for establishing efficacy, but may be acceptable for reimbursement
depending on what safeguards are in place to ensure an acceptable degree
of data quality (e.g. more than 70% missing data is likely to be unacceptable).
However, if these biases can be limited and/or managed, use of RWD has
potential for providing very valuable data on the longer term cost-effective-
ness of many (expensive) cancer drugs.

8.3.2 Internal Validity versus Generalizability


The usual RCT framework provides strong internal validity of clinical find-
ings (efficacy). The ideal conditions and assumptions in which trials are
executed attempt to limit the plausibility of external factors influencing
observed treatment benefit – in other words, yielding unbiased estimates of
treatment effects. In reality, not all biases are quantified, and sometimes if a
bias is there it may not even be known.
Although, strong internal validity is needed for experimental settings, the
restrictive eligibility criteria can exacerbate differences in outcomes between
patients in real practice and ideal settings, and consequently limit external
validity. Hence, external validity needs to be strengthened so that results
from experimental settings can be readily applied for the health of the gen-
eral public (Steckler & McLeroy, 2008). This may also explain the reason why
some reimbursement agencies treat the estimates of treatment effects from
experimental trial conditions as being uncertain when there is a desire to
generalize these effects to a wider population.
It is argued that a “balanced approach based on prospective collection
of RWD can protect against common threats to internal validity” while at
the same time augmenting the external validity of clinical research (Khozin
et al., 2017). It would be misleading to believe, however, the impact of evi-
dence from RCTs alone does not apply to a real-world setting. For example,
Real-World Data in Cost-Effectiveness Studies on Cancer 257

an early phase RCT demonstrating the presence of a food effect, suggests


treatment should be taken with food. If, in a real-world setting, patients do
not take food, whether this is a limitation of the external validity of the trial
or because patients simply don’t follow clinical advice, is something that
should be distinguished. A lack of treatment effect may be due to poorer
drug absorption when taken with/without food. The adage ‘read the label on
the tin’ applies to many products and services, and a treatment should not
be penalized because elementary instructions for taking treatment have not
been followed. This type of criticism of external validity is different to test-
ing treatments in trials with specific populations (e.g. Caucasians) and then
extrapolating to others (ethnic minorities).
A separate question is whether a trial should be designed for external
validity alone. Let us take the above example on the food effect. If, in the
real-world, patients are unlikely to take treatment with food, the question is
whether regulatory agencies, such as the FDA and EMEA, should mandate
food effect studies, because (after all) they don’t reflect the real world. During
Phase III, if patients are not going to follow the protocol, some patients who
take food with the drug may have poorer clinical effects compared to those
who do not take food (e.g. worse effects because the drug might be less well
absorbed); one scenario (taking food) is reflective of the real world and one
is not (take it according to the label). However, both situations (taking the
drug with or without food) are reflective of the real-world setting. There
will be patients who take drug according to the label and those who do not.
Therefore, it may be very difficult to design trials that cover this type of
internal and external validity. A related question is whether real-world stud-
ies themselves have credible internal validity and whether statistical meth-
ods can account for all the biases involved in estimating treatment effects to
strengthen such internal validity.

8.4 Sources for RWD Generation


RWD can be determined from several sources including electronic health
records, disease registries, and other sources of administrative data. RWD
is sometimes used synonymously with electronic health records (EHR).
Ultimately, all or most data is found in some form or another of an EHR. It is
perhaps the way that it is populated in the EHR that is important: A defini-
tion focused on the original intent of data collected at the point of care can
distinguish RWD from conventional clinical trial data. Where the purpose of
collecting data is ‘research,’ then data can be gathered from a clinical trial or
similar experimental design (with or without an intervention). A summary
of the data sources for RWE generation is shown in Table 8.3.
TABLE 8.3
258

Sources of Real-World Data


Source Example Advantages Disadvantages
National registries including Cancer registry SACT Can be linked Biases and confounding
primary care
CPRD Protocol required Government regulations
HES Relevant records Limited/no HRQoL data
THIN Natural disease history Data quality issues
Cheaper than a clinical trial Missing data
Large sample size Costs more to extract large amounts
Real time Limited inference
Useful for rare outcomes Biased estimates
Useful for economics No randomization
Quick access/studies Switching between treatments
Interrogate multiple associations
Audit data/HER Hospital consultants collect data Cheaper than a clinical trial Smaller sample sizes
Based at hospital/point of care Maybe single arm data
Useful for economics Data quality issues
Real time outcomes No randomization
Useful if rare outcome is studied at Missing data
site
Private/commercial companies Private companies charge more
Can pool across hospitals
prospective
Observational studies Prospectively designed Prospective No randomization
Retrospective Defined outcomes Loss to follow up
(Continued)
Economic Evaluation of Cancer Drugs
TABLE 8.3 (CONTINUED)
Sources of Real-World Data
Source Example Advantages Disadvantages
Cheaper than RCTs Sample, observational and recall bias
Useful for economics Can require long follow-up for rare
outcomes
Social media, internet logs Twitter, blogs, internet, meta data Cheap, easy access Complicated/large amount to make
sense
Little HRQoL data
Administrative/claims Medicare/Medicaid Longitudinal Missing data
database
Large database Errors in coding/data quality
Useful for economics Confounding and bias
Useful for rare tumors Switching between treatments
Quick studies – easy to access
Multiple associations
Real-World Data in Cost-Effectiveness Studies on Cancer
259
260 Economic Evaluation of Cancer Drugs

8.4.1 Registries
Registries are national or public electronic health records (EHRs). These are
essentially large repositories of data collected at the patient level. Examples
of these in the UK include the National Cancer Registry (NCR), (see Section
8.5), Hospital Episodes Statistics (HES), systemic anti-cancer therapy (SACT)
data. The HES data is an administrative source of data collected for commis-
sioning purposes. In other countries, such as France or the US however, no
comprehensive national cancer registry exists. Local registries do however
exist (see for example: http:​//inv​s.san​tepub​lique​franc​e.fr/​surve​illan​ce/ca​
ncers​/acte​urs.h​tm.
In these registries/sources, patient-level data recording each episode of an
event (e.g. death, hospitalization, adverse event) may be available. To use the
data, a data dictionary might be needed because conditions are often coded
using the international classification of diseases (ICD) or ICD-O (for oncol-
ogy). It is also possible to link some registries (data) at the patient level. In
the UK, the National Health Service number is a unique variable that allows
merging across data sets such as HES, cancer registry data, cancer treatment
data (SACT), and general practice (primary care) data. Particularly useful
are linkages between cancer registries and official death records to assess
cancer incidence and long-term survival. The registries may have restricted
commercial use and, where used, a protocol (prior to authorization for data
access) is often required. Moreover, charges for data extraction may also be
levied and in many cases only aggregate data are made available because of
national confidentiality regulations.
Some cancer registries increasingly collect important prognostic factors
of survival useful for oncologists, policymakers, and others in order to
make decisions on expected survival rates for subgroups of patients. For
example, calculating the survival probability for an individual (or group of
individuals) who is female, aged 50 (on average), with Stage III cancer, is a
particularly useful statistic for policymakers and planners, especially for
future costing of cancer treatments. However, in many countries, such as
Belgium or France, survival data are not routinely collected/calculated in
the cancer registries. In Section 8.5, cancer registries will be discussed in
more detail.

Example 8.1: Real-World Data on Metastatic


NSCLC from Canadian Cancer Registries
An example of an RWD study with cost-effectiveness implications was
reported by Scherer et al. (2015). This study was undertaken to better
understand the consequences of real-world chemotherapy patterns in
NSCLC. Registries were identified in Canada (using the Ontario Cancer
Registry) and were linked (merged) together. Patients were tracked
using existing cancer registry data over several years (retrospectively).
The data from each patient record included:
Real-World Data in Cost-Effectiveness Studies on Cancer 261

• Diagnosis date
• Institution and region of diagnosis
• Stage
• Pathology
• Age
• Sex
• Date of death

Treatment records included:

• The chemotherapy regimen


• Dose
• Treatment dates
• Treating hospital
• Line of therapy
• Radiotherapy treatment dates and doses

Oral agents, performance status, and comorbidities were not systemati-


cally included in these databases. After linking databases, and perform-
ing statistical analyses, the researchers concluded the following:

• Most patients with (metastatic) NSCLC in the general Canadian


population did not receive systemic therapy.
• Patients selected for first‐ and second‐line systemic treatment,
had survival outcomes comparable to clinical trial results.
• Older patients and patients with squamous histology are less
likely to receive chemotherapy.

The policy implications were:

• To ensure all patients with newly diagnosed advanced NSCLC receive


timely consultation with a trained oncologist.
• The gap between the diagnosis and the initial oncology consultation in
this study was 30 days. In a disease such as metastatic NSCLC, with
often short survival and where patients deteriorate clinically, 30 days
should be reduced; this might improve the number of patients who are
candidates for systemic therapy.
• An increased use of screening methods for NSCLC in at‐risk popula-
tions might be needed.
• To further investigate other unknown factors contributing to low
treatment rates – and consequently to improve treatment access and
outcomes in this population.

8.4.2 Audits
Many researchers collect data on cancer patients at secondary care sites (hospi-
tals). These data are often used for auditing, or assessing the quality or perfor-
mance of, for example, cancer treatment delivery at specific hospitals. Although
data from hospital registries tends to be rich, and often contain key cancer
262 Economic Evaluation of Cancer Drugs

outcomes, unfortunately some audit data sets are limited to small sample sizes,
limiting their generalizability. To strengthen generalizability, similar data sets
would need to be merged together to address a study question. Wilson (1999)
differentiates audit and research: “research is finding out what you ought to be
doing; audit is whether you are doing what you ought to be doing.”
The primary aim of research is to derive knowledge that is new and gen-
eralizable, with clearly defined questions, aims, and objectives for which
there is often a well-written protocol detailing the methodology. An audit,
on the other hand, is a way of understanding whether cancer treatment or
care is reaching a defined standard. The treatment itself is defined (unlike
in research where it could be randomized). Hence, when a new treatment
is licensed and is considered to be the standard of care, data from a clinical
audit might be useful to determine whether the standard of care is being
reached. Outcomes such as survival may also be collected.

Example 8.2: Lenalidomide Alternative Dose


Proposal for Treating Multiple Myeloma
As an example, the lenalidomide alternative dose proposal (Popat et al.,
2015) used audit data to gather evidence in 39 patients on whether alter-
nate dosing was a potentially a more cost-effective dosing regimen com-
pared to daily dosing, without impacting survival benefit. This example
was discussed earlier in the context of modeling (Chapter 4, Example
4.5). Its relevance in the context of RWD shows how it may be possible
for non-trial data to be used to propose a more cost-effective dosing regi-
men with the view that efficacy is not compromised.

8.4.3 Primary Care Databases: CPRD, THIN, QResearch


The Clinical Practice Research Datalink (CPRD) database was established in
1987 in the UK.. It operates as part of the UK Department of Health. It cov-
ers more than 644 general practice databases, exceeding 13 million patient
records and covering about 7% of the general UK population. It offers the
ability to extract anything adequately recorded in primary care.
The Health Improvement Network (THIN) database was established in
around 2003. The database consists of data from over 560 practices exceeding
11 million patients or about 6% of the UK populations. There is some overlap
with the CPRD, although there may be more efficient patient matching for
sociodemographic characteristics.
QResearch is a collaboration with the University of Nottingham contain-
ing data from over 754 practices consisting of over 13 million patients (about
7% of the UK population).
In all the above primary care databases, patient-level data is available on
clinical and demographic characteristics, disease status, and other treatment
details in primary care settings. Typical examples might include data on dia-
betes management. The CPRD can also be linked with cancer registries.
Real-World Data in Cost-Effectiveness Studies on Cancer 263

8.4.4 Insurance Claims Databases


In the US, there are claims databases, or databases that consist of EHRs of
millions of transactions between patients and healthcare providers, includ-
ing hospitals, nursing homes, and pharmacies (Ferver, 2009). In general they
tend to be used by health insurance companies to decide the risks associated
with groups of patients and whether cover should be offered. Ferver et al.
(2009) have provided a very useful summary of how claims data are used,
outlining their strengths (anonymous, cheap, plentiful, widely available in
an electronic format, and useful for identifying rare subgroups) and limita-
tions (the databases were not designed for research, non-essential data for
billing is often excluded, there is missing information from some claims,
health resource use is incorrectly classified, chronic diseases are underre-
ported because only one diagnosis is enough for a reimbursement). The data-
bases nevertheless contain much information on healthcare resource use,
covering many disease areas including cancer, however, as in the case of any
registry, the potential for confounding and bias exists and robust methodol-
ogy should be used in the analysis of this type of data (Ferver et al., 2009).

8.4.5 Digital Data Sources, Social Media and Applications


In recent years, phenomenal growth in information technology solutions has
offered a means to develop an integrated approach for generating RWE from
EHRs and other data sources, such as mobile applications and internet search
logs. Because of the dynamic way in which data processing works, using
digital applications (apps) with web-based analytical tools may enable ear-
lier detection of adverse events or disease progression, than can be obtained
in clinical trials. Since clinical trials collect data at specified discrete time
points, coinciding with scheduled visits, the opportunity for identifying the
consequences of such events is delayed. The type of data available could
include patients taking medication for treating adverse reactions, taking
subsequent therapy, delaying subsequent therapy, missing a potential tumor
progression, or reporting worsening HRQoL (perhaps an indication of dis-
ease progression). These data may have implications for cost-effectiveness.
RCTs that use web-based tools showed potential for several health benefits
including close monitoring of symptoms (Basch et al., 2017; Denis et al., 2017).

Example 8.3: Example of Digital Real-World Web-Mediated


Follow-Up Compared with Routine Surveillance in NSCLC Patients
This example is taken from Denis et al. (2017). Patients with NSCLC were
randomly assigned to web-mediated follow-up (experimental) or usual
follow-up while taking their usual maintenance chemotherapy, or TKI,
or no treatment. Patients were followed-up in both arms every three
months. CT scans were more frequent in the control group compared to
web-mediated follow-up because the web-based application was found
264 Economic Evaluation of Cancer Drugs

to be reliable in detecting patient relapse as a result of self-evaluated


symptoms assessed weekly. The trial results reported in Denis et al.
(2017) showed improved survival in patients using the web-based follow-
up (called e-FAP) compared to patients in the control group. The e-FAP
also reported a decrease in the number of imaging tests (which would
lead to a reduction in health resource costs).

8.4.6 Commercial Data Sources


More recently, private initiatives and collaborations with health research-
ers have developed and enabled wider access to real-world data. However,
there are often strict governance issues on the commercialization of what is
in effect data that belong to the public. Audit data may effectively be used
as research where there is an agreed partnership with local investigators
(e.g. offering small grants), and strong methodology is used to ensure fac-
tors such as bias has been adequately assessed. Some national charities, non-
profit organizations, and patient society networks also collect data. In some
larger national registries, an absence of HRQoL makes their use for an eco-
nomic evaluation limited. National charities and patient networks may have
the potential to offer or collect this type of data.
Access to patients and their records for nonclinical purposes has been and
will continue to remain under scrutiny. Although research ethics commit-
tees (REC) safeguard access to patient data for research purposes, audits
are often below the radar, unless the database is available on a larger scale.
Guidelines published by the UK Royal College of Physicians recommend
submission to a research ethics committee if doubt exists about whether a
project is audit or research.

8.4.7 Pragmatic Clinical Trials


Another source of real-world type data is through the conduct of a pragmatic
RCT. In such a trial, observation and follow-up of patients is carried out
under a routine clinical setting. For example, rather than stipulating in the
trial protocol that scans will occur every two months, scans occur according
to routine practice. Additional tests specified in a trial protocol may not be
undertaken in routine practice. In short, the main difference between a prag-
matic RCT and a highly controlled RCT is the degree of experimental control
involved in conducting the trial. Patients in a pragmatic RCT (PRCT) may
have less restrictive inclusion/exclusion criteria. PRCTs are designed to pro-
duce results that uniquely support clinical decision-making at the point of
care (Kish, 2018; Khozin, 2017). Since PRCTs are conducted in the real world,
data collected may form part of the usual administrative process for record-
ing and entry. A trial conducted in this setting, providing that the technol-
ogy is available, and data security and integrity are preserved, is likely to
yield rich data, including HRQoL (e.g. through the use of mobile phone
applications), and data on health resource use. HRQoL are rarely available in
Real-World Data in Cost-Effectiveness Studies on Cancer 265

cancer registries. A well design PRCT using EHRs can “bring the real-world
evidence base to drug development while driving the focus on improving
quality, patient safety, and value in cancer care delivery” although their cost
may be high (Khozin et al., 2017).

8.4.8 Prospective Observational Research Studies


Observational studies were discussed in Chapter 6. New hypotheses can be
generated from observational studies as supportive evidence for reimburse-
ment agencies (in some cases this is specifically requested by agencies who
might wish to have more data on longer-term outcomes. Longer-term assess-
ment of safety, effectiveness, patients excluded in conventional cancer trials,
patients with poor performance status, history of prior malignancies, organ
dysfunction, or other metastases (e.g. brain) can be evaluated. Evidence also
suggests that well-designed and well-conducted observational studies can
yield results similar to those in RCTs. This might be an opportunity for
building capability and methods in using EHR-based observational research
to enhance generalizability (Anglemyer et al., 2014; Konnerup et al., 2012;
Vázquez et al., 2015). In Section 8.4, an example of how data from a real-world
observational study is analyzed and reported is given.

8.4.9 Case Control Studies


Case control studies make use of EHRs because of their retrospective nature.
Even if a group of patients with the outcome (e.g. disease progression) in ques-
tion are followed up for other data, they can be matched uniquely (1:1 match-
ing) to a patient control (another patient without the disease or outcome, but
who is otherwise ‘identical’ in terms of age, gender, and other characteristics).
A single patient in the group with the disease or outcome of interest (case)
can be matched with several (m:1, many-to-one matching) historical cases.
Alternatively, several cases can be matched to several controls (m:n many-to-
many matching). These studies are less expensive to conduct and more effi-
cient especially for rare diseases or outcomes, and where time to an outcome is
long (e.g. long-term death or toxicity). Clearly, the data will be subject to selec-
tion bias, and information on outcomes and treatments is likely to be subject
to observation bias. In addition, incidence (new events or outcomes) cannot be
computed. Special statistical methods for analyzing such data are needed.

8.5 Using Cancer Registries


Cancer registries have their own unique history. The first-known systematic
collection of cancer data was the general census of cancer in London in 1728
(Hutchinson et al., 2004). Since then registries have developed further (e.g.
266 Economic Evaluation of Cancer Drugs

TABLE 8.4
Cancer Registries in Several Countries
Sample for
Country Years Details Coverage NSCLC Coding
Denmark From 1943 Inpatient/outpatient Nationwide >23,000 ICD-10
oncology
France From 1997 All tumors Regional >900,000 ICD-O-3
Germany From 1998 General population Bavaria >12 million ICD-10
Italy From 2012 NSCLC General >2,500 ICD-10
Norway From 1973 General population General >317,000 ICD-10

1842 in Verona, 1913 in Chicago), such that in the early 1900s cancer regis-
tries were used to systematically track the etiology and survival of cancer
patients. Their ubiquitous capabilities led them to being developed in several
Western European countries. Table 8.4 gives examples of several cancer reg-
istries in several countries (there can be multiple registries in each country).
Merging data is likely to be highly complex. However, the data could be
analyzed locally to furnish evidence across countries. It is often the case that
economic evidence is required at the local level and hence local registries
used. However, it is possible that evidence from a registry in one country
might be admissible in another, especially where the target population is
similar. However, the differences in health systems will make the quantifica-
tion of health resources more challenging.

How Registries Work in Brief


For a national cancer registry, each cancer and its subtype are coded using
ICD low-level codes, of which there are several thousands (updates to these
codes are made yearly). The 2018 ICD-10 codes for cancer, termed ‘neoplasms’
are coded as C00-D49. For example, a code of C00-C14 in the registry will
have information on malignant neoplasms of lip, oral cavity, and pharynx,
which are then further divided into other subtypes.

• C00 Malignant neoplasm of lip


• C01 Malignant neoplasm of base of tongue
• C02 Malignant neoplasm of other and unspecified …
• C03 Malignant neoplasm of gum
• C04 Malignant neoplasm of floor of mouth
• C05 Malignant neoplasm of palate
• C06 Malignant neoplasm of other and unspecified …
Real-World Data in Cost-Effectiveness Studies on Cancer 267

• C07 Malignant neoplasm of parotid gland


• C08 Malignant neoplasm of other and unspecified
• C09 Malignant neoplasm of tonsil
• C10 Malignant neoplasm of oropharynx
• C11 Malignant neoplasm of nasopharynx
• C12 Malignant neoplasm of pyriform sinus
• C13 Malignant neoplasm of hypopharynx

The potentially rich patient-level of data in cancer registries covers a wide


spectrum of tumor types. So, for example, if someone wanted to determine
the incidence of a given cancer, the number of occurrences of a given code
between two consecutive years divided by the population might be extracted.

8.5.1 Examples of Registries in the UK for RWE


In the UK there is a unified healthcare system with multiple sources of rich
data across primary and secondary care settings. There are also strong aca-
demic registries supported politically through ‘big data’ in health philosophy.
EHRs are used extensively in primary care and there is an impetus to use these
extensively in secondary care. The challenges revolve around private access to
public healthcare data, lack of a consistent and clear method of access, inabil-
ity to link databases together, and a complicated governance structure with
a cautious attitude toward RWE by regulators or payers. For example, lung
cancer data can be obtained from the National Cancer Registration Service
(NCRS). Different files exist that contain patient-level data on:

• Radiotherapy
• Chemotherapy
• Imaging (scans)
• Primary care
• Hospital episodes (resource use)
• Specific cancer data
• Genetic data (biobank)
• National Lung Cancer Audit Lung Cancer Data (e.g. previously held
by University of Nottingham), which details chemotherapy
• SACT data set, which consists of five sections (demographics, clinical
status, treatment regimen, number of cycles, details of drugs such as
reductions, and outcomes such as death and disease progression)

In Europe, the commercial availability of RWD is limited. In oncology,


most data is used for market research (e.g. number of prescription sales)
268 Economic Evaluation of Cancer Drugs

and currently not very suitable for evidence generation. Companies are
now creating strategic portals for accessing data to support market access
groups. Some Nordic registries can be accessed through commercial com-
panies, whereas access to some public cancer data sets require academic
partnerships.

8.6 Statistical Analyses of RWD: Addressing Selection Bias


As noted above, RWD has potential for much confounding and bias. Selection
bias might occur when the chance of receiving treatment differs between
patients; and, moreover, the characteristics (e.g. age, gender) are also related
(confounded) with outcomes. Selection bias is likely to lead to incorrect con-
clusions that treatment effects are generalizable. Confounding occurs where
an outcome (death) might be associated with an independent factor (treat-
ment) and consequently thought to be causal (treatment effects from good
quality RCTs are considered to be causal), but if the measure of treatment
effect is influenced by a third factor (ECOG), a valid estimate of treatment
effect cannot be determined, unless we know about ECOG status. In this
case, ECOG status is said to be a confounder. As an example, suppose it was
observed in a cohort of people that many of those who drank alcohol died
from lung cancer. One might think that alcohol is more likely to be related
to liver cancer. However, there is a confounder – a third factor, smoking.
Since those who drink more alcohol also tend to smoke, smoking is the con-
founding factor leading one to incorrectly conclude that alcohol ‘causes’ lung
cancer. Therefore, some specific statistical methods and designs are used for
the analyses of data from registries and observational studies to control for
selection bias, mainly:

(i) Propensity score modeling


(ii) Instrumental variable methods

A general overview of methods for selection bias correction can be found in


Keeble et al. (2015).

8.6.1 Propensity Score Modeling


Comparing treatment differences in RWD studies will be influenced by dif-
ferences in baseline characteristics. The propensity score approach allows
one way of adjusting treatment differences to take into account the (known)
baseline differences. Without delving into the technicality of these models
(see bibliography and references therein), propensity score models (PSM)
Real-World Data in Cost-Effectiveness Studies on Cancer 269

involve modeling the treatment received in terms of the baseline characteris-


tics (Pan & Bai, 2015; Guo & Fraser, 2014).
In the case of two treatments, patients that receive treatment (exposed
to treatment) may be coded as 1 and those that do not (controls) are coded
as 0. These outcomes are then modeled using a multivariate logistic model
(for more than one treatment, a multinomial approach can be used). We are
in fact modeling the chance of receiving treatment given (conditional on)
what we know about baseline characteristics. In an RCT (1:1 allocation), the
chance of receiving treatment is expected to be equal (regardless of patient
characteristic).
In RWD, it is not uncommon to find unequal numbers of patients in a can-
cer registry taking either the experimental treatment (once approved) or the
standard of care. The systematic nature of the unequal numbers receiving
treatment may be related to some baseline characteristic (e.g. Stage III, poor
performance status) – thereby violating the idea that patients have an equal
chance of receiving treatment. It may well be possible that 80% of males
receive treatment A and 90% of females receive treatment B, which compli-
cates a comparison between treatment A and B.

Example 8.3: Propensity Score Modeling


Using Data from a Cancer Registry
The following data (Table 8.5) are reported from a cancer registry. There
are two types of treatment: chemotherapy (C) alone and chemotherapy
in combination radiotherapy (C+R). Five-year survival rates from lung
cancer patients are of interest. Several baseline/clinical characteristics
(age, gender, ECOG, stage) are included in this fictitious example. The
objective is to compare nonrandomized C with C+R in terms of 5-year
survival rates using data from linked registries.
Selection bias is first addressed by a straightforward comparison of
the data in the registry. Some imbalance in the baseline characteristics is
observed (Table 8.5).
As can be observed in Table 8.5, there are statistically significant imbal-
ances in the baseline clinical characteristics that may lead to confound-
ing and potentially misleading conclusions when comparing survival
rates between C versus C+R. It is also important to note that with such
large sample sizes, the p-value will be small. Hence, attention should be
paid to the magnitude of any differences. A propensity score (PS) analy-
sis will be performed that allows for differences between groups.
The first step is to derive the propensity score. Hence, we model the
treatment group (C or C+R) in terms of the covariates using a logistic
regression model. In this case:

Treatment Group (C / C + R ) = Age, Gender , ECOG, Stage

From this model, a propensity score is computed for each patient. We


compare the baseline characteristics and the 5-year survival rates,
270 Economic Evaluation of Cancer Drugs

TABLE 8.5
Comparison of Clinical Characteristics Using Cancer Registry Data
C (N = 4,217) C+R (N = 2,687) p-Value
Age (years) 68.7 74.6 p < 0.0001
Gender:
Male 2,911 (69%) 1,564 (58%) p < 0.0001
Female 1,306 (31%) 1,123 (42%)
ECOG
0 2,145 (51%) 1,852 (69%) p < 0.0001
1 1,347 (32%) 575 (21%)
2 725 (17%) 260 (10%)
Stage
I 1,441 (34%) 1,021 (38%) p = 0.0345
II 2,542 (60%) 1,575 (59%)
III–IV 234 (6%) 91 (3%)

5-year survival rates 25% 39% p < 0.001


Odds ratio = 1.91

adjusting for the PS in the model. The propensity scores can be catego-
rized (e.g. using quantiles) or included as a continuous covariate when
adjusting for the difference between C versus C+R. The results in Table
8.6 show what happens when the propensity scores are included in a
statistical model when comparing 5-year survival rates. The model is of
the form:

Survival ( yes / no) = Treatment , PS, Age, Gender , ECOG, Stage

As can be seen from Table 8.6, without adjusting for differences in


baseline characteristics, the 5-year survival rates appear to be 91% higher
with C+R versus C (OR = 1.91). After adjusting for PS as a continuous
outcome, this difference is no longer statistically significant and falls to
54% higher for C+R versus C. The differences in baseline characteristics
may well have contributed toward a higher favorable treatment for C+R.

Example 8.4: Performing a Propensity Score Model Using Matching


to Compare Health Resource Determined from a Cancer Registry
A researcher wishes to understand how various factors and health
resource use differ between patients treated with existing standard
chemotherapies in multiple myeloma patients. Data from linked regis-
tries were used. A sample of 937,182 patients who received the current
standard of care (control) and 44,215 patients who received an alterna-
tive treatment (CAP: chemotherapy for advanced prostate cancer) were
extracted from the linked Hospital Episodes (HES) database and linked
with cancer registry data. The registry also contained many potential
confounding variables including age, gender, ECOG, and hospital site.
Real-World Data in Cost-Effectiveness Studies on Cancer 271

TABLE 8.6
Comparison of Clinical Characteristics Using Cancer Registry Data
Adjusting for PS
Odds ratio 95% CI p-Value
Unadjusted 1.91 1.06, 3.58 0.028
Adjusted for PS (continuous) 1.54 0.95, 3.12 0.078
Adjusted for PS (categorical) 1.58 0.97, 3.22 0.069

TABLE 8.7
Data Structure for Example 8.3 Prior to Matching
Patient Age Gender ECOG Site Comorbidity Treatment
1 65 Male 2 1 Yes Control
2 59 Female 3 2 Yes Control
3 44 Male 3 3 No Control
937,182 .. .. .. .. .. Control
1 45 Male 3 1 Yes CAP
2 55 Male 2 2 No CAP
3 65 Female 1 3 Yes CAP
.. .. .. .. .. .. ..
44,215 .. .. .. .. .. ..

An important question to answer from the data is how hospital admis-


sions and other health resources (e.g. elective admissions, nonelective
admissions, etc.) differ between CAP and control groups. An example of
the data structure prior to matching is shown in Table 8.7.
In this example, 36,386 CAP patients randomly selected from 44,215
were matched to the same number of controls of which there were
937,182 (Table 8.8).
Step 1: Matching:
In order to perform the matching, several options are available:

(i) 1:1 matching. We could take a sample of patients from the


937,182 controls available in the registry who are identical in
every respect to those that were treated. For each patient who
was treated, say aged 50, with ECOG of 2, and male, we will
seek to match an identical patient who takes the control treat-
ment. The more factors there are to match, the harder it may be
to get a matched patient.
(ii) Many-to-1 matching. Here there may be several control patients
who are similar to the ones treated. Hence, we could have two
patients taking the control treatment for each treated (with a
different treatment) patient.
(iii) m:n matching, A third type of matching is called many-to-many
(m:n) matching). Where there is an abundance of those taking
treatment and control (as is the case in some large national reg-
istries), this might be used.
TABLE 8.8
272

Summary of Baseline Characteristics


Prior to Matching Post-Matching
CAP (N = CAP (N = Control (N = Standardized
44,215) Control (N = 937,182) p-Value 36,386) 36,386) p-Value Difference
Age (years) 70.7 78.6 p < 0.0001 79.1 79.5 0.0327 0.0215
Gender:
Male 23,911 (54%) 525,514 (56%) p < 0.0001 17,432 (48.2%) 17,524 (48%) 0.631 0.0051
Female 20,304 (46%) 411,668 (44%) 18,862 (51.8%) 18,954 (52.1%) 0.622 0.0051
ECOG
0 523 (1%) 12,852 (1%) p < 0.0001 456 (1.3%) 444 (1.2%) 0.689 0.0030
1 1,347 (3%) 37,520 (4%) 969 (2.7%) 1,167 (3.2%) <0.001 0.0322
2 40,206 (91%) 813,570 (87%) 33,210 (91.3%) 33,128 (91%) 0.750 0.0079
3 90 (<1%) 2,733 (<1%) 79 (<1%) 68 (<1%) 0.364 0.0067
>3 2,049 (5%) 70,507 (7%) 1,672 (4.5%) 1,579 (4.3%) 0.128 0.0054

Comorbidity 14,222 (32%) 234,327 (25%) p < 0.0001 11,779 (32.4%) 11,849 (32.6%) 0.648 0.0041
Yes
Economic Evaluation of Cancer Drugs
Real-World Data in Cost-Effectiveness Studies on Cancer 273

Once the matched data sets are formed, the success of matching needs
to be determined, and then followed by estimates of treatment effects
using propensity score methods shown in Example 8.3.
Step 2: Evaluate the successfulness of matching:
Table 8.6 shows the summary statistics for all patients before and after
matching. For such large sample sizes, the p-value is small, even after
matching, where the mean matched age is 79.1 versus 79.5 years (p =
0.0327). Therefore, the standardized difference (SDiff) should be used,
which should be <0.10 for a conclusion of no difference in baseline char-
acteristics between groups as evidence for successfulness of matching.
In this case the SDiff is 0.0215, suggesting patients are matched on age
between CAP versus control groups.
Table 8.8 shows the following:

(a) Prior to matching, the baseline factors were imbalanced show-


ing strong statistical differences (even when the differences
were small, such as gender). There were more comorbidities in
the treated group (p < 0.001).
(b) After taking a matched control group so that we have 36,386
patients in each group who are as similar as possible, the
baseline characteristics are now somewhat balanced. The pro-
pensity scores (Figure 8.1) confirm this. In Figure 8.1b the pro-
pensity scores are practically superimposed between CAP and
control after matching.

This propensity scores in Figure 8.1 are determined by modeling the


treatment group assignment related to covariates as before in Example
8.3. The model generates a propensity score (PS) for each patient. This
score will be used to adjust the differences in outcomes such as survival
at 5 years and health resource use between treatment groups.
Step 3: Use the matched samples to compare groups and draw
inferences.
Under the assumption that matching has been successful so that valid
comparisons from nonrandomized samples can be made, we model the

FIGURE 8.1
(a) Prior to matching, and (b) post-matching propensity scores.
274 Economic Evaluation of Cancer Drugs

response or outcome of interest in the usual way and adjust for the pro-
pensity scores.
Hence, we would use the treatment group (matched samples of 36,386
per group) for our usual statistical analysis. There are several health
resource items of interest: elective admissions, nonelective admissions,
bed stays, outpatient appointments, and emergency attendance. Since we
are modeling the frequency of these occurrences, we will be interested
in either the mean count or incidence of using a given health resource.
That is, we might be interested in the mean number of elective admis-
sions or an incidence rate ratio. If the incidence rate ratio (the ratio of the
mean counts) is, for example, 1.19, this is interpreted as a 19% increase in
the rate of usage of that particular health resource item.
Table 8.9 shows the results using two types of models: a two-part hur-
dle model and a negative binomial model (NBM). The main difference
between the hurdle model and a model such as the NBM is that for count
models (i.e. NBM) values equal to zero and greater than zero are assumed
to come from the same data-generating process. For a hurdle model, these
two processes are not considered to be the same. The hurdle approach
models (as a mixture of two distributions): first whether a health resource
value is zero or not, and second, whether the ‘hurdle’ is crossed, the condi-
tional distribution (conditional on crossing zero) is modeled. However, if it
is the case that data have been collected from hospital records, then it may
be unrealistic to assume that zero health resource use is a plausible value.
Consequently, if zeros are not plausible values, an alternative model, such
as a generalized linear model (GLM), assuming a Gamma distribution
could be used (Khan, 2015). Further details of hurdle models and negative
binomial models can be found in Khan (2015) and Agresti (2013).
The mean number of elective admissions determined from the hurdle
model for those taking treatment was 4.78 elective admissions compared
to about 3.58 elective admissions (p < 0.0001) for the control (Table 8.9).
Hence, the incidence of elective admissions was 19% higher for those
treated compared to those not treated with the chemotherapy of interest.
When using a negative binomial model, the conclusion was the same but
with a slightly higher incidence. Note how the mean (count) number of
elective admissions differs between the models, but the ratio is similar.
This is reflected in the way the models compute effects.
Note also that the propensity score is included as part of the adjustment
into the model. The uncertainty of the incidence of elective admissions
was expressed in terms of the 95% CI: the true increase in the number
of elective admissions for those with treatment compared to untreated
range somewhere between 16% to 21% for the hurdle model and 19%
to 32% for the negative binomial model. The hurdle model appears to
model the within and between subject variability better, resulting in
lower standard errors.

8.6.2 Instrumental Variable Methods


In addition to the various types of propensity score modeling methods for
handling selection bias, another technique called the instrumental variable
TABLE 8.9
Statistical Modeling of Treatment vs. Standard of Care Health Resource Use (Hurdle and Negative Binomial Models) on Matched
Data
Hurdle Model Negative Binomial Model
Health Resource Treated Control Mean Incidence Rate Treated Mean Control Incidence Rate
Item Mean (SE) (SE) Ratio (95% CI) p-Valuea (SE)) Mean (SE) Ratio (95% CI) p-Valueb
Elective Admissions 4.34 (0.024) 3.58 (0.022) 1.19 (1.16, 1.21) <.0001 2.57 (0.121) 2.13 (0.097) 1.25 (1.19, 1.32) <.0001
Nonelective 2.55 (0.002) 2.04 (0.002) 1.24 (1.22, 1.27) <.0001 1.28 (0.018) 1.21 (0.014) 1.27 (1.24, 1.30) <.0001
admissions
Bed stays 24.99 17.06 (0.015) 1.46 (1.45, 1.47) <.0001 3.47 (0.151) 2.10 (0.090) 1.67 (1.61, 1.74) <.0001
(0.023)
Outpatient 8.97 (0.011) 8.10 (0.010) 1.09 (1.09, 1.10) <.0001 4.40 (0.092) 3.71 (0.082) 1.13 (1.11, 1.15) <.0001
appointments
A&E attendances 2.71 (0.003) 2.20 (0.000) 1.23 (1.20, 1.25) <.0001 1.28 (0.018) 1.22 (0.015) 1.25 (1.23, 1.28) <.0001
Real-World Data in Cost-Effectiveness Studies on Cancer

a Notes:  Using a hurdle model adjusted for covariates without adjustment for multiplicity.
b Using a negative binomial model adjusted for covariates.
275
276 Economic Evaluation of Cancer Drugs

FIGURE 8.2
Description of instrumental variables methods.

(IV) method can be used. Methods such as those described above (e.g. pro-
pensity models) use observed variables (age, gender, etc.) to adjust for selec-
tion bias. IV methods assume that there is a set of unobserved factors that
influence treatment received and confounding. Figure 8.2 shows how IVs are
represented.
Observed factors (X) may influence both treatment received and outcome.
Usual methods (regression or propensity score models) can deal with con-
founding for observed factors, but not unobserved factors. IV methods aim
to find instruments correlated with treatment selection but not directly with
outcome. An example is summarized from Faries et al. (2010).

Example 8.4: Example of IV: Comparison of Compliance Rates


This example is adapted from Faries et al. (2010) :

• A number of observed potential confounders (X) were collected


(age, gender, number of previous drugs, number of complete
cycles of treatment). These were different between the two
drugs. Therefore, it was assumed that if differences existed
between treatments A and B for observed variables, such dif-
ferences could also exist for unobserved variables.
• The outcome was compliance rates (cycles of chemotherapy
completed) (Y).
• Instrumental variable (IV) was the prescribing preference of cli-
nicians. The most recent prescription (treatment A or B) was a
dichotomous variable. Those who were newly prescribed to A or B
were likely to be influenced by their cancer consultant/prescriber.
• Patients who had been recently prescribed were extracted from
databases (N = 1,426 of which N = 611 took drug A and N = 815
drug B).
• Selection bias was likely to exist because the two treatments
had different toxicity profiles. This would influence both
patient and clinician choice.
• Traditional methods (regression) may not have estimated the
treatment effect on the outcome properly.
• The objective was to compare compliance rates between two
drugs (cancer drugs) over a 6-month period.
Real-World Data in Cost-Effectiveness Studies on Cancer 277

TABLE 8.10
Summary of Baseline Factors and Compliance
A (N = 611) B (N = 815)
Age (years) 55.5 56.0
Gender:
  Male 56% 54%
  Female 44% 46%
No. of previous therapies 2.1 2.3
No. completed all cycles (mean)a (Y) 66% 59% P = 0.0071
Preference for drug (IV) 47% 53% OR=3.44 (95% CI:
2.76, 4.29)
a Note: For each patient, the number of cycles received divided by the planned number of
cycles expressed as a percentage.

Results
Table 8.10 summarizes the results of the IV method.
The unadjusted mean compliance (%) was statistically different. This could
indicate better cancer management with A compared to B and how prescrib-
ing is done in practice. However, since we suspect that selection bias persists
due to clinician bias in prescribing, an IV modeling approach is used to dis-
entangle persisting bias.
In Table 8.10, the values of 53% and 47% suggest the IV is predictive of
prescriber preference (A appears to be preferred more). The next thing is
to check the relationship between the IV and the observed factors (X) – i.e.
we check the independence assumption (e.g. if prescribers are giving more
females drug A than drug B). The results of a comparison between the mod-
els (raw, adjusted using regression, and IV model are compared in Table
8.11). Compliance rates between drugs for the raw unadjusted values were:
66% versus 59%; after using standard regression techniques adjusted for
observed confounders this was 67% versus 61%; using an IV model this was
73% versus 72% (Table 8.11). Clearly, compared to the unadjusted case where
all patients were assumed to have used drug A, the IV model provided more
comparative compliance rates. In some cases (Brookhart et al., 2006; Landrum
& Ayani, 2001), differences can be much larger between unadjusted and IV
models.
In summary, standard regression methods use observed factors to adjust
for confounding. IV models make use of an IV variable that models both
observed and unobserved factors. This can be very important because,
despite propensity matching appearing successful, a question that often
comes up is “have you controlled for the unobserved confounders?” An
important challenge in using IV models is how an instrument is determined
278 Economic Evaluation of Cancer Drugs

TABLE 8.11
Comparison of Average Compliance for Each Model
Model Treatment Compliance 95% CI (%) p-Value
(Mean) (%)
Unadjusted (raw values)
A 66
B 59
Difference 7 (1.9, 12.1) 0.007
Regression A 67
B 61
Difference 6 (0.9, 11.1) 0.0095
IV A 73
B 72
Difference 1 (–3.6, 5.7) 0.0676

and validated. In the above example, it was assumed that the prescriber’s
last prescription preference affected the subsequent patient’s prescription.
The assumption that the previous patient’s prescription is related to the next
patient’s choice of treatment is likely to be untenable.

Example 8.5: Cost-Effectiveness Analysis of Potentially


Curative and Combination Treatments for Hepatocellular
Carcinoma with Person-Level Data in a Canadian Setting
(Thein et al., 2017; Cancer Medicine, 2017; 6(9):2017–2033).
In this example, the cost-effectiveness of various combination treatments
for hepatocellular carcinoma (HCC) is evaluated using the Ontario
Cancer Registry, using linked administrative data. The sample size used
was 2,222, of which about 11% received radiofrequency ablation (RFA),
14% had surgical resection (SR), and 10% received liver transplantation
(LT) monotherapy; a further 53% received no treatment (control). Hence
the comparison was between RFA, SR, and LT alone (monotherapies), or
in some combination, versus control.
The ICD-9 classification was used to identify patients with the code
155.0 along with other codes for histology (codes 8170–8175). The period
of data extraction was between January 1, 2002 and December 31, 2010.
Hence diagnosis of HCC should be on or after January 1, 2002. In brief, a
summary of this analysis is:

• Health resource use: this was obtained from previously pub-


lished sources. This included items such as outpatient visits,
emergency department visits, acute inpatient hospitalizations,
surgeries, medicines, home visits, and long-term care.
• Effectiveness data: the date between diagnosis and death –
obtained from the registry. The utility data were based on a
separate published sources.
Real-World Data in Cost-Effectiveness Studies on Cancer 279

• Estimation of effectiveness: the statistical methodology used


propensity models to address selection bias and multiple impu-
tation to address missing data.
• Cost-effectiveness: the cost-effectiveness was determined using
a net benefit approach, using the derived QALYs, which were
obtained by weighting survival time with utility (taking into
account disease stage). Other issues such as multicollinear-
ity (when independent variables are correlated) were also
addressed.
• The results show that RFA in combination with transarterial
chemoembolization (TACE) have a high chance of cost-effec-
tiveness (probability of 100%) when compared to no treatment
(cost/QALY of $2,465); while RFA alone versus no treatment
gave a cost/QALY of $15,553.

The above example shows the possibility of using real-world data to


reach an important cost-effectiveness conclusion that alternative treat-
ment options for HCC, which offer good value for money, should be
considered. The study was not free from limitations, such as limited
long-term effectiveness beyond 9 years, and the impact of the stage of
HCC. Hence when data are not available in a registry, the conclusions
will be restricted.

8.7 Summary and Conclusion


The use of RWD has become increasingly common for conducting economic
evaluations (Gansen, 2018). However, there exist a number of methodological
limitations that require addressing, including how a study that uses routine
data is designed, analyzed, and interpreted. The results from this review
only found one cancer indication that used RWD, hence it is difficult to iden-
tify cancer-specific issues. However, the issues identified can be generalized
across diseases, some of which are shown in Table 8.12, but should be con-
sidered along with other criteria for reporting economic evaluations, such as
the Consolidated Health Economic Evaluation Reporting (CHEERS) check-
list (Husereau et al., 2013). The most important consideration is to ensure
wherever possible the myriads of different types of bias. These will need to
be either designed out or adjusted for in the analyses:

• Selection bias: due to variation in access to care; or study population


not representative of the true distributions in the overall population.
• Confounding bias: association between treatment and outcome
being influenced by the presence of extraneous variables; due to
patient characteristics and comorbidities; or EHR diagnostic and
280 Economic Evaluation of Cancer Drugs

TABLE 8.12
Considerations When Using RWD for an Economic Evaluation
Design Considerations
Identify registry Availability of target data, completeness of target data, is linkage
possible? Insurance claims, national registries? Can the data be
collected during an EAMS designation or other early promising
medicine designation?
Identify outcome of Is outcome related to health resource use?
interest Mortality, complications, incidence, HRQoL, QALYs, biomarker
data?
Cost category Inpatient treatment, medication (drug), outpatient treatment,
intervention, medical aids, social care, rehabilitation, sickness
benefits
Comparators Are comparators of interest in the registry – unlikely if the
treatment has not been approved/marketed
Key measures of Can all of these be identified from the registry? Which are the
effectiveness efficacy/safety outcomes? Are these available at the times of
interest?
Completeness of the How much missing data per variable is there?
data
Geographical variables Is inequality an issue?
Time horizon What is the reference point in time – how far back?
Inflation adjustment Past costs to be estimated at current prices
and discounting
Any HRQoL – If not, where will these be found?
preference-based
outcomes
Sample size and How will sample sizes be derived? Will there be any matching,
matching and, if so, how? Will samples be taken at random?
Analysis
Has selection bias been For example, using propensity score models. How is the matching
addressed determined? How much has been stated upfront? Is a statistical
plan available?
Can an economic Is there sufficient data in the registry to populate the model?
model be populated
Assumptions Assumptions around missing health resource use, or other
assumptions on treatment delivery
Uncertainty How uncertain are the estimates from the registry?
Unobserved How will these be addressed? How will instrumental variables be
confounders defined and validated?
Evidence Does the analysis really answer the question of interest? What is
the uncertainty in the conclusions? Has this been quantified (e.g.
using confidence intervals). Any evidence that addresses a safety
concern of a drug must be robust enough to defend not doing a
long-term follow-up study (which is more expensive than using
data registries)
Real-World Data in Cost-Effectiveness Studies on Cancer 281

therapeutic codes; regional variations in standards of care; or avail-


able therapies.
• Compliance bias: due to patient third-party formularies; nonadher-
ence to treatment; or variations in patient adherence to planned
treatment affecting study outcomes.
• Information bias: due to erroneous or inaccurate capture of patient
variables in the EHRs.

8.8 Exercises for Chapter 8


1. Why are registries needed and what advantages do they have over
performing a randomized trial? What are their disadvantages?
2. What is confounding and what could be done to avoid potential con-
founding when comparing treatments for cost-effectiveness?
3. Propensity matching is a replacement for randomization. Discuss.
4. Distinguish between a pragmatic trial, a randomized trial, a case
control study, an observational study, and a registry study.
9
Reporting and Interpreting Results of Cost-
Effectiveness Analyses from Cancer Trials

When looking at the cost-effectiveness of cancer trials there are several


important results that need to be interpreted, both clinically and from a cost-
effectiveness perspective. Some of these concepts were introduced earlier,
and we will now interpret them with practical examples:

(i) Incremental costs


(ii) Incremental QALYs
(iii) Interpreting the ICER and the cost-effectiveness plane
(iv) Uncertainty analyses
(v) Value of information

9.1 Interpreting Incremental Costs and QALYs


For cost-effectiveness analyses we require the mean incremental costs. The
mean incremental cost is computed as the difference in the mean costs
between two groups. In RCTs, the mean of the cumulative costs over their
observation period per patient in each group, is used for this purpose. In
cases of imbalance (e.g. costs differ between groups in terms of their baseline
characteristics), to compute the mean incremental cost the patient-level costs
are often modeled using a statistical model. Many baseline factors can influ-
ence the mean incremental cost, such as age, gender, ECOG, disease stage
(e.g. patients whose illness is more severe may also be associated with the
larger costs). The observed distribution of the patient-level costs will also
influence the type of regression method used (e.g. if the costs are heavily
skewed, a different analysis technique might be needed). In Chapter 5, we
observed patterns of costs that were very skewed and over-dispersed, and
we assumed the costs followed a gamma type distribution (see Figure 5.4).
Usually, for cost-effectiveness, the primary analysis variable of interest in
trials will be the cumulative (total) cost incurred by the patient up to the end
of the trial. Alternatively, costs could be analyzed over a specified period
(say during initial therapy) or a well-specified part of the treatment period

283
284 Economic Evaluation of Cancer Drugs

(say staging, palliative care period, etc.) and broken down by type of cost
items (drugs, radiotherapy, surgery, supportive care, etc.) to visualize the
cost structure of the treatments.
Costs may also be incomplete (right censored) because at the end of the
trial patients might still be alive, but no longer followed up, or because they
are lost to follow-up during the trial period. Hence these costs are censored.
Patients censored before the trial ends will typically have a lower cumulative
cost than those who have not been censored, resulting in a biased estimate of
the total mean cost (see example data generated in Figure 9.1). In such a case,
one needs to apply some statistical method to correct for the bias.

9.1.1 Informative Censoring
A key issue present in most cost data is informative censoring due to the lack
of a common rate of cost accrual over time among patients. Informative cen-
soring means that the reason for censoring may well be related to treatment
(e.g. because of toxicity or patients drop out more in one arm and incur lower
costs) resulting in incomplete costs. To handle non-ignorable censoring, the
popular approaches are either weighting-based (Bang & Tsiatis, 2000; Lin et
al., 1997; Bang, H., & Zhao, H., 2014; Bang, H., & Zhao, H., 2016), or using Kaplan-
Meier estimates as weights. The latter is less popular (Etzioni et al. 1999)
demonstrated that standard survival techniques may yield biased estimates.
Lin et al. (1997) proposed a nonparametric approach that splits the time
period into small intervals and weights mean costs from each interval by
survival probabilities estimated from the Kaplan-Meier curve. An example
of this was shown in Chapter 5.

FIGURE 9.1
Simulated cumulative costs for uncensored patients compared to censored patients in a single arm.
Reporting and Interpreting Results of Cost-Effectiveness Analyses 285

Example 9.1: Modeling Costs from a Myeloma Cancer Trial


Table 9.1 shows the structure of (fictitious) data for 100 patients (50 ran-
domized to treatment A and 50 to treatment B).
Since costs are positive, a skewed gamma distribution was assumed. To
fit a gamma regression model, we added the value of 0.001 to (as in this
example) patient 4 costs because the gamma is only defined for values >0.
This creates a small bias in the estimate of the mean incremental costs but
is of little practical significance. When many zero values are present, zero-
inflated models would be preferable (see Dupuy 2018; Zuur et al. 2017).
Four covariates were used to estimate the mean cost: age, ECOG score,
gender, number of previous chemotherapies, in addition to treatment
arm. The results from the statistical output are shown in Table 9.2.
From the results in Table 9.2 we see that treatment and ECOG are the
only statistically predictive factors (p < 0.05) of the incremental mean
costs. Costs are increasing with all factors, particularly for treatment (A
is more expensive) and ECOG (as patients worsen in the baseline status,
the costs increase by a factor of £100.6 on average). The total costs for
treatment A and B can be estimated as:

Total Cost = 3125 + 625.3 * Treatment group



+23.4 * Gender + 8.90 * Age + 100.6 * ECOG

TABLE 9.1
Example Data Structure for Modeling Costs in Terms of Other Factors
Previous
Patient Cost Age ECOG Gender Chemotherapy Treatment
1 4,380 64 1 Male 2 A
2 6,290 49 2 Male 1 B
3 3,400 62 1 Male 3 A
4 0 75 2 Female 2 A
etc. …
100 4,950 39 1 Female 2 B

TABLE 9.2
Results from Modeling Costs
Factor Estimate Standard Error 95% CI p-Value
Intercept 3,125 233.45
Treatment 625.30 195.52 (243, 1007) <0.001
Gender 23.40 20.94 (–18, 64) 0.345
Age 8.90 8.11 (–7, 24) 0.551
ECOG 100.6 48.11 (6, 195) 0.021
Difference
LSmean A 4,275
LSmean B 3,650 625 (243, 1007)
286 Economic Evaluation of Cancer Drugs

Note that the model predicts the total cost for each patient. The mean of
these predicted costs is called the adjusted mean (or least squares mean,
LSmean for short). The lack of statistical significance of some terms (e.g.
gender) does not imply that these should be dropped. They may still be
used to predict the mean costs. Statistical significance is less of an issue
than generating a reliable estimate of the mean incremental cost. If the
treatment term was not statistically significant, for example, dropping it
would not make sense because we still need a reliable estimate of mean
costs for each treatment group.
Since treatment group takes the value of 1 for treatment A and 0 for
treatment B, the cost of delivering treatment A to males aged 45 (mean
age of the sample) and with an ECOG of 0 or 1 (coded as 1 if ECOG = 0 or
1, and coded as 2 if ECOG > 1) would be:

Total Cost A = 3125 + 625.3 + 23.4 + 8.9 * 45 + 100.6 = £4,275

One can compute the costs for treatment B in a similar way.


In practice, the adjusted mean costs are used to generate the mean
incremental cost of £625. The corresponding 95% CI for this incremen-
tal cost is (£243, £1,007). These values are then used for estimating the
numerator of the ICER:

mean (C1 ) − mean (C2 )


ICERmean =
mean (E1 ) − mean (E2 )

The wide 95% confidence interval for the cost difference between the
two arms in Table 9.2 (£243 to £1,007), shows that there is considerable
uncertainty in the mean cost difference (which is not unusual in cost
data).
One alternative approach to computing the mean cost difference in
case of a skewed distribution might be to compute a bootstrap estimate.
Essentially, this involves executing the above analyses many times and
from each analysis saving the mean incremental costs. This would give
a distribution of mean cost differences from which we could use the 5th
and 95th percentile to estimate the bootstrap confidence interval or use
more technical ways (like bias corrected methods). When there are miss-
ing data (as distinct from zero costs) we can use a multiple imputation
(MI) procedure. The validity of MI depends on the mechanism of miss-
ingness (see Chapter 4). Table 9.3 shows the table again with missing
costs.
A statistical model using MI to predict the missing costs results in a
model (e.g. a type of regression model with covariates) being repeatedly
executed on the data (each bootstrap sample) where missing values are
estimated. These are then essentially averaged over the number of times
the model is executed (Van Buuren, 2018; Carpenter & Kenward, 2013).
The mean costs and mean costs difference are then derived from each
analysis and the uncertainty can be quantified (e.g. through use of confi-
dence intervals). The next step is then to repeat the process for effective-
ness measures.
Reporting and Interpreting Results of Cost-Effectiveness Analyses 287

TABLE 9.3
Data Example Showing Missing Costs for Multiple Imputation-Based Analyses
Previous
Patient Cost Age ECOG Gender Chemotherapy Treatment
1 4380 64 1 Male 2 A
2 . 49 2 Male 1 B
3 . 62 1 Male 3 A
4 0 75 2 Female 2 A
etc.
100 4,950 39 1 Female 2 B

9.2 Interpreting Incremental QALYs


The distribution of QALYs are the result of the patient’s observed (whether
censored or not) survival times and HRQoL (i.e. utility) score(s) over that
period. The derivation and analysis of QALYs in cancer trials are not trivial.
Each patient will have repeated measurements over time. The measures may
be categorized as being observed in a progression-free (PF) period or post-
progression (PP) period, so that we can be sure that utilities over time make
sense (e.g. we expect mean PP utilities to be lower than PF utilities). Utilities
can also be defined for other in-treatment or between-treatment periods and
events (such as adverse events, partial response, or cure).
For each patient profile, we can estimate the QALY using estimates of
the survival rates at each of the specified time points (to generate the QoL-
adjusted survival curve). This was mentioned in Chapter 4 and we show a
practical example below. For now, the QALY is derived over the entire overall
survival time, ignoring the extrapolated survival period.

Example 9.2: Deriving the QALY Using the Kaplan-Meier Curve


We assume that the survival rates over time were observed as shown in
Table 9.4 for one arm of a treatment, along with utilities at three-monthly
time points. Note that utilities were not collected monthly whereas sur-
vival rates are reported monthly in Table 9.4 (this is arbitrary as they could
be reported every two months, but it is useful to report them at time points
where utilities are available). One way of computing the QALY is to derive,
for each patient, the area under the HRQoL curve. So, for example, if a
patient had measures of EQ-5D at 0, 3, 6, 12, and 18 months of 0.52, 0.58,
0.54, 0.43, 0.41, respectively, the QALY between time 0 and 18 months would
be computed. The mean QALY across all patients can then be computed.
An important note is that the utilities used in Table 9.4 are derived
from some external source (e.g. published data). If we have patient level
HRQoL, then for each individual patient we could compute an individ-
ual QALYAUC and perform extrapolation (often drawing a straight line
288 Economic Evaluation of Cancer Drugs

TABLE 9.4
Survival Data with Corresponding Utilities for
Example 9.2
Survival Time
(Months) (t i) % Alive (Si) Mean Utility (Q i)
0 100 0.54
1 85
2 80
3 75 0.52
4 71
5 65
6 60 0.53
7 48
8 35
9 25 0.42
10 20
11 15
12 10 0.39

not only between discrete time points but also between the last observed
time point and some other arbitrary future time point).The assumption
of constancy of utility between time points is strong but practical, as
long as they are not too far apart in time.
To estimate the QALY we use the linear trapezoidal rule (which essen-
tially divides the total area under a (continuous) curve into discrete
intervals (trapeziums) and, using the formula for the area of the trape-
zium, we can compute the QALY) (Figure 9.2).
For the example, in Table 9.4, by 12 months, only 10% of the patients were
alive (taking into account censoring). But the mean utility is only available
at baseline (time 0), 3, 6, 9 and 12 months. Applying the trapezoidal rule, the
mean QALY for all patients (for a given treatment group) is calculated as:

(0.54 + 0.52 ) / 2 * (1.00 + 0.75 ) / 2 * ( 3 − 0 )

+ (0.52 + 0.53) / 2 * ( 0.75 + 0.60 ) / 2 * (6 − 3)

+ (0.53 + 0.42) / 2 * (0.60 + 0.25 ) / 2 * (9 − 6 )

+ (0.42 + 0.39 ) / 2 * (0.25 + 0.10 ) / 2 * (12 − 9 )

= ( left for the reader )


When collected during the trial, mean utility estimates per time point for
different health states can be estimated by statistical methods, such as
a mixed effect (multilevel) regression model that accounts for the num-
ber of time points, possible treatment covariates, and any missing data
that takes into account the intra-patient correlation. This is a separate and
more efficient method to computing patient-level QALYs noted above,
Reporting and Interpreting Results of Cost-Effectiveness Analyses 289

FIGURE 9.2
Using the trapezoidal rule.

because utilities at each of the time points are adjusted for possible con-
founders. In a similar approach to deriving mean incremental costs, non-
parametric bootstrapping methods may be used to derive the confidence
intervals for the QALYs per treatment arm and the incremental QALY.
The utility-adjusted survival will always be lower than the unadjusted
survival (unless the HRQoL is 1 at each time point, in which case it will
be the same as the overall survival curve). Depending on the relationship
between survival length and HRQoL, the QALY value will vary with sur-
vival time (e.g. shorter or longer overall survival time) and possibly other
factors (e.g. age, gender). It may also vary with other post-treatment fac-
tors – for example, non-responders may have a shorter survival time and
a worse HRQoL than longer-term survivors (Figure 9.3).

20

15
Percent

10

0
0 500 1000 1500
days

FIGURE 9.3
Comparison of simulated survival and quality-adjusted survival.
290 Economic Evaluation of Cancer Drugs

Example 9.4: Deriving a QALY with Extrapolated Survival Data


In Example 9.1, 10% of patients were still alive at the end of the trial at 12
months. In the following situation, extrapolation of survival rates beyond
12 months is estimated. However, utility data beyond 12 months needs to
be estimated from published data or some assumptions have to be made
about their behavior. For the purpose of this example, we shall assume a
steady linear decline after 12 months. Table 9.5 shows extrapolated sur-
vival rates with estimated utilities after month 12. Note that after month
12 there is uncertainty in both survival rates and utilities. Using similar
methods as used in Example 9.1, we can then estimate the QALY’s.
The QALYAUC can be computed as follows using the trapezoidal rule:

QALYAUC = (0.632 + 0.588 ) / 2 * (1.00 + 0.80 ) / 2 * ( 3 − 0 )

+ (0.63 + 0.80 ) / 2 * (0.588 + 0.522) / 2 * (6 − 3 )



+ (0.43 + 0.36 ) / 2 * (0.391 + 0.388 ) / 2 * (13 − 12)

+ (0.17 + 0.15) / 2 * (0 + 0.03 ) / 2 * ( 24 − 23 )


Since the estimates of utility are assumed to fall at a constant rate after
month 12, we can directly estimate the utilities at any time point beyond
12 without any need for interpolation. The fact that the utility at 24
months is estimated above zero should not be a surprise because of the
use of a constant falling utility, which varies with time and not with the
proportion alive – and, hence, is a limitation of assuming linear (con-
stant) decline.
Another approach would be to estimate a fixed utility value for the last
month(s) of life the patient was alive. In fact, estimating utility values
beyond the trial observation horizon is fraught with difficulties and will
need some scenario analysis with alternative utility assumptions related
to the longer-term outcome of the still-surviving patients and their
health state(s). For example, as shown in Chapter 3, utility may increase
and then fall depending on whether other anti-cancer treatments have
been given.

9.3 Relationship between Costs and QALYs


In many cases patient-level life-years and QALYs will be (positively) corre-
lated with the patient’s total costs. The longer patients survive, the more care
and monitoring they will receive. In cancer trials it is only after an often long
post-therapy period that the cancer-related costs might fall to zero, if at all.
Even if considered cured, long-term survivors often have life-long sequelae
that still need medical attention. Moreover, HRQoL may also improve over
time as symptoms and treatment-related side effects wane. Generally, one
Reporting and Interpreting Results of Cost-Effectiveness Analyses 291

TABLE 9.5
Survival Data with Extrapolated Survival Rates
for Example 9.3
Survival Time
(months) Alive (%) Utility+
0 100 0.6323866
1 100
2 89
3 80 0.5885334
4 71
5 69
6 63 0.5227535
7 55
8 55
9 55 0.4569736
10 55
11 49
12 43 0.3911938
13* 36 0.3810000
14* 33 0.3600000
15* 32 0.3390000
16* 29 0.3180000
17* 29 0.2970000
18* 25 0.2760000
19* 23 0.2550000
20* 20 0.2340000
21* 18 0.2130000
22* 9 0.1920000
23* 3 0.1710000
24* 0 0.1500000
Notes: * Extrapolated survival rates and utilities.
+ Estimated as a linear (constant rate) decline beyond

month 12.

would expect a U-shaped evolution of HRQoL over time for these patients.
One might also expect the variance of costs to increase in relation to the
length of (quality-adjusted) survival, resulting in a heteroscedastic relation-
ship. Figure 9.4 shows such a typical (simulated) situation.
From Figure 9.4, at the patient level, the cost per QALY ratio or cost-effec-
tiveness ratio (CER) is skewed. This is due to the skewed distribution of the
individual survival times, even after the transformation of survival times to
QALYs as shown in Figure 9.2. The highly right-skewed nature of the patient-
level costs coupled with the similar skewed nature of the individual survival
times can lead to very high cost per QALY ratios for some patients (those
292 Economic Evaluation of Cancer Drugs

1000

800
Cost ( x 1000 Euro)

600

400

200

0
0 200 400 600
qaly

FIGURE 9.4
Relationship between patient-level costs and QALYs.

with large costs and relatively low survival or HRQoL). Plotting these per
trial arm helps to yield some insights at the individual level between the
treatments under study and may be worth investigating further (for exam-
ple: Are the distributions overlapping? Similarly shaped? Any sub-groups?
etc.).

9.4 Interpreting the ICER and the Cost-Effectiveness Plane


Once we have constructed the mean incremental cost and incremental effect,
we need to construct the ICER and present the uncertainty around it. The
‘base’ or ‘reference’ case and (sometimes confusingly and wrongly called)
the ‘baseline’ ICER are considered the key estimates of the mean ICER for
which the uncertainty needs to be quantified. The ICER and its uncertainty,
as presented on a cost-effectiveness plane divided into four quadrants, were
introduced in Chapter 1. Table 9.6 shows the different ways of interpret-
ing the ICER for two treatments (A and B) when the incremental costs and
QALYs are positive or negative.

9.4.1 Uncertainty
Since the ICER is computed as a single value (given that it is based on the dif-
ferences between two means), the uncertainty around its value is calculated
by simulating several thousands of ICERs (in fact by simulating the initial
Reporting and Interpreting Results of Cost-Effectiveness Analyses 293

TABLE 9.6
Interpretation of the ICER for Two Treatments A and B
Incremental Costs Incremental QALYs
(Cost Difference) (Effect Difference) ICER ICER Interpretation
+ve (A > B) +ve (A > B) +ve Cost are higher for A, A
more effective
+ve (A > B) –ve (A < B) –ve Cost are higher for A, A less
effective
–ve (A < B) +ve (A > B) –ve Cost are lower for A, A more
effective
–ve (A < B) –ve (A < B) +ve Cost are lower for A, A less
effective
Notes: +ve = positive; –ve = negative.

trial by bootstrapping methods) to get a feel for their distribution and how
many of these lie above or below the CE threshold.

Example 9.5: ICER and Cost-Effectiveness from a Lung Cancer Trial


Figure 9.5 shows 10,000 simulated incremental costs and effects for
a NSCLC trial comparing erlotinib with BSC (Khan, 2015). The X-axis
shows the mean incremental effectiveness (measured in QALYs) and the
Y-axis the mean incremental costs. In addition, there is a vertical line at
zero, which is presented to help interpret the graph (values to the right
of 0 show the new treatment, erlotinib, is more effective and to the left of
0 show it is less effective). The Y-axis starts at £2,000, which shows that
erlotinib is more costly. We could have started the Y-axis at zero, but
since erlotinib is never cheaper than BSC, starting at £2,000 is fine for the
purposes of this example. The other two reference lines (horizontal and
vertical) show the observed mean incremental costs (horizontal) and the
mean incremental effect (vertical). Where these two lines cross is where
values of the ICER can be determined. In this case, the mean incremen-
tal cost is about £7,891 and the mean incremental effect is 0.139 QALYs,
yielding an ICER of £7,891/0.139 = £56,777. A 95% confidence ellipse
could be added to the graph to highlight the mean ICER of all bootstraps
or the original trial ICER.
In Figure 9.5, there are some outlier incremental costs, which suggest
that erlotinib could be very effective (e.g. incremental QALYs of >0.32)
and in some cases erlotinib worse (incremental QALYs of <–0.03). This
reflects the uncertainty around the true mean incremental costs and
effects. There are also a few observations where the incremental effec-
tiveness ≈ 0, in which case the ICER tends to infinity. However, what is
less certain is that most ICERs are >£30,000. In Figure 9.9 we can visual-
ize the uncertainty in a graph called the cost-effectiveness acceptability
curve (CEAC).
294 Economic Evaluation of Cancer Drugs

FIGURE 9.5
Cost-effectiveness of erlotinib versus BSC.

The more general form of the CE plane can be presented as shown in


Figure 9.6 (reproduced from Chapter 1). We see that most of the simu-
lated ICERs fall in the north eastern quadrant. Clockwise from Figure
9.6, we note these quadrants as NE (++), SE (–+), SW(––) and NW(+–). The
decision in case of the dominant SE quadrant is straightforward (accept
new therapy), as is the decision in case of the NW quadrant (reject new
therapy, Example 9.6). In the case of the SW quadrant, one has to make
a value judgment about the new treatment: are we willing to forego
some clinical efficiency for the sake of a (possibly much) lower cost of
care? This could be a possible decision in very stretched health budgets
whereby the health gain of one group of patients would be (hopefully
slightly) decreased but would allow other patients get some care that
would otherwise not be available to them.
For example, cancer patients would then lose some clinical benefit, but
the money saved could be allocated to fund dialysis units for end-stage
renal disease patients. This is then a policy decision across disease areas.
The most common situation when it comes to funding new treatments
concerns patients with the same indication for which two (or possibly
more) treatments are compared. Here also a value judgment has to be
made: how much more are we willing to pay for an increased clinical
benefit. That is, what is our (i.e. the decision-maker’s) maximum will-
ingness-to-pay (WTP) threshold for one unit increase in clinical benefit?
In cancer, as in other fatal or deadly diseases, the clinical benefit would
at least have to be expressed as a gain in life expectancy (i.e. life-years
gained or LYG) or, even better, given the toxicity of most cancer therapies,
as quality-adjusted life-years (QALY) gained. Another useful plot is to
plot the LYG versus the QALYs, which shows the loss of life-years related
to lower HRQoL. In fact, this is equivalent to an indirect estimation of
Reporting and Interpreting Results of Cost-Effectiveness Analyses 295

FIGURE 9.6
Cost-effectiveness plane.

years of life lost (YLL) from a decrease in quality of life (QoL) due to
treatment toxicity and adverse events and sequelae.
The WTP may differ widely between stakeholder groups such as
patients, physicians, and other health professionals, health economists,
and decision makers (in reimbursement policy committees). This last
group also includes clinicians and administrators at hospital level,
when, for example, the hospital is funded by a fixed annual budget. In
practice the WTP will also depend on other factors, such as the rarity
of the disease, its health impact (morbidity and mortality), the demo-
graphic target group (children, women, the elderly, etc.), the existence
of an alternative treatment or not (as in orphan or genetic diseases), and
the overall wealth level of the country and healthcare system. We can
readily plot this (or several) threshold(s) on the ICER plane once we have
decided upon the WTP threshold(s). Often cited WTP thresholds in
the UK are £15,000, £20,000–£30,000 and £50,000 per QALY gained
(for end of life care). Finally, it should be noted that the ICER is
expressed in an absolute value, i.e. for 1 QALY.

Example 9.6: Impact of Varying WTP Thresholds


In the following example we can readily see that if our WTP threshold is
set at $100,000 per QALY then we would probably accept the new treat-
ment, as most estimates fall below the threshold (a gain of 0.05 QALY =
2.6 weeks at a cost of $4,000 = $1,538 additional expenses per quality-
adjusted week gained, or $80,000 per QALY in Figure 9.7).
296 Economic Evaluation of Cancer Drugs

FIGURE 9.7
Impact of varying WTP (CE thresholds).

9.5 Presenting and Interpreting Results


from Uncertainty Analysis
The purpose of sensitivity analysis in health economic evaluation is to assess
the uncertainty of conclusions (such as those based on the ICER) by vary-
ing the model inputs. Data from a single clinical trial might be of limited
value, especially if it is the only trial that has been carried out for the dis-
ease under investigation (or it is unlikely a similar trial will be conducted
again in the near future). For example, assuming that the treatment-related
adverse event rate is 5% for a particular treatment group, but the reliability
of the rate is questionable (because, for example, the trial was unblinded
and any assessment of treatment relatedness is biased; or the rate has wide
confidence intervals), one might wish to assess the impact on the ICER if the
adverse rate was larger or smaller than 5%.
In decision trees or Markov models and other simulation models used in
HTA, uncertainty is assessed by techniques such as ‘one-way sensitivity anal-
ysis,’ ‘two-way sensitivity analysis,’ and ‘probabilistic sensitivity analysis’
(PSA). One-way sensitivity analysis is where single inputs (costs, or effects,
or model parameters – e.g. estimates of key effects such as response rates)
are varied by a certain amount. If there are multiple parameters, one factor is
Reporting and Interpreting Results of Cost-Effectiveness Analyses 297

varied while the others are held fixed. For example, the cost of drugs, might
be varied by +10% and the impact on the reference ICER observed, while all
other inputs are held constant. Second, the parameters are varied accord-
ing to their lack of accuracy or uncertainty around their (mean) values. In
this approach, different parameters may be varied by different amounts. For
example, if it is not possible to have a response rate below 20%, then one
could vary the parameter as far as 20% only.
Two-way sensitivity analysis is where two inputs are simultaneously varied.
For example, the adverse event rate might increase by 10% and utilities reduce
by 10% (at the same time). The effect on the ICER (i.e. how it has changed from
the base case value) is then observed and plotted on a two-way graph. An alter-
native approach might be to work out the simultaneous percentage changes in
one or two inputs (or sometimes three) simultaneously for a required ICER (i.e.
working backward for a desired ICER), such as changing the price of new drug
to attain an ICER threshold of £30,000 per QALY. With patient-level data, one
can increment inputs (costs, effects) by a certain percentage (so multiplying
each value on one arm by 1.10 would increase these by 10%).
In sensitivity analysis, the decision about the actual amounts to vary the
inputs by is somewhat arbitrary. One could use 95% (or even 90%) confidence
intervals to describe the uncertainty of some input variables. For example,
one could calculate the 95% CI of the mean utility or mean QALYs for a given
treatment group, and also report the 95% CI for the mean costs. The upper
and lower confidence limits from these could then be used to assess the
impact on the ICER in a further sensitivity analysis. The upper and lower
95% confidence intervals of inputs, although plausible, may result in some
extreme but rare ICERs. An alternative approach is to simply present the
95% confidence interval for the mean ICER using a more complex statistical
approach such as Fieller’s theorem (O’Brien & Briggs, 2002) or some other CI
estimation method.
The plausible range of values for the confidence interval of the observed
(simulated) ICERs can lead to different interpretations, especially if the con-
fidence interval covers two or more regions in the cost-effectiveness plane.
It is therefore useful to count the proportion of ICERs in each region of the
cost-effectiveness plane. For example, if the 95% CI for the ICER ranges from
(–£456 to +£899) for a mean ICER of £678/QALY, there are two possible infer-
ences: on the one hand, the new treatment is more expensive (£899) but also
more effective; on the other hand, the new treatment is less effective but
also cheaper (–£456). We need to know the position of incremental costs and
effects (numerator and denominator) in the cost-effectiveness plane to inter-
pret the results properly. In this example, if the cost effectiveness thresh-
old was £1,000, both decisions could be implemented, although it would be
doubtful if a decision-maker, health professional, or member of the public
would accept funding a cheaper but less efficient treatment, certainly for
cancer. This implies that only quadrants A and B are relevant for decision-
making in practice.
298 Economic Evaluation of Cancer Drugs

We now provide examples and interpretations of each of the analyses:

(i) One-way sensitivity analysis


One way of displaying the impact on the ICER in a one-way sensitiv-
ity analysis is through the use of a type of plot called a ‘tornado dia-
gram.’ These are essentially horizontal bar charts that attempt to show
the influence of the factors (for both a positive and negative change per
variable) that impact the ICER. An example of a tornado or influence
plot is shown in Figure 9.8 for some simulated data for two treatments
A and B (not the same as the above example). What is apparent from the
plot is that varying treatment A costs by +10% does not impact the ICER,
whereas varying the costs of treating adverse events on treatment B by
the same amount results in large changes in the ICER.

Example 9.6: One-Way Sensitivity Analysis


Assume the mean cumulative costs for treatments A and B are £3,000
and £4,000 respectively with mean utilities of 1.10 and 1.20. This gives
a base case ICER of (£4,000 – £3,000)/(1.20 – 1.10) = £10,000/QALY for B
versus A as our reference case ICER. This is the calculated mean ICER
prior to any sensitivity analysis. We will now vary the mean cost and
mean utility by + and – 10% one at a time. With all other factors (costs for
treatment B, utilities for A and B) remaining the same. Reducing the cost
of treatment B by 10% has the greatest impact on the ICER.

FIGURE 9.8
Tornado plot for one-way sensitivity analysis.
Reporting and Interpreting Results of Cost-Effectiveness Analyses 299

The impact on the ICER from one-way sensitivity analysis is shown in


Table 9.7 below.

TABLE 9.7
One-Way Sensitivity Analysis for Example 9.4
ICER
Base case £10,000
Costs treatment A +10% £7,000a
Costs treatment B –10% £6,000b
Utility treatment A +10% £9,090c
Notes:  a A 10% increase of £3,000 = £3,300, so that the
revised ICER = £4,000–£3,300/0.1 = £7,000.
b A 10% decrease of £4,000 = £3,600, so that the

revised ICER = £3,600–£3,000/0.1 = £6,000.


c A 10% increase in utility of 1.1 = 1.21, so that the

revised ICER = £4,000–£3,000/0.11 = £9,090.

(ii) Two-Way Sensitivity Analysis

Example 9.7: Two-Way Sensitivity Analysis


Following on from Example 9.6, we now show the impact on the ICER
when two factors are varied simultaneously. In the first few rows of Table
9.9 we note that costs and utilities have been varied by a 10% increase for
treatment A, while simultaneously we assume that costs and utilities for
treatment B have decreases by 10%, resulting in a revised ICER of £2,307/
QALY. In other words, if the costs of treatment A were to rise by 10%
and HRQoL also improved by a similar amount, but treatment B costs
were to fall along with the QALYs by 10%, treatment B would be cheaper
but also less effective. In the second row, the contrary is assumed where
treatment A costs and utilities are assumed to fall by 10%, whereas for
treatment B they are assumed to increase. In this case the ICER rises to
over £5,000/QALY. In Table 9.8 the ICER fell by 30% to 40% as a result of
varying drug costs by +10%.

(iii) Presenting Sensitivity Analysis


In TA299 (NICE TA299, 2013), the treatment for chronic myeloid leuke-
mia with bosutinib was appraised and a one-way sensitivity analysis
was not conducted:
Extensive one-way sensitivity analyses were not performed as Pfizer
believed structural uncertainties were greater than parameter uncer-
tainties. Scenario analyses were performed instead.
The resulting scenarios are shown in Table 9.10 using fictitious data but
following a real situation.
300 Economic Evaluation of Cancer Drugs

TABLE 9.8
Two-Way Sensitivity for Example 9.2
ICER
Treatment Factor Change (Base case £10,000)
A Costs +10% –£2, 307
A Utility +10%
B Costs –10%
B Utility –10%
A Costs –10% £5,151
A Utility –10%
B Costs +10%
B Utility +10%

Example 9.8: Resulting Scenarios from Varying Inputs


What is noticeable from Table 9.9, is that real-world issues, such as taking
additional treatments and higher dosing (e.g. in TA299, it was assumed
the dosing could be increased from 200 mg used in the clinical trial to
500 mg in real-world practice) result in increased ICERs and can reverse
the decision for a drug to be cost-effective.

TABLE 9.9
Scenario Analyses
Revised Impact of New Treatment
Scenario ICER (£) vs. Comparatora
Base case 21,421 New treatment dominant
Reduce cohort age by 10% 23,662 New treatment dominant
Use the lower 95% CI for the hazard ratio 28,656 New treatment dominant
Use the upper 95% CI for the hazard ratio 18,332 New treatment dominant
Fitting exponential curve to both arms 22,843 New treatment dominant
Fitting Weibull model to both arms 24,982 New treatment dominant
Fitting exponential curve to one arm and 25,333 New treatment dominant
Weibull to the other
Assuming mean OS increase by 20% 20,116 New treatment dominant
Time on treatment is increased by 6 months 29,155 New treatment dominant
Dosing in real life increase by 50% 33,213 New treatment dominated
Medical management costs increase by 20% 25,775 New treatment dominant
Additional treatment taken at a cost of £25 per 25,999 New treatment dominant
month
Utilities 20% higher 18,994 New treatment dominant
Utilities 20% lower 31,669 New treatment dominant
a Note:  Threshold is £30,000 per QALY.
Reporting and Interpreting Results of Cost-Effectiveness Analyses 301

(iv) Probabilistic Sensitivity Analysis


In probabilistic sensitivity analysis, the outcomes (mean costs, mean
QALYs, mean OS) and/or inputs are considered to originate from a
probability distribution. Hence, we need to simulate their uncertainty
by drawing from a probability distribution (usually several thousand
times). For example, if the trial size of the study was 300 per group, and
we want to assess how a change in, say, the post-progression utility
impacts the ICER, we could generate 5,000 samples, where each simu-
lated sample consists of 300 values (e.g. utility, survival, costs). For each
of the simulated trials we can then recalculate the mean utility, incre-
mental utility, and ICER. We can then summarize all 5,000 ICERs and
other values (incremental costs, QALYs).
An important feature of the data in an economic evaluation is that some
variables are correlated and so the simulation will need to ensure that this
correlation structure is preserved during the sampling so that the PSA
gives sensible (and less biased) results. This can become complicated in
practice though. If we wish to analyze the impact of increased dosing (as
measured by the average dose received in one of the arms in the trial), we
might expect an increase in drug costs and therefore the overall mean cost
of the treatment. Furthermore, increased dosing would probably increase
the occurrence of adverse events and possibly their severity (i.e. increase
the arm’s treatment toxicity), which in turn would increase the adverse
events treatment costs in the arm. Higher toxicity will in turn impact post-
treatment HRQoL. In some cases, it may even increase the toxicity-related
deaths and therefore decrease the overall survival. These two latter effects
will then decrease the mean QALYs. Hence, a change in one input (average
drug dose) can have positive or negative consequences for other inputs, all
of which would have to be considered. This is why, in practice, researchers
often limit the sensitivity analysis to more aggregate variables.
The usual frequentist approach to PSA uses Monte-Carlo simulation to
estimate the overall uncertainty of cost-effectiveness. In this respect it is
like estimating a population parameter from a single trial as it considers
that the observed trial values are a single realization of a large number
of possible similar trials. This detailed approach is described as follows:

(i) Identify the inputs (variables) to vary (e.g. cost, utilities, sur-
vival, adverse events, etc.).
(ii) Identify the distributional parameters associated with the
inputs (e.g. mean, standard deviation, rates).
(iii) Identify or estimate the correlation or covariance matrix of the
set of variables from which to simulate, if required.
(iv) Determine whether the Monte-Carlo simulation will be from a
univariate or multivariate distribution.
(v) Determine whether a multivariate simulation will take into
account the ‘mixed’ nature of the distributions (i.e. will the sim-
ulation assume data are multivariate normal or a combination
of normally and non-normally distributed data).
(vi) Write software code to simulate the pseudo-trials.
(vii) Compute the ICER for each simulation.
302 Economic Evaluation of Cancer Drugs

(viii) Plot the resulting ICERS on the CE plane and determine the
proportion of ICERs above a certain CE threshold value (λ).
(ix) Plot the cost-effectiveness acceptability curve (CEAC).

Note that with two arms you have only one ICER and one CE plane, if
there are more than two arms then one can overlay two or more compar-
isons on the same CE plane (arm A versus control; arm B versus control)
or the same CEAC.

Example 9.9: Probabilistic Sensitivity Analysis


An example is taken from Lee (2012) for a lung cancer trial. The follow-
ing costs and effects were simulated from a multivariate distribution for
each arm (a joint distribution of several variables with each variable hav-
ing his own distribution):

(a) Total costs – assumed to be normally distributed.


(b) Utilities – assumed to be beta binomial (pre- and post-progres-
sion utility separately).
(c) Overall survival and progression-free survival – assumed to be
exponentially distributed.

Hence, four components of the ICER were to be simulated. The objective


was to simulate from a multivariate distribution where variables (inputs)
had different distributions. The mean and standard deviations for each cost
component is shown in Table 9.10 below for the erlotinib treatment group.
As noted above, it would be incorrect to simulate these independently
and then merge them together. What is needed is a correlation or covari-
ance matrix. We assume that the five components have the correlation
matrix shown in Table 9.11.
Simulating multivariate data from mixed distributions can be very
complex. Several methods exist and we will describe one of them here
that uses the Fleishman power transformation method (Fleishman,
1978). A method using copulas is also possible and interested research-
ers may wish to explore this. Simulation using copulas was successfully
accomplished by Khan (2015) . Details of how this can be achieved are
found in the technical appendix A9.1.

TABLE 9.10
Distributional Assumptions of Inputs for in Probabilistic Sensitivity Analyis
Distribution
Assumed Justification
Total costs Normal Costs > 0 and some extreme values
EQ-5D pre-progression utility Beta-binomial Utilities between 0 and 1
EQ-5D post-progression utility Beta-binomial Utilities between 0 and 1
OS Exponential Survival times assumed exponential
PFS Exponential Progression-free survival
exponential
Reporting and Interpreting Results of Cost-Effectiveness Analyses 303

TABLE 9.11
Correlation Matrix Used for Simulating Multivariate Data (Experimental Arm
Only)
Total Costs PrP-utility PP-utility OS PFS
Total costs 1
Pre-utility 0.61 1
Post-utility 0.65 0.89 1
OS 0.71 0.58 0.62 1
PFS 0.69 0.55 0.66 0.88 1
Notes:  PrP, pre-progression; PP, post-progression.

TABLE 9.12
Output from Multivariate Simulation Using the Fleishman Method for the
Experimental Treatment Group for Example 9.9
Total PFS
Patient Costs (£) Pre-utility Post-utility (Months OS (Months)
1 4,000 0.4 0.2 1.2 1.8
2 8,000 0.8 0.4 2.2 4.4
3 3,500 0.8 0.4 1.1 2.7
Etc. … … … … … …

350 8,500 0.7 0.2 4.9 6.2

For each simulated data set (for each arm), the mean total costs and
QALYs are calculated. It is important that the way the mean total costs
are computed from each simulated sample uses the same methods in the
base case approach. For example, if the mean costs were estimated using
a generalized gamma model in the base case analysis, this should be
repeated for each data set and not using the simple mean. An example of
the structure of the data for each simulation may look like that in Table
9.12 (the data are fictitious). Table 9.13 shows how the data are structured
and simulated for both arms.
The data in Table 9.13 can be used to compute the mean ICER along
with 95% CI as well as the CEAC.
(iv) Cost-Effectiveness Acceptability Curves
The next step will be to determine the proportion of ICERs below a
specified CE threshold (or willingness-to-pay). Recall that an acceptable
ICER is defined as:

∆ C / ∆ e < λ

where λ is the CE willingness-to-pay threshold, ∆c is the difference of


the mean costs per arm and ∆e is the difference of the mean QALYs per
arm. From the 10,000 simulated ICERs, we can calculate the proportion
of simulations that are below any given λ and we can then plot these
304 Economic Evaluation of Cancer Drugs

TABLE 9.13
Simulated ICERs from Each Data Set
Mean
total cost Mean total Mean Mean ICER K
Simulation K cost K QALY K QALY K Erlotinib vs.
K = 1 to 10,000 (erlotinib) (placebo) (erlotinib) (placebo) placebo
1 6,700 5,200 1.80 1.77 50,000
2 8,300 5,200 1.60 1.59 310,000
3 11,700 10,100 1.10 1.0 16,000
Etc…
10,000

proportions in ascending order of λ to generate the CEAC. An alternative


expression introduced earlier is:

INMB = λ * ∆ e − ∆ C

What we require is the probability of the INMB to be >0 for specified


values of λ. In Table 9.14, the proportion of times the ICER is less than the
CE threshold of £1,000 and £10,000 is zero. As the CE increases to £20,000,
one of the simulated ICERs is below £20,000. The process continues and
then the probabilities are plotted on the Y-axis and the CE threshold on
the X-axis. From this, we can generate the CEAC as shown in Figure 9.9.

TABLE 9.14
Proportion of ICERs below the CE Threshold for First 3 Simulated ICERs
CE Is ICER > CE Probability ICER <
Simulation ICER (£) threshold (£) Threshold? CE Threshold
1 50,000 1,000 No 0
2 310,000 1,000 No 0
3 16,000 1,000 No 0

10,000

1 50,000 10,000 No 0
2 310,000 10,000 No 0
3 16,000 10,000 No 0
… Etc.
10,000
1 50,000 20,000 No 0
2 310,000 20,000 No 0
3 16,000 20,000 Yes 1/3

10,000
Reporting and Interpreting Results of Cost-Effectiveness Analyses 305

FIGURE 9.9
CEAC showing probability of cost-effectiveness of erlotinib versus placebo.

The CEAC in Figure 9.9 shows the (empirical cumulative) probabil-


ity of cost-effectiveness for varying thresholds up to £100,000 per QALY.
The horizontal line represents the 80% probability of cost-effectiveness.
Where this line meets the curve, it tells us that for an 80% chance of cost-
effectiveness with the new treatment, the cost per QALY is about £56,000.
At a threshold of £30,000 per QALY, the chance of cost-effectiveness is
<10%. The vertical line at £50,000 is the threshold for end-of-life treat-
ments in the UK. Hence, if erlotinib qualified for an end of life reim-
bursement, the chance of cost-effectiveness approximates 70% if society
was prepared to pay £50,000 per QALY. At some very large value of CE
thresholds there will be a 100% chance that the new treatment (or for that
matter any new treatment) is cost-effective, because ultimately if there
was a very large budget (money is not a concern), λ tends to infinity.
There is however no universal agreement as to what probability level
is acceptable for a new treatment to be cost-effective. The general rule
is the higher the probability, the better. In practice, other factors such
as disease type, target population type and size, availability, and num-
ber of alternative treatments might all play a part in the funding deci-
sion. The CEAC threshold is often based on the primary endpoint of a
trial. Separate CEACs for subgroups and secondary endpoints could be
derived. How results like these are to be interpreted is not always clear
(or easy). For example, consider a CEAC for the primary endpoint (sur-
vival) that is modestly cost effective (e.g. 65% chance of cost-effective at a
threshold of £30,000/QALY). Even if the secondary endpoints generated
strong effects, the CEAC for the primary endpoint would not change.
For this reason, one might argue that decisions should not be made
on the primary endpoint CEAC alone, unless the CEAC encompasses
some secondary efficacy endpoints also (not to mention the correlation
between primary and secondary efficacy endpoints).
306 Economic Evaluation of Cancer Drugs

9.6 Bayesian Sensitivity Analysis


In the above example, Monte-Carlo simulation was used to estimate the (long-
run) probability of cost-effectiveness for varying levels of the CE threshold.
The Bayesian approach is likely to lead to the same conclusions, depending
on prior beliefs about parameters and/or distributions of costs and effects.
For example, if the prior distributions are non-informative (i.e. observed data
dominate the results over the weak prior beliefs), the usual (frequentist) and
Bayesian analyses will give very similar results. If however there is some
previous information available, this can be included in the analysis through
‘informative priors.’
Different choices for priors can lead to different cost-effectiveness conclu-
sions (Maiwenn et al., 2000; Al & Van Hout, 2000). However, Bayesian analy-
sis, despite giving similar results in the case of using weak prior information
gives a different interpretation of the results. In Chapter 4, we noted how,
in hypothesis testing, we simply decide one hypothesis over the other as
being true. The Bayesian philosophy quantifies the probability of a hypoth-
esis being (credible) true. A clinician is more likely to appreciate the idea of
the chance of a hypothesis being true, than deciding one of two hypotheses

TABLE 9.15
Summary of Main Issues in Uncertainty Analyses
Method Situation/Model Main Issues
One-way Decision model/ • Arbitrary amounts varied
clinical trial • Simple
• Can result in extreme ICERS
Two-way Decision model/ • Arbitrary amounts varied
clinical trial • Simple but can become complicated with >2
inputs
Confidence or Aggregate • Based on statistical theory
credible patient-level • Can result in extreme ICERs
Intervals/ dataa • Can lead to different conclusions
bootstrap CIs • In some cases may not be estimable
CEAC Any • Simple to interpret
• Provides an intuitive approach to estimating the
probability of cost-effectiveness
• A bit more complicated with >2 treatments
• Unclear how secondary endpoints information
is incorporated
Scatter plot/CE Any • Shows uncertainty around the individual ICERs
plane • Can see visually how many ICERs might lie in
different quadrants
CE Frontier Any • More appropriate when comparing >2
treatments
a For bootstrapped CI we start from mean cumulative (total) cost and mean QALY from the
observed trial.
Reporting and Interpreting Results of Cost-Effectiveness Analyses 307

to be true. However, unless a prior statistical cost-effectiveness hypothesis is


postulated, the Monte-Carlo simulation (or bootstrap) will provide a similar
interpretation of the results in the context of a probability.
The interpretation of the confidence interval from a Bayesian analysis is
called a credible interval. The credible interval expresses uncertainty about
the (posterior) point estimate of the ICER and is interpreted as offering a
probability of where the parameter of interest lies: “there is a 95% prob-
ability that the credible (confidence) interval will contain the true ICER.”
Compare this with the more wordy frequentist 95% confidence interval “if
you repeated this analysis 1,000 times, then 950 of the confidence intervals
would contain the true ICER.”
Table 9.15 provides a summary of some key issues in uncertainty analysis.

9.6.1 Limitations of the ICER and Using the INMB


There are several limitations of the ICER. One of the major problems with
the ICER, given that it is a ratio, is that when the denominator tends to zero,
the ICER becomes infinitely large. Defining a confidence interval in that case
becomes problematic. Moreover, for negative values of either the numerator
or denominator, the interpretations become trickier. The net monetary ben-
efit (NMB) has been developed as an alternative measure that does not imply
a ratio. It is defined as:

Net monetary benefit ( NMB ) = E * λ − C (9.2)

Its inverse is the net health benefit

(NHB) = − C / λ + E (9.3)
The incremental Net Monetary Benefit (INMB) =NMBT - NMBC = λ * ∆ E − ∆ C
where E = effectiveness (LYG or QALY),
λ = threshold for WTP, and C= total cost of treatment;
∆E and ∆C are the difference in mean effects and difference in mean costs
respectively.

As the NMB and NHB are monotonic linear functions it is statistically


simpler to calculate confidence intervals compared to an ICER. It has also
some nice mathematical properties as highlighted by Messori & Trippoli,
(2017). In contrast to the ICER, the NMB can be calculated for a single
treatment in the absence of any comparison (absolute NMB). Another fea-
ture of NMB is that the incremental NMB (INMB) for the comparison of A
versus B can be estimated as the absolute NMB calculated for treatment A
minus the absolute NMB calculated for treatment B (Messori & Trippoli,
2017, 2018).
308 Economic Evaluation of Cancer Drugs

FIGURE 9.10
Using the NMB.

Source: Maeso et al., 2011.

Graphically, the absolute NMB is shown using straight lines, where a


higher cost decreases the NMB and therefore shifts the entire line down, and
increasing effectiveness increases the slope of the line up from the intercept.
Any treatment that has an NMB above 0 is then by definition a candidate for
funding. Figure 9.10 shows a comparison of the absolute NMB of three treat-
ments with the CCA+CVP+EDM combination superior (more cost-effective)
to the two other modalities (the outer most lines represent 95% CI).
An additional feature of the NMB approach is that it is more amenable
to regression analysis with individual patient data, because it can be disag-
gregated to individual NMBs due to the linearity of the NMB for evaluating
the influence of patient, center, or treatment characteristics, which is not the
case for the ICER . For regressing the ICER, a bivariate regression (such as
seemingly unrelated regression) for deriving ICERs could be used (Willan et
al., 2004) as could equivalent Bayesian approaches that take into account the
correlation between costs and effects (Hernandez, 2009).

9.7 Presenting and Interpreting Results from


Value of Information Analyses
In this and previous chapters, we saw how cost-effectiveness models were
used to estimate the INMB and the ICER. In addition, we discussed how the
Reporting and Interpreting Results of Cost-Effectiveness Analyses 309

model inputs could be subject to uncertainty analysis either using one, two-
way sensitivity analysis and PSA. We also showed how the CEAC was gener-
ated from PSA. Value of information (VOI) can be considered as an extension
of PSA. However, whereas the CEAC yield estimates of the probability of
cost-effectiveness of the new treatment, VOI allows us to quantify the conse-
quences of selecting the wrong treatment and to estimate the expected gain
(reduction) in uncertainty from some (further) data collection. In lay terms,
does the value of additional information outweigh its cost?
Recall the expected net benefit (ENB) is: effects*λ – costs, where λ is the CE
threshold and effects are measures of effectiveness (e.g. QALYs). At the time
of a cost-effectiveness analysis and during a review of the evidence for the
cost-effectiveness of a new treatment by a reimbursement agency, any deci-
sion to reimburse the treatment(s) is based on the highest expected benefit
(expected net benefit). There is always a risk that the decision-maker may
choose the wrong treatment to reimburse based on current evidence. This
is because it is impossible to have complete and perfect information on all
aspects (Fenwick et al., 2008; Claxton, 2008).
Since uncertainty almost always exists, the chance of a wrong decision is
always possible. For example, an economic evaluation might be performed
that results in the new treatment yielding an incremental net benefit (INB) > 0.
However, at the time of marketing authorization, the pharmaceutical company
might be requested to carry out a post-marketing commitment study to evalu-
ate the longer-term effects (e.g. side effects) of the new treatment. It might tran-
spire from the post-marketing study that the adverse event rates were slightly
higher for the experimental/new group or that survival in the real world was
not as long as in the clinical trial (as can sometimes be the case). Consequently,
when the incremental cost-effectiveness ratio and the INMB were recalculated
from the post-marketing study data, they were not as large as initially esti-
mated from the clinical trial, leading to a lower INMB, which would have led
to the rejection of the product had that been known from the start.
A decision at the time of the first analysis (before the post-marketing study)
might be considered too risky if, for example, the available evidence proves
equivocal. In that case, more research to decrease the uncertainty around the
expected result (the incremental net benefit, which is the payoff to be maxi-
mized) has two consequences:

(i) generating new research costs, and


(ii) delaying the availability of the new treatments for patients while
additional research is conducted.

If the new treatment was initially accepted and proved to be more costly
or less effective (i.e. lower survival or QALY) the opportunity cost would
include the extra cost of treatment over the patient population or the differ-
ence in survival over a certain time horizon. The magnitude of the oppor-
tunity loss of the initial decision due to the uncertainty about its payoff (the
expected incremental net (monetary) benefit or EINB) can be quantified and
310 Economic Evaluation of Cancer Drugs

is known as the expected value of perfect information (EVPI). Basically, this


represents the probability of ‘being wrong’ multiplied by the average cost of
being wrong (the opportunity loss).
More formally, the EVPI is the amount the EINB changes (higher or lower)
by eliminating the uncertainty (simultaneously) of all parameters/variables
(e.g. costs, effects, utilities, adverse event rates, etc.) used in a decision to
choose a particular treatment for reimbursement. In some cases, additional
information (through more research) on some specific parameters (e.g. effi-
cacy endpoints, like survival or HRQoL) might be sought. Specific param-
eters might be those whose uncertainty is largest (because there was very
little data available before the trial started) and the value (in terms of changes
to the EINB) of reducing this will be estimated. When only one parameter
is assessed this is called the expected value of partially perfect information
(EVPPI) or expected value of perfect parameter estimation. When the per
patient EVPI is multiplied by the expected future patient population over a
number of years it is called the ‘population EVPI.’ As such it represents the
maximum potential value of further research. If this is close to zero then this
means that it is unlikely that the value brought by additional research (to
reduce uncertainty) will exceed the cost of that research. Hence, a first step
in analyzing the decision is to calculate its population EVPI.
However, in order to have ‘perfect information’ (i.e. eliminate all sources of
uncertainty) we would need a very large, in fact infinite, sample size. Hence,
‘perfect information’ is an ideal (in as much as having an unlimited sample
size is in a clinical trial) to answer a specific question. Therefore, in practice,
we try to quantify the uncertainty of decisions (of choosing treatments) by
research based on some finite sample size. As such we are interested in the
expected value of sample information (EVSI).
The EVSI compares the INB based on trial results given a current sample
size (imperfect information based on current data) with that obtained if more
data were collected (approaching perfect information – since the greater the
sample size, the closer to perfect information one gets).
We may calculate the EVSI for a range of sample sizes for a given research
design and compare it to the cost of conducting that research, including the
opportunity cost for the patients that will receive the ‘inferior’ treatment
during that study (typically lower survival or QALYs). This is called the
expected net benefit of sampling (ENBS). As for the EVPI it can be calculated
on a per patient or population level, for all sources of uncertainty or for only
some parameters (partial ENBS).
Hence the EVSI is considered to be another approach to estimating the
optimal sample size for a future study (Figure 9.11) [22].
The question therefore becomes, for example: “How much value (based on
the INB) is there in having more data (with possibly a larger sample size than
the one we currently have) so that the current decision does (or does not) not
change?” We now show an example of how the EVSI can be calculated and
used in the context of sample size calculation. The EVSI is considered to be
Reporting and Interpreting Results of Cost-Effectiveness Analyses 311

FIGURE 9.11
Sample sizes for varying values of EVSI.

Source: Maeso et al., 2011.

a fully Bayesian approach, but we have removed technical details to ensure


focus is on understanding the concept.

Example 9.10: Simple Description of EVSI


We adapt an example from Ades et al. (2004) following their algorithms
to compute the EVSI.
A clinical trial has been performed (n = 600; 300 per treatment group).
The experimental treatment group (treatment A) was shown to be more
cost-effective than the standard treatment (treatment B) with the current
expected net monetary benefit (ENMB) of £30,000 for treatment A and
£20,000 for treatment B (Table 9.16).
However, it may be the case that a reimbursement agency requests
more data in order to better estimate the uncertainty of long-term toxic-
ity data (which can have a significant cost burden). Although the initial
decision looks favorable for treatment A (i.e. a higher expected NMB),
the payer review group (who are involved in reimbursement) has sug-
gested the sponsor continue to investigate whether AE rates (perhaps
using a two arm observational study) might change the current decision
(that A is cost-effective compared to B). One question is how much more
information should we collect (i.e. what sample size should be used) so
that a (currently favorable) decision remains favorable in the future. At
what point should we be concerned that the payer will switch a decision
(from favorable to unfavorable, or vice versa).
As a starting point, let us assume the payer requests additional data
because they believe, from clinical trial data, the longer-term toxicity of
chemotherapy is too uncertain. We decide to proceed with a sample size
of n = 200 (100 per group), with the primary objective of toxicity, since
efficacy has been already demonstrated and there are no major concerns
around the uncertainty of OS or PFS.
312 Economic Evaluation of Cancer Drugs

TABLE 9.16
Model Inputs for EVSI Computations
Treatment PAE Cost of treatment (£) QALY ENMBa (£)
A 0.25 150,000 6 30,000
B 0.10 100,000 4.3 29,000
a Note: ENMB = –Cost + λ*Effect: ENMB-A = –£150,000 + 6*£30,000; ENMB-B = –£100,000 +
4.3*£30,000 (assuming a CE threshold, λ of £30,000). PAE: probability of adverse event

TABLE 9.17
Example Realization of Value of Sample Information After One Simulation of PAAE
and PBAE, the Adverse Event Rates for Treatments A and B, Respectively
Posterior
Current decision decision VSI
Simulation
Number PAAE PBAE ENBA ENBB [Max] ENBA ENBB [max]
Prior (trial) 25% 10% 33,000 30,000 33,000 – 37,000
1 28%, 15% 33,000 30,000 33,000 31,000 37,000 – 4,000+
2 … … … …

10,000
Mean (EVSI) 6,120

The parameter of particular interest was the (long-term) probability of


an adverse event (PAE) , estimated as 25% for treatment A and 10% for B
in the initial RCT (which had a sample size of n = 600 patients). The ques-
tion is whether collecting additional data impacts the ENMB. Currently,
the decision is in favor of treatment A (ENMB-A > ENMB-B = £30,000 –
£29,000 = £1,000) as shown in Table 9.17. A step-by-step approach to a VOI
is relegated to the technical appendix (A9.3). We interpret the results.
After the technical process of simulating the possibilities of future
long-term toxicity response rates, we compute the maximum of the
ENMB between each treatment. For example, if, after the first simula-
tion, we might believe the AE rate is going to be 28% and 15% (higher
than that in the trial) for treatments A and B respectively, the posterior
mean ENMB for treatment A is now £37,000 and for treatment B is now
£31,000 (before these were £33,000 and £30,000 for treatments A and B
respectively).
We retain the maximum of these values (i.e. £37,000) – sometimes the
ENMB for B may be higher and sometimes (occasionally) lower because
we are randomly sampling the AE rate so the costs will vary. This maxi-
mum is then compared (subtracted) with the ENMB under the current
decision (i.e. £33,000) as shown in Table 9.18. Table 9.18 shows how the
data structure might look like after one simulation.
Note: VSI is the value of sample information. The EVSI is the expected
value, which is calculated from the average of all the 10,000 VSI values.
The VSI is calculated as 33,000 – 37,000 = –4,000.
Reporting and Interpreting Results of Cost-Effectiveness Analyses 313

TABLE 9.18
EVSI and Chance that the ENMB Under the Plan of
Additional Data is > ENB Under the Current
Decision
Pr[ENBn=ni]B >
Sample Size (ni ) EVSI (£) Pr[ENBcurrent]A
50 1,300 0.15
100 2,400 0.22
200 5,280 0.27
400 6,090 0.29
600 6,120 0.33
1,000 6,180 0.38
2,000 6,185 0.39
5,000 6,190 0.40
10,000 6,192 0.40

Based on Table 9.18, after one simulation, a decision-maker (payer)


may switch from the initial decision that treatment A was cost-effec-
tive (£33,000 vs. £30,000) to it now being not cost-effective (£31,000 vs.
£37,000): Treatment B (old, or standard of care) has a greater (posterior)
ENMB than treatment A. The process continues for each cycle of the
simulation so that we generate a posterior distribution of differences.
For all the 10,000 simulated differences between current ENMB and
posterior ENMB, we can compute the mean of all the differences (Table
9.18) to generate the EVSI, which is £6,120. This was evaluated for a sam-
ple size of n = 150 per group. This can be repeated for other sample sizes
(Table 9.18).
In other words, if we continued to collect further data (e.g. about 300
patients) on long-term toxicity, the ENMB for treatment A will exceed
that of treatment B, on average, about 70% of the time (Table 9.19). There
may be some chance that treatment B will be less cost-effective than A
(Table 9.19) about 27% to 29% of the time. That is, the decision-maker
might switch the decision with about a 30% (27% exactly) chance if more
data were collected on longer-term toxicity. Hence, the payer may request
additional data (on toxicity) to be more certain the ENMB for treatment
A is higher than treatment B. Conversely, there is about a 70% chance
the payer has made the correct decision and the chance of switching is
low. What the threshold chance of switching should be to request further
data is unclear and may vary from payer to payer. If collecting further
data does not have much chance of changing the decision, then there is
less justification for a payer to delay a decision for reimbursement.
The chance of switching a decision appears to plateau after 1,000
patients (Table 9.18). Hence, any further data collected from either clini-
cal trials or observational studies are likely to result in ‘erroneous’ deci-
sions not greater than 40% of the time, if we had much more information
(e.g. data from 10,000 patients). That is, with perfect information of
long-term toxicity, a decision-maker could switch their decision with a
314 Economic Evaluation of Cancer Drugs

TABLE 9.19
Example of a Bootstrap Simulation Showing
Probability of Cost-Effectiveness >80%
Simulated CER (Cost/
Sample QALY) Below £30,000?
1 19,445 Yes
2 34,215 No
3 33,111 No
4 28,666 Yes
5 19,114 Yes
6 22,875 Yes
7 24,934 Yes
8 27,143 Yes
9 26,165 Yes
10 28,888 Yes

probability of 40%. To reduce this uncertainty, the payer might be justi-


fied in requesting more data on toxicity (or even ask the manufacturer to
change the price so that value is demonstrated!).
A separate question is whether a probability of 30% (27%) is an accept-
able risk level for the decision-maker and the patient. In comparison
with a Type I error of 5% for marketing approval, this value might be
high. One (perhaps ethical) issue is whether patients should continue to
be treated with the new treatment even if there is a risk that the payer
might be wrong 30% of the time. The risk is not that the new treatment
is not efficacious, but rather that it may not be cost-effective. This risk to
the public (declaring that the new treatment is efficacious when in real-
ity it is not) set at 5% (5% Type I error) is unlikely to be given the same
weight as a wrong decision on economic grounds. Whether this is right
or wrong is a matter for debate.

Example 9.11: Value of Information from Bevacizumab Trial


We now present an example of a value of information analysis reported
for bevacizumab used for the treatment of platinum-resistant, recurrent
ovarian cancer in a Canadian population (Ball et al., 2018).

BACKGROUND
A cost-utility analysis was undertaken over a 7-year time horizon using
a three-state partitioned survival model to compare the effectiveness
of bevacizumab plus chemotherapy (BEV) compared to chemotherapy
alone. The original trial results were published from the AURELIA
Phase III RCT (n = 361). which had a primary endpoint of PFS. The pub-
lished KM curves were taken and using special software, the survival
rates for each time point were digitally determined. These were used to
determine the transition probabilities for a three-state partitioned sur-
vival cost-effectiveness model.
Reporting and Interpreting Results of Cost-Effectiveness Analyses 315

The survival times were assumed to follow a log normal distribution


(see Chapter 7) as these fitted the Kaplan-Meier curve estimates best
using statistical tests (e.g. Akaike’s information criterion) and visual
inspection. By 7 years, the expected survival rates are around 0 (the
median OS was 13.3 and 16.6 months for chemotherapy and bevaci-
zumab, respectively), which provides some justification for the proposed
model.

RESULTS
The results reported the incremental cost per QALY (ICER) as Can
$213,424 (£123,064; Can $1/£1 = 0.58) based on a mean incremental QALY
of 0.1129. A threshold cost/QALY in oncology reported by the authors
was considered to be Can $100,000 (£58,000); and to ensure this threshold
could be reached, the authors advised the price (of bevacizumab) should
be reduced by 39%.
A value of information analysis was undertaken. The object of this
VOI was not, as in the example above, to compute a probability of switch-
ing, but rather, to estimate the value of future research when the WTP
(cost-effectiveness) threshold was fixed at Can $100,000. The description
of the VOI described by the authors is given below:

(i) First, the mean net benefit was calculated over all of the Monte-
Carlo simulations in the probabilistic sensitivity analysis.
(ii) Second, the net benefit for each individual Monte-Carlo simu-
lation in each of the treatment arms was calculated, and the
maximum net benefit across the treatment arms for each simu-
lation was identified.
(iii) Third, the mean of these maximized net benefit values was
then taken.
(iv) Finally, the difference between the mean of the maximized net
benefits and the maximum of the mean values was calculated.
(v) The EVPI was estimated to explore the value of conducting
future research given a willingness-to-pay ceiling ratio of Can
$100,000 (£58,000) per QALY gained and was calculated by mul-
tiplying per-patient EVPI by the effective population.
(vi) The estimated 2015 incidence of ovarian cancer multiplied
by the proportion of patients with recurrent ovarian cancer
yielded an effective per annum population of 2,380.
(vii) In addition, expected value of perfect partial information
(EVPPI) analyses were conducted to identify specific param-
eters for which additional data collection may be worthwhile.
(viii) The EVPI was estimated to be Can $804,818 (£466,794) at a will-
ingness-to-pay threshold of Can $100,000 per QALY gained.
Hence, further research may be worth conducting up to a maxi-
mum expected cost of Can $804,818 (£466,794).
(ix) Results of the EVPPI analyses showed EVPPI to be the highest
for the OS parameter values, suggesting that additional data
collection could be worthwhile for collecting further informa-
tion on OS – for example on longer-term survival.
316 Economic Evaluation of Cancer Drugs

9.8 Challenges of VOI Analysis in Healthcare Decisions


In practice, few VOI analyses have been conducted up to now in health policy
decisions for cancer treatment. One reason is that VOI analysis requires sophis-
ticated simulation methods and presents computational challenges, especially
for EVPPI and EVSI (long computation times because of multiple random sam-
pling loops). There is also a lack of automatized dedicated software to make it
easier to perform for nonspecialists. It also requires some assumptions about
the distribution of the INB (normal or non-normal) and its variance. Most
published VOI studies are based on a simulation model (such as the Markov
model) rather than on individual patient data (from single or pooled trials), as
it is felt that models are able to incorporate more evidence than a single trial.
However, with limited evidence when new treatments are assessed (typi-
cally one or two pivotal Phase III trials or a single Phase II trial) VOI analysis
can be performed on individual patient data as well. Previous results from
Phase II trials or observational studies could then be incorporated in the
choice of priors in a fully Bayesian analysis of the pivotal trial(s). In addition,
from a policy perspective the decision to adopt (and pay for) the new tech-
nology and the cost of conducting additional research are often not borne by
the same economic actor (typically a healthcare ministry or agency on the
one hand, with a remit for adoption or reimbursement, and a private drug
or medical device company, or another official agency, on the other hand).
Furthermore, once an adoption/reimbursement decision has been made,
reversing it proves very difficult in practice.
As an example, one might cite the Belgian law of 2010 that introduced the
possibility of temporary reimbursement for mostly orphan and innovative
drugs (so-called Class 1 drugs) and drugs with a high social or therapeutic
purpose. Only Class 1 drugs need to complete a cost-effectiveness analysis.
The reimbursement price is then, in principle, granted for a limited period of
up to three years (renewable twice) during which additional (generally real-
life observational) data are to be collected by (and at the cost) of the applicant.
Since the year 2010, about 169 contracts have been initiated of which about
60% are active (some firms may be dropped during the procedure because of
a lack of agreement between the parties, or for some other reason). Most con-
tracts (called ‘article 81 agreement ’) find their origin in a difference between
the company’s asking price for reimbursement and the price proposed by the
Drug Reimbursement Commission; or because of drug budget impact conse-
quences (Gerkens, 2017). Not all conventions include an additional research
requirement. Firms are generally also reluctant to start a new RCT if they
have already performed one, or several, Phase III trials.
Furthermore, it appears that few of these conventions led to much new data
collection in practice, although no formal systematic assessment has been
published to our knowledge (the details of the conventions are confidential by
law in Belgium). In cases where further data collection is requested, VOI and
Reporting and Interpreting Results of Cost-Effectiveness Analyses 317

especially (partial) EVSI approaches could be helpful tools. A potential appli-


cation of VOI could be for an interim analysis decision where the magnitude
of the observed effect size is uncertain. VOI using the expected net benefit
could be used to evaluate whether collecting additional data is likely to reach
a different value proposition based on alternative possibilities of the effect
size (in short, is continuing the trial based on the observed effect size likely to
lead to a different future cost-effectiveness decision compared to the present).

9.9 Summary
In this chapter we discussed how to present and interpret incremental cost-
effectiveness ratios and gave an example of calculating incremental QALYs from
a Kaplan-Meier survival curve using the trapezoid rule. We derived QALYs
with extrapolated survival data and discussed the relationship between indi-
vidual costs and QALYs using lung cancer data. Various univariate and mul-
tivariate sensitivity analyses were presented in the form of cost-effectiveness
acceptability curves (CEACs). We also introduced Bayesian sensitivity analysis
techniques and discussed the limitations of ICERs. Finally we introduced the
value of information (VOI) concept and its calculation from clinical trial data,
giving a detailed example from a bevacizumab trial. We finally discussed, in
brief, the challenges of VOI analysis in practice for healthcare decisions.

9.10 Exercises for Chapter 9


1. How would you decide whether a new treatment was cost-effective
in the following situations (assume two treatments being compared
against each other)?
a. The primary endpoint was very positive (i.e. a good outcome for
the new treatment) and all secondary outcomes were also better
for the new treatment.
b. The primary endpoint was very positive (i.e. a good outcome)
and all secondary outcomes were worse for the new treatment.
c. The primary endpoint was negative and all secondary outcomes
were worse for the new treatment.
d. The primary endpoint was no different between treatments but
all secondary outcomes were superior for the new treatment.
e. Is there a limitation in the way current economic evaluation is
performed, based on your answers to the above?
318 Economic Evaluation of Cancer Drugs

2. What is value of information? When should it be used? Could value


of information be used in an interim analysis and, if so, under what
circumstances?
3. What is a CEAC and what does it show? What should be an appro-
priate threshold for a chance of cost-effectiveness in your opinion,
and why? justify your answer.
4. What does a cost-effectiveness plane show?

Technical Appendix for Chapter 9


A9.1 Simulation
The process of simulating multivariate correlated data from any distribution
requires the use of Fleishman coefficients for pairwise variables to gener-
ate the intermediate correlations. For example, with two inputs, total costs,
and pre-progression utility, these can be relabeled as X1 and X2 respectively.
Hence:

X1 = a1 + b1Z1 + c1Z12 + d1Z13 (9.1)

X 2 = a2 + b2Z2 + c2Z22 + d2Z32 (9.2)

The values of ai, bi, ci, and di for (i = 1, 2) are estimated through the power
transformation and may be found in Fleishman tables using higher-order
moments. Once the values of ai, bi, ci, and di are estimated, the intermediate
correlation between X1 and X2 is determined by using the equation from Vale
and Maurelli (1983):

R x1x 2 = ρ ( b1 b2 + 3 b1d 2 + 3d1 b2 + 9d1d 2 ) + ρ2 ( 2c1c 2 ) + ρ3 (6d1d 2 )

This is then repeated for each of the pairwise variables to form an ‘interme-
diate correlation matrix.’ SAS code based on Fan et al., (2002) can be used
to simulate multivariate correlated data where each variable is from any
distribution:
The SAS code uses PROC IML for a Newton-Raphson iterative procedure
to find the values of a, b, c and d in (9.1) and (9.2). Using the data from Lee et al.
(2012), with a sample size of 670, a data set of n = 670 patients was simulated
with corresponding costs and effects. This was then repeated 10,000 times
(10,000 data sets of size n = 670 with n = 350 on erlotinib and n = 320 on pla-
cebo). For each data set the ICER was computed.
Reporting and Interpreting Results of Cost-Effectiveness Analyses 319

A9.2 Bayesian PSA
PSA under a Bayesian context is best understood starting with the bivari-
ate general linear model (GLM). We start with each patient having two
responses, one for costs, ci and one for effects, ei, contained in the matrix
Y. In addition, there are two treatment groups (although it can be extended
to more than two groups). This is the matrix X, where the first column is
the intercept and the second column an indicator variable for the treatment
group. We also have the parameter matrix β and the error vector ε.
Hence: Yij = µ + τ i + ε ij is a standard form of a GLM, where the subscript i
= 1, 2 indicates the treatment group and j is the number of observations for
each patient in the trial.
 µc 
Furthermore, µ is a vector   of the mean costs and effects and τ is a
 µe 
 τc 
vector of treatment effects for each of costs and effects:   ; Ɛij is a matrix of
 τe 
 ε11 ε 21 
residual errors:     where the Ɛij ~ MVN (0, β).
 
 ε1n ε 2 n 
 c11 e21 
If Y is     , a matrix (of n × 2) of responses for costs and effects,
 
 c1n e2 n 
then the model (in a frequentist framework) can be written as below:

Y= X β+ ε
Costs Effects Intercept group Parameters Error
2,000 1.2 11 µ1 µ2 Ɛ11 Ɛ21
3,000 1.6 11 τ11 τ12 Ɛ12 Ɛ22
8,000 0.8 11 Ɛ13 Ɛ23
etc. etc. .. ..
.. ..
.0 ..
10 Ɛ1n Ɛ2n

 µc 
The parameters to be estimated are µ =   and the variance – covariance
 µe 
 µc 
matrix ∑. In a Bayesian context, we assume prior distributions for µ =   ,
which we can call  µe 
 µ 0c  æ s c2 rs cs e ö
µ 0 =   and also a prior distribution for ∑,t med å 0 = çç ÷
 µ0e  è rs cs e s e2 ÷ø
320 Economic Evaluation of Cancer Drugs

TABLE A1
Model Inputs for EVSI Computations
Treatment PAE Cost of Treatment (£) QALY ENBa (£)
A 0.25 150,000 6 30,000
B 0.10 100,000 4.3 29,000
a ENB = –Cost + λ*Effect: ENBA = –£150,000 + 6*£30,000; ENBB = –£100,000 + 4.3*£30,000 (assum-
ing a CE threshold, λ of £30,000).

The objective is to combine the prior information with the likelihood func-
tions to determine an (updated) estimate of the mean costs and effects. These
are called posterior means. Using the posterior means, the (posterior) ICER
is then derived. Simulations from the posterior distributions (i.e. posterior
mean costs and effects) are carried out (similar to the frequentist method)
resulting in 10,000 (for example) posterior mean costs and effects. With this
data, the CEAC can be generated as before.
The software in which Bayesian modeling is often carried out in is called
WINBUGS, however the PROC MCMC and a PROC GENMOD in SAS are
also available for Bayesian analysis.

A9.3 Value of Information
Step 1: First calculate the ENB for each treatment group (Table A1).
Step 2: Determine which parameters will be subject to uncertainty and
determine the distribution to be used. The main reason for a future obser-
vational study is to get a better estimate of the values of the parameter PAE,
the probability of an adverse event (since only one RCT has ever been con-
ducted in this indication). One concern was that data from which the current
adverse event rate probability is uncertain and a further observational study
with a sample size of n = 300 (150 per group) may help to reduce the uncer-
tainty for the purposes of decision making. Therefore, PAE will be simulated
10,000 times from a beta binomial (BB) distribution. The BB distribution has
parameters (α, β), so we need to choose a set of parameters whose mean will
result in about 0.25 for treatment A and 0.10 for B, using the fact that:

(( ) )
α = µ *  µ * (1 − µ ) / σ 2 − 1 (9.3)
 

(( ) )
β = (1 − µ ) *  µ * (1 − µ ) / σ 2 − 1  (9.4)
 
In this example, an estimate of α = 3 and β = 9 for treatment A (this combina-
tion of α and β yields an estimate with mean 0.25, using mean = α/α+β for
treatment A, from (9.3)). Simulation from a BB is carried out for each treat-
ment group separately. Data from a BB can be simulated in SAS using the fact
that the ratio of two gamma distributed variables is a beta:
Reporting and Interpreting Results of Cost-Effectiveness Analyses 321

Z1 = rangam (seed,alpha ) ;
Z2 = rangam (seed,beta ) ;

X = Z1 / ( Z1 + Z 2) ; * *gamma (alpha, beta )

Step 3: Once 10,000 values of PAE for each of the treatments have been gener-
ated, for each simulated PAE, we then generate data from: binomial (n = 150,
PAE), each simulated data set being of sample size n = 150. The likelihood was
therefore generated from a binomial (n = 150, P*AE).
Step 4: The next step is to compute the posterior mean adverse event rate.
The prior BB combined with a binomial likelihood yields a posterior mean
of P*AE of the form:

a * /a * + b *

Example A9.3.1: Beta-Binomial Prior and Binomial Likelihood


Before Any Data is Observed
Before observing any data, θ (i.e. PAE for treatment A) was assumed to be a
Beta (2, 2). Since the mean of a Beta (a,b) = a/(a + b), this corresponds to a prior
mean of (2/(2 + 2) = 50%).
1
The form of a prior Beta (1, 3) is : θ (1 − θ)
Beta (1, 3 )
1
θ a −1 (1 − θ )
b −1
In general, a Beta (a,b) density is:
Beta ( a, b )

After Data Have Been Observed


Now assume that 10 further patients are observed for any adverse events.
These outcomes are assumed to be dichotomous (Yes/No type outcomes).
Assume that the 10 subjects have the following observed data:

X = (1, 0, 1, 0, 0, 0, 0, 1, 0, 0 )

where xi is an indicator variable for 1 if an AE is observed and 0 if not.


For the above realization of AEs, the PAE rate can now be updated by com-
puting the posterior mean. In order to do this we need the likelihood of the
10 outcomes.
10

L(θ; ×) = ∏θ
i =1
xi
(1 − θ)1− x i
= θ 3 (1 − θ )
7
322 Economic Evaluation of Cancer Drugs

We now combine the prior distribution with the likelihood to form the pos-
terior distribution of PAE:

P(θ; x) ∝ θ 3 (1 − θ ) θ (1 − θ ) = θ 4 (1 − θ )
7 8

1
qa-1 ( 1 - q ) (a posterior Beta (5, 9))
b -1
Compare this with:
Beta ( a , b )
Here a – 1 = 4 and b – 1 = 8, giving a = 5 and b = 9.

Prior Beta (2, 2) 2/(2 + 2) = 50%


Posterior Beta (5, 9) 5/(5 + 9) = 36%

The proportionality symbol (α) means that the full computation of the like-
lihood with the prior is determined through integrating out the prior × like-
lihood such that some terms cancel out.
From the computations (integrations), the above results in a posterior Beta
(5,9). The mean of a Beta (a,b) = a/(a+b). Therefore, the posterior (mean) PAE,
after having observed a further 10 outcomes is 5/(5+9) = 36%. The new (addi-
tional) data has revised the estimate of PAE from 50% to 36%.
Hence, we will generate 10,000 posterior mean values Pi*AE, where the sub-
script i is from 1 to 10,000 for each simulated value.
Step 5: For each simulated posterior mean PAE, compute the ENB for each
treatment group. That is, we would have 10,000 ENB values for each of treat-
ments A and B. Hence, by formulating a prior distribution based on the
original n = 600 from the RCT (which is a large amount of data to have an
informative prior), and further simulate n = 300 (n = 150 per treatment group
if a two group observational study), we now have 10,000 (updated) ENBs for
each of the treatments. We are now in a position for each simulated ENB to
compute the impact of the additional n = 300 patients through the EVSI.
10
Factors Predictive of HTA Success
and the Global Landscape

10.1 Introduction
In this chapter we will discuss reimbursement strategies and experiences
across several countries within Europe. In the UK, NICE appears to have one
of the most comprehensive and transparent approaches to evaluating cost-
effectiveness. We will then discuss briefly some approaches for HTA in other
countries, including the US. We consider the potential factors predictive of
successful HTAs using available data. Some practical issues in HTAs are also
discussed when designing cancer trials for cost-effectiveness.

10.2 Cancer Drugs Rejected by NICE


The National Institute for Health and Care Excellence (NICE) became a legal
entity in April 1999 with the aim to create consistent guidelines and to end
the rationing of treatment through ‘postcode lotteries’ (i.e. a random process
where some regions could get access to cancer treatments and others could
not, thereby increasing inequality) across the UK. NICE provides guidance
to the NHS in England on the clinical- and cost-effectiveness of selected new
and established technologies. The Institute undertakes appraisals of health
technologies at the request of the UK Department of Health. Guidance pro-
duced by the Institute on health technologies is also applied selectively in
Northern Ireland, Scotland, and Wales (Timmins, Rawlins & Appleby, 2016).
Initially, the technology appraisal was based on examining the values from
multiple health technologies (drugs, devices, or other health interventions
in the NHS, including advice) for any one condition. The single technology
appraisal (STA) process was introduced in early 2005 to assess a single drug
or treatment for a single indication. Both the multiple technology appraisal
(MTA) and the STA are processes designed to provide recommendations, in

323
324 Economic Evaluation of Cancer Drugs

69

36

23
17

2 3

Not Opmised Opmised Recommended Recommended Terminated-


Recommended (CDF) (CDF) Non Submission

FIGURE 10.1
Breakdown of HTA decisions from 150 cancer products (NICE HTA, April 2018). Note: CDF:
cancer drug fund; optimized: the reimbursement guideline only covers a subpopulation of
the licensed population; terminated non-submission: manufacturer decided not to submit the
evidence.

the form of NICE guidance, on the use of new and existing medicines, prod-
ucts, and treatments in the NHS. The STA process was introduced as a mech-
anism to provide a prompt appraisal of technologies for use within the NHS
in England and Wales so that national guidance for new products could be
provided as closely as possible to their launch (i.e. when they would be offi-
cially available for prescription or access by patients), and more quickly than
the existing MTA process.
According to the data available, as of October 2017, 150 cancer medicines
(technologies) were appraised by NICE following the STA process. Figure 10.1
provides a breakdown of outcomes for different decisions reached by NICE.
Of the 150 single technology assessment submissions, almost one-quarter
(36/150 or 24%) were not recommended for routine clinical use in England
and Wales; 23 appraisals were terminated due to non-submission of evidence
by the manufacturers. Reimbursement was granted for 91 out of 150 (61%) of
the technologies, either as a recommendation in line with marketing autho-
rization (n = 69) for routine use through the cancer drug fund (CDF), or opti-
mized recommendation, which cover only a subgroup.
Table 10.1 lists the drugs not recommended for reimbursement by NICE
since 2001, including both STA and MTA recommendations.

10.3 Summary of Criticisms of Economic Models of Cancer


The availability of appropriate and robust data to populate model parameters
requires efficacy and safety data regarding disease symptoms, disease pro-
gression, adverse events, death, HRQoL, health resource utilization, as well
TABLE 10.1
List of Drugs Not Recommended by NICE between 2001 and 2017
Appraisal
Number Year Process Drug Condition
TA025 2001 MTA Gemcitabine (second line) Pancreatic cancer
TA030 2001 MTA Docetaxel in combination with an anthracycline Advanced breast cancer
(first line)
TA033 2002 MTA Irinotecan in combination with 5-fluorouracil and Colorectal cancer (advanced)
folinic acid (5FU/FA) (first-line)
TA037 2002 MTA Rituximab (third and subsequent lines of Lymphoma (follicular non-Hodgkin's)
treatment)
TA054 2002 MTA Vinorelbine combination therapies Breast cancer
TA108 2006 STA Paclitaxel Breast cancer (early)
TA118 2007 MTA Bevacizumab in combination with 5-fluorouracil Colorectal cancer (metastatic)
plus folinic acid (with or without irinotecan)
TA118 2007 MTA Cetuximab in combination with irinotecan Colorectal cancer (metastatic)
TA119 2007 STA Fludarabine (monotherapy) Leukemia (lymphocytic)
TA124 2007 STA Pemetrexed (second line) Non-small-cell lung cancer
TA162 2008 STA Erlotinib (second-line treatment in patients for Non-small-cell lung cancer
whom docetaxel is unsuitable; or as a third-line
treatment after docetaxel therapy)
TA172 2009 STA Cetuximab Head and neck
Factors Predictive of HTA Success and the Global Landscape

TA178 2009 MTA Bevacizumab (first line) Advanced and/or metastatic renal cell carcinoma (RCC)
TA178 2009 MTA Sorafenib (first line) RCC
TA178 2009 MTA Temsirolimus (first line) RCC
TA178 2009 MTA Sunitinib (second line) RCC
TA178 2009 MTA Sorafenib (second line) RCC
(Continued)
325
TABLE 10.1 (CONTINUED)
326

List of Drugs Not Recommended by NICE between 2001 and 2017


Appraisal
Number Year Process Drug Condition
TA184 2009 MTA Intravenous topotecan Relapsed small-cell lung cancer
TA189 2010 STA Sorafenib (first line) Hepatocellular carcinoma
TA196 2010 STA Imatinib adjuvant Gastrointestinal stromal tumors (resectable)
TA202 2010 STA Ofatumumab CLL
TA209 2010 MTA Imatinib (dose escalation to 600 mg/day or Gastrointestinal stromal tumors
800 mg/day following disease progression on
400 mg/day imatinib)
TA212 2010 STA Bevacizumab in combination with oxaliplatin and Metastatic colorectal cancer
either fluorouracil plus folinic acid or
capecitabine
TA214 2011 STA Bevacizumab in combination with a taxane (first Metastatic breast cancer
line)
TA219 2011 STA Everolimus Advanced renal cell carcinoma
TA227 2011 STA Erlotinib (first line maintenance treatment) Advanced/ metastatic NSCLC
TA239 2011 STA Fulvestrant Locally advanced or metastatic breast cancer
TA241 2012 MTA Dasatinib Chronic myeloid leukemia
TA241 2012 MTA High dose imatinib Chronic myeloid leukemia
TA242 2012 MTA Cetuximab monotherapy or in combination with Metastatic colorectal cancer
chemotherapy
TA242 2012 MTA Bevacizumab + non-oxaliplatin chemotherapy Metastatic colorectal cancer
TA242 2012 MTA Panitumumab monotherapy Metastatic colorectal cancer
TA250 2012 STA Eribulin Advanced or metastatic breast cancer
(Continued)
Economic Evaluation of Cancer Drugs
TABLE 10.1 (CONTINUED)
List of Drugs Not Recommended by NICE between 2001 and 2017
Appraisal
Number Year Process Drug Condition
TA251 2012 MTA Dasatinib (first-line) Chronic phase Philadelphia-chromosome-positive
chronic myeloid leukemia
TA255 2012 STA Cabazitaxel in combination with prednisone or Metastatic prostate cancer
prednisolone
TA257 2012 MTA Trastuzumab in combination with an aromatase Metastatic breast cancer
inhibitor (first line)
TA263 2012 STA Bevacizumab in combination with capecitabine Metastatic breast cancer
(first line)
TA272 2013 STA Vinflunine Advanced or metastatic transitional cell carcinoma of the
urothelial tract
TA284 2013 STA Bevacizumab in combination with paclitaxel and Advanced ovarian cancer
carboplatin
TA285 2013 STA Bevacizumab in combination with gemcitabine Advanced ovarian cancer
and carboplatin
TA289 2013 STA Ruxolitinib Myelofibrosis
TA295 2013 STA Everolimus in combination with exemestane Advanced breast cancer
TA296 2013 STA Crizotinib Non-small-cell lung cancer (NSCLC)/anaplastic
lymphoma
Factors Predictive of HTA Success and the Global Landscape

TA299 2013 STA Bosutinib Chronic myeloid leukemia


TA307 2014 STA Aflibercept in combination with irinotecan and Metastatic colorectal cancer
fluorouracil-based therapy
TA309 2014 STA Pemetrexed NSCLC
TA338 2015 STA Pomalidomide in combination with Multiple myeloma
dexamethasone
(Continued)
327
TABLE 10.1 (CONTINUED)
328

List of Drugs Not Recommended by NICE between 2001 and 2017


Appraisal
Number Year Process Drug Condition
TA360 2015 STA Paclitaxel as albuminbound nanoparticles in Pancreatic cancer
combination with gemcitabine
TA371 2015 STA Trastuzumab emtansine Metastatic breast cancer
TA374 2015 MTA Gefitinib NSCLC
TA378 2016 STA Ramucirumab alone or with paclitaxel Gastric cancer or gastro-oesophageal
TA389 2016 MTA Gemcitabine in combination with carboplatin Recurrent ovarian cancer
TA389 2016 MTA Trabectedin in combination with PLDH Recurrent ovarian cancer
TA389 2016 MTA Topotecan Recurrent ovarian cancer
TA399 2016 STA Azacitidine Acute myeloid leukemia
TA403 2016 STA Ramucirumab in combination with docetaxel NSCLC
TA411 2016 STA Necitumumab, in combination with gemcitabine NSCLC
and cisplatin
TA414 2016 STA Cobimetinib in combination with vemurafenib Melanoma
TA425 2016 STA High-dose imatinib Chronic myeloid leukemia
TA440 2017 STA Pegylated liposomal irinotecan in combination Pancreatic cancer
with 5fluorouracil and leucovorin
STA: Single Technology Appraisal; MTA: Multiple Technology Appraisal.
Economic Evaluation of Cancer Drugs
Factors Predictive of HTA Success and the Global Landscape 329

as data regarding patient drug utilization behaviors (e.g. therapy discontinu-


ation or switching). These inputs are needed to generate cost-effectiveness
analyses. Not all of this data, as noted from previous chapters, are routinely
collected during clinical trials; data from other diverse sources are used to
complement or ‘fill in the gaps’ in order to build a robust health economic
model. More often than not, economic data for health economic models in
cancer come from alternative sources such as naturalistic, non-interventional
studies (including both prospective and retrospective observational studies).
For these reasons, the quality of the final health economic model may not
always meet the standards expected for a positive recommendation by reim-
bursement bodies.
Given that approximately 25% of oncology drugs were not recommended
by NICE for a variety of different cancer subtypes and indications, the com-
mon criticisms of health economic models that led to their rejection are worth-
while reviewing. In subsequent sections of this chapter, we summarize the
research carried out to identify potential factors predictive of successful HTA
submissions. We also present the major areas of criticism of health economic
models used for cancer drugs. The criticisms are categorized as follows:

(i) Criticism of clinical evidence. There is a debate as to whether this is


the remit of NICE given that the multiple licensing authorities have
already given an approval.
(ii) Criticism of utility data. The use of external data based on unreason-
able assumptions is often cited. see recent ISPOR publication on 29
March 2019 http://press.ispor.org/index.php/global-expert-panel-
publishes-new-recommendations-on-the-use-of-health-state-utili-
ties-in-cost-effectiveness-models/
(iii) Criticism of the health economic model and/or analysis. The type of
model, its assumptions, and failure to take into account fundamen-
tal aspects (e.g. treatment switching) are often cited.

Given the large number of HTAs not recommended for reimbursement, it is


difficult to detail every particular feature and the reasons for rejection. Table
10.2 shows the variety of criticisms from a number of NICE-appraised HTAs.
There were some common themes:

(a) Absence of EQ-5D utility, or inadequate reporting, or assumptions


around it.
(b) Modeling survival data was found problematic, and also based on
unjustifiable assumptions.
(c) Evidence synthesis often inadequate.
(d) Reliable or immature evidence from overall survival (often the pri-
mary endpoint).

(i) Summary of Criticisms of Clinical Evidence from NICE HTAs


330 Economic Evaluation of Cancer Drugs

TABLE 10.2
Summary of Criticisms Across Most HTAs
Example
Review
Feature Main Criticisms Cited
Clinical evidence
Population • Worse performance patients not presented adequately TA374
• Differences in baseline characteristics TA374
• Target population likely to be older than trial sample TA307
Intervention
Comparator • Nivolumab and criotinib were excluded (this is despite TA403
the fact that nivolumab was undergoing appraisal at
the same time)
Outcomes • Absence of benefit in OS TA374
• Mean estimate of OS unreliable due to small numbers TA304
• Lack of information on censoring. Suggestion that TA389
censoring may be informative and reduce validity of
Kaplan-Meier and other survival estimates
Trial design
Experimental Trial design good, but unlikely to reflect clinical practice TA374
design
Interim analyses No mature data during interim analyses TA 202
Health resource/ • Regimen used in trials unlikely to be used in practice TA374
costs • Determined from a retrospective study TA304
• Acquisition and administration costs of drug TA304
inappropriate TA304
• Median values of health resource use applied; whereas TA389
means used in cost-effectiveness
• Assumption that patients will receive a low number of
cycles (which reduces costs); assuming efficacy is not
impacted
HRoL (utility) • Unpublished or lack of peer review TA374
• Treatment-related AE utilities used not related to TA374
population TA374
• Estimated from a separate cross-section study TA304
• Utility value in progressive disease state too high TA304
• No adjustment of utility of patient aging over time TA389
• Insufficient review of utility data TA389
• Utility estimated from a separate trial TA403
• Validity of EQ-5D translation TA403
• Small differences in EQ-5D TA403
• Statistical comparisons of EQ-5D not provided TA403
• External EQ-5D data (mis-reported) TA403
• Contradictory assumptions on EQ-5D: on the one
hand, assuming a constant EQ-5D post-progression
while, on the other hand, using systematic reviews
showing utility declined during subsequent therapy
(Continued)
Factors Predictive of HTA Success and the Global Landscape 331

TABLE 10.2 (CONTINUED)


Summary of Criticisms Across Most HTAs
Example
Review
Feature Main Criticisms Cited
Economic model
Assumptions • Nonproportional hazards not accepted: survival curves TA304
considered to converge after 5 years. TA304
• Assumptions around model too simplistic TA389,
• Proportional hazards assumption violated TA403
• Similarity of treatment effects between studies in TA403
meta-analyses not justified TA403
• Unjustifiable assumption of PH: “no theoretical reason
to believe PH assumption holds for two treatments that
differ in their mode of action”1
Model structure • Partitioned survival model TA374,
• 3 state Markov model TA403,
TA304,
TA374
TA304
Confounding Post-progression therapy use/crossover bias TA389
Prediction of • Piecewise model preferred TA374
survival • Extrapolation is uncertain (small numbers): projected TA304
curve separation is unreliable TA304
• Truncation of projected OS advised
Estimate of Hazard ratios were adjusted when used in the economic TA389
treatment effects model, ERG used unadjusted
Subgroup analyses Lack of statistical significance TA307
New comparator Nivolumab not yet appraised, but expected as a TA403
comparator
Systematic review • Insufficient evidence or further evidence needed TA374
• No previous published cost-effectiveness analyses TA304
• Insufficient data for a pairwise meta-analysis TA389
• Network meta-analyses (NMA) performed, but limited TA389
data TA403
• Exclusion of some studies in the NMA
Analysis • Statistical heterogeneity across the different studies not TA403
adequately addressed TA403
• Sparse data resulting in lack of convergence for
statistical models
Note: 1 One normally tests for a rejection of PH (i.e. in the absence of data we assume PH). Here
the ERG assumes a starting position of non-PH unless evidence is bought forward con-
trary to this hypothesis. For example, for both cytotoxic (drugs that kill cancer cells)
and cytostatic drug s (drugs that arrest the growth of cancer cells), the relationship
between OS and tumor response might be poor; it does not follow that it is a necessary
condition that the survival rates of the two drugs when compared are proportional (i.e.
parallel survival curves).
332 Economic Evaluation of Cancer Drugs

The robustness of clinical evidence is frequently questioned by NICE


appraisal committees and ‘independent’ evidence review groups (ERG). As
discussed in detail in Chapter 2, the endpoints in oncology clinical trials
are often well defined and primarily related to survival. Most regulatory
authorities require evidence of clinical efficacy for new drug registration in
terms of benefits from OS, either directly or indirectly using surrogate end-
points such as PFS.
On many occasions, the evidence for improvement in OS and/or HRQoL
is limited at the time of initial drug registration. Only 57% (39/68) of cancer
drugs approved by the European Medicines Agency (EMA) between 2009
and 2013 showed evidence of improved survival or HRQoL at the time of
market entry (Davis, 2017). Showing benefits in OS, simply put, takes longer
and it is more expensive to follow up patients until death. Kim and Prasad
(2015, 2016) show something similar. Hence, it should come as no surprise
that the clinical benefit based on OS at the time of initial regulatory approval
is inadequate or uncertain at best. In some cases, it may even be difficult to
measure meaningful OS improvements, because patients are likely to expe-
rience crossover and receive multiple lines of treatment following disease
progression.
PFS is an often used surrogate endpoint accepted by several regulatory
authorities and also some reimbursement bodies. In Germany, the Institute
for Quality and Efficiency in Health Care (IQWiG) is likely to endorse the
combination of PFS with improved HRQoL (not necessarily the EQ-5D) as
an acceptable ‘benefit’ for their assessments. However, for countries where
cost-effectiveness assessments are carried out, such as England and Wales
(NICE), Scotland (SMC), and Australia (PBAC), both OS and HRQoL data
may be required. In the absence of OS and HRQoL data (e.g. using tumor
response), drug manufacturers are likely to be required to conduct economic
modeling in order to extrapolate available clinical data to show a meaningful
improvement in survival and HRQoL.
A lack of head-to-head clinical data comparing investigational treat-
ments with all possible comparators is one of the larger problems in devel-
oping economic models, as noted in Table 10.2. For example, most payers
and reimbursement bodies around the world require comparisons with
the most appropriate comparators for their health economic evaluations. In
some cases, for an emerging treatment, no published data are available, yet
the decision-making body will request it. This might seem like an unfair
request, however it will be important at some point to ensure optimal deci-
sions can be made on cost-effectiveness. Hence relegating the comparison to
a future date only delays the inevitable.
Local decision-makers are often interested to know what extent the trial
data are reflected in or applicable to the target population. Multinational tri-
als should try to account for some features of the target population. For some
rare tumors this might be difficult, because each country will only have a
few patients. Oncology clinical trials recruit fewer patients than some of the
Factors Predictive of HTA Success and the Global Landscape 333

other non-oncology indications such as respiratory or cardiovascular dis-


ease. According to one study, based on data from Clinicaltrials.gov, 98% of
oncology-related clinical trials registered from 2007 to 2010 have 1,000 par-
ticipants or fewer, and 75% have 100 or fewer participants (Miller et al., 2014).
Due to the global nature of these trials, there may only be a limited number
of patients enrolled from a particular country or region.
Some multinational trials may be nonrandomized, single arm, Phase I/II
studies, with no comparators. In some cases, RCTs may not include all the
potential treatment options that may be considered the standard of care in
a particular country. In fact, the NICE scoping document (NICE – Guide to
Technology Appraisal Process, January 2019), which is prepared in collabo-
ration with the manufacturer prior to the STA submission, often includes
more comparators than those included in the control arm of registration
clinical studies. In the absence of head-to-head trials versus all possible
comparators, published data is used for clinical and cost-effectiveness mod-
eling, which leaves the models open to criticisms and increased uncertainty
(see Table 10.2).

Example 10.1: Case Study – TA428 (NICE, TA428,


2017): Pembrolizumab for PD-L1-Positive Non-
Small-Cell Lung Cancer after Chemotherapy
BACKGROUND
Pembrolizumab received marketing authorization for treating locally
advanced or metastatic NSCLC in adults whose tumors express PD-L1
(defined as a tumor proportion score (TPS) ≥ 1%) and who have had at
least one chemotherapy regimen. Patients with the actionable tumor
mutations positive (EGFR positive or anaplastic lymphoma kinase (ALK)
positive tumor) should also have had an approved therapy for these
mutations (e.g. erlotinib, which is used for EGFR +ve patients) before
having pembrolizumab.

CLINICAL TRIAL EVIDENCE


The manufacturers submission was based on two trials:

(i) The KEYNOTE-010, and


(ii) KEYNOTE-001

The KEYNOTE-010 population consisted only of patients with PD-L1-


positive NSCLC. KEYNOTE-010 was powered to detect a difference
between pembrolizumab and docetaxel in the population with a TPS of
50% or more, and in the overall TPS 1% or more population, but not for
the TPS 1 to 49% population. The TPS is a biomarker to score patients at
different levels of expression of the program cell death ligand-1 (PD-L1)
expression on the cancer cells. The patients, whose tumors express high
levels of PD-L1 are scored high on the TPS, are more likely to respond to
pembrolizumab treatment.
334 Economic Evaluation of Cancer Drugs

The inclusion criteria in KEYNOTE-010 required patients to have an


Eastern Cooperative Oncology Group (ECOG) performance status of 0
or 1. The median overall survival was 10.5 months for pembrolizumab
compared with 8.6 months for docetaxel in the ITT population. This dif-
ference was statistically significant.
KEYNOTE-001 was a nonrandomized cohort study of pembrolizumab
that retrospectively identified PD-L1 status and used the docetaxel arm
of KEYNOTE-010 as a comparator.
The manufacturer’s base case ICER using a simple discount (confiden-
tial discount from the list price) was £48,667 per QALY. In the pivotal
trial, treatment continued until disease progression and, given the long-
term benefit of immunotherapy drugs such as pembrolizumab, some
patient continued to benefit from treatment for a longer period of time,
incurring costs to the NHS. However, NICE recognized that with no
treatment stopping rule (i.e. a hard stop once a patient received treat-
ment for a certain number of cycles) the ICER was likely to be higher.
Following further analysis to address uncertainties in the model, the
NICE appraisal committee concluded that based on the trial data, pem-
brolizumab had an important extension-to-life benefit for people with
locally advanced or metastatic NSCLC whose tumors express PD-L1
compared with docetaxel.
In order to meet the NICE cost-effectiveness threshold, the manu-
facturer proposed that the treatment should be stopped at two years
instead of the treatment-till-progression approach employed in the clini-
cal trials. During the appraisal committee meeting, NICE identified the
following areas of uncertainty:

• Optimal duration of treatment


• Magnitude of OS gain
• Long-term treatment effect

NICE concluded that it was aware of several ongoing clinical trials that
could reduce this uncertainty and, if pembrolizumab was recommended
for routine commissioning, relevant data would be collected by the sys-
temic anti-cancer therapy (SACT) data set (see Chapter 8 on RWD). NICE
concluded that uncertainty about the long-term treatment effect would
fall as more information (data) became available on the optimal duration
of treatment of PD-1 inhibitors in the next two years.

WHAT HAPPENED NEXT?


NICE will review the data available from ongoing clinical trials and the
data collected within the SACT data set in two years’ time to issue the
final guidance whether pemobrolizumab should be made available for
routine use outside CDF. In this example, too, reliance on data outside a
clinical trial (the SACT database) is of interest as noted in Chapter 8 on
real-world evidence.
Factors Predictive of HTA Success and the Global Landscape 335

10.4 Factors Predictive of Successful HTAs in Cancer


We noted earlier in Section 10.1, NICE issued not-recommended guidance
for 24% of STA cancer drugs. In contrast, during the same period, the over-
all not-recommended guidance for oncology and non-oncology technologies
was 18%. This gives one the impression that oncology drugs are more likely
to be issued with a negative reimbursement decision and there may be some
bias, systematic or otherwise, in how cancer drugs are viewed for reimburse-
ment in the UK.
Although a shadow price of a QALY is not described as a single threshold,
according to the most recent and definitive statement from NICE (2013), tech-
nologies costing less than £20,000 cost per QALY gained are usually consid-
ered cost-effective. Treatments for children, disadvantaged populations, and
severe diseases may be treated more favorably (NICE 2008, 2013; Rawlins et
al., 2010). A drug with orphan designation does not play any role in the NICE
cost-effectiveness assessment (Littlejohns and Rawlins, 2009). Although
NICE has become increasingly explicit and transparent about its decision-
making process, areas of considerable uncertainty remain. This issue is not
limited to NICE but also to some European countries:

there remains a lack of transparency around critical elements, such


as how multiple factors or criteria are weighed during committee
deliberations.
(Stafiniski et al. 2011)

For example, Bossers et al. (2015) , analyzed single technology assessments


conducted by NICE, Scottish Medicines Consortium (SMC), Zorginstituut
Nederland (ZIN), Haute Autorité de Santé (HAS), and Gemeinsamer
Bundesausschuss (G-BA) as well as the clinical, economic, and societal data
underlying these assessments. This analysis revealed that in most countries
clinical factors appeared to be predictive factors for successful reimburse-
ment, whereas in countries such as the UK, where cost-effectiveness analyses
are performed, economic factors appeared to be predictive. These findings
confirm the previous results of Devlin and Parkin (2004), who showed cost-
effectiveness was a key driver for NICE decisions – and not necessarily the
clinical rationale, although other factors such as uncertainty and burden of
disease were also significant. Table 10.3 presents the results of research in
this area.
Dakin et al. (2015), investigated the following factors: clinical and eco-
nomic evidence; characteristics of patients, disease, or treatment; and con-
textual factors potentially affecting decision-making. Their analysis showed
that the ICER alone correctly predicted 82% of all NICE decisions between
1999 and 2011. There was no evidence that the cost per QALY threshold
336 Economic Evaluation of Cancer Drugs

TABLE 10.3
Research Findings on Factors Predictive of Cost-Effectiveness
HTA Body Variables Reference
NICE Cost-effectiveness, clinical evidence, technology Dakin et al. (2006)
type, and patient group
NICE Cost-effectiveness, statistical superiority of primary Cerri et al. (2014)
endpoint, number of pharmaceuticals, and the
appraisal year
NICE ICER, uncertainty, availability of other therapies, Tappenden et al.
and severity of illness (2007)
AWMSG Cost-effectiveness Linley and Hughes
(2012)
SMC Cost-effectiveness Msheila et al. (2013)
APBAC Cost-effectiveness Harris et al. (2008)
Note: AWMSG: All Wales Medicines Strategy Group; SMC: Scottish Medicines Consortium;
APBAC: Australian Pharmaceutical Benefits Advisory Committee

has significantly changed over time. No other variables were statistically


predictive of a decision to reimburse. This essentially means that all focus
should be to reduce the ICER. This is an expected conclusion as it is logi-
cal that the lower (or higher) the ICER, the higher (or lower) the chance of a
reimbursement decision. Technologies costing £27,000, £40,000, and £52,000
per QALY were reported to have probabilities of rejection at around 25%,
50%, and 75% respectively (which is also logical, as the higher the ICER,
the greater the likelihood that it will be rejected). A more interesting ques-
tion however, is what the predictors of successful reimbursement for cancer
drugs are, based on a range of evidence. In particular, are there any features,
such as lower incremental costs, QALYs, or the size of the study, that are
predictive of this?
Table 10.4 shows data extracted from HTA reports showing the decision
(recommend/not recommend) in relation to:

(i) ICER
(ii) Incremental QALY
(iii) Incremental cost
(iv) Hazard ratio
(v) Performance status (ECOG)
(vi) Sample size

Other factors could also be included, such as mean survival difference, type
of model (partitioned survival or Markov model), HRQoL (condition-spe-
cific), year of appraisal, and tumor type. However, for simplicity we used the
six factors above. The submissions cover multiple tumor types and in some
TABLE 10.4
An Extract of UK HTA Decisions for Cancer Drugs
INC_ Sample
Drug Condition Categorization ICER INC_QALY COST_best Hros ECOG Size
Temozolomide Brain cancer Recommended 42920 0.2 3863 1.44 n/a n/a
(recurrent)
Bevacizumab (first Advanced and/or Not Recommended 171301 0.26 45435 1.27 0-1 649
line) metastatic renal
cell carcinoma
Temsirolimus (first Advanced and/or Not Recommended 81687 0.24 19276 1.28 n/a 626
line) metastatic renal
cell carcinoma
Sunitinib (second Advanced and/or Not Recommended 71462 0.44 31185 1.54 0-1 750
line) metastatic renal
cell carcinoma
Sorafenib (second Advanced and/or Not Recommended 102498 0.23 24001 1.38 0-2 903
line) metastatic renal
cell carcinoma
Sunitinib Unresectable and/ Recommended 32636 0.5 16337 1.14 0-4 361
or metastatic
Pemetrexed in Locally advanced Recommended 33065 0.041 1364 1.19 0-1 1725
combination with or metastatic
Factors Predictive of HTA Success and the Global Landscape

cisplatin (first line)


Sorafenib (first line) Advanced and Not Recommended 64754 0.36 23232 1.45 0-2 602
metastatic
hepatocellular
carcinoma
(Continued)
337
TABLE 10.4 (CONTINUED)
338

An Extract of UK HTA Decisions for Cancer Drugs


INC_ Sample
Drug Condition Categorization ICER INC_QALY COST_best Hros ECOG Size
Pemetrexed Non-small-cell Recommended 33732 0.271 9137 1.43 0-1 663
(maintenance lung cancer
treatment)
Gefitinib (first line) Locally advanced Recommended 20744 0.177 3666 1.09 0-2 1217
or metastatic
Gefitinib (first line) Locally advanced Recommended 19402 0.187 3637 1.09 0-2 1217
or metastatic
Gefitinib (first line) Locally advanced Recommended 35992 0.223 8023 1.09 0-2 1217
or metastatic
Gefitinib (first line) Locally advanced Recommended 28663 0.145 4138 1.09 0-2 1217
or metastatic
Bevacizumab in Metastatic breast Not Recommended 77314 0.259 30469 1.15 n/a 736
combination with a cancer
taxane (first line)
Bevacizumab in Metastatic breast Not Recommended 57753 0.273 31416 1.15 n/a 736
combination with a cancer
taxane (first line)
Bevacizumab in Metastatic breast Not Recommended 60101 0.259 27358 1.15 n/a 736
combination with a cancer
taxane (first line)
Erlotinib (first-line Advanced or Not Recommended 44812 0.1591 7129 1.23 0-1 487
maintenance metastatic
treatment) non-small cell
lung cancer
(Continued)
Economic Evaluation of Cancer Drugs
TABLE 10.4 (CONTINUED)
An Extract of UK HTA Decisions for Cancer Drugs
INC_ Sample
Drug Condition Categorization ICER INC_QALY COST_best Hros ECOG Size
Fulvestrant Treatment of Not Recommended 31982 0.383 12239 1.19 n/a 736
locally advanced
breast cancer
Cetuximab Treatment of Not Recommended 88000 0.6 53100 1.29 0-2 572
monotherapy or in metastatic
combination with colorectal
chemotherapy carcinoma
Bevacizumab in Treatment of Not Recommended 98000 0.25 24500 1.29 0-2 572
combination with metastatic
non-oxaliplatin colorectal
(fluoropyr carcinoma
Panitumumab Treatment of Not Recommended 150000 0.19 29000 1.29 0-2 572
monotherapy metastatic
colorectal
carcinoma
Eribulin Treatment of Not Recommended 46050 0.1213 5586 1.23 0-2 488
locally advanced
or metastatic
Factors Predictive of HTA Success and the Global Landscape

breast cancer
Eribulin Treatment of Not Recommended 27183 0.1904 5177 1.23 0-2 78
locally advanced
or metastatic
breast cancer
(Continued)
339
TABLE 10.4 (CONTINUED)
340

An Extract of UK HTA Decisions for Cancer Drugs


INC_ Sample
Drug Condition Categorization ICER INC_QALY COST_best Hros ECOG Size
Eribulin Treatment of Not Recommended 35602 0.1136 4041 1.23 0-2 139
locally advanced
or metastatic
breast cancer
Eribulin Treatment of Not Recommended 47631 0.2683 12779 1.23 0-2 43
locally advanced
or metastatic
breast cancer
Ipilimumab Previously Recommended 31559 0.75 23766 1.38 0-1 502
untreated
advanced
melanoma
Dabrafenib Unresectable or Recommended 112727 0.45 55000 1.31 0-1 250
metastatic BRAF
melanoma
Pembrolizumab Treating advanced Recommended 46662 n/a n/a 1.13 0-1 540
melanoma
Cabozantinib Medullary thyroid Recommended 31546 1.34 42215 1.01 n/a 331
cancer
Source: Data from https://www.nice.org.uk/guidance.
Economic Evaluation of Cancer Drugs
Factors Predictive of HTA Success and the Global Landscape 341

cases several comparisons. We used the HTA-accepted model whenever pos-


sible. In some cases data were not available (n/a).
If we summarize the average (mean) of each parameter by whether the
drug was recommended or not, we observe from Table 10.5 the mean values
that separate recommended from non-recommended drugs for each of the
parameters. So, for example, we might expect that for recommended drugs,
the mean ICER should be around £39,000, and the median incremental QALY
around 0.389. For incremental cost and the treatment effect (the hazard
ratios were inverted to reflect the treatment effect of the experimental drug)
the average values were £15,558 and 0.794 respectively. Larger studies also
seemed more strongly associated with a positive recommendation, on aver-
age. If we model the probability of recommendation for these parameters
(Table 10.6), we note that, as expected, the ICER is statistically predictive with
small p-values for other parameters. Interestingly, larger sample sizes seem
to influence a recommendation. This might be a reason to consider power-
ing trials for cost-effectiveness. Due to the small sample of HTAs used in
this analysis, there remains much uncertainty. As future analyses will use
more data, we should expect to have some patterns emerging (this is an area
of current research). This example is merely intended to show how data can
be used to provide a more informed framework for investigating reimburse-
ment decisions.

10.5 The Changing Pace of the Reimbursement Environment


There have been large increases in the costs of cancer care worldwide and
there is real concern that cancer treatment is becoming unsustainable. Drug
budgets are being tightened, resulting in increased payer management of
drug pricing and restricted reimbursement decisions. The oncology clinical
development market has become crowded, and therefore clear differentia-
tion is essential to demonstrate value. Most reimbursement bodies around
the world routinely reject drugs that fail to demonstrate benefit in terms of
survival and/or HRQoL (see Table 10.1 for example).
In Germany, the ability to negotiate price is based on assessment of ‘bene-
fit’ and drugs without assigned ‘benefit’ are automatically forced to be priced
at the cost of the existing standard of care, without any negotiation. Even
in countries such as the US, where drug prices can be set without govern-
ment restrictions, the cost of cancer is rapidly becoming a hot political topic.
According to the American Institute of Cancer Research, cancer costs the
world more money than any other disease group – about $895 billion a year.
In addition to the cost of drugs, we must add the costs of diagnosis, radio-
therapy, imaging, pathology, surgery, and end-of-life care. The US National
Bureau of Economic Research reports that the prices of cancer drugs have
342

TABLE 10.5
Sample Parameters Related to Recommendation
Decision #HTAs Parameter n Mean Median Min. Max.
Recommended 12 ICER 12 39,137 32,850 19,402 112,727
INC QALY 11 0.389 0.223 0.041 1.340
INC cost 11 15,558 8,023 1,364 55,000
Sample size 11 840 663 250 1,725
HR for OS 12 0.794 0.813 0.649 0.869
Not recommended 18 ICER 18 71,804 62,427 27,183 171,301
INC QALY 17 0.270 0.259 0.113 0.60
INC cost 17 22,701 24,001 4,041 53,100
Sample size 18 601 614 43 1,401
HR for OS 18 0.845 0.881 0.694 0.99
Notes: INC: incremental; HR: hazard ratio for overall survival (OS).
Economic Evaluation of Cancer Drugs
Factors Predictive of HTA Success and the Global Landscape 343

TABLE 10.6
Statistical Predictors of a Recommendation
Parameter p-Value
Incremental cost 0.242
Incremental QALY 0.261
ICER 0.039
HR 0.101
Sample size 0.130

increased 10% every year between 1995 and 2013. In the US, the freedom to
price means top-selling drugs are on average three times higher than in the
UK. With new immunotherapy drugs given as monotherapy or in combina-
tion with other drugs, the costs increase (you have to pay for more than one
drug) to more than £100,000 per year per patient. The ‘financial toxicity’ of
these drugs is a real concern for patients and healthcare systems alike across
the world.
Since 2012, there has been an increased focus in the US on spiraling cancer
drug prices, with oncologists calling for new regulations to keep prices in
check. Even American patients are concerned – 77% believe drug costs are
unreasonable and 73% think the pharmaceutical industry cares more about
profits than people. Professional bodies such as the American Society of
Clinical Oncology (ASCO) and the European Society of Medical Oncology
(ESMO) are becoming more vocal about the price and effectiveness of new
therapies. In June 2015, ASCO unveiled its conceptual framework to assess
the value of new cancer treatment options, and ESMO issued guidance on
assessment of meaningful clinical benefit of new anti-cancer therapies.
In contrast to other countries, the US has a more fragmented approach
to HTA, with different organizations applying varying methodologies.
Recently, the Institute of Clinical and Economic Review initiated an emerg-
ing therapy assessment program with the goal of creating a transparent
method for analyzing and judging value. The institute’s budget impact
assessment considers the effect of a drug on net health spending over five
years, taking into account assumptions about the product’s projected uptake.
Given those assumptions, it then calculates a drug price such that annual net
spending on that drug would not exceed roughly US $900 million, a number
derived from assumptions about how fast the US economy is growing and
the number of new medications approved each year. The greatest impact of
these reports thus far has been to exert pressure on manufacturers when
drug prices exceed the institute’s threshold of value and societal afford-
ability, although not all the drugs evaluated so far have been determined
to be overpriced. Although, a not-for-profit organization, without any legal
authority to set prices, its assessments have come under increasing criticism
from the drug manufacturers.
344 Economic Evaluation of Cancer Drugs

10.6 Reimbursement and Payer Evidence


Requirements across Different Countries
Over the last three decades, HTA has become more visible and dissemi-
nated in Europe, North America, Australia, and, more recently, in devel-
oping countries where health policies are being revised (Banta et al., 2009).
Almost all of these HTA agencies require health economic evaluation to
support reimbursement decisions. Some agencies recommend cost utility
analyses while others require cost-effectiveness analyses. In terms of evi-
dence requirements, almost all agencies recommend at least one main com-
parator. This comparator is often the standard of care or current practice.
Recommendations regarding the time horizon depend on the duration of
disease, but this is often only useful for a societal perspective, although some
HTA agencies such as NICE recommend choosing a time horizon depending
on relevant costs and benefits. There are also considerable variations regard-
ing costs that should be included and these vary depending on the chosen
perspective. Many agencies, such as NICE, Canadian Agency for Drugs and
Technologies in Health (CADTH), and Agency for Healthcare Research and
Quality (AHRQ) also include direct costs outside the healthcare system.
Additionally, indirect costs included in the societal perspective differ.
Some agencies include only the productivity loss (e.g. IQWiG), while others
also include the time cost of families (e.g. Pharmaceutical Benefits Board,
Sweden). In Europe, some countries (e.g. the UK, the Netherlands, and
Sweden) have well-established procedures for considering cost-effectiveness
in reimbursement decisions and require cost per QALY, while others (e.g.
France) consider health economic information less formally although cost-
effectiveness analysis may be recommended in manufacturers’ submissions
(Sorenson et al., 2008). IQWiG specifies the type of clinical outcomes, mortal-
ity, morbidity, and validated surrogates as the outcomes required, and not
the type of analysis.
In the US, while there has been some progress, the trend toward using
cost-effectiveness analysis has been slower to develop. American HTA orga-
nizations tend to separate the evaluation of clinical evidence from economic
evidence. Outside Europe, PBAC in Australia, CADTH in Canada, and
AHRQ in the US also conduct economic evaluation of new drugs for their
respective territories.
Understanding payers and payer systems is a key element to understand-
ing global market access, drug pricing, and reimbursement. There are a
number of different payer systems and price control mechanisms around
the world that range from national to regional bodies. NICE (UK) provides
national level guidance regarding reimbursement of new cancer drugs
within England and Wales, while regional budgets are controlled by clini-
cal commissioning groups (CCGs). Similarly, in Canada, national agencies
Factors Predictive of HTA Success and the Global Landscape 345

control certain aspects of drug pricing, and regional provinces are left to
decide budget impact and inclusion of new drugs in formularies. In Italy
and Spain, there is tight control exerted by the national authorities over drug
pricing and reimbursement, however, regional bodies can exert additional
control over drug utilization in their own regional healthcare budgets. In
Germany and the Netherlands, a large number of independent sick funds
control the implementation of healthcare budgets in addition to the national
funding and control structure.
For payers worldwide, open consideration of economic efficiency raises
challenges. Nevertheless, the lack of procedures for considering economic
evidence in a transparent way also creates problems. Recently, the govern-
ments of Belgium, the Netherlands, Luxembourg and Austria have joined
forces to try to find a more sustainable way to provide access to costly drugs
(Beneluxa.org, accessed January 2019). The coalition aims to provide inter-
ested parties with a central information point about collaboration and its
different strands: horizon scanning, information sharing, health technol-
ogy appraisals, and joint price negotiations. In the future, this site intends to
make available documents, such as the terms of reference of the coalition and
scientific reports, and will be regularly updated.

10.6.1 Canada
Since 2003, new drugs are reviewed by the Common Drug Review (CDR)
process to evaluate if they qualify for provincial formulary considerations.
Oncology drug review is carried out by the pan-Canadian Oncology Drug
Review (pCODR). Both CDR and pCODR review drugs through expert
committees and are managed by CADTH. Cost-effectiveness assessment
is required in addition to comparative drug effectiveness as well as safety
assessment and potential budget impact.

10.6.2 France
In theory, France has a free pricing system for non-reimbursed drugs (see
https​://ww​w.isp​or.or​g/HTA​RoadM​aps/F​rance​.asp)​, however, prices for
reimbursed drugs are controlled through the Economic Committee of Health
Products (CESP). Medical/economic reviews are required for all new drugs
for which the company claims an ASMR (medical improvement score) rating
of I, II or III, has expected sales above 20 million euros, and is evaluated by
the CEESP (Commission d’Évaluation Économique et de Santé Publique) of
the HAS. The ASMR is established by the transparency commission (commis-
sion de la transparence) as part of the remit of the HAS. The CESP considers
assessments in euros per QALY but there are no predefined ICER thresholds
(Figure 10.2).
346 Economic Evaluation of Cancer Drugs

FIGURE 10.2
French system.

Source: Adapted from ISPOR. Accessed from https​: //to​ols.i​spor.​org/h​taroa​dmaps​/ Fran​


ce.as​p.

10.6.3 Germany
Outlines the decision-making process in Germany (ISPOR, 2009). Following
dossier submission, G-BA commissions IQWiG to prepare an added benefit
of new drug versus one or more existing comparators. Drugs without added
benefit are price referenced against the identified comparative benchmarks,
without negotiation. Health economic evaluations are not mandated for
reimbursement evaluation, but can be undertaken by the manufacturers at
their discretion Figure 10.3.
As stated by IQWiG: first, because

The G-BA is responsible for the overall procedure of early benefit assess-
ment and the pharmaceutical companies submit their dossiers to the
G-BA. The G-BA usually commissions IQWiG with the scientific report.
After publication of the report, the G-BA conducts a commenting pro-
cedure. This can provide supplementary information and can conse-
quently also lead to a modified result of the assessment. The assessment
procedure is only complete with a formal decision by the G-BA on the
added benefit and on the extent of added benefit. The further procedure
depends on this decision and can, in simple terms, take two directions. If
no added benefit can be determined, a reference price is allocated to the
new drug (or a price that is not allowed to be higher than that of the com-
parator therapy). The latter is the case if no suitable reference price group
exists. If an added benefit has been determined, then price negotiations
Factors Predictive of HTA Success and the Global Landscape 347

FIGURE 10.3
German process adapted from ISPOR.

Source: IQWiG https​: //ww​w.iqw​ig.de​/en/a​bout-​us/10​-year​s-of-​iqwig​/amno​g-sin​ce-20​11.63​


33.ht​ml.

are conducted between the Umbrella Organization of Statutory Health


Insurance (‘GKV-Spitzenverband’) and the pharmaceutical company.
Since the introduction of AMNOG, a health economic evaluation is (only)
planned for the situation where, after a regular early benefit assessment,
price negotiations fail, and an arbitral verdict does not lead to the set-
ting of a price either. Then the pharmaceutical company or the Umbrella
Organization of Statutory Health Insurance (‘GKV-Spitzenverband’) can
apply for a health economic evaluation.

10.6.4 Italy
Health economic evaluations in Italy have only limited impact on pricing
and reimbursement decisions. However, the budget impact model as well as
patient and clinical outcome data play a more important role. At the regional
and hospital level, budget impact analyses are particularly important. Another
unique aspect of the reimbursement mechanism in Italy is the preference for
risk-sharing schemes for oncology drugs. For example, Bayer agreed to pro-
vide 50% discount on nexevar for the initial two months of treatment. Once
response to treatment is established, only patients who have been deemed to
respond to treatment with nexevar will be covered for 100% reimbursement.

10.6.5 Spain
The Spanish National Health Service (Sistema Nacional de Salud, SNS) pro-
vides universal health coverage to, essentially, the whole Spanish population.
348 Economic Evaluation of Cancer Drugs

However, there is limited influence of health economic data on pricing and


reimbursement. A recent EUnetHTA report describes it as:

The Agencia Española de Medicamentos y Productos Sanitarios (AEMPS)


is the Spanish Medicines Agency which is part of the Spanish Ministry
of Health, Social Services and Equality. As part of its remit it produces
health technology assessments of medicinal products called therapeutic
positioning reports. Therapeutic positioning reports identify the thera-
peutic value of a product compared to alternatives. The reports provide
advice to the DG Pharmacy to inform national pricing and reimburse-
ment decisions. The reports also provide advice to the 17 regional health
authorities about procurement and selection of medicinal products and
to other decision makers at a healthcare level (hospitals, prescribers, etc.)
about the use of medicinal products.

About 50 therapeutic positioning reports are produced by AEMPS each


year:

Reports produced by AEMPS include clinical effectiveness. The process


is completed sequentially where clinical effectiveness information is
compiled and an economic assessment is performed after price setting.
Because AEMPS is a medicines agency supporting licensing of medi-
cines, they have access to the regulatory documents that underpin the
marketing authorisation procedure. AEMPS use these documents to
complete the assessment of therapeutic effectiveness themselves with-
out using additional evidence, unless significant evidence exists outside
the regulatory submission identified by the organisation itself, provided
by the marketing authorisation holder, or other stakeholders involved in
the procedure (including scientific societies and patients associations).
(Source: EUnetHTA WP7 research and analysis activity 1: Annex 2
Case studies)

10.6.6 Australia
Australia was one of the first countries to implement a cost-effective-
ness requirement as a part of pricing and reimbursement approval.
Australia’s Medicare provides comprehensive healthcare coverage for all
residents and prices of drugs that are reimbursed are controlled by the
Pharmaceutical Benefits Scheme (PBS). Listing on PBS is subject to review
by the Pharmaceutical Benefits Advisory Committee (PBAC). The subcom-
mittee of the PBAC reviews pharmacoeconomic submissions that manu-
facturers need to submit in order to qualify for reimbursement. Based on
the innovation status of the drug, the committee determines whether the
manufacturer is required to submit incremental cost-effectiveness or cost-
minimization in comparison with the appropriate reference comparator(s).
Factors Predictive of HTA Success and the Global Landscape 349

Generally, manufacturers of a new drug with substantial incremental costs


are required to justify this higher cost by demonstrating robust cost-effec-
tiveness. Australian reimbursement authorities may use higher cost-effec-
tiveness thresholds for cost-effectiveness assessment of oncology drugs.

10.6.7 United Kingdom
Under intense pressure from the public, clinicians, and the pharmaceutical
industry, the NHS in England has made a number of attempts to reassess
its approach to cancer drugs reimbursement. The first of these changes was
the introduction of end-of-life criteria. In January 2009, NICE introduced
supplementary advice to improve NHS access to end-of-life treatments. The
advice meant that treatments for patients with a short life expectancy can
exceed NICE’s cost effectiveness threshold of £30,000 per QALY, provided
that: they are for patients with a short life expectancy; they extend life by at
least three months compared with current NHS treatment; and they apply
to small patient populations. In addition to end-of-life criteria, the Cancer
Drug Fund (CDF) was established in April 2011, which provided a budget
and funding approval mechanism for cancer drugs rejected by NICE. The
CDF came under intense criticism for lack of value and was subsequently
reformed in 2016 (Agrawal et al., 2017). The new arrangements put it on a
more sustainable footing with three key objectives:

• Patients have faster access to the most promising new cancer


treatments.
• Taxpayers get better value for money in drug expenditure.
• Pharmaceutical companies that are willing to price their products
responsibly can access a new, fast-track route to NHS funding for the
best and most promising drugs.

10.7 Pricing and Reimbursement


Environment in the United States
Healthcare in the US is administered through private insurance or managed
care organizations (MCO). Managed care plans are a type of health insur-
ance. They have contracts with healthcare providers and medical facilities
to provide care for members at reduced costs. In addition, government-run
Medicare and Medicaid bodies also administer healthcare in the US.
The US does not have a centralized reimbursement system and drug prices
can be set without government restrictions, although there are some post-
launch limitations in pricing freedom for the government-managed sectors.
350 Economic Evaluation of Cancer Drugs

In the private sector, each drug manufacturer has the freedom to determine
the optimal price for their drugs. They can also adjust prices over time, for
example linking them to inflation rates or other factors (e.g. competitor drugs
or new data availability).
The Institute for Clinical and Economic Review (ICERev) (not to be con-
fused with the incremental cost-effectiveness ratio!) is the first organization
in the US to address drug prices using cost-effectiveness methods, and to
gain the attention of important stakeholders. An ICERev report is composed
of six main parts: comparative clinical-effectiveness, incremental cost-effec-
tiveness, potential benefits or disadvantages that lie outside the scope of clin-
ical- or cost-effectiveness, contextual considerations, budget impact analysis,
and a section in which the value-based price benchmark is calculated (see:
https://icer-review.org/methodology/). However, it must be pointed out that
ICERev is a non-profit organization and does not have any influence over
stakeholders involved in setting drug prices in the US.

10.8 Value-Based Pricing (VBP) for Cancer Drugs


Under the PPRS reform discussions in the UK from 2010 to 2013 there was
a call for a move to a value-based pricing system, where price is directly
linked to the value a drug provides. The idea gained tremendous traction
with health economists around the world because of the underlying appeal
of linking drug price to the value of drug.
The initial VBP proposal included the following elements (DOH, White
Paper, 2010).

1. Flexibility to establish value by incorporating burden of illness and


societal benefits as modifiers to the cost-effectiveness cut-off criteria
operated by NICE.
2. Price control at the established value of the new drug instead of a
limit on reimbursement at that established value under the existing
system.

Such a system was due to be introduced in the UK but complexity in the


negotiations has seen this, and its iteration, value-based assessment, shelved.
The underlying reasons included lack of agreement on the ‘value’ (DOH,
White Paper, 2010; Cohen, 2017). Fundamentally, value is defined as the
health outcomes achieved per unit of currency spent, but there is no univer-
sally accepted definition of what good value—or even acceptable value—
means in cancer care. Value means something different to each stakeholder,
whether a patient with cancer or a caregiver, a physician treating patients
with cancer, a healthcare payer, or another medical decision-maker. Another
Factors Predictive of HTA Success and the Global Landscape 351

important consideration is that the oncology drug development paradigm


requires drug manufacturers to demonstrate efficacy and safety in later
lines of treatment first because it is considered unethical to experiment new
treatment options in earlier lines. The incremental benefit in later lines of
treatment is much smaller and therefore a new drug may not be able to dem-
onstrate its ‘value’ at the time of initial approval and launch, restricting reim-
bursement decisions.
Table 10.7 shows a case study outlining the approval history of rituximab
in patients in follicular lymphoma. Rituximab was evaluated and approved
initially in patients who were resistant to prior treatment options, and later
trials gradually expanded the indication to earlier lines of treatment and
regimens. It is also evident from the data that efficacy benefit was greatest in
the first-line setting; however, these data did not become available until more
than 12 years after the initial approval.

Example 10.2: Case Study – Evolution of Rituximab


Approvals from Last-Line to Front-Line Treatment
in Patients with Follicular Lymphoma
This creates a major problem for manufacturers, as a lack of demon-
strated benefit at the time of initial approval may force reimbursement
and price below the ultimate value, and increasing price later to reflect
‘value’ is only theoretically possible. For example, the UK has introduced

TABLE 10.7
Approval History of Rituximab in Patients in Follicular Lymphoma (Rituximab
Was Evaluated and Approved Initially)
Approval Summary of Clinical Data From
Date Patient Population Approved Label (SPC)
June 1998 Chemoresistant or in second or Response rate: 48%
higher relapse after
chemotherapy
March 2004 Previously untreated (in Median time to progression: 14.7
combination with one particular months
type of chemotherapy only)
July 2006 Maintenance treatment in Median PFS was 42.2 months in the
relapsed or refractory, but rituximab maintenance arm compared
responding to induction to 14.3 months in the observation arm
chemotherapy
January 2008 Previously untreated To expand use to combination with all
types of chemotherapy based on three
different trials in combination with
chemotherapy
October 2010 Previously untreated Median PFS: not reached in rituximab
arm vs. 48 months for control arm
Note: SPC: summary of product characteristics.
352 Economic Evaluation of Cancer Drugs

a formal mechanism to reprice a drug on that basis; however it has never


led to an actual increase (Table 10.7).
At the time of publishing of this book, the future of VBP in the UK is
still unknown. The terminology seems to have shifted to ‘value-based
assessment,’ however, there is no progress to report on that front either.
The uncertainties around the exit of the UK from the European Union
(‘Brexit’) has created uncertainties for both reimbursement and market
authorization bodies.

10.9 Risk-Sharing Scheme


Risk-sharing schemes operate on the basic principle that in return for drug
manufacturers’ guarantee of a price reduction, the new drug will be added
to the list of reimbursed drugs (Carlson et al., 2017; Carlson et al., 2010;
Gonclaves et al., 2018). These schemes are increasingly common in European
countries and Australia where there are centralized reimbursement mecha-
nisms in place, giving payers powers over market access.
In Italy, the risk-sharing deals are used to avoid paying for patients who do
not respond to treatment. In the UK, the term patient-access scheme (PAS) is
routinely used to describe these arrangements. Table 10.8, lists the oncology
treatments approved by NICE with associated patient-access schemes. It is
evident from the table that the majority of currently reimbursed drugs have
a PAS associated as the condition for reimbursement. As of October 2017,
65 oncology treatments have a PAS associated with positive reimbursement
decisions, representing almost two-thirds of total treatments reimbursed by
NICE to date. Companies offer these deals in order to bring the ICER within
accepted cost-effectiveness threshold.
Figure 10.4 presents the breakdown of different types of PASs. PASs
are usually initiated by the drug manufacturers in order to resolve the

60 56
50
40
30
20
10
3 2 1 1 1 1
0
Simple Dose cap Free stock Discount+ Single fixed Time cap Response
discount poss rebate price scheme

FIGURE 10.4
Breakdown of different types of PAS.
Factors Predictive of HTA Success and the Global Landscape 353

TABLE 10.8
A List of Oncology Treatments Approved by NICE with Associated Patient-Access
Schemes
Treatment Indication Type of scheme
Bortezomib Multiple myeloma Response scheme
Sunitinib Renal cell carcinoma Free stock
Lenalidomide Multiple myeloma Dose cap
Sunitinib Gastrointestinal stromal tumor Free stock
Trabectedin Advanced soft tissue sarcoma Dose cap
Gefitinib Non-small-cell lung cancer Single fixed price
Pazopanib Advanced renal cell carcinoma Discount plus
rebate
Azacitidine Myelodysplastic syndromes, chronic Simple discount
myelomonocytic leukemia and acute myeloid
leukemia
Mifamurtide High grade resectable non-metastatic osteosarcoma Simple discount
Nilotinib Imatinib-resistant chronic myeloid leukemia Simple discount
Nilotinib First-line treatment of chronic myeloid leukemia Simple discount
Fingolimod Highly active relapsing-remitting multiple sclerosis Simple discount
Erlotinib First-line treatment of locally advanced or metastatic Simple discount
EGFR-TK mutation-positive non-small-cell lung cancer
Denosumab Skeletal related events in adults with bone Simple discount
metastases from solid tumors
Ipilimumab Advanced melanoma, second line Simple discount
Vemurafenib Metastatic mutation positive melanoma Simple discount
Pixantrone Multiple relapsed or refractory aggressive non- Simple discount
Hodgkin's B-cell lymphoma
Afatinib Locally advanced or metastatic non-small-cell lung Simple discount
cancer with activating epidermal growth factor
Enzalutamide Metastatic hormone-relapsed prostate cancer in Simple discount
adults whose disease has progressed during or after
docetaxel-containing chemotherapy
Ipilimumab Adults with previously untreated advanced Simple discount
(unresectable or metastatic) melanoma
Dabrafenib Unresectable or metastatic melanoma with a Simple discount
BRAFV600 mutation
Lenalidomide Myelodysplastic syndromes associated with an Dose cap
isolated deletion 5q cytogenetic abnormality
Axitinib Advanced renal cell carcinoma after failure of prior Simple discount
systemic treatment
Obinutuzumab Untreated chronic lymphocytic leukemia Simple discount
Ofatumumab Untreated chronic lymphocytic leukemia Simple discount
Nintedanib Previously treated locally advanced, metastatic, or Simple discount
locally recurrent non-small-cell lung cancer
Pembrolizumab Advanced melanoma after disease progression with Simple discount
ipilimumab
(Continued)
354 Economic Evaluation of Cancer Drugs

TABLE 10.8 (CONTINUED)


A List of Oncology Treatments Approved by NICE with Associated Patient-Access
Schemes
Treatment Indication Type of scheme
Pembrolizumab Advanced melanoma not previously treated with Simple discount
ipilimumab
Erlotinib Non-small-cell lung cancer that has progressed after Simple discount
prior chemotherapy
Enzalutamide Metastatic hormone-relapsed prostate cancer before Simple discount
chemotherapy is indicated
Panobinostat Multiple myeloma after at least 2 previous Simple discount
treatments
Olaparib Maintenance treatment of relapsed, platinum- Time cap
sensitive, BRCA mutation-positive ovarian,
fallopian tube, and peritoneal cancer after response
to second-line or subsequent platinum-based
chemotherapy
Ruxolitinib Disease-related splenomegaly or symptoms in adults Simple discount
with myelofibrosis.
Cabazitaxel Hormone-relapsed metastatic prostate cancer treated Simple discount
with docetaxel.
Ceritinib Previously treated anaplastic lymphoma kinase Simple discount
positive non-small-cell lung cancer
Trametinib In combination with dabrafenib for treating Simple discount
unresectable or metastatic melanoma
Dabrafenib In combination with trametinib for treating Simple discount
unresectable or metastatic melanoma
Bosutinib Previously treated chronic myeloid leukemia Simple discount
Trifluridine and Previously treated metastatic colorectal cancer Simple discount
tipiracil
hydrochloride
Crizotinib Untreated anaplastic lymphoma kinase-positive Simple discount
advanced non-small-cell lung cancer
Talimogene Unresectable metastatic melanoma Simple discount
laherparepvec
Radium-223 Hormone-relapsed prostate cancer with bone Simple discount
dichloride metastases
Nivolumab Previously treated advanced renal cell carcinoma in Simple discount
adults
Everolimus Advanced breast cancer after endocrine therapy Simple discount
Crizotinib Untreated anaplastic lymphoma kinase-positive Simple discount
advanced non-small-cell lung cancer
Eribulin Breast cancer (locally advanced, metastatic) Simple discount
Pertuzumab Neoadjuvant treatment of breast cancer Simple discount
Dasatinib Imatinib-resistant or intolerant chronic myeloid Simple discount
leukemia
(Continued)
Factors Predictive of HTA Success and the Global Landscape 355

TABLE 10.8 (CONTINUED)


A List of Oncology Treatments Approved by NICE with Associated Patient-Access
Schemes
Treatment Indication Type of scheme
Nilotinib Imatinib-resistant or intolerant chronic myeloid Simple discount
leukemia
Dasatinib Untreated chronic myeloid leukemia Simple discount
Nilotinib Untreated chronic myeloid leukemia Simple discount
Pomalidomide Multiple myeloma previously treated with Simple discount
lenalidomide and bortezomib
Pembrolizumab PD-L1-positive non-small-cell lung cancer after Simple discount
chemotherapy
Everolimus Renal cell carcinoma Simple discount
Ibrutinib Previously treated chronic lymphocytic leukemia Simple discount
and untreated chronic lymphocytic leukemia with
17p deletion or TP53 mutation
Cetuximab Previously untreated metastatic colorectal cancer Simple discount
Panitumumab Previously untreated metastatic colorectal cancer Simple discount
Everolimus Unresectable or metastatic neuroendocrine tumors in Simple discount
people with progressive disease
Blinatumomab Previously treated Philadelphia-chromosome- Simple discount
negative acute lymphoblastic leukemia
Ponatinib Chronic myeloid leukemia and acute lymphoblastic Simple discount
leukemia
Carfilzomib Previously treated multiple myeloma Simple discount
Nivolumab Relapsed or refractory classical Hodgkin lymphoma Simple discount
Cabozantinib Previously treated advanced renal cell carcinoma Simple discount
Obinutuzumab Rituximab refractory follicular lymphoma Simple discount
Nab-paclitaxel Adenocarcinoma of the pancreas Simple discount

cost-effectiveness issues following initial negative advanced consultation


document (ACD) issued by NICE. It is evident from Table 10.8 and Figure 10.4
that almost all of these negotiations involved price reductions by offering
confidential simple discounts. However, some of the treatments approved by
NICE have innovative PASs associated with the approval.

Example 10.3: Case Study 1 – Bortezomib for


Multiple Myeloma – Response-Based PAS
In 2006, NICE declined to recommend bortezomib for the treatment of
patients with multiple myeloma, as bortezomib wasn’t considered cost-
effective (cost per QALY > £30,000). Under the risk-sharing scheme,
the company proposed to reimburse the treatment cost for patients
with a more than 50% reduction. The M-spike is a sign of abnormal
gamma immunoglobin 50% reduction in serum M-protein (a criteria
356 Economic Evaluation of Cancer Drugs

for measuring response to treatment in these patients). Following this


arrangement, NICE reversed their initial negative reimbursement ruling
and made the drug available for routine use within the NHS.

Example 10.4: Case Study 2 – Lenalidomide


for Multiple Myeloma – Dose Cap
Another innovative deal also involved patients with multiple myeloma
where, in 2009, NICE declined to reimburse lenalidomide for patients
who previously received at least two prior treatments, based on the high
cost of drug acquisition (as patients with lenalidomide are treatment-
until-progression). The company proposed to provide the drug free of
charge beyond two years, i.e. the NHS will pay for first two years of
treatment and the company will provide free drugs for patients who
continue to derive benefit following two years of treatment.

10.10 The Future of Cost-Effectiveness of Cancer Treatments


There are several areas of future research in cost-effectiveness in cancer.
These can be split into methodological aspects, in terms of improving techni-
cal methodology in economic evaluation, and how the reimbursement envi-
ronment may impact on how economic evaluation is conducted.

10.10.1 Future Research: Methodology


Cost-effectiveness in clinical trials still offers many challenges for research-
ers in health economic evaluation. The methods used in health economic
evaluation are not always understood by researchers and payer review
groups. For example, the methods described for handling crossover in can-
cer trials can be very complex, and can take time for even an experienced
statistician to appreciate. The use of Bayesian methods in clinical trials still
poses problems, since many statisticians are used to designing clinical trials
using classical (frequentist) methods and are accustomed to using software
such as SAS®. It is only relatively recently that Bayesian computation has
become slightly more feasible. Figures 10.5 and 10.6 give some examples of
challenges in trial design and analysis for the future.
Statistical support for payer and reimbursement activities are often sepa-
rated from the clinical drug development programme. This should start from
early phase trials. Moreover, statistical knowledge and experience of payer
evidence requirements is invested in a few statisticians and clinical trial staff
whereas the concept of demonstrating value of treatments is more of a ‘mind-
set’ about adding value and not just an isolated (statistical) function. Often,
Factors Predictive of HTA Success and the Global Landscape 357

FIGURE 10.5
Some challenges in designing trials for health economic evaluation.

FIGURE 10.6
Some challenges in analyzing data for health economic evaluation.
358 Economic Evaluation of Cancer Drugs

payer evidence considerations are thought about at the start of the Phase III
trial, whereas there is a need to think about what combination of possible
studies will result in a high probability of market access; or what combination
of studies will be likely to demonstrate value. The other challenge is that the
statistical tools for estimating value are not clear or are unknown to statisti-
cians – unfamiliarity with G-estimation techniques for handling crossover
or VOI methods are areas where training and courses have only become
available relatively recently for applications in clinical trials. Value analysis/
reimbursement analysis/health economic analysis plans (HEAPs), in addi-
tion to statistical analysis plans (SAPs), which identify clearly those endpoints
necessary for demonstrating value and those as direct inputs into a health
economic model, are needed to be written collaboratively.
Several other technical issues or interesting areas of research that require
additional work include sample sizes based on value of information (VOI),
value-based pricing models, cost-effectiveness based on more than one end-
point, extrapolation in the presence of switching treatment (in cancer trials),
and also simulation in the presence of crossover for PSA. Applications of
flexible parametric models are still limited; improvements in mapping func-
tions for HRQoL and, particularly, some generic measures, where the ability
of generic instruments to detect meaningful treatment effects, is of concern.
The use of platform and basket trials and the challenges these bring for eco-
nomic evaluation is also an area of research where much methodological
innovation is required. In short, there is no shortage of methodological areas
for statisticians, health economists, and researchers in health economic eval-
uation to develop, whether in academia or industry. More recently, Neumann
et al. (2018) identify some areas:

(i) Modeling (Comparative Modeling and Model Transparency).


(ii) Health outcomes (valuing temporary health and path states, as well
as health effects on caregivers).
(iii) Incorporating societal perspectives.
(iv) Evidence synthesis (developing theory on learning across studies
and combining data from clinical trials and observational studies).
(v) Estimating and using cost-effectiveness thresholds (empirically rep-
resenting two broad concepts: opportunity costs and public willing-
ness to pay).
(vi) Reporting and communicating CEAs (written protocols and a qual-
ity scoring system).

10.10.2 Future Reimbursement Landscape


In October 2016, NICE and NHS England (NHS-E) put out new proposals
for consultation on how they would like to work closely to “simplify and
Factors Predictive of HTA Success and the Global Landscape 359

speed up” new technology appraisals. The document includes the following
proposals:

(a) A new budget impact threshold.


(b) Restructuring of QALY assessment for highly specialized treatments
for very rare diseases.
(c) Fast-track appraisals for drugs that have a cost per QALY of below
£10,000.

Budget Impact Threshold


The “special arrangements” will be triggered when a drug appraised by
NICE is expected to exceed a budget impact threshold of £20 million. NICE
would calculate the drug’s potential budget impact by estimating its net
annual cost to the NHS. NHS-E is responsible for allocating funding for new
products, some of which have a high budget impact. In order to deal with this
responsibility and avoid risking access to other forms of care, NHS-E wants to
put in place new arrangements for managing the budget impact. If an agree-
ment was struck that lowered the drug’s budget impact below the threshold,
the standard 90-day funding requirement for a NICE-assessed drug would
apply. If not, then NHS England could ask NICE to alter the standard funding
requirement to allow for a longer period of phased introduction. According
to current regulations NICE recommendations have to be complied with no
more than three months after the final guidance is published.

10.10.2.1 Automatic Funding for Highly Specialized Drugs for Rare Diseases


The document suggests that treatments for very rare conditions would be
automatically funded if they have an incremental cost-effectiveness ratio
of up to £100,000 per QALY. The proposed limit is five times higher than
the lower end of NICE’s standard threshold of less than £20,000 per QALY.
Treatments deemed to have a high value and low cost are likely to be the
most successful, while those with a low benefit and higher cost will be less
likely to win a recommendation. If the drug’s ICER did exceed £100,000 per
QALY, it would not be subject to the funding requirement, but would be
considered for funding through NHS England’s annual specialized commis-
sioning process (Adkins et al., 2017).

10.10.2.2 Fast-Track Appraisals
According to the proposal, any products with £10,000 or less per QALY will
be fast-tracked with 11 weeks’ reduction (32 weeks, down from 43 weeks) to
the final guidance. However, between 2007 and 2014, around 15% of NICE’s
technology appraisals fell at or below £10,000 per QALY. The introduction of
360 Economic Evaluation of Cancer Drugs

a fast-track process would enable them to be routed through a lighter touch


appraisal process, “speeding up access for patients,” as stated in the docu-
ment. Any policy changes as a result of this consultation may have profound
implications for the pricing and reimbursement of technologies assessed by
NICE in future.

10.11 Summary
An analysis of NICE Single Technology Assessments in Oncology since 2001
shows that 61% were granted complete or partial reimbursement. Criticism
of the rejected submissions covered a wide range of aspects including health
resources/costs, HRQoL measures, clinical-effect predictions, and other lack
of evidence. The vast majority, if not all, of submissions relied upon simu-
lation modeling. Problems or lack of sufficient evidence about OS and PFS
were often highlighted by the review groups. An analysis of the accepted
oncology submissions showed a mean of £39,000 and a median incremental
QALY of 0.39 or about 4.7 months. A short review of the payer requirements
across different countries shows a highly variable situation, with the admin-
istrative processes, evidence, and cost criteria and requirements with no
common trend. A general trend was, however, the increase in risk-sharing
schemes (often including a discount price) for new, costly cancer drugs in
various European countries.

10.12 Exercises for Chapter 10


1. Explain the difference between an MTA and an STA. What factors
does NICE consider when making HTAs?
2. What factors might predict the recommendation of a cancer drug by
reimbursement authorities?
3. Compare and contrast the different reimbursement strategies across
countries worldwide. Is there a general pattern?
4. Identify some future areas of research in cost-effectiveness. Why are
some of these important for cancer drugs in particular?
References

Abernethy, A., Abrahams, E., Barker, A. et al. (2014). Turning the tide against can-
cer through sustained medical innovation: The pathway to progress. Clinical
Cancer Research; 20(5):1081–1086.
Ades, A.E., Lu, G., & Claxton, K. (2004). Expected value of sample information cal-
culations in medical decision modeling. Medical Decision Making; 24(2):207–227.
Ades, A.E., Lu, G., & Madan, J.J. (2013). Which health-related quality-of-life outcome
when planning randomized trials: Disease-specific or generic, or both? A com-
mon factor model. Value in Health; 16(1):185–194.
Ades, A.E., Madan, J., & Welton, N.J. (2011). Indirect and mixed treatment compari-
sons in arthritis research. Rheumatology; 50(suppl 4):iv5–iv9.
Adkins, E.M. , Nicholson, L., Floyd, D. et al. (2017). Oncology drugs for orphan
indications: How are HTA processes evolving for this specific drug category?
ClinicoEconomics and Outcomes Research; 9:327–342.
A’Hern, R.P. (2016). Restricted mean survival time: An obligatory end point for time-
to-event analysis in cancer trials? Journal of Clinical Oncology; 34(28):3474–3476.
Aggarwal, A., Fojo, T., Chamberlain, C. et al. (2017). Do patient access schemes for
high-cost cancer drugs deliver value to society?—lessons from the NHS Cancer
Drugs Fund. Annals of Oncology; 28(8):1738–1750.
Agresti, A. (2013). Categorical Adata Analysis, 3rd Edition. John Wiley Press.
Ajani, J.A. (2007). The area between the curves gets no respect: Is it because of the
median madness? Journal of Clinical Oncology; 25(34):5531.
Al, M.J., & Van Hout, B.A. (2000). A Bayesian approach to economic analyses of clini-
cal trials: The case of stenting versus balloon angioplasty. Health Economics;
9(7):599–609.
Albertsen, P.C., Hanley, J.A., Gleason, D.F. et al. (1998). Competing risk analysis of
men aged 55 to 74 years at diagnosis managed conservatively for clinically
localized prostate cancer. JAMA; 280(11):975–980.
Almirall, D., Ten Have, T.T., & Murphy, S.A. (2010). Structural nested mean models for
assessing time-varying effect moderation. Biometrics; 66(1):131–139.
Amdahl, J., Manson, S.C., Isbell, R. et al. (2014). Cost-effectiveness of pazopanib in
advanced soft tissue sarcoma in the United Kingdom. Sarcoma; 2014:481071.
American Lung Association. (n.d.). Lung cancer fact sheet. Retrieved from: www.
lung.org/lung-health-and-diseases/lung-disease-lookup/lung-cancer/
resource-library/lung-cancer-fact-sheet.html.
American Cancer Society. (2018). Breast cancer survival rates. Retrieved from: www.
cancer.org/cancer/breast-cancer/understanding-a-breast-cancer-diagnosis/
breast-cancer-survival-rates.html.
Amico, M., & Van Keilegom, I. (2018). Cure models in survival analysis. Annual
Review of Statistics and Its Application; 5(1):311–342.
Andersen, P.K., Esbjerg, S., & Sorensen, T.I. (2000). Multi-state models for bleeding
episodes and mortality in liver cirrhosis. Statistics in Medicine; 19(4):587–599.
Andersen, P.K., Hansen, L.S., & Keiding, N. (1991). Assessing the influence of revers-
ible disease indicators on survival. Statistics in Medicine; 10(7):1061–1067.

361
362 References

Andersen, P.K., Hansen, M.G., & Klein, J.P. (2004). Regression analysis of restricted
mean survival time based on pseudo-observations. Lifetime Data Analysis;
10(4):335–350.
Andersen, P.K., & Keiding, N. (2002). Multi-state models for event history analysis.
Statistical Methods in Medical Research; 11(2):91–115.
Anderson, D.F., & Kurtz, T. (2015). Stochastic analysis of biochemical systems, Stochastics
in biological systems series, Volume 1.2. Springer International Publishing,
p. 592.
Anglemyer, A., Horvath, H.T., & Bero, L. (2014). Healthcare outcomes assessed with
nonexperimental designs compared with those assessed in randomised trials.
Cochrane Database of Systematic Reviews; 4:MR000034.
Ara, R., & Wailoo, A. (n.d.). NICE DSU technical support document 12: The use of
health state utility values in decision models. Retrieved from: www.nicedsu.
org.uk/.
Araújo, A., Parente, B., Sotto-Mayor, R. et al. (2008). An economic analysis of erlotinib,
docetaxel, pemetrexed and best supportive care as second or third line treat-
ment of non-small cell lung cancer. Revista Portuguesa de Pneumologia (English
Edition); 14(6):803–827.
Armero, C., Cabras, S., Castellanos, M.E., et al. (2016). Bayesian analysis of a disability
model for lung cancer survival. Statistical Methods in Medical Research; 25(1):336–
351. Epub 2012 July 5.
Asukai, Y., Valladares, A., Camps, C. et al. (2010). Cost-effectiveness analysis of
pemetrexed versus docetaxel in the second-line treatment of non-small cell
lung cancer in Spain: Results for the non-squamous histology population. BMC
Cancer; 10(1):26.
Australian Government Department of Health. (2010). Pemetrexed disodium, powder
for I.V. infusion, 100 mg (base) and 500 mg (base), Alimta®. The Pharmaceutical
Benefits Scheme. Retrieved from: www.pbs.gov.au/info/industry/listing/
elements/pbac-meetings/psd/2010-03/pbac-psd-Pemetrexed-mar10.
Australian Government Department of Health. (2013a). Afatinib, tablet, 20 mg, 30
mg, 40 mg and 50 mg, (as dimaleate), Giotrif® (first line) – July 2013. The phar-
maceutical benefits scheme. Retrieved from: www.pbs.gov.au/info/industry/
listing/elements/pbac-meetings/psd/2013-07/afatinib-first-line.
Australian Government Department of Health. (2013b). Erlotinib, tablets, 25 mg, 100
mg, 150 mg (as hydrochloride), Tarceva® – July 2013. The pharmaceutical bene-
fits scheme. Retrieved from: www.pbs.gov.au/info/industry/listing/elements/
pbac-meetings/psd/2013-07/erlotinib.
Australian Government Department of Health. (2013c). Gefitinib, tablet, 250 mg,
Iressa® – July 2013. The pharmaceutical benefits scheme. Retrieved from: www.
pbs.gov.au/info/industry/listing/elements/pbac-meetings/psd/2013-07/
gefitinib.
Bagust, A., & Beale, S. (2014). Survival analysis and extrapolation modeling of time-
to-event clinical trial data for economic evaluation: An alternative approach.
Medical Decision Making; 34(3):343–351. Epub July 30.
Ball, G., Xie, F., & Tarride, J.E. (2018). Economic evaluation of bevacizumab for treatment
of platinum-resistant recurrent ovarian cancer in Canada. PharmacoEconomics
Open; 2(1):19–29.
Bang, H., & Tsiatis, A.A. (2000). Estimating medical costs with censored data.
Biometrika; 87(2):329–343.
References 363

Bang, H., & Zhao, H. (2014). Cost-effectiveness analysis: A proposal of new reporting
standards in statistical analysis. Journal of Biopharmaceutical Statistics.
Bang, H., & Zhao, H. (2016). Median-based incremental cost-effectiveness ratios with cen-
sored data. Journal of Biopharmaceutical Statistics; 26(3):552–564. Epub 2015 May 26.
Banta, D., & Almeida, R.T. (2009). The development of health technology assessment
in Brazil. International Journal of Technology Assessment in Health Care; 25(Suppl
1):255–259.
Bartha, E., Arfwedson, C., Imnell, A. et al. (2013). Randomized controlled trial of
goal-directed haemodynamic treatment in patients withproximal femoral frac-
ture. British Journal of Anaesthesia; 110(4):545–553.
Basch, E., Deal, A.M., Dueck, A.C. et al. (2017). Overall survival results of a trial
assessing patient-reported outcomes for symptom monitoring during routine
cancer treatment. JAMA; 318(2):197–198.
Batchelor, T.T., Mulholland, P., Neyns, B. et al. (2013). Phase III randomized trial
comparing the efficacy of cediranib as monotherapy, and in combination with
lomustine, versus lomustine alone in patients with recurrent glioblastoma.
Journal of Clinical Oncology; 31(26):3212–3218.
Batty, A., Winn, B., Lebmeier, M. et al. (2012). A comparison of patient and general-
population utility values for advanced melanoma in health economic model-
ling. Value in Health; 15:A277–575.
Bebu, I., Luta, G., Mathew, T. et al. (2016). Parametric cost-effectiveness inference with
skewed data. Computational Statistics and Data Analysis; 94:210–220.
Beck, N., & Jackman, S. (1998). Beyond linearity by default: Generalized additive
models. American Journal of Political Science; 42(2):596–627.
Beckett, P., Calman, L., & Darlison, L. (2012). 91 Follow-up of patients with advanced
NSCLC following 1st line chemotherapy – A British Thoracic Oncology Group
National survey; L. Darlison, P. Beckett, L. Calman, C. Mulatero, K. O’Byrne, M.
Peake, D. Talbot; Lung Cancer; 75(Suppl 1): S30–S31.
Benelexua Initiative on Pharmaceutical Policy. (n.d.). Retrieved from: www.
beneluxa.org/.
Berthelot, J.M., Will, B.P., Evans, W.K. et al. (2000). Decision framework for chemo-
therapeutic interventions for metastatic non-small-cell lung cancer. Journal of
the National Cancer Institute; 92(16):1321–1329.
Beusterien, K.M., Szabo, S.M., Kotapati, S. et al. (2009). Societal preference values for
advanced melanoma health states in the United Kingdom and Australia. British
Journal of Cancer; 101(3):387–389.
Billingham, L.J., Bathers, S., Burton, A. et al. (2002). Patterns, costs and cost-effective-
ness of care in a trial of chemotherapy for advanced non-small cell lung cancer.
Lung Cancer; 37(2):219–225.
Biomarkers Definitions Working Group. (2001). Biomarkers and surrogate endpoints:
Preferred definitions and conceptual framework. Clinical Pharmacology and
Therapeutics; 69(3):89–95.
Blazeby, J.M., Avery, K., Sprangers, M. et al. (2006). Health-related quality of life mea-
surement in randomized clinical trials in surgical oncology. Journal of Clinical
Oncology; 24(19):3178–3186.
BNF Publications. (n.d.). Retrieved from: www.bnf.org/.
Bodrogi, J., & Kaló, Z. (2010). Principles of pharmacoeconomics and their impact
on strategic imperatives of pharmaceutical research and development. British
Journal of Pharmacology; 159(7):1367–1373.
364 References

Bongers, M., Coupe, V., Jansma, E. et al. (2011). PCN93 Cost-Effectiveness of Treatment
with New Agents in Advanced Non-Small-Cell Lung Cancer: A Systematic
Review. Value in Health. 2011; 14(7): A451.
Bongers, M.L., de Ruysscher, D., Oberije, C. et al. (2016). Multistate statistical model-
ing: A tool to build a lung cancer microsimulation model that includes parame-
ter uncertainty and patient heterogeneity. Medical Decision Making; 36(1):86–100.
Bossers, N., Van Engen, A., & Heemstra, L. (2015). Understanding key drivers of suc-
cessful Hta submission — Developing a model. Value in Health; 18(7):A342.
Brada, M., Stenning, S., Gabe, R. et al. (2010). Temozolomide versus procarbazine,
lomustine, and vincristine in recurrent high-grade glioma. Journal of Clinical
Oncology; 28(30):4601–4608.
Bradbury, P.A., Tu, D., Seymour, L. et al. & the NCIC Clinical Trials Working Group
on Economic Analysis. (2010). Economic analysis: Randomized placebo-con-
trolled clinical trial of erlotinib in advanced non-small cell lung cancer. Journal
of the National Cancer Institute; 102(5):298–306. Epub 2010 February 16.
Brandes, A.A., Finocchiaro, G., Zagonel, V. et al. (2016). AVAREG: A phase II, random-
ized, noncomparative study of fotemustine or bevacizumab for patients with
recurrent glioblastoma. Neuro-Oncology; 18(9):1304–1312.
Bragg, R.H. & Packer, C.M. (1962). Orientation Dependence of Structure in Pyrolitic
Graphite. Nature; 195: 1080–1082.
Brard, C., Le Teuff, G., Le Deley, M.C. et al. (2017). Bayesian survival analysis in clini-
cal trials: What methods are used in practice? Clinical Trials; 14(1):78–87.
Bray, F., Ferlay, J., Soerjomataram, I. et al. (2018). Global cancer statistics 2018:
GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in
185 countries. CA: A Cancer Journal for Clinicians; 68(6):394–424.
Brazier, J., & Longworth, L. (2011). NICE DSU Technical Support Document 8:
An introduction to the measurement and valuation of health for NICE sub-
missions. https://www.ncbi.nlm.nih.gov/books/NBK425820/pdf/Bookshelf_
NBK425820.pdf
Brazier, J., & Longworth, L. (2013). Mapping to obtain EQ-5D utility values for use in
NICE health technology appraisals. Value in Health; 16(1):202–210.
Brazier, J., Ratcliffe, J., Salomon, J. et al. (2016). Measuring and valuing health benefits for
economic evaluation. Oxford: Oxford University Press.
Brazier, J., Rowen, D. et al. (2012). NICE DSU Technical Support Document 11:
Alternatives to EQ-5D for generating health state utility values. Report by the
decision support unit, March 2011. London: National Institute for Health and
Care Excellence (NICE)
Brazier, J., Yang, Y., Tsuchiya, A. et al. (2009). A review of studies mapping (or cross
walking) non-preference based measures of health to generic preference-based
measures. The European Journal of Health Economics; 11(2):215–225.
Brent, R. (2014). Cost-Benefit and Health Care Evaluations, 2nd Edition. Edward Elgar.
Briggs, A., & Tambour, M. (2001). The design and analysis of stochastic cost-effective-
ness studies for the evaluation of health care interventions. Drug Information
Journal; 35(4):1455–1468.
Brito, M., Esteves, S., André, R. et al. (2016). Comparison of effectiveness of biosimilar fil-
grastim (Nivestim), reference Amgen filgrastim and pegfilgrastim in febrile neu-
tropenia primary prevention in breast cancer patients treated with neo(adjuvant)
TAC: A non-interventional cohort study. Supportive Care in Cancer; 24(2):597–603.
References 365

Brookhart, M.A., Wang, P.S., Solomon, D.H. et al. (2006). Evaluating short-term drug
effects using a physician-specific prescribing preference as an instrumental
variable. Epidemiology; 17(3):268–275.
Brooks, R., Rosalind, R., & de Charro, F. (2013). The measurement and valuation of health
status using EQ-5D: A European perspective: Evidence from the EuroQol BIOMED
research programme. Springer Science & Business Media.
Brown, T., Boland, A., Bagust, A. et al. (2010). Gefitinib for the first-line treatment
of locally advanced or metastatic non-small cell lung cancer. Health Technology
Assessment; 14(Suppl 2):71–79.
Bryant, J., & Day, R. (1995). Incorporating toxicity considerations into the design of
two-stage phase II clinical trials. Biometrics; 51(4):1372–1383.
Buchholz, I., Thielker, K., Feng, Y.S. et al. (2015). Measuring changes in health over
time using the EQ-5D 3L and 5L: A head-to-head comparison of measurement
properties and sensitivity to change in a German inpatient rehabilitation sam-
ple. Quality of Life Research; 24(4):829–835.
Burau, V., & Blank, R.H. (2006). Comparing health policy: An assessment of typol-
ogies of health systems. Journal of Comparative Policy Analysis: Research and
Practice; 8(1):63–76. Published online: 24 Jan 2007.
Calman, K.C. (1984). Quality of life in cancer patients—An hypothesis. Journal of
Medical Ethics; 10(3):124–127.
Calvo, E., Escudier, B., Motzer, R.J. et al. (2012). Everolimus in metastatic renal cell
carcinoma: Subgroup analysis of patients with 1 or 2 previous vascular endo-
thelial growth factor receptor-tyrosine kinase inhibitor therapies enrolled in
the phase III RECORD-1 study. European Journal of Cancer; 48(3):333–339.
Camm, A.J., & Fox, K.A.A. (2018). Strengths and weaknesses of ‘real-world’ stud-
ies involving non-vitamin K antagonist oral anticoagulants. Open Heart;
5(1):e000788.
Canadian Agency for Drugs and Technologies in Health. (n.d.). CADTH pan-Cana-
dian oncology drug review. Retrieved from: https://cadth.ca/pcodr.
Canadian Agency for Drugs and Technologies in Health. (2004). Recommendation
on reconsideration and reason for reconsideration. Retrieved from: www.
cadth.ca/media/cdr/complete/cdr_complete_iressa_06-23-04.pdf.
Canadian Agency for Drugs and Technologies in Health. (2005). CEDAC final recom-
mendation and reason for recommendation. Retrieved from: www.cadth.ca/
media/cdr/complete/cdr_complete_Tarceva_Dec605.pdf.
Cancer Research UK. (n.d.). Cancer incidence for all cancers combined. Retrieved from:
www.cancerresearchuk.org/health-professional/cancer-statistics/incidence/
all-cancers-combined#heading-One.
Cancer Facts & Figures (2011). (2010). American cancer society report. Retrieved from:
https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-
statistics/annual-cancer-facts-and-figures/2011/cancer-facts-and-figures-2011.
pdf.
Cancer Patients Alliance. (n.d.). Pancreatic Cancer Prognosis & Survival.
Pancreatica. Retrieved from: https://pancreatica.org/pancreatic-cancer/
pancreatic-cancer-prognosis/.
Cancer Research UK. (n.d.) Cancer statistics for the UK. Retrieved from: www.cancerre-
searchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/
lung-cancer
366 References

Cancer Research UK. (n.d). UK Lung Cancer and Smoking Statistics.


Cancer Research UK. (n.d) . World Wide Cancer Statistics. Retrieved from: www.
cancerresearchuk.org/health-professional/cancer-statistics/worldwide-cancer.
Cappuzzo, F., Ciuleanu, T., Stelmakh, L., et al. (2010). Erlotinib as maintenance treat-
ment in advanced non-small-cell lung cancer: a multicentre, randomised, pla-
cebo-controlled phase 3 study. Lancet Oncol.; 11(6): 521–529.
Carlson, J.J., Chen, S., & Garrison, L.P. Jr. (2017). Performance-based risk-shar-
ing arrangements: An updated international review. Pharmacoeconomics;
35(10):1063–1072.
Carlson, J.J., Reyes, C., Oestreicher, N. et al. (2008). Comparative clinical and economic
outcomes of treatments for refractory non-small cell lung cancer (NSCLC).
Lung Cancer; 61(3):405–415.
Carlson, J.J., Sullivan, S.D., Garrison, L.P. et al. (2010). Linking payment to health
outcomes: A taxonomy and examination of performance-based reimburse-
ment schemes between healthcare payers and manufacturers. Health Policy;
96(3):179–190.
Caro, J., Möller, J., Karnon, J. et al. (2016). Discrete event simulation for health technology
assessment. New York: Chapman and Hall/CRC.
Carpenter, J., & Kenward, M. (2013). Multiple imputation and its application. John Wiley
Publications.
Carreon, L.Y., Berven, S.H., Djurasovic, M. et al. (2013). The discriminative properties
of the SF−6D compared With the SF−36 and ODI. Spine; 38(1):60–64.
Center for Medical Technology Policy. (n.d.). Best practices for the design, implemen-
tation, analysis, and reporting of oncology trials with high rates of treatment
switching. Resource Center. Retrieved from: www.cmtpnet.org/resource-center/
view/new-guidance-for-treatment-switching-in-oncology-drug-trials/.
Collet, D. (2017). Modelling Survival Data in Medical Research, Second Edition.
Chapman & Hall/CRC Texts in Statistical Science.
Cerri, K.H., Knapp, M., Fernández, J.L. (2014). Decision making by NICE: Examining
the influences of evidence, process and context. Health Econ. Policy Law;
9(2):119–141.
Cesarec, A., & Likić, R. (2017). Budget impact analysis of biosimilar trastuzumab for
the treatment of breast cancer in Croatia. Applied Health Economics and Health
Policy; 15(2):277–286.
Chabot, I., & Rocchi, A. (2010). How do cost-effectiveness analyses inform reim-
bursement decisions for oncology medicines in Canada? The example of suni-
tinib for first-line treatment of metastatic renal cell carcinoma. Value in Health;
13(6):837–845.
Chamberlain, C., Collin, S.M., Hounsome, L. et al. (2015). Equity of access to treat-
ment on the Cancer Drugs Fund: A missed opportunity for cancer research?
Journal of Cancer Policy; 5:25–30.
Chhatwal, J., He, T., & Lopez-Olivo, M.A. (2016). Systematic review of model-
ling approaches for the cost effectiveness of hepatitis C treatment with
direct-acting antivirals. Pharmacoeconomics; 34(6):551–67. doi: 10.1007/
s40273-015-0373-9.
Chiou, V.L., & Burotto, M. (2015). Pseudoprogression and immune-related response
in solid tumors. Journal of Clinical Oncology; 33(31):3541–3543.
Chouaid. (2012). Health-related quality of life in advanced non-small cell lung cancer
(NSCLC) patients. Value in Health; 15(4):A227.
References 367

Chouaid, C., Le Caer, H., Locher, C. et al. & GFPC 0504 Team. (2012). Cost effectivenes
of erlotinib versus chemotherapy for first-line treatment of non small cell lung
cancer (NSCLC) in fit elderly patients participating in a prospective phase 2
study (GFPC 0504). BMC Cancer; 12:301.
Ciani, O., Buyse, M., Drummond, M. et al. (2016). Use of surrogate end points in
healthcare policy: A proposal for adoption of a validation framework. Nature
Reviews Drug Discovery; 15(7):516.
Ciani, O., Buyse, M., Garside, R. et al. (2013). Comparison of treatment effect sizes
associated with surrogate and final patient relevant outcomes in randomised
controlled trials: Meta-epidemiological study. BMJ; 346:f457.
Clauser, S.B. (2004). Use of cancer performance measures in population health: A
macro-level perspective. Journal of the National Cancer Institute Monographs;
(33):142–154.
Claxton, K. (2008). Exploring uncertainty in cost-effectiveness analysis.
PharmacoEconomics; 26(9):781–798.
Claxton, K. (2011). Heterogeneity in cost-effectiveness of medical interventions: The
Cochrane Review 2008; challenges of matching patients to appropriate care.
14th Annual ISPOR Congress, 1–8 November 2011.
Claxton, K., Martin, S., Soares, M., (2015). Methods for the estimation of the NICE cost
effectiveness threshold. Health Technology Assessment; 19(14):1–503.
Claxton, K., Paulden, M., Gravelle, H. et al. (2011). Discounting and decision mak-
ing in the economic evaluation of health-care technologies. Health Economics;
20(1):2–15.
Clement, F.M., Ghali, W.A., Donaldson, C. et al. (2009). The impact of using differ-
ent costing methods on the results of an economic evaluation of cardiac care:
Microcosting vs. gross-costing approaches. Health Economics; 18(4):377–388.
Clement, F.M., Harris, A., Li, J.J. et al. (2009). Using effectiveness and cost-effective-
ness to make drug coverage decisions: A comparison of Britain, Australia, and
Canada. JAMA; 302(13):1437–1443.
Cohen, D. (2017). Most drugs paid for by £1.27bn Cancer Drugs Fund had no “mean-
ingful benefit”. BMJ; 357:j2097.
Collet, D. (2014). Modelling survival data, 3rd Edition. Chapman & Hall.
Comabella, C.C.I., Gibbons, E., & Fitzpatrick, R. (2010). A structured review of patient-
reported outcome measures (PROMs) for lung cancer. Oxford: University of Oxford.
Courtney, D., Huseyin, N., Evrim, G. et al. (2017). Availability of evidence of benefits
on overall survival and quality of life of cancer drugs approved by European
Medicines Agency: Retrospective cohort study of drug approvals 2009–13. BMJ;
359:j4530.
Coyle, D., & Coyle, K. (2014). The inherent bias from using partitioned survival mod-
els in economic evaluation. Value in Health; 17(3):A194.
Coyle, T. (2017). Sentinel system overview. Center for Drug Evaluation and Research.
United States Food and Drug Administration. Retrieved from: www.fda.gov/
downloads/ForPatients/About/UCM595420.pdf.
Cromwell, I., van der Hoek, K., Melosky, B. et al. (2011). Erlotinib or docetaxel for
second-line treatment of non-small cell lung cancer: A real-world cost-effec-
tiveness analysis. Journal of Thoracic Oncology; 6(12):2097–2103.
Cronin, A. (2016). STRMST2: Stata module to compare restricted mean survival
time. Statistical software components S458154. Boston, MA, : Boston College
Department of Economics.
368 References

Crott, R., & Briggs, A. (2010). Mapping the QLQ-C30 quality of life cancer ques-
tionnaire to EQ-5D patient preferences. European Journal of Health Economics;
11(4):427–434.
Crott, R., Versteegh, M., & Uyl-de-Groot, C. (2013). An assessment of the external
validity of mapping QLQ-C30 to EQ-5D preferences. Quality of Life Research;
22(5):1045–1054.
Crowther, L. (2016). Multi-state survival analysis in Stata, Stata UK Meeting 8th–
9th September 2016. Retrieved from: www.stata.com/meeting/uk16/slides/
crowther_uk16.pdf.
Cykert, S., Kissling, G., & Hansen, C.J. (2000). Patient preferences regarding possible
outcomes of lung resection: What outcomes should preoperative evaluations
target? Chest; 117(6):1551–1559.
Dakin, H., Devlin, N., Feng, Y. et al. (2015). The influence of cost‐effectiveness and
other factors on NICE decisions. Health Economics; 24(10):1256–1271.
Dakin, H., Devlin, N.J., Odeyemi, I.A. (2006). “Yes”, “No” or “Yes, but”? Multinomial
modelling of NICE decision-making. Health Policy; 77(3): 352–367.
Damm, K., Roeske, N., & Jacob, C. (2013). Health-related quality of life questionnaires
in lung cancer trials: A systematic literature review. Health Economics Review;
3(1):15.
Davis, C., Naci, H., Gurpinar, E., (2017). Availability of evidence of benefits on overall
survival and quality of life of cancer drugs approved by European Medicines
Agency: Retrospective cohort study of drug approvals 2009–13. BMJ; 359:j4530.
Day, S. (2002). Dictionary of Clinical Trials, First Ed. John Wiley.
de Bock, G.H., Putter, H., Bonnema, J. et al. (2009). The impact of loco-regional recur-
rences on metastatic progression in early-stage breast cancer: A multistate
model. Breast Cancer Research and Treatment; 117(2):401–408.
de Glas, N.A., Kiderlen, M., Vandenbroucke, J.P. et al. (2016). Performing survival
analyses in the presence of competing risks: A clinical example in older breast
cancer patients. Journal of the National Cancer Institute; 108(5):djv366.
de WreedeL.C., & Fiocco, P. (2010). The mistate package for estimation and prediction
in non- and semi-parametric multi-state and competing risks models. Computer
Methods and Programs in Biomedicine, 261–274.
de Wreede, L.C., Fiocco, M., & Putter, H. (2011). Mstate: An R package for the analysis of
competing risks and multi-state models. Journal of Statistical Software; 38(7):1–30.
Dehbi, H.M., Royston, P., & Hackshaw, A. (2017). Life expectancy difference and life
expectancy ratio: Two measures of treatment effects in randomised trials with
non-proportional hazards. BMJ; 357:j2250.
Del Paggio, J.C., Azariah, B., Sullivan, R. et al. (2017). Do contemporary randomized
controlled trials meet ESMO thresholds for meaningful clinical benefit? Annals
of Oncology; 28(1):157–162.
Denis, F., Lethrosne, C., Pourel, N. et al. (2017). Randomized trial comparing a web
mediated follow-up with routine surveillance in lung cancer patients. Journal of
the National Cancer Institute; 109(9).
Department of Health and Social Care. (n.d.). NHS prescription services. Retrieved
from: www.nhsbsa.nhs.uk/nhs-prescription-services.
Department of Health and Social Care. (n.d.). Publications by Department of Health
and Social Care. Retrieved from: www.gov.uk/government/publications?depa
rtments%5B%5D=department-of-health-and-social-care.
References 369

Department of Health and Social Care. (2011). Equity and excellence:


Liberating the NHS. Retrieved from: www.gov.uk/government/news/
equity-and-excellence-liberating-the-nhs.
DeVine, J., Norvell, D.C., Ecker, E. et al. (2011). Evaluating the correlation and respon-
siveness of patient-reported pain with function and quality-of-life outcomes
after spine surgery. Spine; 36(21 Suppl):S69–S74.
Devlin, N., & Parkin, D. (2004). Does NICE have a cost-effectiveness threshold and
what other factors influence its decisions? A binary choice analysis. Health
Economics; 13(5):437–452.
Dewar, R., & Khan, I. (2015). A new SAS macro for flexible parametric survival mod-
eling: Applications to clinical trials and surveillance data. Clinical Investigation
London; 5(12):00–00.
Díaz-Ordaz, K., Kenward, M.G., & Grieve, R. (2014). Handling missing values in cost-
effectiveness analyses that use data from cluster randomised trials. Journal of
the Royal Statistical Society: Series A; 177(2):457–474.
Doble, B., & Lorgelly, P. (2016). Mapping the EORTC QLQ-C30 onto the EQ-5D-3L:
Assessing the external validity of existing mapping algorithms. Quality of Life
Research; 25(4):891–911.
Dobrez, D., Cella, D., Pickard, A.S. et al. (2007). Estimation of patient preference-
based utility weights from the functional assessment of cancer therapy—gen-
eral. Value in Health; 10(4):266–272.
Dolan, P. (1997). Modelling valuations for EuroQol health states. Medical Care;
35(11):1095–1108.
Dobrez, D.G., Mathes, A, Amdahl, M., et al. (2004). Paricalcitol-treated patients expe-
rience improved hospitalization outcomes compared with calcitriol-treated
patients in real-world clinical settings. Nephrology Dialysis Transplantation; 19(5):
1174–1181.
Doyle, S., Lloyd, A., & Walker, M. (2008). Health state utility scores in advanced non-
small cell lung cancer. Lung Cancer; 62(3):374–380.
Drummond, M., & McGuire, A. (2001). Economic evaluation in health care : merging
theory with practice. Oxford University Press. Retrieved from https://books.
google.fr/books?hl=en&lr=&id=_fJK15F75-EC&oi=fnd&pg=PP2&dq=drumm
ond+McGuire&ots=JUaIBpVA-F&sig=rNq0WR4nVWcvI0q8d5ufL4s9z8s#v=on
epage&q=drummond McGuire&f=false.
Drummond, M.F., & McGuire, A. (2002). Economic evaluation in health care: Merging
theory with practice. Oxford: Oxford University Press.
Drummond, M.F., & O’Brien, B. (1993). Clinical importance, statistical significance
and the assessment of economic and quality-of-life outcomes. Health Economics;
2(3):205–212.
Drummond, M.F., Sculpher, M.J., Torrance, G.W. et al. (2005). Methods for the economic
evaluation of health care programmes, 3rd Edition. Oxford: Oxford University
Press.
Drummond, M.F., Sculpher, M.J., Torrance, G.W. et al. (2015). Methods for the economic
evaluation of health care programmes, 4th Edition. Oxford University Press.
Dukes, O., Vansteelandt, S. (2018). A note of G-estimation of causal risk ratios.
American Journal of Epidemiology; 187(5): 1079–1084.
Duan, N. (1983). Smearing estimate: A nonparametric retransformation method.
Journal of the American Statistical Association; 78(383):605–610.
370 References

Dukhovny, D., Lorch, S.A., Schmidt, B. et al. & Caffeine for Apnea of Prematurity
Trial Group. (2011). Economic evaluation of caffeine for apnea of prematurity.
Pediatrics; 127(1):e146–e155.
Dunlop, W., Iqbal, I., Khan, I. et al. (2013). Cost-effectiveness of modified-release
prednisone in the treatment of moderate to severe rheumatoid arthritis
with morning stiffness based on directly elicited public preference values.
ClinicoEconomics and Outcomes Research; 5:555–564.
Dukhobny, D., Lorch, S.A., Schmidt, B. (2012). Economic evaluation of caffeine for
apnea of prematurity; caffeine for apnea of prematurity trial group. Pediatrics;
127(1): e146–155.
Dunlop, W., Uhl, R., Khan, I. et al. (2012). Quality of life benefits and cost impact of
prolonged release oxycodone/naloxone versus prolonged release oxycodone in
patients with moderate-to-severe non-malignant pain and opioid-induced con-
stipation: A UK cost-utility analysis. Journal of Medical Economics; 15(3):564–575.
Dunn, A., Grosse, S.D., & Zuvekas, S.H. (2018). Adjusting health expenditures for
inflation: A review of measures for health services research in the United
States. Health Services Research; 53(1):175–196.
Dvortsin, E., Gout-Zwart, J., Eijssen, E. L. M., Van Brussel, J., & Postma, M. J. (2016).
Comparative cost-effectiveness of drugs in early versus late stages of cancer;
Review of the literature and a case study in breast cancer. PLoS ONE. https://
doi.org/10.1371/journal.pone.0146551
Eddy, D.M., Hollingworth, W., Caro, J.J. et al., & ISPOR-SMDM Modeling Good
Research Practices Task Force. (2012). Model transparency and validation: A
report of the ISPOR-SMDM Modeling Good Research Practices Task Force-7.
Medical Decision Making; 32(5):733–743.
Eisenhauer, E.A., Therasse, P., Bogaerts, J. et al. (2009). New response evaluation cri-
teria in solid tumours: Revised recist guideline (Version 1.1). European Journal of
Cancer; 45(1):228–247.
Ekelund, R.B. Jr., & Hébert, R.F. (1999). Secret origins of modern microeconomics: Dupuit
and the engineers. Chicago: University of Chicago Press.
Elbasha, E.H., & Chhatwal, J. (2016). Myths and misconceptions of within-cycle correc-
tion: A guide for modelers and decision makers. Pharmacoeconomics; 34(1):13–22.
EMA (2012). Methodological consideration for using progression-free survival (PFS)
or disease-free survival (DFS) in confirmatory trials; https://www.ema.europa.
eu/en/appendix-1-guideline-evaluation-anticancer-medicinal-products-man-
methodological-consideration-using.
Etzioni, R.D., Feuer, E.J., Sullivan, S.D. et al. (1999). On the use of survival analysis tech-
niques to estimate medical care costs. Journal of Health Economics; 18(3):365–380.
EUnetHTA (2018). European Network for Health Technology Assessment. Retrieved
from: www.eunethta.eu.
European Medicines Agency. (2017). ICH E9 (R1) addendum on estimates and sen-
sitivity analysis in clinical trials to the guideline on statistical principles for
clinical trials. Retrieved from: www.ema.europa.eu/documents/scientific-
guideline/draft-ich-e9-r1-addendum-estimands-sensitivity-analysis-clinical-
trials-guideline-statistical_en.pdf.
European Organisation for Research and Treatment of Cancer. (n.d.). Manuals.
EORCT Quality of Life. Retrieved from: http://groups.eortc.be/qol/manuals.
European Society for Medical Oncology. (2015). ESMO Press release: ESMO
announces a scale to stratify the magnitude of clinical benefit of anti-cancer
References 371

medicines. Retrieved from: www.esmo.org/Press-Office/Press-Releases/


ESMO-Announces-a-Scale-to-Stratify-the-Magnitude-of-Clinical-Benefit-of-
Anti-Cancer-Medicines.
European Union. (n.d.). The EU general data protection regulation. Retrieved from:
https://eugdpr.org/.
Evidence Review Group. (2008). The clinical- and cost-effectiveness of lenalido-
mide for multiple myeloma in people who have received at least one prior
therapy: An evidence review of the submission from celgene. PenTAG on
behalf of NICE. Retrieved from: www.nice.org.uk/guidance/ta171/resources/
multiple-myeloma-lenalidomide-evidence-review-group-report2
Fabrini, M.G., Silvano, G., Lolli, I. et al. (2009). A multi-institutional phase II study
on second-line fotemustine chemotherapy in recurrent glioblastoma. Journal of
Neuro-Oncology; 92(1):79–86.
FACIT. (n.d.). Questionnaires. FACIT measurement system. Retrieved from: www.
facit.org/facitorg/questionnaires.
Fairclough, L.D. (2010). Design and analysis of quality of life studies in clinical trials, 2nd
Edition. London: Chapman & Hall/CRC Interdisciplinary Statistics.
Fan, M.Y., & Zhou, X.H. (2007). A simulation study to compare methods for con-
structing confidence intervals for the incremental cost-effectiveness ratio.
Health Services and Outcomes Research Methodology; 7(1–2):57–77.
Fan, X. Sivo, S., Keenan, S. (2002). SAS for Monte Carlo Studies: A guide for Quantitative
Researchers. Cary, NC: SAS Institute.
Faria, R., Gomes, M., Epstein, D. et al. (2014). A Guide to handling missing data in
cost-effectiveness analysis conducted within randomised controlled trials.
PharmacoEconomics; 32(12):1157–1170.
Faries, D.E., Leon, A.C., Haro, J.M. et al. (2010). Analsis of observational health care data
using SAS. SAS Publications.
Fayers, P.M., Aaronson, N.K., Bjordal, K. et al. (2001). The EORTC QLQ-C30 scor-
ing manual, 3rd Edition. Brussels: European Organisation for Research and
Treatment of Cancer.
FDA (2018). Guidance to Industry. Retrieved from https://www.fda.gov/downloads/
Drugs/Guidances/ucm071590.pdf.
Federal Ministry of Health (Germany). (n.d.). German System of Healthcare.
Retrieved from: www.bundesgesundheitsministerium.de/
Fenwick, E., Claxton, K., & Sculpher, M. (2008). The value of implementation and
the value of information: Combined and uneven development. Medical Decision
Making; 28(1):21–32.
Ferlay, J., Shin, H.R., Bray, F. et al. (2010). Estimates of worldwide burden of cancer in
2008: GLOBOCAN 2008. International Journal of Cancer; 127(12):2893–2917.
Ferver, K., Burton, B., & Jesilow, P. (2009).The use of claims data in healthcare research.
Open Public Health Journal; 2(1):11–24.
Fiteni, F., Westeel, V., Pivot, X. et al. (2014). Endpoints in cancer clinical trials. Journal
Visceral Surgeon; 151(1):17–22.
Fleishman, A.I. (1978). A method for simulating non-normal distributions.
Psychometrika; 43(4):521–532.
Fojo, T., Mailankody, S., & Lo, A. (2014). Unintended consequences of expensive
cancer therapeutics—The pursuit of marginal indications and a me-too men-
tality that stifles innovation and creativity: The John Conley Lecture. JAMA
Otolaryngology Head and Neck Surgery; 140(12):1225–1236.
372 References

Folland, S., Goodman, A.C., & Stano, M. (2012). The economics of health and health care,
7th Edition. Boston, MA.Prentice Hall.
Food and Drug Administration. (2017) Sentinel System Overview. Retrieved from:
https://www.fda.gov/downloads/ForPatients/About/UCM595420.pdf.
Food and Drug Administration. (2018). Information Exchange and Data
Transformation (INFORMED). Retrieved from: https://www.fda.gov/aboutfda/
centersoffices/officeofmedicalproductsandtobacco/oce/ucm543768.htm.
Fragoulakis, V., Pallis, A., & Georgoulias, M. (2014). Economic evaluation of peme-
trexed versus erlotinib as second-line treatment of patients with advanced/
metastatic non-small cell lung cancer in Greece: A cost minimization analysis.
Lung Cancer: Targets and Therapy;21(7) 43–51.
Friedman, H.S., Prados, M.D., Wen, P.Y. et al. (2009). Bevacizumab alone and in com-
bination with irinotecan in recurrent glioblastoma. Journal of Clinical Oncology;
27(28):4733–4740.
Fukuoka, M., Wu, Y.L., Thongprasert, S. et al. (2011). Biomarker analyses and final
overall survival results from a phase III, randomized, open-label, first-line
study of gefitinib versus carboplatin/paclitaxel in clinically selected patients
with advanced non-small-cell lung cancer in Asia (IPASS). Journal of Clinical
Oncology; 29(21):2866–2874.
Fundamental Finance. (n.d.). Marginal Cost (MC) & Average Total Cost (ATC).
Retrieved from: http://economics.fundamentalfinance.com/micro_atc_mc.php.
Gaafar, R.M., Surmont, V.F., Van Klaveren, R.J., et al. (2011). A double-blind, ran-
domised, placebo-controlled phase III intergroup study of gefitinib in patients
with advanced NSCLC, non-progressing after first line platinum-based chemo-
therapy (EORTC 08021/ILCP 01/03). Eur. J Cancer; 47(15): 2331–2340.
Gansen, F.M. (2018). Health economic evaluations based on routine data in Germany:
A systematic review. BMC Health Services Research; 18(1):268.
Garber, A.M., & Phelps, C.E. (1997). Economic foundations of cost-effectiveness anal-
ysis. Journal of Health Economics; 16(1):1–31.
Gardiner, J.C. (2010). Survival Analysis: Overview of parametric, nonparametric
and semiparametric approaches and new developments, Paper 252-2010, SAS
Global Forum 2010, Seattle, WA, April 11–14. Retrieved from: https://support.
sas.com/resources/papers/proceedings10/252-2010.pdf.
Gazdar, A.F. (2009). Activating and resistance mutations of EGFR in non-small-
cell lung cancer: Role in clinical response to EGFR tyrosine kinase inhibitors.
Oncogene; 28(Suppl 1):S24–S31.
Gelber, R.D., Goldhirsch, A., Cole, B.F. et al. (1996). A quality-adjusted time without
symptoms or toxicity (Q-TWiST) analysis of adjuvant radiation therapy and
chemotherapy for resectable rectal cancer. Journal of the National Cancer Institute;
88(15):1039–1045.
Gerard, K., Ryan, M., & Amaya-Amaya, M. (2008). Introduction. In: M. Ryan, K.
Gerard, & M. Amaya-Amaya (Eds.). Using discrete choice experiments to value
health and health care. Dordrecht, Netherlands: Springer, 1–10.
Gerkens, S., Neyt, M., San Miguel, L. et al. (2017). KCE report 288. Health Service
Research: How to improve the Belgian process for Managed Entry Agreements?
An analysis of the Belgian and international experience. Brussels: Belgian
Health Care Knowledge Centre (KCE). see: https://kce.fgov.be/sites/default/
files/atoms/files/KCE_288_Improve_Belgian_process_managed_entry_agree-
ments_Report.pdf
References 373

Gheorghe, A., Roberts, T., Hemming, K. et al. (2015). Evaluating the generalisabil-
ity of trial results: Introducing a centre- and trial-level generalisability index.
Pharmacoeconomics; 33(11):1195–1214.
Gibbons, E.C., Casañas i Comabella, C., & Fitzpatrick, R. (2013). A structured review
of patient‐reported outcome measures for patients with skin cancer, 2013. British
Journal of Dermatology (2013); 168(6):1176–1186.
Gilbert, M.R., Dignam, J.J., Armstrong, T.S. et al. (2014). A randomized trial of beva-
cizumab for newly diagnosed glioblastoma. New England Journal of Medicine;
370(8):699–708.
Gilbert, M.R., Wang, M., Aldape, K.D. et al. (2013). Dose-dense temozolomide for
newly diagnosed glioblastoma: A randomized phase III clinical trial. Journal of
Clinical Oncology; 31(32):4085–4091.
Gilberto de Lima, L.G., Segel, J., Tan, D. et al. (2011). Cost-effectiveness of epider-
mal growth factor receptor mutation testing and first-line treatment with
gefitinib for patients with advanced adenocarcinoma of the lung. Cancer;
118(4):1032–1039.
Glasziou, P.P., Simes, R.J., & Gelber, R.D. (1990). Quality adjusted survival analysis.
Statistics in Medicine; 9(11):1259–1276.
Glick, H.A. (2011). Sample size and power for cost-effectiveness analysis (Part 1).
Pharmacoeconomics; 29(3):189–198.
Glick, H.A., Doshi, J.A., Sonnad, S.S., & Polsky, D. (2014). Economic evaluation in clinical
trials. Oxford University Press.
Goldberg, S.B., Supko, J.G., Neal, J.W. et al. (2012). A phase I study of erlotinib and
hydroxychloroquine in advanced non-small-cell lung cancer. Journal of Thoracic
Oncology; 7(10):1602–1608.
Goldhirsch, A., Gelber, R.D., Simes, R.J. et al. (1989). Costs and benefits of adjuvant
therapy in breast cancer: A quality-adjusted survival analysis. Journal of Clinical
Oncology; 7(1):36–44.
Gonçalves, F.R., Santos, S., Silva, C. et al. (2018). Risk-sharing agreements, present and
future. Ecancermedicalscience; 12:823.
Goodwin, P.J., Black, J.T., Bordeleau, L.J. et al. (2003). Health-related quality-of-life
measurement in randomized clinical trials in breast cancer—Taking stock.
Journal of the National Cancer Institute; 95(4):263–281.
Goozner, M. (2012). Drug approvals 2011: Focus on companion diagnostics. Journal of
the National Cancer Institute; 104(2):84–86.
Goulart, B., & Ramsey, S. (2011). A trial-based assessment of the cost-utility of bevaci-
zumab and chemotherapy versus chemotherapy alone for advanced non-small
cell lung cancer. Value in Health; 14(6):836–845.
Government of UK. Medicines and Healthcare products Regulatory Agency. (2014).
Apply for the early access to medicines scheme (EAMS). Retrieved from: www.
gov.uk/guidance/apply-for-the-early-access-to-medicines-scheme-eams.
Graham, C.N., Hechmati, G., & Hjelmgren, J. (2014). Cost-effectiveness analysis of
panitumumab plus mFOLFOX6 compared with bevacizumab plus mFOLFOX6
for first-line treatment of patients with wild-type RAS metastatic colorectal
cancer. Journal of Cancer; 50(16):2791–2801.
Gray, A.M., Clarke, P.M., Wolstenholme, J.L. et al. (2011). Applied methods of cost-effec-
tiveness in health care. Oxford: Oxford University Press.
Green Park Collaborative. (2016). Retrieved from: http://www.cmtpnet.org/resource-
center/view/new-guidance-for-treatment-switching-in-oncology-drug-trials/.
374 References

Greenhalgh, J., McLeod, C., Bagust, A. et al. (2010). Pemetrexed for the maintenance
treatment of locally advanced or metastatic non-small cell lung cancer. Health
Technology Assessment; 14(Suppl 2):33–39.
Greenland, S., Lanes, S., & Jara, M. (2008). Estimating effects from randomized trials
with discontinuations: The need for intent-to-treat design and G-estimation.
Clinical Trials; 5(1):5–13.
Gridelli, C., Ciardiello, F., Gallo, C. et al. (2012). First-line erlotinib followed by second-
line cisplatin-gemcitabine chemotherapy in advanced non-small-cell lung can-
cer: The TORCH randomized trial. Journal of Clinical Oncology; 30(24):3002–3011.
Grieve, R., Nixon, R., Thompson, S.G. et al. (2005). Using multilevel models for assess-
ing the variability of multinational resource use and cost data.Health Economics;
14(2):185–196.
Grieve, R., Nixon, R., Thompson, S.G. et al. (2007). Multilevel models for estimat-
ing incremental net benefits in multinational studies. Health Economics;
16(8):815–826.
Grieve, R., Nixon, R., & Thompson, S.G. (2014). Bayesian hierarchical models for cost-
effectiveness analyses that use data from cluster randomized trials; healthcare:
A review of principles and applications. Journal of Medical Economics; 20(2):
163‒175.
Guo, S., & Fraser, M.E. (2014). Propensity score analysis: Statistical methods and applica-
tions. Sage Publications.
Hall, A.E., & Highfill, T. (2013). Calculating disease-based medical care expenditure
indexes for Medicare beneficiaries: A comparison of method and data choices.
nber.org.
Hao, Y., Wolfram, V., & Cook, J.S. (2016). A structured review of health utility mea-
sures and elicitation in advanced/metastatic breast cancer. ClinicoEconomics and
outcomes research.
Harris, A.H., Hill, S.R., Chin, G., et al. (2008). The role of value for money in public
insurance coverage decisions for drugs in Australia: a retrospective analysis
1994-2004. Decis. Making; 28(5): 713–722.
Hashim, D., Boffetta, P., La Vecchia, C. et al. (2016). The global decrease in cancer
mortality: trends and disparitiesAnnals of Oncology; 27(5):926–933.
Haycox, A., Drummond, M., & Walley, T. (1997). Pharmacoeconomics: Integrating
economic evaluation into clinical trials. British Journal of Clinical Pharmacology;
43(6):559–562.
Henry, J., & Kaise Family Foundation. (2018). Public opinion on prescription drugs
and their prices. KFF health tracking poll. Retrieved from: www.kff.org/
slideshow/public-opinion-on-prescription-drugs-and-their-prices/.
Herbst, R.S., Prager, D., Hermann, R.L. et al. (2005). Tribute: A phase III trial of erlo-
tinib hydrochloride (OSI-774) combined with carboplatin and paclitaxel cho-
metherapy in advanced non-small-cell lung cancer. Journal of Clinical Oncology;
23(25):5892–5899. Epub 2005 July 25.
Herdman, M., Gudex, C., Lloyd, A. et al. (2011). Development and preliminary test-
ing of the new five-level version of EQ-5D (EQ-5D-5L). Quality of Life Research;
20(10):1727–1736. Epub 2011 April 9.
Hernández, M.A., Vázquez-Polo, F.J., González-Torre, F.J. et al. (2009). Complementing
the net benefit approach: A new framework for Bayesian cost-effective-
ness analysis. International Journal of Technology Assessment in Health Care;
25(4):537–545.
References 375

Herrlinger, U., Schäfer, N., Steinbach, J.P. et al. (2016). Bevacizumab plus irinotecan
versus temozolomide in newly diagnosed O6-Methylguanine-DNA methyl-
transferase nonmethylated glioblastoma: The randomized GLARIUS trial.
Journal of Clinical Oncology; 34(14):1611–1619.
Hettle, R., Posnett, J., & Borrill, J. (2015). Challenges in economic modeling of antican-
cer therapies: An example of modeling the survival benefit of olaparib main-
tenance therapy for patients with BRCA-mutated platinum-sensitive relapsed
ovarian cancer. Journal of Medical Economics; 18(7):516–524.
Hlatky, M.A., Boothroyd, D.B., & Johnstone, I.M. (2002). Economic evaluation in long-
term clinical trials. Statistics in Medicine; 21(19):2879–2888.
Hoaglin, D.C., Hawkins, N., Jansen, J.P. et al. (2011). Conducting indirect-treatment-
comparison and network-meta-analysis studies: Report of the ISPOR Task
Force on indirect treatment comparisons good research practices: Part 2. Value
in Health; 14(4):429–437.
Hoch, J.S., & Dewa, C.S. (2014). Advantages of the net benefit regression framework
for economic evaluations of interventions in the workplace: A case study of
the cost-effectiveness of a collaborative mental health care program for peo-
ple receiving short-term disability benefits for psychiatric disorders. Journal of
Occupational and Environmental Medicine; 56(4):441–445.
Hodi, F.S., Ribas, A., Daud, A. et al. (2014). Evaluation of immune-related response
criteria (irRC) in patients (pts) with advanced melanoma (MEL) treated with the
anti-PD-1 monoclonal antibody MK-3475. Journal of Clinical Oncology; 32(suppl
15s; abstr3006):10.
Hoefman, R.J., van Exel, J., & Brouwer, W. (2013). How to include informal care in
economic evaluations. Pharmacoeconomics; 31(12):1105–1119.
Hollen, P.J., Gralla, R.J., Cox, C. et al. (1997). A dilemma in analysis: Issues in the serial
measurement of quality of life in patients with advanced lung cancer. Lung
Cancer; 18(2):119–136.
Holmes, J., Dunlop, D., Hemmett, L. et al. (2004). A cost-effectiveness analy-
sis of docetaxel in the second-line treatment of non-small cell lung cancer.
Pharmacoeconomics; 22(9):581–589.
Howard, D.H., Bach, P.B., Berndt, E.R. et al. (2015.) Pricing in the market for anticancer
drugs. Journal of Economic Perspectives; 29(1):139–162.
Huang, Y. (2009). Cost analysis with censored data. Medical Care; 47(7 Suppl 1):
S115–S119.
Hughes, D.A., Bagust, A., Haycox, A. et al. (2001). The impact of non-compliance on
the cost-effectiveness of pharmaceuticals: A review of the literature. Health
Economics; 10(7):601–615.
Hurst, N.P. (1997). Re: Quality of life measures. Rheumatology; 36(1):147–148.
Husereau, D., Drummond, M.F., Petrou, S. et al. (2013). Consolidated health economic
evaluation reporting standards (CHEERS)—Explanation and elaboration: A
report of the ISPOR health economic evaluations publication guidelines good
reporting practices task force. Value in Health; 16(2):231–250.
Husson, O., Steenbergen, L.N., Koldewijn, E.L. et al. (2014). Excess mortality in
patients with prostate cancer. BJU International; 114:691–697.
Hutchinson, C.L., Menck, H.R., Burch, M. et al. (2004). Cancer Registry Management
Principles & Practice, 2nd Edition. Kendall & Hunt Publishing.
Institute for Clinical and Economic Review. (n.d.). Methodology. Retrieved from:
https://icer-review.org/methodology/.
376 References

Institute for Quality and Efficiency in Healthcare. (n.d.). Retrieved from: www.iqwig.
de/en/home.2724.html.
Irwin, J.O. (1949). The standard error of an estimate of expectation of life, with special
reference to expectation of tumourless life in experiments with mice. Journal of
Hygiene; 47(2):188.
Ishak, K.J., Proskorovsky, I., Korytowsky, B. et al. (2014). Methods for adjusting for
bias due to crossover in oncology trials. Pharmacoeconomics; 32(6):533–546.
Janne, P.A., Ramalingam, S.S., Yang, J.C. et al. (2014). Clinical activity of the mutant-
selective EGFR inhibitor AZD9291 in patients (pts) with EGFR inhibitor–resis-
tant non-small cell lung cancer (NSCLC). [ASCO abstract 8009]. Journal of
Clinical Oncology; 32(5_suppl): 8009.
Jansen, J.P., Fleurence, R., Devine, B. et al. (2011). Interpreting indirect treatment com-
parisons and network meta-analysis for health-care decision making: Report
of the ISPOR task force on indirect treatment comparisons good research prac-
tices: Part 1. Value Health; 14(4):417–428.
Jemal, A., Bray, F., Center, M.M. et al. (2011). Global cancer statistics. CA: A Cancer
Journal for Clinicians; 61(2):69–90.
Jiaqi, L. (2016). Modeling approaches for cost and cost-effectiveness estimation using
observational data. Publicly accessible Penn dissertations. 1858. Retrieved
from: http://repository.upenn.edu/edissertations/1858.
Jones, B., & Kenward, M.G. (2014). Design and analysis of cross-over trials, 3rd Edition.
Chapman and Hall/CRC.
Jönsson, B., Ramsey, S., & Wilking, N. (2014). Cost effectiveness in practice and its
effect on clinical outcomes. Journal of Cancer Policy; 2(1):12–21.
Jönsson, L., Sandin, R., Ekman, M. et al. (2014). Analyzing overall survival in ran-
domized controlled trials with crossover and implications for economic evalu-
ation. Value in Health; 17(6):707–713.
Kaplan, E.L., & Meier, P. (1958). Nonparametric estimation from incomplete observa-
tions. Journal of the American Statistical Association; 53(282):457–481.
Karrison, T. (1987). Restricted mean life with adjustment for covariates. Journal of the
American Statistical Association; 82(400):1169–1176.
Katakami, N., Atagi, S., Goto, K. et al. (2013). LUX-Lung 4: A phase II trial of afatinib
in patients with advanced non-small-cell lung cancer who progressed during
prior treatment with erlotinib, gefitinib, or both. Journal of Clinical Oncology;
31(27):3335–3341.
Keeble, G.R.L., Barber, S., & Baxter, P.D. (2015). Choosing a method to reduce selection
bias: A tool for researchers. Open Journal of Epidemiology; 5:155–162.
Kenneth, C., Park, B.S., Schwimmer, J.E. et al. (2001). Decision analysis for the
cost-effective management of recurrent colorectal cancer. Annals of Surgery;
233(3):310–319.
Khan, I. (2015). Design & analysis of clinical trials for economic evaluation & reimburse-
ment: An applied approach using SAS & STATA. Chapman & Hall.
Khan, I. (2019). Modelling and extrapolating post progression utility in NSCLC
patients; a modelling approach (Medical Decision Making), [in press].
Khan, I., Bashir, Z., & Forster, M. (2015). Interpreting small treatment differences from
quality of life data in cancer trials: An alternative measure of treatment benefit and
effect size for the EORTC-QLQ-C30. Health and Quality of Life Outcomes; 13(1):180.
Khan, I., Morris, S., Pashayan, N. et al. (2016). Comparing the mapping between
EQ-5D-5L, EQ-5D-3L and the EORTC-QLQ-C30 in non-small cell lung cancer
patient. Health and Quality of Life Outcomes; 14:60.
References 377

Khan, I., Sarker, S.J., & Hackshaw, A. (2012). Smaller sample sizes for phase II trials
based on exact tests with actual error rates by trading-off their nominal levels
of significance and power. British Journal of Cancer; 107(11):1801–1809.
Khan, I., Stavros, P., Khan, K. et al. (2018). Does exercise improve cognitive impairment in
people with mild to moderate dementia? A cost-effectiveness analysis from a con-
firmatory randomised controlled trial (DAPA Trial). PharmacoEconomics; [in Press].
Khozin, S., Blumenthal, G.M., & Pazdur, R. (2017). Real-world data for clinical evidence
generation in oncology. JNCI: Journal of the National Cancer Institute; 109(11).
Kim, C., & Prasad, V. (2015). Cancer drugs approved on the basis of a surrogate end
point and subsequent overall survival: An analysis of 5 years of US Food and
Drug Administration approvals. JAMA Internal Medicine; 175(12):1992–1994.
Kim, C., & Prasad, V. (2016). Strength of validation for surrogate end points used
in the US Food and Drug Administration’s approval of oncology drugs. Mayo
Clinic Proceedings; 91(6):713–725.
King, M.T. (1996). The interpretation of scores from the EORTC quality of life ques-
tionnaire QLQ-C30. Quality of Life Research; 5(6):555–567.
Kish, J.K., Ward, M.A., Garofalo, D. et al. (2018). Real-world evidence analysis of pal-
bociclib prescribing patterns for patients with advanced/ metastatic breast can-
cer treated in community oncology practice in the USA one year post approval.
Breast Cancer Research; 20(3):1–8.
Klaxton, C. (1999). The irrelevance of inference: A decision-making approach to the
stochastic evaluation of health care technologies. Journal of Health Economics;
18(3):341–364.
Klein, J.P., & Moeschberger, M. (2005). Survival analysis: Techniques for censored and
truncated data, 2nd Edition. New York: Springer.
Klein, J.P., Gerster, M., Andersen, P.K. et al. (2008). SAS and R functions to compute
pseudo-values for censored data regression. Computer Methods and Programs in
Biomedicine; 89(3):289–300.
Klein, R., Muehlenbein, C., Liepa, A.M. et al. (2009). Cost-effectiveness of pemetrexed
plus cisplatin as first-line therapy for advanced nonsquamous non-small cell
lung cancer. Journal of Thoracic Oncology; 4(11):1404–1414.
Kong, D.S., Lee, J.I., Kim, J.H. et al. (2010). Phase II trial of low-dose continuous
(metronomic) treatment of temozolomide for recurrent glioblastoma. Neuro-
Oncology; 12(3):289–296.
Konnerup, M. & Kongsted, H.C., (2012). Are more observational studies being
included in Cochrane Reviews? BMC Research Notes; 5:570.
Krahn, M., Bremner, K.E., Tomlinson, G. et al. (2007). Responsiveness of disease-spe-
cific and generic utility instruments in prostate cancer patients. Quality of Life
Research; 16(3):509–522.
Kruger, C.J.C. (2004). Constrained cubic spline interpolation for chemical engineer-
ing applications. Retrieved from: www.korf.co.uk/spline.pdf.
Kumar, H., Fojo, T., & Mailankody, S. (2016). An appraisal of clinically meaningful
outcomes guidelines for oncology clinical trials. JAMA Oncology; 2(9):1238–1240.
Kuntz, K.M., & Weinstein, M.C. (2001). Modeling in economic evaluation. In: M.
Drummond, & A. McGuire (Eds.). Economic evaluation in health care: Merging
theory with practice. New York: Oxford University Press, 141–171.
Kunz, R., Vist, G., & Oxman, A.D. (2007). Randomisation to protect against selection
bias in healthcare trials. Cochrane Database of Systematic Reviews; 2(2):MR000012.
Lai, X., & Zee, B.C.Y. (2015). Mixed response and time-to-event endpoints for multi-
stage single-arm phase II design. Trials; 16:250.
378 References

Landrum, M.B., & Ayanian, J.Z. (2001). Causal effect of ambulatory specialty care
on mortality following myocardial infarction: A comparison of propensity
score and instrumental variable analyses. Health Services and Outcomes Research
Methodology; 2(3–4):221–245.
Latimer, N.R., Siebert, U., Henshall, C. et al. Treatment switching: Statistical and
decision-making challenges and approaches. International Journal of Technology
Assessment in Health Care; 32(2): 160–166.
Latimer, N.R. (2011). NICE DSU Technical Support Document 14: Survival analysis
for economic evaluations alongside clinical trials – Extrapolation with patient-
level data. Report by the Decision Support Unit, June 2011 (last updated March
2013). Retrieved from: http://nicedsu.org.uk/wp-content/uploads/2016/03/
NICE-DSU-TSD-Survival-analysis.updated-March-2013.v2.pdf.
Latimer, N.R. (2015) Treatment switching in oncology trials and the acceptability of
adjustment methods. Expert Review of Pharmacoeconomics & Outcomes Research;
15(4): 561–564.
Latimer, N.R., & Abrams, K.R. (2014). NICE DSU Technical Support Document 16:
Adjusting survival time estimates in the presence of treatment switching.
Retrieved from: http://nicedsu.org.uk/wp-content/uploads/2016/03/TSD16_
Treatment_Switching.pdf.
Latimer, N.R., Abrams, K.R., Lambert, P.C. et al. (2017). Adjusting for treatment
switching in randomised controlled trials – A simulation study and a simpli-
fied two-stage method. Statistical Methods in Medical Research; 26(2):724–751.
Latimer, N.R., White, I.R., Abrams, K.R. et al. (2018). Causal inference for long-term
survival in randomised trials with treatment switching: Should re-censoring
be applied when estimating counterfactual survival times? Statistical Methods
in Medical Research. Retrieved from: www.sheffield.ac.uk/scharr/sections/
heds/staff/latimer_n_publications.
Ledermann, J.A., Embleton, A.C., Perren, T. et al. (2017). Overall survival results of
ICON6: A trial of chemotherapy and cediranib in relapsed ovarian cancer.
Journal of Clinical Oncology; 35(15_suppl):5506–5506. Retrieved from: www.
icon6.org/media/1390/asco_2017.pdf.
Lee, C.F., Luo, N., Ng, R. et al. (2013). Comparison of the measurement properties
between a short and generic instrument, the 5-level EuroQoL Group’s 5-dimen-
sion (EQ-5D-5L) questionnaire, and a longer and disease-specific instrument,
the Functional Assessment of Cancer Therapy—Breast (FACT-B), in Asian
breast cancer patients. Quality of Life Research; 22(7):1745–1751.
Lee, L., Wang, L., & Crump, M. (2011). Identification of potential surrogate endpoints
in randomized clinical trials of aggressive and indolent non-Hodgkin’s lym-
phoma: Correlation of complete response, time-to-even and overall survival
endpoints. Annals of Oncology; 22(6):1392–1403.
Lee, S.M., Khan, I., Upadhyay, S. et al. (2012). First-line erlotinib in patients
with advanced non-small-cell lung cancer unsuitable for chemotherapy
(TOPICAL): A double-blind, placebo-controlled, phase 3 trial. Lancet Oncology;
13(11):1161–1170.
Lewis, G., Peake, M., Aultman, R. et al. (2010). Cost-effectiveness of erlotinib versus
docetaxel for second-Line treatment of advanced non-small-cell lung cancer in
the United Kingdom. Journal of International Medical Research; 38(1):9–21.
Li, H., Han, D., Hou, Y. et al. (2015). Statistical interference methods for two crossing
survival curves: A comparison of methods. PLoS One; 10(1):1–18.
References 379

Lin, D.Y. (2000). Proportional means regression for censored medical costs. Biometrics;
56(3):775–778.
Lin, D.Y., Feuer, E.J., Etzioni, R. et al. (1997). Estimating medical costs from incomplete
follow-up data. Biometrics; 53(2):419–434.
Linley, W.G., Hughes, D.A. (2012). Reimbursement decisions of the All Wales
Medicines Strategy Group: influence of policy and clinical and economic fac-
tors. Pharmacoeconomics; 30(9): 779–794.
Littlejohns, P., & Rawlins, M. (2009). Patients, the public and priorities in healthcare, 1st
Edition. CRC Press.
Long, E.R. (1928). A history of pathology. London: Baillière, Tindall & Cox.
Lorgelly, P.K., Doble,B., Rowen, D. et al., & Cancer 2015 Investigators. (2017). Condition-
specific or generic preference-based measures in oncology? A comparison of
the EORTC-8D and the EQ-5D-3L. Quality of Life Research; 26(5):1163–1176.
Maeso, S., Callejo, D., Hernández, R. et al. (2011). Esophageal Doppler monitoring
during colorectal resection offers cost-effective improvement of hemodynamic
control. Value in Health; 14(6):818–826.
Maguire, J., Khan, I., McMenemin, R. et al. (2014). SOCCAR: A randomised phase II
trial comparing sequential versus concurrent chemotherapy and radical hypo-
fractionated radiotherapy in patients with inoperable stage III non-small cell lung
cancer and good performance status. European Journal of Cancer; 50(17):2939–2949.
Maisonneuve, H., & Floret, D. (2012). Wakefield’s affair: 12 years of uncertainty
whereas no link between autism and mmr vaccine has been proved. Presse
Medicale; 41(9 Pt 1):827–834.
Maiwenn, J., & Van Hout, B. (2000). A Bayesian approach to economic analyses of
clinical trials: The case of stenting versus balloon angioplasty. Health Economics;
9(7):599–609.
Malkin, A.G., Goldstein, J.E., Perlmutter, M.S. et al. (2013). Responsiveness of the
EQ-5D to the effects of low vision rehabilitation. Optometry and Vision Science;
90(8):799–805.
Manca, A., Rice, N., Sculpher, M.J. et al. (2005). Assessing generalisability by location
in trial-based cost-effectiveness analysis: The use of multilevel models. Health
Economics; 14(5):471–485; Erratum in: Health Economics; 14(5):486.
Maniadakis, N., Fragoulakis, V., Pallis, A.G. et al. (2010). Economic evaluation of
docetaxel-gemcitabine versus vinorelbine-cisplatin combination as front-line
treatment of patients with advanced/metastatic non-small-cell lung cancer in
Greece: A cost-minimization analysis. Annals of Oncology; 21(7):1462–1467.
Manzini, G., Ettrich, T.J., Kremer, M. et al. (2018). Advantages of a multi-state
approach in surgical research: How intermediate events and risk factor pro-
file affect the prognosis of a patient with locally advanced rectal cancer. BMC
Medical Research Methodology; 18(1):23.
Maringwa, J., Quinten, C., King, M. et al. (2011). Minimal clinically meaningful dif-
ferences for the EORTC QLQ-C30 and EORTC QLQ-BN20 scales in brain cancer
patients. Annals of Oncology; 22(9):2107–2112.
Martell, R.E., Sermer, D., Getz, K. et al. (2013). Oncology drug development
and approval of systemic anticancer therapy by the U.S. Food and Drug
Administration. Oncologist; 18(1):104–111.
Matulonis, U.A., Oza, A., Ho, M. et al. (2015). Intermediate clinical endpoints: A
bridge between progression-free survival and overall survival in ovarian can-
cer trials. Cancer; 121(11):1737–1746.
380 References

McCabe, C., Claxton, K., & Culyer, A.J. (2008). The NICE cost-effectiveness threshold:
What it is and what that means. Pharmacoeconomics; 26(9):733–744.
McIntosh, E., Louviere, J.J., Frew, E. et al. (2010). Applied methods of cost–benefit analysis
in health care. Oxford University Press.
Meads, D.M., Marshall, A., Hulme, C.T. et al. (2016). The cost effectiveness of docetaxel
and active symptom control versus active symptom control alone for refrac-
tory oesophagogastric adenocarcinoma: Economic analysis of the COUGAR-02
trial. Pharmacoeconomics; 34(1):33–42.
Meier-Hirmer, C., & Schumacher, M. (2013). Multi-state model for studying an inter-
mediate event using time-dependent covariates: Application to breast cancer.
BMC Medical Research Methodology; 13:80.
Menon, U., McGuire, A.J., Raikou, M. et al. (2017). The cost-effectiveness of screening
for ovarian cancer: Results from the UK Collaborative Trial of Ovarian Cancer
Screening (UKCTOCS). British Journal of Cancer; 117(5):619–627.
Messori, A., & Trippoli, S. (2017). The results of a pharmacoeconomic study:
Incrementalcost-effectiveness ratio versus net monetary benefit. Heart; 103(21):1746.
Messori, A., & Trippoli, S. (2018). Incremental cost-effectiveness ratio and net mon-
etary benefit, promoting the application of value-based pricing to medical
devices. Therapeutic Innovation & Regulatory Science; 52(6):755–756.
Miller, R.G. (1981). Survival Analysis. New York: Wiley.
Miller, V.A., Hirsh, V., Cadranel, J. et al. (2012). Afatinib versus placebo for patients
with advanced, metastatic non-small-cell lung cancer after failure of erlotinib,
gefitinib, or both, and one or two lines of chemotherapy (LUX-Lung 1): A phase
2b/3 randomised trial. Lancet. Oncology; 13(5):528–538.
Miller, J.D., Foley, K.A., Russel,l M.W. (2014).; Current challenges in health economic
modeling of cancer therapies: a research inquiry. Am. Health Drug Benefits; 7(3):
153–162.
Montazeri, A., Harirchi, I., Vahdani, M. et al. (2000). The EORTC breast cancer-spe-
cific quality of life questionnaire (EORTC QLQ-BR23): Translation and valida-
tion study of the Iranian version. Quality of Life Research; 9(2):177–184.
Montazeri, A., Milroy, R., Hole, D. et al. (2001). Quality of life in lung cancer patients:
As an important prognostic factor. Lung Cancer; 31(2–3):233–240.
Morden, J.P., Lambert, P.C., Latimer, N. et al. (2011). Assessing methods for dealing
with treatment switching in randomised controlled trials: A simulation study.
BMC Medical Research Methodology; 11(1): 4.
Moreno, E., Girón, F.J., Martínez, M.L. et al. (2013). Optimal treatments in cost-effec-
tiveness analysis in the presence of covariates: Improving patient subgroup
definition. European Journal of Operational Research; 226(1):173–182.
Morris, S., Devlin, N., & Spencer, A. (Eds.). Economic analysis in health care, 2nd Edition.
Chichester, UK: John Wiley & Sons.
Msheilla, I., White, R., & Mukku, S.R. An Investigation into the Key Drivers
Influencing the Decision Making of the Scottish Medicines Consortium. Value
in Health; 16(3): A264.
Mullins, C.D., Montgomery, R., & Tunis, S. (2010). Uncertainty in assessing value of
oncology treatments. Oncologist; 15:58–64.
Murray, T.A., Thall, P.F., Yuan, Y. et al. (2017). Robust treatment comparison based
on utilities of semi-competing risks in non-small-cell lung cancer. Journal of the
American Statistical Association; 112:11–23.
References 381

Nafees, B., Stafford, M., Gavriel, S. et al. (2008). Health state utilities for non-small cell
lung cancer. Health and Quality of Life Outcomes; 6(1):84.
National Cancer Institute. (n.d.). Dictionary of cancer terms. Retrieved from: www.
cancer.gov/publications/dictionaries.
National Comprehensive Care Network. (2015). NCCN guidelines & clinical
resources. NCCN clinical practice guidelines in oncology: Non-small cell lung
cancer. National Comprehensive Cancer Network. Retrieved from: https://
improvement.nhs.uk/resources/reference-costs/#rc1718.
National Health Service. (n.d.). Reference costs. NHS improvement. Retrieved from:
https://improvement.nhs.uk/resources/reference-costs/.
National Health Service. (n.d.). Technology appraisal data. NHS improvement.
Retrieved from: www.nice.org.uk/about/what-we-do/our-programmes/
nice-guidance/nice-technology-appraisal-guidance/data.
National Health Service England. (n.d.). Cancer Drugs Fund. Retrieved from: www.
england.nhs.uk/cancer/cdf/.
National Health Service England. (2013). Appraisal and funding of cancer drugs
from July 2016 (including the new Cancer Drugs Fund). A new deal for patients,
taxpayers and industry. Retrieved from: www.england.nhs.uk/wp-content/
uploads/2013/04/cdf-sop.pdf.
National Health Service England. (2015). NHS increases budget for cancer drugs fund
from £280 million in 2014/15 to an expected £340 million in 2015/16. Retrieved
from: www.england.nhs.uk/2015/01/12/cancer-drug-budget/.
National Health Service (NHS). (2015). Statistics. Retrieved from: https://www.
england.nhs.uk/2015/01/cancer-drug-budget/.
National Institute for Health and Care Excellence. (n.d.). How we work. Retrieved from:
www.nice.org.uk/aboutnice/howwework/devnicetech/technologyappraisal
processguides/GuideToMethodsTA201112.jsp.
NIH National Cancer Institute. (n.d). NCI dictionary of cancer terms. Retrieved from:
www.cancer.gov/publications/dictionaries/cancer-terms?cdrid=45333.
NIH National Cancer Institute. (n.d.) Retrieved from: www.cancer.gov/about-cancer/
understanding/what-is-cancer.
National Institutes of Health (2001). Considerations in the evaluation of surrogate
endpoints in clinical trials. Summary of a National Institutes of Health work-
shop. Control Clin Trials. 2001 Oct; 22(5): 485–502.
National Institute for Health and Care Excellence. (n.d.). NICE TA269. Retrieved
from: https://www.nice.org.uk/Guidance/TA269.
National Institute for Health and Care Excellence. (n.d.). Search results. Retrieved
from: www.nice.org.uk/Guidance/TA202.
National Institute for Health and Care Excellence. (n.d.). Utilities TSD series.
Retrieved from: http://nicedsu.org.uk/technical-support-documents/
utilities-tsd-series/.
National Institute for Health and Care Excellence. (2007a). Bortezomib monother-
apy for relapsed multiple myeloma. Technology appraisal guidance [TA129].
Retrieved from: www.nice.org.uk/guidance/ta129.
National Institute for Health and Care Excellence. (2007b). Pemetrexed for the treat-
ment of non-small-cell lung cancer. Technology appraisal guidance [TA124].
Retrieved from: www.nice.org.uk/guidance/ta124.
382 References

National Institute for Health and Care Excellence. (2008a). Erlotinib for the treat-
ment of non-small-cell lung cancer. Technology appraisal guidance [TA162].
Retrieved from: www.nice.org.uk/guidance/ta162.
National Institute for Health and Care Excellence. (2008b). TA171. Retrieved from:
https://www.nice.org.uk/guidance/ta171/resources/multiple-myeloma-
lenalidomide-evidence-review-group-report2.
National Institute for Health and Care Excellence. (2008c). TA162: Erlotinib for the
treatment of non-small-cell lung cancer. Retrieved from: http://www.nice.org.
uk/guidance/TA162.
National Institute for Health and Care Excellence. (2009a). Pemetrexed for the first-
line treatment of non-small-cell lung cancer. Technology appraisal guidance
[TA181]. Retrieved from: www.nice.org.uk/guidance/ta181.
National Institute for Health and Care Excellence. (2009b). Rituximab for the first-
line treatment of chronic lymphocytic leukaemia. Technology appraisal guid-
ance [TA174]. Retrieved from: www.nice.org.uk/guidance/ta174.
National Institute for Health and Care Excellence. (2010a). Gefitinib for the first-line treat-
ment of locally advanced or metastatic non-small-cell lung cancer. Technology
appraisal guidance [TA192]. Retrieved from: www.nice.org.uk/guidance/TA192.
National Institute of Health and Care Excellence. (2009c). Sunitinib for the treatment
of gastrointestinal stromal tumours - guidance (TA179). HTA TA179. Retrieved
from: https://www.nice.org.uk/guidance/ta179.
National Institute for Health and Care Excellence. (2010b). Ofatumumab (Arzerra®)
for the treatment of chronic lymphocytic leukaemia in patients who are
refractory to fludarabine and alemtuzumab. National Institute for Health
and Clinical Excellence. Retrieved from: www.nice.org.uk/guidance/ta202/
documents/chronic-lymphocytic-leukaemia-ofatumumab-manufacturers-
submission2.
National Institute for Health and Care Excellence. (2010c). Pemetrexed for the main-
tenance treatment of non-small-cell lung cancer. Technology appraisal guid-
ance [TA190]. Retrieved from: www.nice.org.uk/guidance/TA190.
National Institute for Health and Care Excellence. (2010d). Sorafenib for the treat-
ment of advanced hepatocellular carcinoma. Technology appraisal guidance
[TA189]. Retrieved from: www.nice.org.uk/guidance/TA189.
National Institute for Health and Care Excellence. (2010e). HTA TA189. Retrieved
from: https://www.nice.org.uk/guidance/ta189.
National Institutes for Health and Care Excellence. (2010f). TA189. Retrieved from:
https://www.nice.org.uk/guidance/ta189.
National Institute for Health and Care Excellence. (2010g). HTA TA202. Retrieved from:
https://www.nice.org.uk/guidance/ta202/documents/chronic-lymphocytic-
leukaemia-ofatumumab-manufacturers-submission2.
National Institute for Health and Care Excellence. (2011a). Erlotinib monotherapy
for maintenance treatment of non-small-cell lung cancer. Technology appraisal
guidance [TA227]. Retrieved from: www.nice.org.uk/guidance/TA227.
National Institute for Health and Care Excellence. (2011b). Lung cancer: Diagnosis
and management. Retrieved from: www.nice.org.uk/guidance/cg121.000.
National Institute for Health and Care Excellence. (2011c). Advanced or metastatic
NSCLC treatment pathway based on NICE guidance CG121 23. Retrieved from:
www.nice.org.uk/guidance/cg121.
References 383

National Institute for Health and Care Excellence. (2011d). TA227: Erlotinib mono-
therapy for maintenance treatment of non-small-cell lung cancer. Retrieved
from: http://www.nice.org.uk/guidance/TA227.
National Institute for Health and Care Excellence. (2012a). Erlotinib for the first-line
treatment of locally advanced or metastatic EGFR-TK mutation-positive non-
small-cell lung cancer. Technology appraisal guidance [TA258]. Retrieved
from: www.nice.org.uk/guidance/TA258.
National Institute for Health and Care Excellence. (2012b). Erlotinib for the first-line
treatment of locally advanced or metastatic EGFR-TK mutation-positive non-
small-cell lung cancer. Retrieved from: http://www.nice.org.uk/guidance/
TA258.
National Institute for Health and Care Excellence. (2013a). Bosutinib for previously
treated chronic myeloid leukaemia. Technology appraisal guidance [TA299].
Retrieved from: www.nice.org.uk/guidance/TA299.
National Institute for Health and Care Excellence. (2013b). Pertuzumab for the neo-
adjuvant treatment of HER2-positive breast cancer. Technology appraisal guid-
ance [TA424]. Retrieved from: www.nice.org.uk/guidance/TA424.
National Institute for Health and Care Excellence. (2013c). Olaparib for maintenance
treatment of relapsed, platinum-sensitive, BRCA mutation-positive ovarian,
fallopian tube and peritoneal cancer after response to second-line or subse-
quent platinum-based chemotherapy. Technology appraisal guidance [TA381].
Retrieved from: www.nice.org.uk/guidance/TA381.
National Institute for Health and Care Excellence. (2013d). Crizotinib for previously
treated non-small-cell lung cancer associated with an anaplastic lymphoma
kinase fusion gene. Technology appraisal guidance [TA296]. Retrieved from:
www.nice.org.uk/guidance/TA296.
National Institute for Health and Care Excellence. (2013e). How NICE measures
value for money in relation to public health interventions. Local government
briefing. Retrieved from: www.nice.org.uk/media/default/guidance/lgb10-
briefing-20150126.pdf.
National Institute for Health and Care Excellence. (2014a). Afatinib for treating epi-
dermal growth factor receptor mutation-positive locally advanced or metastatic
non-small-cell lung cancer. Technology appraisal guidance [TA310]. Retrieved
from: www.nice.org.uk/guidance/TA310.
National Institute for Health and Care Excellence (2014b). Pemetrexed maintenance
treatment following induction therapy with pemetrexed and cisplatin for non-
squamous non-small-cell lung cancer. Technology appraisal guidance [TA309].
Retrieved from: www.nice.org.uk/guidance/TA309.
National Institute for Health and Care Excellence. (2017a). Pembrolizumab for treat-
ing PD-L1-positive non-small-cell lung cancer after chemotherapy. Technology
appraisal guidance [TA447]. Retrieved from: www.nice.org.uk/guidance/
TA447.
National Institute for Health and Care Excellence. (2017b). Pembrolizumab for
untreated PD-L1-positive metastatic non-small-cell lung cancer. Technology
appraisal guidance [TA428]. Retrieved from: www.nice.org.uk/guidance/TA428.
National Institute for Health and Care Excellence. (2017c). Nivolumab for previously
treated squamous non-small-cell lung cancer. Technology appraisal guidance
[TA483]. Retrieved from: www.nice.org.uk/guidance/TA483.
384 References

National Institute for Health and Care Excellence. (2017d). Venetoclax for treating
chronic lymphocytic leukaemia. Technology appraisal guidance [TA487].
Retrieved from: www.nice.org.uk/guidance/ta487.
National Institute for Health and Care Excellence. (2017e). TA487. Retrieved from:
https://www.nice.org.uk/guidance/ta487.
National Institute for Health and Care Excellence. (2018a). Atezolizumab for treating
locally advanced or metastatic non-small-cell lung cancer after chemotherapy.
Technology appraisal guidance [TA520]. Retrieved from: www.nice.org.uk/
guidance/TA520.
National Institute for Health and Care Excellence. (2018b). Cabozantinib for treating
medullary thyroid cancer. Technology appraisal guidance [TA516]. Retrieved
from: www.nice.org.uk/guidance/TA516.
National Institute for Health and Care Excellence. (2018c). TA516. Retrieved from:
https://www.nice.org.uk/guidance/ta516.
National Institute for Health and Care Excellence. (2019). NICE Technology
Appraisal Data. Retrievede from: https://www.nice.org.uk/about/what-we-
do/our-programmes/nice-guidance/nice-technology-appraisal-guidance/
data/cancer-appraisal-recommendations.
National Institute for Sickness and Disability Insurance. (n.d.). INAMI home page.
Retrieved from: www.inami.fgov.be/fr/Pages/default.aspx.
Neumann, P.J., Kim, D.D., Trikalinos, T.A. et al. (2018). Future directions for cost-
effectiveness analyses in health and medicine. Medical Decision Making;
38(7):767–777.
Ng, R., Kornas, K., Sutradhar, R. et al. (2018). The current application of the Royston-
Parmar model for prognostic modeling in health research: A scoping review.
Diagnostic and Prognostic Research; 2(1):4.
Nguyen, V.T., & Dupuy, J.F. (2018). Zero-inflated Poisson regression with right-cen-
sored data. Retrieved from: https://hal.archives-ouvertes.fr/hal-01811949.
Nishino, M., Jagannathan, J.P., Krajewski, K.M. et al. (2012). Personalized tumor
response assessment in the era of molecular medicine: Cancer-specific and
therapy-specific response criteria to complement pitfalls of RECIST. American
Journal of Roentgenology; 198(4):737–745.
Noble, S.M., Hollingworth, W., & Tilling, K. (2012). Missing data in trial-based cost-
effectiveness analysis: The current state of play. Health Economics; 21(2):187–200.
Norden, A.D., Lesser, G.J., Drappatz, J. et al. (2013). Phase 2 study of dose-intense
temozolomide in recurrent glioblastoma. Neuro-Oncology; 15(7):930–935.
Oaknin, A. (2015). XVII Simposio de Revisiones en Cancer. Madrid 11–13 February
2015; accessed March 2018.
O’Brien, B.J., & Briggs, A.H. (2002). Analysis of uncertainty in health care cost-effec-
tiveness studies: An introduction to statistical issues and methods. Statistical
Methods in Medical Research; 11(6):455–468.
O’Connor, R.D., O’Donnell, J.C., Pinto, L.A. et al. (2002). Two-year retrospective eco-
nomic evaluation of three dual-controller therapies used in the treatment of
asthma. Chest; 121(4):1028–1035.
Olchanski, N., Zhong, Y., Cohen, J.T. et al. (2015). The peculiar economics of life-
extending therapies: A review of costing methods in health economic evalu-
ations in oncology. Expert Review of Pharmacoeconomics and Outcomes Research;
15(6):931–940.
References 385

O’Mahony, J.F., Newall, A.T., & van Rosmalen, J. (2015). Dealing with time in health
economic evaluation: Methodological issues and recommendations for prac-
tice. PharmacoEconomics; 33(12):1255–1268.
Omuro, A., Chan, T.A., Abrey, L.E. et al. (2013). Phase II trial of continuous low-dose
temozolomide for patients with recurrent malignant glioma. Neuro-Oncology;
15(2):242–250.
Oppe, M., Devlin, N.J., van Hout, B. et al. (2014). A program of methodological
research to arrive at the new international EQ-5D-5L valuation protocol. Value
in Health; 17(4):445–453.
Osoba, D. (2007). Translating the science of patient-reported outcomes assess-
ment into clinical practice. Journal of the National Cancer Institute Monographs;
37(37):5–11.
O’Sullivan, A.K., Thompson, D., & Drummond, M.F. (2005). Collection of health-eco-
nomic data alongside clinical trials: Is there a future for piggyback evaluations?
Value in Health; 8(1):67–79.
Oxnard, G.R., Morris, M.J., Hodi, F.S. et al. (2012). When progressive disease does not
mean treatment failure: Reconsidering the criteria for progression. JNCI Journal
of the National Cancer Institute; 104(20):1534–1541.
Pan, W., & Bai, H. (2015). Propensity score interval matching: Using bootstrap con-
fidence intervals for accommodating estimation errors of propensity scores.
BMC Medical Research Methodology; 15:53.
Papaioannou, D., Brazier, J., & Paisley, S. (2013). Systematic searching and selection
of health state utility values from the literature. Value in Health; 16(4):686–695.
Parikh, R.C., Du, X.L., Morgan, R.O. et al. (2016). Patterns of treatment sequences in
chemotherapy and targeted biologics for metastatic colorectal cancer: Findings
from a large community-based cohort of elderly patients. Drugs Real World
Outcomes; 3(1):69–82.
Parikh, R.C., Du, X.L., Robert, M.O. et al. (2017). Cost-effectiveness of treatment
sequences of chemotherapies and targeted biologics for elderly metastatic colorec-
tal cancer patients. Journal of Managed Care and Specialty Pharmacy; 23(1):64–73.
Parkin, M. (2016). Opportunity cost: A re-examination. Journal of Economic Education;
47(1):12–22.
Parner, E.T., & Andersen, P.K. (2010). Regression analysis of censored data using
pseudo-observations. The STATA Journal; 10(3):408–422.
pBAC. (2013c). 07-2013: Gefitinib, tablet, 250 mg, Iressa®. Retrieved from: http://www.
pbs.gov.au/info/industry/listing/elements/pbac-meetings/psd/2013-07/
gefitinib.
pCODR. (2013a). Crizotinib (Xalkori) Resubmission for Advanced Non-Small
Cell Lung Cancer. Retrieved from: www.pcodr.ca/wcpc/portal/Home/
FindaReview/XalkoriAdvNSCLCResub?_afrLoop=457986569182000&amp;_
afrWindowMode=0&amp;_adf.ctrl-state=17jia3apey_276.
pCODR. (2013b). Pemetrexed (Alimta) for Non-Squamous Non-Small Cell
Lung Cancer. Retrieved from: http://www.pcodr.ca/wcpc/portal/Home/
Fi ndaReview/Al i mtaNS -NSCLC?_ af rLoop=458044314492000&amp;_
afrWindowMode=0&amp;_adf.ctrl-state=17jia3apey_347.
Peng, Y., & Taylor, J.M.G. (2014). Chapter 6: Cure models. In: J. Klein, H. van Houwelingen,
J.G. Ibrahim, T.H. Scheike (Eds.). Handbooks of modern statistical methods series:
Handbook of survival analysis, Boca Raton, FL: Chapman & Hall, 113–134.
386 References

Penn. Medicine. (n.d.). Division of general internal medicine research. Retrieved from:
www.pennmedicine.org/departments-and-centers/department-of-medicine/
divisions/general-internal-medicine/research.
Petrou, S.A.G., & Gray, A. (2011a). Economic evaluation alongside randomised con-
trolled trials: Design, conduct, analysis, and reporting. BMJ; 342:d1548.
Petrou, S.A.G., & Gray, A. (2011b). Economic evaluation using decision analytical
modelling: Design, conduct, analysis, and reporting. BMJ; 342:d1766.
Pickard, A.S., Neary, M.P., & Cella, D. (2007). Estimation of minimally important dif-
ferences in EQ-5D utility and VAS scores in cancer. Health and Quality of Life
Outcomes; 5(1):70.
Pocock, S.J. (1983). Clinical trials: A practical approach. New York: Wiley.
Polley, W.J. (2015). The rhetoric of opportunity cost. American Economist; 60(1):9–19.
Popat, R., Khan, I., Dickson, J. et al. (2015). An alternative dosing strategy of lenalido-
mide for patients with relapsed multiple myeloma. British Journal of Haematology;
168(1):148–151.
Porter, M.E. (2010). What is value in health care? New England Journal of Medicine;
363(26):2477–2481.
Public health France. (n.d.). Epidemiological surveillance of cancers in France.
Institute for Public Health Surveillance (InVS). Retrieved from: http://invs.san-
tepubliquefrance.fr/surveillance/cancers/acteurs.htm.
Putter, H., Fiocco, M., & Geskus, R.B. (2007). Tutorial in biostatistics: Competing risks
and multi-state models. Statistics in Medicine; 26(11):2389–2430.
Putter, H., van der Hage, J., de Bock, G.H. et al. (2006). Estimation and prediction in a
multi-state model for breast cancer. Biometrical Journal. Biometrische Zeitschrift;
48(3):366–380.
Rabin, R., & de Charro, Fd (2001). EQ-5D: A measure of health status from the
EuroQol Group. Annals of Medicine; 33(5):337–343.
Ramsey, S.D, Willke, R.J, Briggs, A. et al. (2005). Good research practices for cost-
effectiveness analysis alongside clinical trials: The ISPOR RCT-CEA task force
report. Value in Health; 8(5):521–533.
Ramsey, S.D., Willke, R.J., Glick, H. et al. (2015). Cost-effectiveness analysis alongside
clinical trials II—An ISPOR good research practices task force report. Value in
Health; 18(2):161–172.
Rappange, D.R., van Baal, P.H., van Exel, N.J. et al. (2008). Unrelated medical costs in
life-years gained: Should they be included in economic evaluations of health-
care interventions? Pharmacoeconomics; 26(10):815–830.
Rascati, K.L. (2009). Essentials of Pharmacoeconomics. Philadelphia, PA: Lippincott
Williams & Wilkins.
Ratowsky D. & Ratkowsky, A. (1989). Handbook of nonlinear regression models, New
York : M. Dekker.
Rawlins, M., Barnett, D., & Stevens, A. (2010). Pharmacoeconomics: NICE’s approach
to decision-making. British Journal of Clinical Pharmacology; 70(3):346–349.
Richardson, J., Khan, M.A., Iezzi, A. et al. (2015). Comparing and explaining differ-
ences in the magnitude, content, and sensitivity of utilities predicted by the
EQ-5D, SF−6D, HUI 3, 15D, QWB, and AQoL-8D multi-attribute utility instru-
ments. Medical Decision Making; 35(3):276–291.
Rivera, F., Valladares, M., Gea, S. et al. (2017). Cost-effectiveness analysis in the Spanish
setting of the PEAK trial of panitumumab plus mFOLFOX6 compared with bev-
acizumab plus mFOLFOX6 for first-line treatment of patients with wild-type
RAS metastatic colorectal cancer. Journal of Medical Economics; 20(6):574–584.
References 387

Robert J. Brent; A Simple Method for Converting a Cost-Effectiveness Analysis


into a Cost-Benefit Analysis with an Application to State Mental Health
Expenditures First Published March 1, 2002 Research Article; https://doi.org
/10.1177/109114210203000204 https://journals.sagepub.com/doi/abs/10.1177/
109114210203000204
Robins, J.M. (1986). A new approach to causal inference in mortality studies with sus-
tained exposure periods –Application to control the healthy worker survivor
effect. Mathematical Modelling; 7:1392–1512.
Robins, J.M., & Finkelstein, D.M. (2000). Correcting for noncompliance and depen-
dent censoring in an AIDS Clinical Trial with inverse probability of censoring
weighted (IPCW) log-rank tests. Biometrics; 56(3):779–788.
Robins, J.M., Rotnitzky, A., & Zhao, L.P. (1994). Estimation of regression coefficients
when some regressors are not always observed. Journal of the American Statistical
Association; 89(427): 846‒866.
Rodriguez, G. (2010). Parametric survival models. Retrieved from: ata.princeton.
edu/pop509/ParametricSurvival.pdf.
Rowen, D., Brazier, J., & Van Hout, B. (2015). A comparison of methods for convert-
ing DCE values onto the full health-dead QALY scale. Medical Decision Making;
35(3):328–340.
Rowen, D., Brazier, J., Young, T. et al. (2011). Deriving a preference-based measure for
cancer using the EORTC QLQ-C30. Value Health; 14(5):721–731.
Royal College of Physicians. (n.d.). Organising an audit or quality improve-
ment project: Tips for beginners. Student and foundation doctor network.
Retrieved from: www.rcplondon.ac.uk/projects/outputs/organising-audit-or-
quality-improvement-project-tips-beginners.
Royston, P. (2015). Restricted mean survival time: Calculation and some applications
in trials and prognostic studies. Flexible parametric survival models work-
shop. Stockholm, November 2011.
Royston, P., & Lambert, P.C. (2011). Flexible parametric survival analysis using Stata:
Beyond the Cox Model. College Station, TX: Stata Press.
Royston, P., & Parmar, M.K.B. (2002). Flexible proportional-hazards and propor-
tional odds models for censored survival data, with application to prog-
nostic modelling and estimation of treatment effects. Statistics in Medicine;
21(15):2175–2197.
Royston, P., & Parmar, M.K.B. (2013). Restricted mean survival time: An alternative to
the hazard ratio for the design and analysis of randomized trials with a time-
to-event outcome. BMC Medical Research Methodology; 13(1):15.
Royston, P., Parmar, M.K.B., & Qian, W. (2003). Novel designs for multi-arm clinical
trials with survival outcomes with an application in ovarian cancer. Statistics in
Medicine; 22(14):2239–2256.
Rubin, D.B. (2004). Multiple imputation for nonresponse in surveys. Hoboken, NJ: Wiley.
Saad, E.D., Zalcberg, J.R., Péron, J. et al. (2018). Understanding and communicat-
ing measures of treatment effect on survival: Can we do better? Journal of the
National Cancer Institute; 110(3):232–240.
Sacco, E., Tienforti, D., D’Addessi, A. et al. (2010). Social, economic, and health utility
considerations in the treatment of overactive bladder. Open Access. Journal of
Urology; 2:11–24.
Sacher, A.G., Le, L.W., Lau, A. et al. (2015). Real-world chemotherapy treatment pat-
terns in metastatic non-small cell lung cancer: Are patients undertreated?
Cancer; 121(15):2562–2569.
388 References

Saint-Pierre, P. (2016). Multi-state models and cost-effectiveness analysis. Journal de


Gestion et d’Économie Médicales; 34(2):133–144.
Salleh, S., Thokala, P., Brennan, A. et al. (2017). Discrete event simulation-based
resource modelling in health technology assessment. Pharmacoeconomics;
35(10):989–1006.
Sandmann, F.G., Robotham, J.V., Deeny, S.R. et al. (2018). Estimating the opportunity
costs of bed-days. Health Economics; 27(3):592–605.
Santerre, R.E., & Neun, S.P. (2000). Health economics: Theories, insights, and industry
studies. Fort Worth, TX: Dryden Press.
Sartor, O., & Halabi, U. (2015). Independent data monitoring committees: An update
and overview. Urologic Oncology; 33(3):143–148.
Schnipper, L.E., Davidson, N.E., Wollins, D.S. et al. (2015). American Society of
Clinical Oncology statement: A conceptual framework to assess the value
of cancer treatment options. Journal of Clinical Oncology; 33(23):2563–2577.
Retrieved from: http://ascopubs.org/doi/abs/10.1200/jco.2015.61.6706.
Scuffham, P.A., Whitty, J.A., Mitchell, A. et al. (2008). The use of QALY weights for
QALY calculations. Pharmacoeconomics; 26(4):297–310.
Sculpher, M.J., Claxton, K., Drummond, M. et al. (2006). Whither trial-based economic
evaluation for health care decision making? Health Economics; 15(7):677–687.
Seruga, B., Pond, G.R., Hertz, P.C. et al. (2012). Comparison of absolute benefits of
anticancer therapies determined by snapshot and area methods. Annals of
Oncology; 23(11):2977–2982.
Shaw, J.W., Johnson, J.A., & Coons, S.J. (2005). US valuation of the EQ-5D health states:
Development and testing of the D1 valuation model. Medical Care; 43(3):203–220.
Shepherd, F.A., Rodrigues Pereira, J.R., Ciuleanu, T. et al. (2005). Erlotinib in pre-
viously treated non-small-cell lung cancer. New England Journal of Medicine;
353(2):123–132.
Simes, R.J., & Coates, A.S. (2001). Patient preferences for adjuvant chemotherapy of
early breast cancer: How much benefit is needed? Journal of the National Cancer
Institute Monographs; 30(30):146–152.
Sleeper, L.A., & Harrington, D.P. (1990). Regression splines in the Cox Model with
application to covariate effects in liver disease. Journal of the American Statistical
Association; 85(412):941–949.
Slevin, M.L., Plant, H., Lynch, D. et al. (1988). Who should measure quality of life, the
doctor or the patient? British Journal of Cancer; 57(1):109–112.
SMC (2006). 220/05: Erlotinib (Tarceva®) re-submission. Retrieved from: https://www.
scottishmedicines.org.uk/SMC_Advice/Advice/Erlotinib__Tarceva__
Resubmission/Erlotinib__Tarceva__.
SMC (2012). 749/11: erlotinib (Tarceva). Retrieved from: https://www.scottishmedi-
cines.org.uk/SMC_Advice/Advice/749_11_erlotinib_Tarceva/erlotinib_Tarceva.
SMC (2013). Crizotinib (Xalkori). Retrieved from: http://www.scottishmedicines.
org.uk/SMC _Advice/Advice/865_13_cri zot i n ib_ Xalkori/cri zot i n ib_
Xalkori_Resubmission.
SMC (2014). SMC. 920/13: afatinib (Giotrif). Retrieved from: https://www.scot-
tishmedicines.org.uk/SMC_Advice/Advice/920_13_afatinib_Giotrif/
afatinib_Giotrif.
Sorenson, C., Kanavos, P., & Drummond, M. (2008). Ensuring value for money in health
care: The role of HTA in the European Union. Copenhagen: European Observatory
on Health Systems and Policies, World Health Organisation.
References 389

Spiegelhalter, D.J., & Best, N.G. (2003). Bayesian approaches to multiple sources of
evidence and uncertainty in complex cost‐effectiveness modelling. Statistics in
Medicine; 22(23):3687–3709.
Spruance, S.L., Reid, J.E., Grace, M. et al. (2004). Hazard ratio in clinical trials.
Antimicrobial Agents and Chemotherapy; 48(8):2787–2792.
Stadtmauer, E., Weber, D., Dimopolous, M. et al. (2006). Lenalidomide in combination
with dexamethasone is more effective Than dexamethasone at first relapse in
relapsed multiple myeloma. ASH Annual Meeting Abstracts; 108(11):3552.
Stafinski, T., Menon, D., Davis, C. et al. (2011). Role of centralized review processes
for making reimbursement decisions on new health technologies in Europe;
ClinicoEconomics and Outcomes Research; 3:117–186.
Stare, J., & Boulch, D.M. (2016). Odds ratio, hazard ratio and relative risk. Metodoloski
Zvezki; 13(1):59–67. Retrieved from: www.stat-d.si/mz/mz13.1/p4.pdf.
Steckler, A., & McLeroy, K.R. (2008). The importance of external validity. American
Journal of Public Health; 98(1):9–10.
Stinnett, A.A., & Mullahy, J. (1998). Net health benefits. Medical Decision Making; 18
(2 Suppl):S68–S80.
Sullivan, S.D., Mauskopf, J.A., Augustovski, F. et al. (2015). Budget impact analysis:
Principles of good practice. Report of the ISPOR working group on good prac-
tices for budget impact analysis II, 2012. Kachestvennaya Klinicheskaya Praktika;
2:104–118.
Susarla, V., & Van Rizyn, J. (1980). Large sample theory for an estimator of the mean
survival time from censored samples. The Annals of Statistics; 8(5):1002–1016.
Susarla, V., & Van Rizyn, J. (1984). A Buckley-James-type estimator for the mean with
censored data. Biometrika; 71(3):624–629.
Sydes, M.R., Parmar, M.K.B., James, N.D. et al. (2009). Issues in applying multi-
arm multi-stage methodology to a clinical trial in prostate cancer: The MRC
STAMPEDE trial. Trials; 10:39.
Tai, B.C., Wee, J., & Machin, D. (2011). Analysis and design of randomised clinical tri-
als involving competing risks outcomes. Trials; 12:127.
Taphoorn, M.J.B., Henriksson, R., Bottomley, A. et al. (2015). Health-related quality
of life in a randomized phase III study of bevacizumab, temozolomide, and
radiotherapy in newly diagnosed glioblastoma. Journal of Clinical Oncology;
33:2166–2175.
Tappenden, P., Brazier, J., Ratcliffe, J., Chilcott, J. (2007). A stated preference binary
choice experiment to explore NICE decision making. Pharmacoeconomics; 25(8):
685–693.
Temel, J.S., Greer, J.A., Muzikansky, A. et al. (2010). Early palliative care for patients
with metastatic non–small-cell lung cancer. New England Journal of Medicine;
363(8):733–742.
Thatcher, N., Chang, A., Parikh, P. et al. (2005). Gefitinib plus best supportive care in
previously treated patients with re-fractory advanced non-small-cell lung can-
cer: Results from a randomised, placebo-controlled, multicentre study (Iressa
Survival Evaluation in Lung Cancer). Lancet; 366(9496):1527–1537.
The Professional Society for Health Economics and Outcomes Research. (2009).
Germany: Pharmaceutical. Global health technology assessment road map.
Retrieved from: https://tools.ispor.org/htaroadmaps/Germany.asp.
The World Bank. (n.d.). Current health expenditure (% of GDP). Retrieved from:
https://data.worldbank.org/indicator/SH.XPD.CHEX.GD.ZS.
390 References

Thein, H.H., Isaranuwatchai, W., Qiao, Y. et al. (2017). Cost-effectiveness analysis of


potentially curative and combination treatments for hepatocellular carcinoma
with person-level data in a Canadian setting. Cancer Medicine; 6(9):2017–2033.
Thompson, S.G., Nixon, R.M., & Grieve, R. (2006). Addressing the issues that arise
in analysing multicentre cost data, with application to a multinational study.
Journal of Health Economics; 25(6):1015–1028.
Thongprasert, S., Tinmanee, S., & Permsuwan, U. (2012). Cost-utility and budget
impact analyses of gefitinib in second-line treatment for advanced non-small
cell lung cancer from Thai payer perspective. Asia-Pacific Journal of Clinical
Oncology; 8(1):53–61.
Timmins, N., Rawlins, M., & Appleby, J. (2016). A terrible beauty. A short history of NICE,
the National Institute for Health and Care Excellence. Retrieved from: www.
idsihealth.org/wp-content/uploads/2016/02/A-TERRIBLE-BEAUTY_resize.pdf.
Titman, A. (2016). Multi-state models: An overview. Presentation. Retrieved from:
www.maths.lancs.ac.uk/~titman/leeds_seminar.pdf.
Toms, J.R. (2004). CancerStats monograph 2004: Cancer incidence, survival and mortality in
the UK and EU. London: Cancer Research UK.
Trinquart, L., Jacot, J., Conner, S.C. et al. (2016). Comparison of treatment effects
measured by the hazard ratio and by the ratio of restricted mean survival
times in oncology randomized controlled trials. Journal of Clinical Oncology;
34(15):1813–1819.
Truven https://truvenhealth.com/Portals/1/.../INTL_12543_0413_RedbookPS_WEB1.
pdf, accessed 31 Jan 2018.
Tuffaha, H.W., Gordon, L.G., & Scuffham, P.A. (2016). Efficient value of information
calculation using a nonparametric regression approach: An applied perspec-
tive. Value Health; 19(4):505–509. Epub 2016.
Tuffaha, H.W., Reynolds, H., Gordon, L.G. et al. (2014). Value of information analy-
sis optimizing future trial design from a pilot study on catheter securement
devices. Society for Clinical Trials; 11(6):648‒656.
Uhry, Z., Hédelin, G., Colonna, M. et al. (2010). Multi-state Markov models in can-
cer screening evaluation: A brief review and case study. Statistical Methods in
Medical Research; 19(5):463–486.
Ulmeanu, R., Antohe, I., Anisie, E. et al. (2016). Nivolumab for advanced non-small
cell lung cancer: An evaluation of a phase III study. Expert Review of Anticancer
Therapy; 16(2)165–167.
U.S. Department of Health and Human Services. Food and Drug Administration.
(2018). Clinical trial endpoints for the approval of cancer drugs and biolog-
ics: Guidance for industry. Retrieved from: www.fda.gov/downloads/Drugs/
Guidances/ucm071590.pdf.
University of Kent. (n.d.). Unit costs of health and social care. Personal social services
reesearch unit. Retrieved from: www.pssru.ac.uk/project-pages/unit-costs/.
University of Texas at Dallas. (n.d.). Markov chains. Retrieved from: www.utdallas.
edu/~jjue/cs6352/markov/markov.html.
U.S. Department of Health and Human Services. Food and Drug Administration.
(2007). Clinical trial endpoints for the approval of cancer drugs and biolog-
ics: Guidance for industry. Center for Drug Evaluation and Research (CDER),
Center for Biologics Evaluation and Research (CBER). Retrieved from: www.
fda.gov/downloads/Drugs/Guidances/ucm071590.pdf.
References 391

U.S. Food and Drug Administration. (n.d.). Information exchange and data transfor-
mation (INFORMED). Retrieved from: www.fda.gov/aboutfda/centersoffices/
officeofmedicalproductsandtobacco/oce/ucm543768.htm.
U.S. Food and Drug Administration. (n.d.). 21st century cures act. Retrieved from:
www.fda.gov/regulatoryinformation/lawsenforcedbyfda/significantamendm
entstothefdcact/21stcenturycuresact/default.htm.
van Agt, H.M.E., Essink-Bot, M., Krabbe, P.F.M. et al. (1994). Test-retest reliability of
health state valuations collected with the EuroQol questionnaire. Social Science
and Medicine; 39(11):1537–1544.
van Agt, H.M.E., van der Stege, H.A., de Ridder-Sluiter, H.A. et al. (2005). Quality
of life of children with language delays. Qual Life Res. 2005 Jun; 14(5): 1345–55.
Vale, C.D., Maurelli, V.A. (1983). Simulating multivariate nonnormal distributions.
Psychometrika; 48: 465–471.
van Baal, P.H., Feenstra, T.L., Polder, J.J. et al. (2011). Economic evaluation and the
postponement of health care costs. Health Economics; 20(4):432–445.
van Baal, P.H., Morton, A., Brouwer, W. et al. (2017). Should cost effectiveness analy-
ses for NICE always consider future unrelated medical costs? BMJ; 359:j5096.
Van Buuren, S. (2018). Flexible imputation of missing data, 2nd Edition. Chapman & Hall.
van Erning, F.N., van Steenbergen, L.N., Lemmens, V.E.P.P. et al. (2014). Conditional
survival for long-term colorectal cancer survivors in the Netherlands: Who do
best? European Journal of Cancer; 50(10):1731–1739.
Van Harten, W., & IJzerman, M.J. (2017). Responsible pricing in value-based assess-
ment of cancer drugs: Real-world data are an inevitable addition to select
meaningful new cancer treatments. Ecancermedicalscience; 11:ed71.
Van Hout, B., Janssen, M.F., Feng, Y.S. et al. (2012). Interim scoring for the EQ-5D-5L:
Mapping the EQ-5D-5L to EQ-5D-3L value sets. Value in Health; 15(5):708–715.
Van Houwelingen, H., & Putter, H. (2012). Dynamic prediction in clinical survival analy-
sis. Boca Raton, FL: CRC Press, Taylor & Francis.
Vázquez, G.H., Holtzman, J.N., Lolich, M. et al. (2015). Recurrence rates in bipolar
disorder: Systematic comparison of long-term prospective, naturalistic stud-
ies versus randomized controlled trials. European Neuropsychopharmacology;
25(10):1501–1512.
Vergnenegre, A., Corre, R., Berard, H. et al. (2011). Cost-effectiveness of second-line
chemotherapy for non-small cell lung cancer: An economic, randomized, pro-
spective, multicenter Phase III trial comparing docetaxel and pemetrexed: The
GFPC 05–06 study. Journal of Thoracic Oncology; 6(1):161–168.
Versteegh, M., Knies, S., & Brouwer, W. (2016). From good to better: New Dutch
guidelines for economic evaluations in healthcare. Pharmacoeconomics;
34(11):1071–1074.
Vredenburgh, J.J., Desjardins, A., Reardon, D.A. et al. (2010). Bevacizumab (BEV) in
combination with temozolomide (TMZ) and radiation therapy (XRT) followed
by BEV, TMZ, and irinotecan for newly diagnosed glioblastoma multiforme
(GBM). Journal of Clinical Oncology; 28(15_suppl):2023–2023.
Wailoo, A., Alava, M.H., Grimm, S. et al. (2017). Comparing the EQ-5D-3L and 5L ver-
sions. What are the implications for cost effectiveness estimates? Report by the
Decision Support Unit.Retrieved from: http://scharr.dept.shef.ac.uk/nicedsu/
wp-content/uploads/sites/7/2017/05/DSU_3L-to-5L-FINAL.pdf.
392 References

Walker, A.S., White, I.R., & Babiker, A.G. (2004). Parametric randomization-based
methods for correcting for treatment changes in the assessment of the causal
effect of treatment. Statistics in Medicine; 23(4):571–590.
Wang, Y-.W. & Li, N. (2010). Statistical Analysis for Treatment Crossover or
Nonsynchronized Interval-Censoring Data in a Mortality Trial. Statistics in
Biopharmaceutical Research; 2(2): 175–181.
Wang, S., Peng, L., Li, J. et al. (2013). A trial-based cost-effectiveness analysis of erlo-
tinib alone versus platinum-based doublet chemotherapy as first-line therapy
for eastern asian nonsquamous non–small-cell lung cancer. PLoS One; 8(3).
Weinstein, M.C., Fineberg, H.V., Elstein, A.S. et al. (1980). Clinical decision analysis.
Philadelphia, PA: W. B. Saunders.
Weinstein, M.C., & Manning, W.G. Jr. (1997). Theoretical issues in cost-effectiveness
analysis. Journal of Health Economics; 16(1):121–128.
Weintraub, W.S., Daniels, S.R., Burke, L.E. et al. (2011). Value of primordial and
primary prevention for cardiovascular disease: A policy statement from the
American Heart Association. Circulation; 124(8):967–990. Retrieved from: http://
circ.ahajournals.org/content/124/8/967/F2.
Wendt, C., Frisina, L., & Rothgang, H. (2009). Social policy & healthcare system types:
A conceptual framework for comparison. Administration; 43(1):70–90.
White, I.R., Babiker, A.G., Walker, S. et al. (1999). Randomization-based methods for
correcting for treatment changes: Examples from the Concorde trial. Statistics
in Medicine; 18(19):2617–2634.
Wijeysundera, H.C., Wang, X., Tomlinson, G. et al. (2012). Techniques for estimat-
ing health care costs with censored data: An overview for the health services
researcher. ClinicoEconomics and Outcomes Research; 4:145–155.
Willan, A.R., & Briggs, A.H. (2006). Statistical analysis of cost-effectiveness data. New
York: John Wiley Publications.
Willan, A.R., Briggs, A.H., & Hoch, J.S. (2004). Regression methods for covariate
adjustment and subgroup analysis for non-censored cost-effectiveness data.
Health Economics; 13(5):461–475.
Willekens, F.J., & Putter, H. (2014). Software for multistate analysis. Demographic
Research; 31:381–420.
Williams, C., Lewsey, J.D., Briggs, A.H. et al. (2017). Cost-effectiveness analysis in R
using a multi-state modeling survival analysis framework: A tutorial. Medical
Decision Making; 37(4):340–352.
Williams, C., Lewsey, J.D., Mackay, D.F. et al. (2017). Estimation of survival prob-
abilities for use in cost-effectiveness analyses: A comparison of a multi-state
modeling survival analysis approach with partitioned survival and Markov
decision-analytic modeling. Medical Decision Making; 37(4):427–439.
Willke, R.J., Glick, H.A., Polsky, D. et al. (1998). Estimating country-specific cost-effec-
tiveness from multinational clinical trials. Health Economics; 7(6):481–493.
Wilson, A., Grimshaw, G., Baker, R. et al. (1999). Differentiating between audit and
research: Postal survey of health authorities’ views. BMJ; 319(7219):1235.
Wilson, E.C. (2015). A practical guide to value of information analysis.
Pharmacoeconomics; 33(2):105–121.
Wilson, E.C., Mugford, M., Barton, G. et al. (2016). Efficient research design: Using;
value-of-information analysis to estimate the optimal mix of top-down and
bottom-up costing approaches in an economic evaluation alongside a clinical
trial. Medical Decision Making; 36(3):335–348.
References 393

Winsor, S., Smith, A., Vanstone, M. et al. (2013). Experiences of patient-centredness


with specialized community-based care: A systematic review and qualitative
meta-synthesis. Ontario Health Technology Assessment Series; 13(17):1–33.
Wolchok, J.D., Hoos, A., O’Day, S. et al. (2009). Guidelines for the evaluation of
immune therapy activity in solid tumors: Immune-related response criteria.
Clinical Cancer Research; 15(23):7412–7420.
Woods, B., Sideris, E., Palmer, S. et al. (2017). NICE DSU document 19: Partitioned
survival analysis for decision modelling in health care: A critical review report
by the decision support unit. ScHARR, University of Sheffield.Retrieved from:
http://scharr.dept.shef.ac.uk/nicedsu/wp-content/uploads/sites/7/2017/06/
Partitioned-Survival-Analysis-final-report.pdf.
World Health Organization. (1947). The constitution of the World Health Organization.
WHO Chronicle; 1:29.
World Health Organization (2014). Who we are. Retrieved from: www.who.int/
about/who-we-are/constitution.
Yamaguchi, T., & Ohashi, Y. (2004). Adjusting for differential proportions of second-
line treatment in cancer clinical trials. Part I: Structural nested models and
marginal structural models to test and estimate treatment arm effects. Statistics
in Medicine; 23(13):1991–2003.
Yanni, H., Wolfram, V., & Cook, J.S. (2016). A structured review of health utility mea-
sures and elicitation in advanced/metastatic breast cancer. ClinicoEconomics and
Outcomes Research; 8(1):293–303.
Yap, C., Pettitt, A., & Billingham, K. (2013). Screened selection design for randomised
phase II oncology trials: An example in chronic lymphocytic leukaemia. BMC
Medical Research Methodology; 13(1):87.
Zahid, K.F., Burney, I., Ahmed, T. et al. (2015). Clinicopathological features and
treatment outcomes of breast cancer patients with brain metastases. Annals of
Oncology; 26(Suppl 9):22–23.
Zeng, X., Li, J., Peng, L. et al. (2014). Economic outcomes of maintenance gefitinib for
locally advanced/metastatic non-small-cell lung cancer with unknown EGFR
mutations: A semi-Markov model analysis. PLoS One; 9(2):e88881. eCollection 2014.
Zhang, L., Ma, S., Song, X. et al. (2012). Gefitinib versus placebo as maintenance ther-
apy in patients with locally advanced or metastatic non-small-cell lung can-
cer (INFORM; C-Tong 0804): A multi-centre, double-blind randomised phase 3
trial. Lancet; 13(5):466–475.
Zhao, H., & Bang, H. (2012). Pharmacoeconomics. 2015 February; 33(2):105–121. doi:
10.1007/s40273-014-0219-x. PubMed PMID: 24194511.
Zhao, H., Zuo, C., Chen, S. et al. (2012). Nonparametric inference for median costs
with censored data. Biometrics; 68(3):717–725.
Zhao, Y., Li, S.P., Liu, L. et al. (2017). Does the choice of tariff matter?: A comparison
of EQ-5D-5L utility scores using Chinese, UK, and Japanese tariffs on patients
with psoriasis vulgaris in Central South China Chinese. Medicine (Baltimore);
96(34):e7840.
Zhu, J., Li, T., Wang, X. et al. (2013). Gene-guided gefitinib switch maintenance ther-
apy for patients with advanced EGFR mutation-positive non-small cell lung
cancer: An economic analysis. BMC Cancer; 13(1):39.
Zuur, A.F., Ieno, E.N., & Saveliev, A.A. (2017). Beginner’s guide to spatial, temporal and
spatial-temporal ecological data analysis with R-INLA. Newburgh, UK: Highland
Statistics.
394 References

Additional Bibliography

Chapter 1
Araújo, A., Parente, B., Sotto-Mayor, R. et al. (2008). An economic analysis of erlotinib,
docetaxel, pemetrexed and best supportive care as second or third line treat-
ment of non-small cell lung cancer. Revista Portuguesa de Pneumologia (English
Edition); 14(6):803–827.
Asukai, Y., Valladares, A., Camps, C. et al. (2010). Cost-effectiveness analysis of
pemetrexed versus docetaxel in the second-line treatment of non-small cell
lung cancer in Spain: Results for the non-squamous histology population. BMC
Cancer; 10(1):26.
Berthelot, J.M., Will, B.P., Evans, W.K. et al. (2000). Decision framework for chemo-
therapeutic interventions for metastatic non-small-cell lung cancer. Journal of
the National Cancer Institute; 92(16):1321–1329.
Bradbury, P.A., Tu, D., Seymour, L. et al. (2010). Economic analysis: Randomized pla-
cebo-controlled clinical trial of erlotinib in advanced non-small cell lung can-
cer. Journal of the National Cancer Institute; 102(5):298–306. Epub 2010 February.
Brown, T., Boland, A., Bagust, A. et al. (2010). Gefitinib for the first-line treatment
of locally advanced or metastatic non-small cell lung cancer. Health Technology
Assessment; 14(Suppl 2):71–79.
Carlson, J.J., Reyes, C., Oestreicher, N. et al. (2008). Comparative clinical and economic
outcomes of treatments for refractory non-small cell lung cancer (NSCLC).
Lung Cancer; 61(3):405–415.
Chouaid, C., Le Caer, H., Locher, C. et al., & GFPC 0504 Team. (2012). Cost effec-
tivenes of erlotinib versus chemotherapy for first-line treatment of non small
cell lung cancer (NSCLC) in fit elderly patients participating in a prospective
phase 2 study (GFPC 0504). BMC Cancer; 12:301.
Cromwell, I., van der Hoek, K., Melosky, B. et al. (2011). Erlotinib or docetaxel for
second-line treatment of non-small cell lung cancer: A real-world cost-effec-
tiveness analysis. Journal of Thoracic Oncology; 6(12):2097–2103.
Fragoulakis, V., Pallis, A., & Georgoulias, M. (2014). Economic evaluation of peme-
trexed versus erlotinib as second-line treatment of patients with advanced/
metastatic non-small cell lung cancer in Greece: A cost minimization analysis.
Lung Cancer: Targets and Therapy; 3:43–51.
Gilberto de Lima, L.G., Segel, J., Tan, D. et al. (2011). Cost-effectiveness of epider-
mal growth factor receptor mutation testing and first-line treatment with
gefitinib for patients with advanced adenocarcinoma of the lung. Cancer;
118(4):1032–1039.
Goulart, B., & Ramsey, S. (2011). A trial-based assessment of the cost-utility of bevaci-
zumab and chemotherapy versus chemotherapy alone for advanced non-small
cell lung cancer. Value in Health; 14(6):836–845.
Greenhalgh, J., McLeod, C., Bagust, A. et al. (2010). Pemetrexed for the maintenance
treatment of locally advanced or metastatic non-small cell lung cancer. Health
Technology Assessment; 14(Suppl 2):33–39.
References 395

Maniadakis, N., Fragoulakis, V., Pallis, A.G. et al. (2010). Economic evaluation of
docetaxel-gemcitabine versus vinorelbine-cisplatin combination as front-line
treatment of patients with advanced/metastatic non-small-cell lung cancer in
Greece: A cost-minimization analysis. Annals of Oncology; 21(7):1462–1467.
Thongprasert, S., Tinmanee, S., & Permsuwan, U. (2012). Cost-utility and budget
impact analyses of gefitinib in second-line treatment for advanced non-small
cell lung cancer from Thai payer perspective. Asia-Pacific Journal of Clinical
Oncology; 8(1):53–61.
Vergnenegre, A., Corre, R., Berard, H. et al., & GFPC 0506 Team. (2011). Cost-
effectiveness of second-line chemotherapy for non-small cell lung cancer: An
economic, randomized, prospective, multicenter phase III trial comparing
docetaxel and pemetrexed: The GFPC 05–06 study. Journal of Thoracic Oncology;
6(1):161–168.
Wang, S., Peng, L., Li, J. et al. (2013). A trial-based cost-effectiveness analysis of erlo-
tinib alone versus platinum-based doublet chemotherapy as first-line therapy for
Eastern Asian nonsquamous non–small-cell lung cancer. PLoS One; 8(3):e55917.
Winsor, S., Smith, A., Vanstone, M. et al. (2013). Experiences of patient-centredness
With specialized community-based care: A systematic review and qualitative
meta-synthesis. Ontario Health Technology Assessment Series; 13(17):1–33.
Zhu, J., Li, T., Wang, X. et al. (2013). Gene-guided gefitinib switch maintenance ther-
apy for patients with advanced EGFR mutation-positive Non-small cell lung
cancer: An economic analysis. BMC Cancer; 13(1):39.

Chapter 3
Brooks, R., Rosalind, R., & de Charro, F. (Eds.) (2013). The measurement and valuation
of health status using EQ-5D: A European perspective: Evidence from the EuroQol
BIOMED research programme. Dordrecht, Netherlands: Springer Science &
Business Media.
Cella, D.F., Wiklund, I., Shumaker, S.A. et al. (1993). Integrating health-related qual-
ity of life into cross-national clinical trials. Quality of Life Research; 2(6):433–440.
European Organisation for Research and Treatment of Cancer. (n.d.). Manuals.
EORCT Quality of Life. Retrieved from: http://groups.eortc.be/qol/manuals.
Hollen, P.J., Gralla, R.J., Cox, C. et al. (1997). A dilemma in analysis: Issues in the serial
measurement of quality of life in patients with advanced lung cancer. Lung
Cancer; 18(2):119–136.

Chapter 4
A’Hern, R.P. (2016). Restricted mean survival time: An obligatory end point for time-
to-event analysis in cancer trials? Journal of Clinical Oncology; 34(28):3474–3476.
Ajani, J.A. (2007). The area between the curves gets no respect: Is it because of the
median madness? Journal of Clinical Oncology; 25(34):5531.
Albertsen, P.C., Hanley, J.A., Gleason, D.F. et al. (1998). Competing risk analysis of
men aged 55 to 74 years at diagnosis managed conservatively for clinically
localized prostate cancer. JAMA; 280(11):975–980.
Amdahl, J., Manson, S.C., Isbell, R. et al. (2014). Cost-effectiveness of pazopanib in
advanced soft tissue sarcoma in the United Kingdom. Sarcoma; 2014:481071.
396 References

American Cancer Society. (2018). Breast cancer survival rates. Retrieved from: www.
cancer.org/cancer/breast-cancer/understanding-a-breast-cancer-diagnosis/
breast-cancer-survival-rates.html.
Amico, M., & Van Keilegom, I. (2018). Cure models in survival analysis. Annual
Review of Statistics and Its Application; 5(1):311–342.
Andersen, P.K., Esbjerg, S., & Sorensen, T.I. (2000). Multi-state models for bleeding
episodes and mortality in liver cirrhosis. Statistics in Medicine; 19(4):587–599.
Andersen, P.K., & Keiding, N. (2002). Multi-state models for event history analysis.
Statistical Methods in Medical Research; 11(2):91–115.
Armero, C., Cabras, S., Castellanos, M.E., et al. (2016). Bayesian analysis of a dis-
ability model for lung cancer survival. Statistical Methods in Medical Research;
25(1):336–351.
Bongers, M.L., de Ruysscher, D., Oberije, C., et al. (2016). Multistate statistical model-
ing: A tool to build a lung cancer microsimulation model that includes parame-
ter uncertainty and patient heterogeneity. Medical Decision Making; 36(1):86–100.
Cancer Patients Alliance. (n.d.). Pancreatic cancer prognosis & survival. Pancreatica.
Retrieved from: https://pancreatica.org/pancreatic-cancer/pancreatic-cancer-
prognosis/.
Coyle, D., & Coyle, K. (2014). The inherent bias from using partitioned survival mod-
els in economic evaluation. Value in Health; 17(3):A194.
Cronin, A. (2016). STRMST2: Stata module to compare restricted mean survival
time. Statistical software components S458154. Boston, MA: Boston College
Department of Economics.
de Bock, G.H., Putter, H., Bonnema, J. et al. (2009). The impact of loco-regional recur-
rences on metastatic progression in early-stage breast cancer: A multistate
model. Breast Cancer Research and Treatment; 117(2):401–408.
de Glas, N.A., Kiderlen, M., Vandenbroucke, J.P. et al. (2016). Performing survival
analyses in the presence of competing risks: A clinical example in older breast
cancer patients. Journal of the National Cancer Institute; 108(5):djv366.
de Wreede, L.C., & Fiocco, M.,(2010). The mstate package for estimation and pre-
diction in non- and semi-parametric multi-state and competing risks models.
Computers Methods and Programs in Biomedicine, 261–274.
de Wreede, L.C., Fiocco, M., & Putter, H. (2011). Mstate: An R package for the analysis
of competing risks and multi-state Models. Journal of Statistical Software; 38(7).
Elgalta, R., Putter, H., van der Hage, J. et al. (2006). Estimation and prediction in a
multi-state model for breast cancer. Biomedical Journal; 48:366–380.
Gardiner, J.C. (2010). Survival analysis: Overview of parametric, nonparametric and
semiparametric approachesand new developments, Paper 252-2010, SAS Global
Forum 2010, Seattle WA, April 11–14. Retrieved from: https://support.sas.com/
resources/papers/proceedings10/252-2010.pdf.
Gelber, R.D., Goldhirsch, A., Cole, B.F. et al. (1996). A quality-adjusted time without
symptoms or toxicity (Q-TWiST) analysis of adjuvant radiation therapy and
chemotherapy for resectable rectal cancer. Journal of the National Cancer Institute;
88(15):1039–1045.
Glasziou, P.P., Simes, R.J., & Gelber, R.D. (1990). Quality adjusted survival analysis.
Statistics in Medicine; 9(11):1259–1276.
Goldhirsch, A., Gelber, R.D., Simes, R.J. et al. (1989). Costs and benefits of adjuvant
therapy in breast cancer: A quality-adjusted survival analysis. Journal of Clinical
Oncology; 7(1):36–44.
References 397

Hansen, L.S., Andersen, P.K., & Keiding, N. (1991). Assessing the influence of revers-
ible disease indicators on survival. Statistics in Medicine; 10(7):1061–1067.
Husson, O., Steenbergen, L.N., Koldewijn, E.L. et al. (2014). Excess mortality in
patients with prostate cancer. BJU International; 114:691–697.
Irwin, J.O. (1949). The standard error of an estimate of expectation of life, with special
reference to expectation of tumourless life in experiments with mice. Journal of
Hygiene; 47(2):188.
Karrison, T. (1987). Restricted mean life with adjustment for covariates. Journal of the
American Statistical Association; 82(400):1169–1176.
Lai, X., & Zee, B.C.Y. (2015). Mixed response and time-to-event endpoints for multi-
stage single-arm phase II design. Trials; 16:250.
Ledermann, J.A. (2013). Ecco 2013. Retrieved from: ww.cancernetwork.com/
conference.../slide-show-2013-european-cancer-congress.
Ledermann, J.A., Embleton, A.C., Perren, T. et al. (2017). Overall survival results of
ICON6: A trial of chemotherapy and cediranib in relapsed ovarian cancer.
Journal of Clinical Oncology; 35(15_suppl):5506–5506. Retrieved from: www.
icon6.org/media/1390/asco_2017.pdf.
Lewsey, J.D., Williams, C., Mackay, D.F. et al. (2017). Estimation of survival prob-
abilities for use in cost-effectiveness analyses: A comparison of a multi-state
modeling survival analysis approach with partitioned survival and Markov
decision-analytic modeling. Medical Decision Making; 37(4):427–439.
Manzini, G., Ettrich, T.J., Kremer, M. et al. (2018). Advantages of a multi-state
approach in surgical research: How intermediate events and risk factor pro-
file affect the prognosis of a patient with locally advanced rectal cancer. BMC
Medical Research Methodology; 18(1):23.
Meier-Hirmer, C., & Schumacher, M. (2013). Multi-state model for studying an inter-
mediate event using time-dependent covariates: Application to breast cancer.
BMC Medical Research Methodology; 13:80.
Murray, T.A., Thall, P.F., Yuan, Y. et al. (2017). Robust treatment comparison based
on utilities of semi-competing risks in non-small-cell lung cancer. Journal of the
American Statistical Association; 112:11–23.
National Cancer Institute. (n.d.). Dictionary of cancer terms. Retrieved from: www.
cancer.gov/publications/dictionaries/cancer-terms/.
National Institute for Health and Care Excellence. (2009b). Rituximab for the first-
line treatment of chronic lymphocytic leukaemia. Technology appraisal guid-
ance [TA174]. Retrieved from: www.nice.org.uk/guidance/TA174.
Ng, R., Kornas, K., Sutradhar, R. et al. (2018). The current application of the Royston-
Parmar model for prognostic modeling in health research: A scoping review.
Diagnostic and Prognostic Research; 2(1):4.
Nishino, M., Jagannathan, J.P., Krajewski, K.M. et al. (2012). Personalized tumor
response assessment in the era of molecular medicine: Cancer-specific and
therapy-specific response criteria to complement pitfalls of RECIST. AJR.
American Journal of Roentgenology; 198(4):737–745.
Oaknin, A. (2015). XVII Simposio de Revisiones en Cancer. Madrid 11–13 February
2015; accessed March 2018.https://seom.org/agenda2/106803-xxi-simposio-
de-revisiones-en-cancer-2019.
Oxnard, G.R., Morris, M.J., Hodi, F.S. et al. (2012). When progressive disease does not
mean treatment failure: Reconsidering the criteria for progression. JNCI Journal
of the National Cancer Institute; 104(20):1534–1541.
398 References

Parikh, R.C., Du, X.L., Morgan, R.O. et al. (2016). Patterns of treatment sequences in
chemotherapy and targeted biologics for metastatic colorectal cancer: Findings
from a large community-based cohort of elderly patients. Drugs Real World
Outcomes; 3(1):69–82.
Parmar, M.K.B., & Royston, F. (2013). Restricted mean survival time: An alternative to
the hazard ratio for the design and analysis of randomized trials with a time-
to-event outcome. BMC Medical Research Methodology; 13(1):15.
Parner, E.T., & Andersen, P.K. (2010). Regression analysis of censored data using
pseudo-observations. The STATA Journal; 10(3):408–422.
Peng, Y., & Taylor, J.M.G. (2014). Chapter 6: Cure models. In: J. Klein, H. van
Houwelingen, J.G. Ibrahim, T.H. Scheike (Eds.). Handbooks of modern statistical
methods series: Handbook of survival analysis. Boca Raton, FL: Chapman & Hall.
Putter, H., Fiocco, M., & Geskus, R.B. (2007). Tutorial in biostatistics: Competing risks
and multi-state models. Statistics in Medicine; 26(11):2389–2430.
Ratkowsky, D.A. (1989). Handbook of nonlinear regression models. New York: Marcel
Dekker.
Rodriguez, G. (2010). Parametric survival models. Retrieved from: ata.princeton.
edu/pop509/ParametricSurvival.pdf.
Royston, F. (2015). Restricted mean survival time: Calculation and some applications
in trials and prognostic studies. Flexible parametric survival models workshop.
Stockholm 10 November 2011. Retrieved from: www2.le.ac.uk/Members/pl4/
workshop2011-1/Royston-Stockholm-10nov2011b.pdf.
Seruga, B., Pond, G.R., Hertz, P.C. et al. (2012). Comparison of absolute benefits of
anticancer therapies determined by snapshot and area methods. Annals of
Oncology; 23(11):2977–2982.
Tai, B.C., Wee, J., & Machin, D. (2011). Analysis and design of randomised clinical tri-
als involving competing risks outcomes. Trials; 12:127.
Titman, A. (2016). Multi-state models: An overview. Presentation. Retrieved from:
www.maths.lancs.ac.uk/~titman/leeds_seminar.pdf.
Trinquart, L., Jacot, J., Conner, S.C. et al. (2016). Comparison of treatment effects
measured by the hazard ratio and by the ratio of restricted mean survival
times in oncology randomized controlled trials. Journal of Clinical Oncology;
34(15):1813–1819.
Uhry, Z., Hédelin, G., Colonna, M. et al. (2010). Multi-state Markov models in can-
cer screening evaluation: A brief review and case study. Statistical Methods in
Medical Research; 19(5):463–486.
University of Texas at Dallas. (n.d.). Markov chains. Retrieved from: www.utdallas.
edu/~jjue/cs6352/markov/markov.html.
U.S. Department of Health and Human Services. Food and Drug Administration.
(2007). Clinical trial endpoints for the approval of cancer drugs and biolog-
ics: Guidance for industry. Center for Drug Evaluation and Research (CDER),
Center for Biologics Evaluation and Research (CBER). Retrieved from: www.
fda.gov/downloads/Drugs/Guidances/ucm071590.pdf.
Van Erning, F.N., van Steenbergen, L.N., Lemmens, V.E.P.P. et al. (2014). Conditional
survival for long-term colorectal cancer survivors in the Netherlands: Who do
best? European Journal of Cancer; 50(10):1731–1739.
Van Houwelingen, H., & Putter, H. (2012). Dynamic prediction in clinical survival analy-
sis. Boca Raton, FL: CRC Press, Taylor & Francis.
References 399

Willekens, F.J., & Putter, H. (2014). Software for multistate analysis. Demographic
Research; 31:381–420.
Williams, C., Lewsey, J.D., Briggs, A.H. et al. (2017). Cost-effectiveness analysis in R
using a multi-state modeling survival analysis framework: A tutorial. Medical
Decision Making; 37(4):340–352.

Chapter 5
Department of Health and Social Care. (n.d.). NHS prescription services. Retrieved
from: www.nhsbsa.nhs.uk/nhs-prescription-services.
Fundamental Finance. (n.d.). Marginal Cost (MC) & Average Total Cost (ATC).
Retrieved from: http://economics.fundamentalfinance.com/micro_atc_mc.php.
National Institute for Sickness and Disability Insurance. (n.d.). INAMI home page.
Retrieved from: www.inami.fgov.be/fr/Pages/default.aspx.

Chapter 7
Anderson, D.F., & Kurtz, T. (2015). Stochastic analysis of biochemical systems, Stochastics
in Biological Systems Series, Volume 1.2. Springer International Publishing.
Caro, J., Möller, J., Karnon, J. et al. (2016). Discrete event simulation for health technology
assessment. New York: Chapman and Hall/CRC.
Parikh, R.C., Du, X.L., Robert, M.O. et al. (2017). Cost-effectiveness of treatment
sequences of chemotherapies and targeted biologics for elderly metastatic
colorectal cancer patients. Journal of Managed Care and Specialty Pharmacy; 23(1):64.
Saint-Pierre, P. (2016). Multi-state models and cost-effectiveness analysis. Journal de
Gestion et d’Économie Médicales; 34(2):133.
Salleh, S., Thokala, P., Brennan, A. et al. (2017). Discrete event simulation-based
resource modelling in health technology assessment. Pharmacoeconomics;
35(10):989.
Zeng, X., Li, J., Peng, L. et al. (2014). Economic outcomes of maintenance gefitinib for
locally advanced/metastatic non-small-cell lung cancer with unknown EGFR
mutations: A semi-Markov model analysis. PLoS One; 9(2):e88881. eCollection 2014.

Chapter 9
Bang, H., & Zhao, H. (2014). Cost-effectiveness analysis: A proposal of new reporting
standards in statistical analysis. Journal of Biopharmaceutical Statistics. Taylor &
Francis.
Bang, H., & Zhao, H. (2016). Median-based incremental cost-effectiveness ratios with
censored data. Journal of Biopharmaceutical Statistics; 26(3):552–564.
Bebu, I., Luta, G., Mathew, T. et al. (2016). Parametric cost-effectiveness inference with
skewed data. Computational Statistics and Data Analysis; 94(February):210–220.
Brard, C., Le Teuff, G., Le Deley, M.C. et al. (2017). Bayesian survival analysis in clini-
cal trials: What methods are used in practice? Clinical Trials; 14(1):78–87.
Fan, M.Y., & Zhou, X.H. (2007). A simulation study to compare methods for con-
structing confidence intervals for the incremental cost-effectiveness ratio.
Health Services and Outcomes Research Methodology; 7(1–2):57–77.
400 References

Gordon, L.G., Tuffaha, H.W., & Scuffham, P.A. (2016). Efficient value of information
calculation using a nonparametric regression approach: An applied perspec-
tive. Value Health; 19(4):505–509. Epub 2016.
Grieve, R., Nixon, R., & Thompson, S.G. (2014). Bayesian hierarchical models for cost-
effectiveness analyses that use data from cluster randomized trials; healthcare:
A review of principles and applications. Journal of Medical Economics.
Hoch, J.S., & Dewa, C.S. (2014). Advantages of the net benefit regression framework
for economic evaluations of interventions in the workplace: A case study of
the cost-effectiveness of a collaborative mental health care program for peo-
ple receiving short-term disability benefits for psychiatric disorders. Journal of
Occupational and Environmental Medicine; 56(4):441–445.
Meads, D.M., Marshall, A., Hulme, C.T. et al. (2016). The cost effectiveness of docetaxel
and active symptom control versus active symptom control alone for refrac-
tory oesophagogastric adenocarcinoma: Economic analysis of the COUGAR-02
trial. Pharmacoeconomics; 34(1):33–42.
Stinnett, A.A., & Mullahy, J. (1998). Net health benefits. Medical Decision Making; 18(2
Suppl):S68–80.
Tuffaha, H.W., Reynolds, H., Gordon, L.G. et al. (2014). Value of information analy-
sis optimizing future trial design from a pilot study on catheter securement
devices. Society for Clinical Trials.11(6):648‒658
Weintraub, W.S., Daniels, S.R., Burke, L.E. et al. (2011). Value of primordial and
primary prevention for cardiovascular disease: A policy statement from the
American Heart Association. Circulation; 124(8):967–990; originally published
online. Retrieved from: http://circ.ahajournals.org/content/124/8/967/F2.
Wilson, E.C. (2015). A practical guide to value of information analysis.
Pharmacoeconomics; 33(2):105–121.
Wilson, E.C., Mugford, M., Barton, G. et al. (2016). Efficient research design: Using;
value-of-information analysis to estimate the optimal mix of top-down and
bottom-up costing approaches in an economic evaluation alongside a clinical
trial. Medical Decision Making; 36(3):335–348.
Zhao, H., & Bang, H. (2012). Pharmacoeconomics. 2015 February; 33(2):105–121. doi:
10.1007/s40273-014-0219-x. PubMed PMID: 24194511. PMID: 25336432. PubMed
PMID: 24650041.
Zhao, H., Zuo, C., Chen, S., & Bang, H. (2012). Nonparametric inference for median
costs with censored data. Biometrics; 68(3):717–725.
Index

1:1 matching, 271 Australia, 332, 344, 348–349, 352


21st Century Cures Act (2016) (US), 255 Austria, 57, 345
Automatic funding, for highly
Accelerated failure time (AFT), 123, specialized drugs for rare
129, 131 diseases, 359
ACD, see Advanced consultation Average total cost (ATC), 151
document Average versus marginal and
Adjusting for crossover, 45 incremental cost, 151–152
Advanced consultation document Axitinib, 353
(ACD), 355 Azacitidine, 328, 353
AEMPS, see Agencia Espanola de
Medicamentos y Productos Baseline characteristics, 272
Sanitarios and health resource use, 143–144
Afatinib, 87, 216, 239, 240, 243, 244, 353 Baseline hazard function, 112
Aflibercept, 327 Basket trials, 358
AFT, see Accelerated failure time Bayesian sensitivity analysis, 306–307
Agencia Espanola de Medicamentos y ICER and INMB limitations and,
Productos Sanitarios 307–308
(AEMPS), 348 PSA, 319–320
Agency for Healthcare Research and BB, see Beta binomial (BB) distribution
Quality (AHRQ), 344 Belgium, 57, 260, 316, 345
AHRQ, see Agency for Healthcare Best supportive care (BSC), 194, 233, 234,
Research and Quality 293, 294
Akaike’s Information Criterion, 84 Beta binomial (BB) distribution, 320
Alemtuzumab, 181, 182 prior, and binomial likelihood,
Allocative efficiency, 14–15 321–322
American Institute of Cancer Bevacizumab, 314, 325, 326, 327, 337,
Research, 341 338, 339
American Society of Clinical Oncology Bilateral switch, 124
(ASCO), 7, 24, 343 BIMs, see Budget impact models
Aminoglycoside, 223 Bioequivalence trials, 29
AMNOG law (Germany) (2011), 24, 347 Biologics, 31
Anastrazole, 55 Biomarker, 5–6, 15, 252
Anti-cancer therapies, confounding Biosimilars, see Equivalence trials
from, 43–44 Blinatumomab, 355
Area under the curve (AUC), 72, 73, 74 Blood cancer, 10
Article 81 agreement, 316 BNF, see British National Formulary
ASCO, see American Society of Clinical Bootstrapping, 108–110, 286
Oncology Bortezomib, 152, 197, 355, 353
ATC, see Average total cost Bosutinib, 152, 299, 327, 354
Atezolizumab, 52, 55 Bottom-up approach, 150
AUC, see Area under the curve Bowel cancer
Audits, 261–262 cost of treating, 8

401
402 Index

Bragg and Packer model, 83 Capecitabine, 326


Breast cancer, 2, 4, 31, 60, 64, 65 Carboplatin, 241, 327, 328
cost of treating, 8 Carfilzomib, 355
typical health resource use in trial Case report form (CRF), 158, 164, 165,
of, 142 166, 178, 191, 192
Brexit, 352 design, 199
British National Formulary (BNF), 16 CCGs, see Clinical commissioning
BSC, see Best supportive care groups
Budget impact models (BIMs), 13 CCyR, see Cytogenetic response
Budget impact threshold, 359 CDF, see Cancer drug fund
CDR, see Common Drug Review
Cabazitaxel, 327, 354 CEA, see Cost-effectiveness analysis
Cabozanitib, 210–212, 232, 233, 234, 235, CEAC, see Cost-effectiveness
340, 355 acceptability curve
CADTH, see Canadian Agency for Drugs Censored and missing costs, 158–160,
and Technologies in Health 171–172
Canada, 154, 344 costs data analysis strategies, 160–161
Canadian Agency for Drugs and imputation methods, 161–162
Technologies in Health missing resource data avoiding
(CADTH), 344, 345 strategies, 160
Cancer; see also individual entries Center for Medicare Services
definition of, 1 (CMS), 155
drug development in practice, and Ceritinib, 354
health economic evaluation, Cervical cancers, 2
23–24 CESP, see Economic Committee of
modern paradigm, 24–26 Health Products (CESP)
economic burden of Cetuximab, 152, 325, 326, 339, 355
healthcare expenditure on drugs, Charges, 154
7–10 cost-to-charge ratios, 155
health expenditure, 6–7 non-medical costs, 155
epidemiology of, 1–2 CHEERS, see Consolidated Health
cancer trends, 2, 5 Economic Evaluation
interventions, important economic Reporting
concepts for cost effectiveness Chemoradiotherapy (chemo-RT), 40
of, 12 Chemo-RT, see Chemoradiotherapy
economics, health economics, Chemotherapy side effects, costs and
economic evaluation, and effects of treating, 223
pharmacoeconomics, 12–23 Chronic lymphocytic leukaemia,
outcomes, prognostic factors 47, 176, 181
associated with, 5–6 Chronic myeloid leukemia (CML), 3,
treatments for, 10–11 53, 299
Cancer drug fund (CDF), 8, 324, 349 CI, see Confidence interval
Cancer economic models, criticisms of, Cisplatin, 242, 328, 337
324, 329–334 Client services receipt inventory
Cancer registries and real-world data, (CSRI), 156
265–266 Clinical commissioning groups
working of, 266–268 (CCGs), 344
Cancer surveillance, applications in, Clinical Practice Research Datalink
119–122 (CPRD), 262
Index 403

Clinical trial and real-world setting, perspective, and cost collection,


42–43 145–146
Clinicaltrials.gov, 333 statistical analysis and, 165–172
CML, see Chronic myeloid leukemia types, typical of cancer trials, 141
CMS, see Center for Medicare Services baseline characteristics and health
Cobimetinib, 328 resource use, 143–144
Colorectal carcinoma, 3, 4 costs determined by study
Commercial data sources, 264 protocol, 144–145
Common Drug Review (CDR), 345 health resource use
Complete response (CR), 55 categorization, 142
Compliance bias, 281 health resource use monitoring,
Compliance rates, comparison of, 276 142–143
Condition-specific preference measure Cost-effectiveness acceptability curve
(CS-PBM), 70 (CEAC), 293, 305, 309, 320
Confidence interval (CI), 30, 109, 307 Cost-effectiveness analysis (CEA), 15, 54,
Confounding bias, 263, 268, 269, 276, 277, 57, 92, 123
279, 281 switching impact on, 127–128
Consolidated Health Economic Cost-effectiveness designing
Evaluation Reporting case study
(CHEERS), 279 TA516 cabozanitib + vandetinib,
Copulas, 302 210–212
Correlation, significance of, 109 clinical trial designs
Cost analysis adaptive designs, 183, 185–186
case report forms (CRF) and health multi-arm, multi-stage (MAMS),
resource use, 164, 165, 166 186–187
censored and missing costs and, observational studies, 189–190
158–160, 171–172 open label extensions (OLE), 189
costs data analysis strategies, parallel groups and crossover
160–161 designs, 178–179, 180
imputation methods, 161–162 retrospective case control studies,
missing resource data avoiding 190
strategies, 160 single arm phase II designs,
charges and, 154 179–183, 184
cost-to-charge ratios, 155 CRF design, 199
non-medical costs, 155 economic data collection reasons in
cost distribution and, 155–157 clinical trial, 175–178
cost data transformation, health economic evaluation planning
157–158 in clinical trial
future costs and, 162–164 designing considerations,
health resource use collection, across 191–194
treatment pathway, 146–148 early ICER/INMB, 206–207
time horizon, 148–150 economic evaluation integration
methods, micro versus macro in clinical trial, 194–195
approach, 150 endpoints and outcomes, 196–204
average versus marginal and generic/competition entry
incremental cost, 151–152 time, 204
inflation, 152–153 multicenter trials, 207–209
time preference and discounting, subgroups/heterogeneity
153–154 identification, 206
404 Index

treatment compliance, 205–206 Decision tree models, 215, 222–223


treatment pathways, 204 outcome valuation, 224–226
measurement timings and, 198 Denmark, 266
sample size methods for cost- Denosumab, 353
effectiveness, 199–201 DES, see Discrete event simulation, 243
formulae, 201–204 Dexamethasone, 116, 117, 118, 206, 327
trial design, 198 DFS, see Disease-free survival
Cost-effectiveness plane, 19–22 Diagnostic-related group (DRG), 154
Cost-effectiveness threshold, changing, Diary data, 199
21–22 DICE, see Discretely integrated
Cost-minimisation analysis (CMA), condition event (DICE)
216, 240 Dictionary of Cancer Terms (National
Cost-to-charge ratios, 155 Cancer Institute), 1
Cost-utility analysis (CUA), 15, 72, 216, 221 Diffuse large B-cell lymphoma
Cox proportional hazard (Cox PH) (DLBCL), 3, 204, 205
model, 111–112, 114 Digital data sources, social media and
adjusting for covariates with, 112 applications, 263–264
results interpretation from, 112–113 Direct comparison, 133
CPRD, see Clinical Practice Research Disability-adjusted life-years (DALYs),
Datalink 197
CPT, see Current procedural Discounting, 17–18; see also Future costs
terminology Discrete event simulation (DES), 243
CR, see Complete response Discretely integrated condition event
Credible interval, 307 (DICE), 212
CRF, see Case report form Disease-free survival (DFS), 37
Criotinib, 330 Distribution of costs, 94, 141, 155–158
Crizotinib, 87, 216, 217, 239, 240, 245, DLBCL, see Diffuse large B-cell
327, 354 lymphoma
Crossover, see Treatment switching Docetaxel, 9, 241, 325, 328
Cross-walking, 75 Dolan UK tariff, 68
CS-PBM, see Condition-specific DoR, see Duration of response
preference measure Doxorubicin, 204
CSRI, see Client services receipt DRG, see Diagnostic-related group
inventory (CSRI) (DRG)
CUA, see Cost-utility analysis Drug Reimbursement Commission, 316
Current procedural terminology Dual-price system, 16
(CPT), 155 Dupuit, Jules, 14
Cyclophosphamide, 204 Duration of response (DoR), 38
Cytogenetic response (CCyR), 53
EAMS, see Early access to medicines
Dabrafenib, 340, 353, 354 scheme
DALYs, see Disability-adjusted life-years Early access to medicines scheme
Dasatinib, 326, 327, 354, 355 (EAMS), 175, 182, 252
Data collection form (DCF), see Case Eastern Cooperative Oncology Group
report form (ECOG), 5, 6, 143, 144, 156,
Data synthesis and network 268, 334
meta-analyses, 132 ECOG, see Eastern Cooperative
mixed treatment comparisons Oncology Group
(MTC), 132–139 Economic burden of cancer, see Cancer
Index 405

Economic Committee of Health HTAs with, 53–55


Products (CESP), 345 tumor-centred, 36, 37
Economic evaluation, importance of, 13, immune-related response criteria
61; see also individual entries (irRC), 55–56
Economic evaluation models minimal residual disease
continuous time Markov models, 230 (MRD), 56
decision tree models, 215, 222–223 pathological complete response
outcome valuation, 224–226 (pCR), 56–57
health economic model types and, value demonstration, 57–58
215, 216–221 ENMB, see Expected net monetary
HTA submissions and cost- benefit
effectiveness models, 239, Enzalutamide, 353, 354
244–247 EORTC, see European Organization for
Markov models, 226–229 Research and Treatment of
partitioned survival model, 231 Cancer
case study, 232–236 Epidermal growth factor receptor
crossover, 236–238 (EGFR), 5–6, 244, 246
economic model using patient- EQ-5D, 22, 242, 330
level data, 231–232 sensitivity and responsiveness
Economic versus clinical hypotheses, 29 versus QLQ-C30, 76–77
equivalence hypothesis, 31–32 Equivalence trials, 29, 30, 31–32
non-inferior equivalence Eribulin, 326, 339, 340, 354
hypothesis, 32 Erlotinib, 6, 9, 87, 192, 206, 217, 218, 241,
superiority hypothesis, 29–31 245, 293, 305, 325, 326, 338,
e-FAP, 264 353, 354
Efficacy versus effectiveness, 26–27 comparison with gefitinib, 135–139
EFS, see Event-free survival Estimands, 125, 126, 237
EGFR, see Epidermal growth factor EUnetHTA, see European Network of
receptor Health Technology assessment
EINB, see Expected incremental net European Medicines Agency (EMA), 15,
benefit 31, 47, 48, 332
Electronic health records (EHR), 250, European Medicines Evaluation Agency
255–256, 257, 260, 263, 265, 267 (EMEA), 255
EMA, see European Medicines Agency European Network of Health
EMEA, see European Medicines Technology assessment
Evaluation Agency (EUnetHTA), 47, 348
ENB, see Expected net benefit European Organization for Research
ENBS, see Expected net benefit of and Treatment of Cancer
sampling (EORTC), 63
End-of-life treatment, 305 European Society of Medical Oncology
Endpoints, 35 (ESMO), 343
overall survival (OS), 36, 37, 39–41, 48 EuroQol
and economic evaluation, 41–46 EQ-5D-3L, 23, 68–69
relationship with ORR, 48, 50 EQ-5D-5L, 68, 69–70
relationship with PFS, 50–51 Event-free survival (EFS), 38
relationship with response, 48 Everolimus, 326, 327, 354, 355
patient-centred, 36, 37 Evidence review group (ERG), see under
surrogate, 46–53 National Institute of Health
advantage of, 47 and Care Excellence (NICE)
406 Index

EVPI, see Expected value of perfect Follicular lymphoma, 351, 352


information Food and Drug Administration (FDA),
EVPPI, see Expected value of partially 15, 56
perfect information Amendments Act (FDAAA), 253
EVSI, see Expected value of sample France, 57, 260, 266, 344, 345–346
information Free pricing, 24, 176
EXAM trial, 233 Fulvestrant, 326, 339
Exchangeability, 135 Functional Assessment of Cancer
Exemestane, 327 Therapy (FACT), 62, 63
Expected incremental net benefit Functional domains, 66
(EINB), 309, 310 Future costs, 149, 150, 162–164; see also
Expected net benefit (ENB), 309, 320 Discounting
Expected net benefit of sampling
(ENBS), 310 Gastric cancer, 3
Expected net monetary benefit (ENMB), G-BA, 346
311, 312, 313 GDPR, see General Data Protection
Expected value of partially perfect Regulation
information (EVPPI), 310, 315 Gefitinib, 9, 88, 177, 218, 219, 241, 246, 328,
Expected value of perfect information 338, 353
(EVPI), 310, 315 comparison with erlotinib, 135–139
Expected value of sample information Gemcitabine, 9, 55, 242, 325, 327, 328
(EVSI), 310–311 General Data Protection Regulation
description of, 311–314 (GDPR), 42
Expenditure, 6–7, 32, 162, 188 Generalized linear models (GLMs), 166,
Exponential model, 114, 115 274, 319
Extrapolation, 47, 52, 54, 78, 85, 86, 121, Generic preference-based instrument
193, 212, 216–220, 245, 332 (G-PBM), 70
of costs, 163 Germany, 24, 57, 176, 196, 197, 266, 332,
of survival data, 114–118, 119, 290 341, 345, 346–347
G-estimation, 130
FACT, see Functional Assessment of Glioblastoma, 3, 48, 49
Cancer Therapy trials, 50, 51, 149
FACT-B, 65 GLMs, see Generalized linear models
FACT-G, 63, 64, 66, 237 Global Cancer Incidence, Mortality and
FACT-Lymp, 65 Prevalence (GLOBOCAN), 1
FACT-Ntx, 65 GLOBOCAN, see Global Cancer
FACT-O, 65 Incidence, Mortality and
Failure rate, see Hazard rate Prevalence
Fast-track appraisals, 359–360 Gompertz model, 123
FDA, see Food and Drug Administration G-PBM, see Generic preference-based
Fingolimod, 353 instrument
Five-parameter model, 84 Group sequential designs, 183, 185–186
Fleishman method, 302, 303, 318
Flexible parametric survival models, 118 Half-cycle correction (HCC), 229
censoring and, 120–122 Hawthorne effect, 205
in lung cancer trial, 119–120 Hazard rate, 98–99, 109
Fludarabine, 55, 181, 182, 325 relationship with survival rate, 102–103
Fluorouracil, 325, 326, 328 Hazard ratio (HR), 30, 99–101, 109
Folinic acid, 325, 326 to predict survival rates, 113–114
Index 407

HCA, see Human capital approach (HCA) Health technology assessments


HCC, see Half-cycle correction (HTAs), 44, 45, 176; see also
Healthcare resource, 6, 12, 63, 143, 153, Reimbursement
176, 253, 263 cost-effectiveness future, 356
Health economic analysis plans future reimbursement landscape,
(HEAPs), 358 358–359
Health economics, 5, 12–13, 15, 16–17, research methodology, 356–358
23–26, 71, 188, 191–209, 254, 346, criticism across, 330–331
347–348, 356, 357 factors predictive of successful,
models, 2215, 216–221, 239, 240–242, 335–341
244–247, 329 risk-sharing scheme and, 352–356
Health Products Pricing Committee submissions and cost-effectiveness
(Comité Economique des models, 239, 244–247
Produits de Santé) (France), 16 with surrogate endpoints, 53–55
Health-related quality of life (HRQoL), value-based pricing (VBP) for cancer
1, 10, 17, 22–23, 36, 37, 48, 51, drugs and, 350–352
59, 192, 196, 198, 237, 263, Health utility index (HUI), 67
264–265, 358 HEAPs, see Health economic analysis
anti-cancer treatment limitations, plans
59–60 Hematological toxicity, 116
challenges with, 61–62 Hepatocellular carcinoma, 3, 113
condition-specific measures of, 62–63 cost-effectiveness analysis of
common general measures, 63–67 treatments for, 278
data collection reasons, 60–61 OS in, 100
economic evaluation in utility data HER, see Electronic health records (EHR)
absence, 74–75 Herceptin, 31, 56
EQ-5D sensitivity and HES, see Hospital Episodes Statistics
responsiveness versus QLQ- Hospital Episodes Statistics (HES),
C30, 76–77 260, 270
instruments, 64–65 HR, see Hazard ratio
issues, in cancer drugs health HRQoL, see Health-related quality of
technology appraisals, 87–89 life
measuring, for economic evaluation, HTAs, see Health technology
67–68 assessments
EuroQol EQ-5D-3L, 68–69 HUI, see Health utility index
EuroQol EQ-5D-5L, 69–70 Human capital approach (HCA), 155
post-progression (PP) utility Hurdle model, 274, 275
measurement, 77–78 Hypothesis testing, 91, 93, 306
non-linear methods, 82–86
plausible post-progression utility Ibrutinib, 355
behavior, 80–81 ICD-9 classification, 278
utility behavior in cancer patients, ICER, see Incremental cost-effectiveness
79–80 ratio
utility estimation between disease ICH E9 Addendum, 125
progression and death, 78–79 IDMC, see Interim Data monitoring
QALYs and, 72–73, 289, 290–292 committees (DMC)
calculation in cancer trials, 73–74 Imatinib, 326, 328
reduced, 57 Immune-related response criteria
utilities construction, 70–72 (irRC), 55–56
408 Index

Improvement.nhs.uk, 239 Inverse probability-of-censoring


Imputation methods, 161–162 weighting (IPCW), 126,
Incidence, meaning of, 98 128–129, 237
Incremental cost, 20 IPCW, see Inverse probability-of-
Incremental cost-effectiveness ratio censoring weighting
(ICER), 18–19, 24, 27, 44, 45, Ipilimumab, 340, 353
48, 92, 108, 132, 145, 157, 164, IQWiG, see Institute for Quality and
192, 207, 210, 245, 247, 286, 315, Efficiency
335–336, 341, 343, 350, 359 Irinotecan, 325, 327, 328
acceptable, 303 irRC, see Immune-related response
interpreting, and cost-effectiveness criteria
plane, 292 ISPOR, see International Society of
uncertainty, 292–296 (see also Pharmacoeconomics and
Sensitivity analysis) Outcomes Research
value of information (VOI) analyses Italy, 57, 154, 207, 266, 345, 347, 352
and results, 308–314, 320–321 ITC, see Indirect treatment comparison
bevacizumab trial, 314–315 ITT, see Intention-to-treat
challenges in healthcare
decisions, 316–317 Kaplan Meier (KM) curve/plot, 36, 40,
Incremental costs, interpreting, 283–284 41, 73, 95–98, 101, 104, 107, 111,
informative censoring and, 284–287 114, 116, 120, 171, 212, 231, 236,
Incremental effectiveness, 20 284, 314, 315
Incremental net monetary benefit QALY derivation using, 287–289
(INMB), 19, 67, 93, 168, 206–207, KM, see Kaplan Meier (KM) curve/plot
208–209, 304, 307 Kolmogorov-Smirnov test, 157
Indirect treatment comparison
(ITC), 133 Last observation carried forward
Inflation, 152–153 (LOCF), 149, 161
Information bias, 281 Lenalidomide, 16, 116, 197, 206, 262, 353
Information Exchange and Data cost-effectiveness of, 115–116
Transformation (INFORMED), Leptokurtic distribution, 156
255 Leucovorin, 328
INFORMED, see Information Exchange Liver cancer, 2, 4
and Data Transformation LOCF, see Last observation carried
INMB, see Incremental net monetary forward
benefit Log rank test, 109
Institute for Quality and Efficiency Lung cancer, 2, 3, 4, 42, 63, 64
(IQWiG), 24, 332, 344, 346 cost of treating, 8
Institute of Clinical and Economic cost-effectiveness models used in,
Review, 343 240–242
Instrumental variable methods, 274, 276 data, 136
Insurance claims databases, 263 HTA submissions key criticisms,
Intention-to-treat (ITT), 27, 44, 103, 244–247
127–128, 159, 161, 237 incidence, 5
Interim analyses and Data Monitoring published costs and QALYs in, 9–10
Committees (IDMC), 188 trial, ICER and cost-effectiveness
International Society of from, 293–295
Pharmacoeconomics and Luxembourg, 345
Outcomes Research (ISPOR), 132 Lymphoma/myeloma, 64, 65
Index 409

M:n matching, 271 Model fitting, issues in, 122–123


Major molecular response (MMR), 53 Modified intention-to-treat (mITT), 27
MAMS, see Multi-arm, multi-stage Monte-Carlo sampling, 111
Managed care organizations (MCO), 349 Monte-Carlo simulation, 301, 306
Many-to-one matching, 271 MPFS, see Medicare Physician Fee
MAR, see Missing at random Schedule
Marginal structural model (MSM), 128 MRD, see Minimal residual disease
Markovian process, 103 MSM, see Marginal structural model
Markov models, 226–229 MTA, see Multiple technology appraisal
continuous time, 230 MTC, see Mixed treatment comparisons
Maximum tolerated dose (MTD), 35 MTD, see Maximum tolerated dose
MCAR, see Missing completely at Multi-arm, multi-stage (MAMS),
random 186–187
MCO, see Managed care organizations Multicenter trials, 207
MDS, see Myelodysplastic syndrome Multidisciplinary team (MDT), 24–25
MDT, see Multidisciplinary team Multilevel models (MLM), 208
Mean, 109 Multinational trials, 332, 333
Mean health resource use, 167 Multiple imputation (MI), 161, 286
Mean incremental cost, 161 Multiple myeloma, 355
Mean survival and restricted mean, bortezomib for, 355–356
106–108 lenalidomide for, 356
MED, see Minimum effective dose Multiple technology appraisal (MTA),
Median, 109 323, 324
Median survival, 96–98 Myelodysplastic syndrome (MDS), 25
Medicare Physician Fee Schedule Myeloma cancer trial, modeling costs
(MPFS), 155 from, 285
Melanoma, 3
Meta-analysis, 134 National Cancer Institute, 1
MI, see Multiple imputation National Cancer Registration Service
Mifamurtide, 353 (NCRS), 267
Minimal residual disease (MRD), 56 National Cancer Registry (NCR), 28, 260
Minimum effective dose (MED), 35 National Comprehensive Cancer
Missing at random (MAR), 159 Network (NCCN), 24
Missing completely at random National Health Service (NHS), 7, 145,
(MCAR), 159 176, 182, 239
Missing data, 52, 62, 125, 156, 158–159, England (NHS-E), 358, 359
160, 161, 199, 237, 255, 256, 286 National Health Service number, 260
Missing not at random (MNAR), 159 National Institute for Health Research
MITT, see Modified intention-to-treat (NIHR), 176
Mixed effect models, 166 National Institute of Health (NIH), 46
Mixed treatment comparisons (MTC), National Institute of Health and Care
132–134, 194, 212, 244 Excellence (NICE), 8, 13, 25,
assumptions for carrying out, 45, 53, 57, 113, 145, 175, 176, 200,
134–139 212, 239, 334, 335, 344, 349, 352,
Mixed treatment comparisons (MTC), 355, 358, 359
132–139 cancer drugs rejected by, 323–324,
MLM, see Multilevel models 325–328
MMR, see Major molecular response Economic Review Group (ERG), 113,
MNAR, see Missing not at random 181, 182, 186, 244, 247
410 Index

Methods Guide, 47 Norway, 266


oncology treatments approved by, NPV, see Net present value
353–355 NSCLC, see Non-small-cell lung cancer
Single Technology Assessments in
Oncology, 360 Obinutuzumab, 353, 355
technical support document (TSD), 118 Objective response rate (ORR), 38, 46,
National Lung Cancer Audit Lung 48, 182
Cancer Data, 267 relationship with survival, 48, 50
NBM, see Negative binomial model O’Brien and Fleming rule, 186
NCCN, see National Comprehensive Observational research studies,
Cancer Network prospective, 265
NCR, see National Cancer Registry Observational studies, 189–190
NCRS, see National Cancer Registration Ofatumumab, 176, 181, 182, 184, 326, 353
Service Olaparib, 45, 354
Necitumumab, 328 OLE, see Open label extensions (OLE)
Negative binomial model (NBM), 274, 275 OLS, see Ordinary least squares
Neoadjuvant therapy, 56 One-way sensitivity analysis, 296–297,
Net health benefit (NHB), 307 298–299
The Netherlands, 57, 344, 345 Open label extensions (OLE), 189
Net monetary benefit (NMB), 307, Opportunity cost, 16–17
307–308 Ordinary least squares (OLS), 165, 168
Net present value (NPV), 153 ORR, see Objective response rate
Network meta-analysis (NMA), 134, 137 Outcomes curse, 224
Network of evidence, 134 Ovarian cancer, 64, 65
Neurological toxicity, 65 Overall survival (OS), 36, 37, 39–41,
Neutropenia, 251 48, 111, 113, 116, 127, 135,
Nexevar, 347 231–232, 236
NHB, see Net health benefit (NHB) and economic evaluation, 41–46
NHS, see National Health Service relationship with ORR, 48, 50
NICE, see National Institute of Health relationship with PFS, 50–51
and Care Excellence relationship with response, 48, 182
NIH, see National Institute of Health Oxaliplatin, 326
(NIH)
NIHR, see National Institute for Health Paclitaxel, 9, 241, 325, 327, 328, 355
Research PAE, see Potential adverse event
Nilotinib, 353, 355 Palbociclib, 251
Nintedanib, 353 Palliative care, 147, 148, 163
Nivolumab, 52, 79, 194, 330, 331, 354, 355 Palliative therapy, 59
NMA, see Network meta-analysis (NMA) Pan-Canadian Oncology Drug Review
NMB, see Net monetary benefit (pCODR), 345
Non-inferiority hypothesis, 30, 32 Panitumumab, 326, 339, 355
Non-medical costs, 155 Panobinostat, 354
Nonproportional hazard, 39 Parametric methods, 114–118
Non-small-cell lung cancer (NSCLC), 10, Pareto-type model, for post-progression
63, 77, 135–139, 177, 260–262, 333 utility, 83
digital real-world web-mediated Partial response (PR), 55
follow-up compared with Partitioned survival model, 231
routine surveillance in, 263–264 case study, 232–236
health economic models in, 216–221 crossover, 236–238
Index 411

economic model using patient-level missing data between death and, 52


data, 231–232 Post-treatment period and cancer trial,
PAS, see Patient-access scheme 147, 148
Pathological complete response (pCR), Post-trial period, 163
56–57 and cancer trial, 147, 148
Patient-access scheme (PAS), 352 Potential adverse event (PAE), 312, 320, 322
breakdown of, 352 PP, see Per protocol (PP)
response-based, 355–356 PP, see Post-progression
Pazopanib, 353 PPRS, see Pharmaceutical Price
PBAC, see Pharmaceutical Benefits Regulation Scheme
Advisory Committee PPS, see Post-progression survival
PBM, see Preference-based measures PR, see Partial response
PBS, see Pharmaceutical Benefits Scheme Pragmatic clinical trials, 264–265
PCODR, see Pan-Canadian Oncology Pragmatic RCT (PRCT), 264–265
Drug Review Prednisolone, 327
pCR, see Pathological complete response Prednisone, 204, 327
PD, see Progressive disease Preference-based measures (PBM), 67
Pembrolizumab, 52, 194, 333–334, 340, Premium price, 16, 25
353, 354, 355 Pre-staging and cancer trial, 146, 147
Pemetrexed, 9, 55, 88, 219–221, 241, 242, Primary care databases, 262
245, 247, 325, 327, 337, 338 Probabilistic sensitivity analysis (PSA),
Per protocol (PP) analysis, 128 132, 301–305, 309, 358
Pertuzumab, 55, 56, 354 Bayesian, 319–320
PFS, see Progression-free survival Prognostic factors, see under Cancer
Pharmaceutical Benefits Advisory Progression-free survival (PFS), 37,
Committee (PBAC), 348 43–44, 46, 47, 48, 53, 84, 96, 111,
Pharmaceutical Benefits Scheme 116, 135, 179, 192, 231, 232, 236,
(PBS), 348 245, 314, 332
Pharmaceutical Price Regulation assessment of, 52
Scheme (PPRS), 16, 350 relationship with overall survival,
Pharmacoeconomics, 13 50–51
PIM, see Promising innovative medicine Progressive disease (PD), 46–47, 48, 51,
Pixantrone, 353 78, 79, 142
Placebo, 100, 102, 113, 137, 234, 241 prolonged treatment until, 51–52
Platform trials, 358 Promising innovative medicine (PIM), 175
Pomalidomide, 327, 355 Propensity score, 190, 269–270, 273–274
Ponatinib, 355 Propensity score modeling (PSM),
Population EVPI, 310 268–274, 275
Post-progression (PP), 238 Proportional hazards, 106
utility measurement, 77–78 Prostate cancer, 1–2, 4, 60, 64, 65
non-linear methods, 82–86 cost of treating, 8
plausible post-progression utility Proxy price, 17
behavior, 80–81 PSA, see Probabilistic sensitivity
utility behavior in cancer patients, analysis
79–80 PSM, see Propensity score modeling
utility estimation between disease p-value, 109, 187
progression and death, 78–79
Post-progression survival (PPS), 36, 51, QALY, see Quality-adjusted life-years
84, 231, 232 QLQ, see Quality of Life Questionnaire
412 Index

QLQ-8D, 63 digital data sources, social media


QLQ-BR23, 64 and applications, 263–264
QLQ-C30, 63, 64, 66, 68, 197 insurance claims databases, 263
QLQ-LC13, 63, 64 pragmatic clinical trials, 264–265
QLQ-MY20, 64 primary care databases, 262
QLQ-OV28, 64 prospective observational
QLQ-PR25, 64 research studies, 265
QResearch, 262 registries, 260–262
Quality-adjusted life-years (QALY), internal validity versus
8–12, 22–23, 36, 42, 44, 45, 48, generalizability and, 256–257
57, 61, 63, 67, 69, 72–73, 78, 81, limitations of, 255–256
116, 123, 125, 196, 197, 295, 305, on metastatic NSCLC from Canadian
335, 359 cancer registries, 260–262
calculation in cancer trials, 73–74 statistical analyses of, 268
cost and, 8, 10 instrumental variable methods,
incremental, interpreting, 287–290 274, 276
post-progression, 84–86 propensity score modeling (PSM),
reduced, 51 268–274, 275
relationship with costs, 290–292 strengths of, 253, 255
Quality of life, definition of, 59 to support cost-effectiveness
Quality of Life Questionnaire (QLQ), 62, analysis, 251–253, 254
63, 64–65 Real-world evidence, see Real-world
Quality of Time Spent Without data (RWD)
Symptoms of Disease and REC, see Research ethics committees
Toxicity (QTWiST), 197 Re-censoring, 131
Quantiles, 109 RECIST, see Response Evaluation
Criteria in Solid Tumors
Radium-223 dichloride, 354 Registries, 260–262
Ramucirumab, 328 Reimbursement, 15–16, 25 26, 42, 45, 48,
Randomized clinical trials (RCT), 15, 26, 50, 57, 119, 126, 132, 146, 154,
27, 52, 60, 70, 132, 136, 178, 189, 155, 175–176, 178, 182, 183, 194,
250, 256–257, 264, 283, 333 197, 198, 200, 204, 206–208, 231,
Rank-preserving structural failure 256, 263, 265, 295, 305, 309–311,
time models (RPSFTMs), 126, 313, 316, 323, 324, 329, 332, 335,
129–131, 212, 236 336, 341, 351, 352, 356, 358, 360
RBRVS, see Resource-based relative changing pace of, 341, 343
value system environment, and pricing in United
RCC, see Renal cell cancer (RCC) States, 349–350
R-CHOP, 204 and payer evidence requirements
RCT, see Randomized clinical trials across countries, 344–349
Real-world data (RWD), 27–29, Renal cell cancer (RCC), 45
249–250, 280 Research and audit, difference
cancer registries and, 265–266 between, 262
working of, 266–268 Research ethics committees (REC), 264
and clinical trials compared, 250 Resource-based relative value system
generation sources, 257–259 (RBRVS), 155
audits, 261–262 Response Evaluation Criteria in Solid
case control studies, 265 Tumors (RECIST), 46, 55, 56
commercial data sources, 264 Restricted mean, 106, 109
Index 413

preference over median estimate, 107 Single technology appraisal (STA),


Rituximab, 183, 204, 205, 325, 351, 352 323, 324
for follicular lymphoma, 351–352 Single technology assessment, 324, 335
Royston-Parmar (RP) model, 120, 121 Skewed distribution, 95
RP, see Royston-Parmar (RP) model SMC, see Scottish Medicines
RPSFTMs, see Rank-preserving Consortium
structural failure time models Smearing factor, 158
Ruxolitinib, 327, 354 SNNM, see Structural nested mean
models
SACT, see Systemic anti-cancer therapies Sorafenib, 55, 100, 113, 325, 326, 337
SAPs, see Statistical analysis plans Spain, 57, 154, 207, 345, 347–348
SAS® software, 356 Spanish National Health Service, 347
code, 318, 320 STA, see Single technology appraisal
Scottish Medicines Consortium Stable disease (SD), 55, 142
(SMC), 13 Staging, and cancer trial, 146
SD, see Stable disease STAMPEDE MAMs design, 187
Selection bias, 190, 279 Standard deviation, 111
real-world data and, 268 Standard gamble (SG), 71
instrumental variable methods, Standardized differences, 273
274, 276 Statistical analysis plans (SAPs), 358
propensity score modeling (PSM), Statistical methods, for economic
268–274, 275 evaluation, 91
Semi-parametric methods, 111–112 bootstrapping and, 108–110
Sensitivity analysis, 296–300 cost, utility, and survival data and
Bayesian, 306–307 distributions, 93–95
ICER and INMB limitations and, data analysis, 111
307–308 applications in cancer
one way, 296–297, 298–299 surveillance, 119–122
probabilistic, 301–305 Cox proportional hazard (Cox
two way, 297, 299–300 PH) model, 111–114
Sentinel Initiative, 253 survival data advanced modelling
SF, see Shapiro-Francia test techniques, 118
SG, see Standard gamble survival data modeling for
Shadow price, 17 extrapolation, 114–118
Shapiro-Francia (SF) test, 157 data synthesis and network meta-
Shapiro-Wilks (SW) test, 157 analyses, 132
Silo effect, 146 mixed treatment comparisons
Simulation, 108–111, 117, 192, 193, 202, (MTC), 132–139
215, 243, 296, 301, 302, 306, 314, hazard rate and, 98–99
316, 318, 320 relationship with survival rate,
Single arm trials, 47, 179–181, 252, 255 102–103
with ofatumumab, for chronic hazard ratio and, 99–101
lymphocytic leukaemia, to predict survival rates, 113–114
181–182 issues in fitting models, 122–123
with venetoclax for chronic mean survival and restricted mean
lymphocytic leukaemia, and, 106–108
182–183, 184 median survival and, 96–98
Single imputation methods, 161 Monte-Carlo sampling and, 111
Single-stage design, 179 proportional hazards, 106
414 Index

survival rates and proportions, Time-to-event endpoints, 95–96


101–102 Time-to-next treatment (TTNT), 38
time-to-event endpoints and, 95–96 Time-to-progression (TTP), 116, 118
transition probability and matrix, Time-to-treatment failure (TTF), 38
103–104 Time trade off (TTO), 71, 237
transition probability and survival Tipiracil hydrochloride, 354
rates, relationship between, TKIs, see Tyrosine kinase inhibitors (TKIs)
104–106 TOI, see Trial Outcome Index
treatment switching, 123–124 Top-down approach, 150
implications of, 124–126 Topotecan, 326, 328
switching handling in statistical Tornado diagram, 298
analyses, 126–132 TPS, see Tumor proportion score
types of, 124 Trabectedin, 328, 353
uncertainty and, 91–92 Trametinib, 354
variability and, 92–93 Transarterial chemoembolization
hypothesis testing, 93 (TACE), 279
Stomach cancer, 4 Transition probability, 102, 105, 106, 109
Structural nested mean models Transition probability and matrix,
(SNNM), 131–132 103–104, 227, 251
Subscales, 63 Transition probability and survival rates,
Sunitinib, 102, 325, 337, 353 relationship between, 104–106
Superiority trial, 29–31 Trapezoidal rule, 288, 290
Surrogate endpoints, 46–53 Trastuzumab, 31, 327, 328
advantage of, 47 Treatment emergent adverse events
HTAs with, 53–55 (TEAE), 164
Survival data advanced modelling Treatment switching, 123–124
techniques, 118 implications of, 124–126
Survival data modelling, for switching handling in statistical
extrapolation, 114–118 analyses, 126–132
Survival rates and proportions, 101–102 types of, 124
SW, see Shapiro-Wilks (SW) Trial Outcome Index (TOI), 66
Sweden, 57, 344 Trifluridine, 354
Switching, see Treatment switching TTF, see Time-to-treatment failure
Systemic anti-cancer therapies (SACT), TTNT, see Time-to-next treatment
28, 260, 267, 334 TTO, see Time trade off
TTP, see Time-to-progression (TTP)
TACE, see Transarterial Tumor-centred endpoints, 36, 37
chemoembolization immune-related response criteria
Talimogene laherparepvec, 354 (irRC), 55–56
Taxane, 326, 338 minimal residual disease (MRD), 56
Technical efficiency, 15–16 pathological complete response
Temozolomide, 337 (pCR), 56–57
Temsirolimus, 325, 337 Tumor proportion score (TPS), 333
The Health Improvement Network Two-part models, 167
(THIN), 262 Two-stage adjustment model, 131
THIN, see The Health Improvement Two-stage design, 179, 183
Network Two-way sensitivity analysis, 297,
Time preference and discounting, 299–300
153–154 Tyrosine kinase inhibitors (TKIs), 53
Index 415

UK, see United Kingdom challenges in healthcare decisions,


UK Department of Health and 316–317
Healthcare, 16 Value of sample information (VSI), 312
UK Royal College of Physicians, 264 Valuing, of health, 13–14
Umbrella Organization of Vandetinib, 210–212, 232, 233, 234,
Statutory Health 237, 239
Insurance, 347 Variability, 92–93
Uncertainty, 7, 24, 28, 41, 48, 54, 85, 91, VBP, see Value-based pricing
92, 93, 188, 193, 225, 231, 274, Vemurafenib, 45, 328, 353
292–307, 309–310, 314, 320, 334, Venetoclax, 47, 182–183, 184
335, 341, 352 Vincristine, 204
Unilateral switch, 124 Vinorelbine, 9, 325
United Kingdom (UK), 57, 68, 145, 146, VOI, see Value of information
188, 207, 262, 305, 332, 344, 349, VSI, see Value of sample information
350, 351
registries for RWE in, 267–268 Web-based follow-up, see e-FAP
United States (US), 155, 260, 263, 343, Weibull model, 114–115, 117, 120
344, 349 WHO, see World Health Organization
US, see United States Willingness pay (WTP), 18, 21, 295
US National Bureau of Economic thresholds, impact of varying, 295–296
Research, 341 WINBUGS software, 139, 140, 320
Uterine cancer, 4 World Health Organization (WHO), 59
Utility function, 71–72 WTP, see Willingness pay

Value-based pricing (VBP), 350–352 Zero costs, 157


Value of information (VOI), 308–314, ZETA trial, 233
320–321, 358 trial design and cost-effectiveness
bevacizumab trial, 314–315 features of, 234–235

You might also like