Supplementary Appendix
Supplementary Appendix
Supplement to: Smolen JS, Feist E, Fatenejad S, et al. Olokizumab versus placebo or adalimumab in rheumatoid
arthritis. N Engl J Med 2022;387:715-26. DOI: 10.1056/NEJMoa2201302
This appendix has been provided by the authors to give readers additional information about the work.
TABLE OF CONTENTS
1
STUDY INVESTIGATORS
2
Dr Edit Drescher Vital-Medicina Kft. Veszprem; Hungary
Dr Filipe Martins de Mello Clínica de Neoplasias Litoral Ltda Santa Catarina; Brazil
Spitalul Clinic Judetean de Urgenta
Dr Ana Maria Mihaela Ramazan Constanta; Romania
"Sf Apostol Andrei" Constanta
Ciudad Autonoma Buenos Aires;
Dr Cecilia Asnal Instituto Centenario
Argentina
Dr Gisela Constanza Subils Instituto Medico DAMIC Cordoba; Argentina
Instituto de Investigaciones Mar del Plata; Buenos Aires;
Dr Juan Pablo Gulin
Clinicas Argentina
Dr Eleonora del Valle Centro de Investigaciones San Miguel de Tucumán; Tucuman;
Lucero Reumatológicas Argentina
3
Dr Pablo Alejandro Mannucci APRILLUS Asistencia e Ciudad Autonoma Buenos Aires;
Walter Investigacion Argentina
CER San Juan Centro Polivalente
Dr Jose Luis Cristian Moreno Este; San Juan; Argentina
de Asistencia e Inv. Clinica
Centro Medico Privado de San Miguel de Tucuman; Tucuman;
Dr Alberto Jorge Spindler
Reumatologia Argentina
Centro de Investigaciones Medicas Mar del Plata; Buenos Aires;
Dr Gladys Alicia Testa
Mar del Plata Argentina
Atencion Integral en Reumatologia Ciudad Autonoma Buenos Aires;
Dr Horacio Oscar Venarotti
(AIR) Argentina
Dr Benito Jorge Velasco Zamora Instituto Medico CER Quilmes; Buenos Aires; Argentina
Hospital General de Agudos Dr. J. Ciudad Autonoma Buenos Aires;
Dr Eduardo Kerzberg
M. Ramos Mejia Argentina
Organizacion Medica de Ciudad Autonoma Buenos Aires;
Dr Patricio Tate
Investigacion (OMI) Argentina
Instituto de Investigaciones
Dr Jose Luis Velasco Zamora Quilmes; Buenos Aires; Argentina
Clinicas Quilmes
Venado Tuerto; Santa Fe;
Dr Rosana Gallo Sanatorio San Martin
Argentina
Clinica de Higado y Aparato
Dr Judith Carrio Rosario Santa Fe; Argentina
Digestivo
Hospital Privado Centro Medico de
Dr Alejandro Jose Alvarellos Cordoba; Argentina
Cordoba S.A
Siauliai Republican Hospital,
Dr Vida Basijokiene Siauliai; Lithuania
Public Institution
Klaipeda University Hospital,
Dr Loreta Bukauskiene Klaipeda; Lithuania
Public Institution
Center Outpatient Clinic, Public
Dr Ruta Lauciuviene Vilnius; Lithuania
Institution
Alytus Regional S. Kudirkos
Dr Rita Kvedaraviciene Alytus, Lithuania
Hospital
Republican Kaunas Hospital,
Dr Joana Dambrauskiene Kaunas; Lithuania
Public Institution
Vilnius University Hospital
Dr Jelena Ranceva Santariskiu Clinics, Public Vilnius; Lithuania
Institution
Dr Hung-An Chen Chi Mei Medical Center Tainan; Taiwan, Republic of China
Chang Gung Memorial Hospital, Guishan District; Taoyuan; Taiwan,
Dr Lieh-Bang Liou
Linkou Republic of China
Fundacion Instituto de
Dr Monique Rose Chalem Choueka Bogota; Colombia
Reumatologia Fernando Chalem
Dr Alvaro Arbelaez Cortes Clinica de Artritis Temprana S.A.S Cali; Colombia
Centro de Investigacion en
Dr Patricia Julieta Velez Sanchez Reumatologia y Especialidades Bogotá; Colombia
Medicas SAS. CIREEM
Centro de Investigacion Medico
Dr Juan Jose Jaller Raad Barranquilla; Colombia
Asistencial S.A.S
Dr Diego Luis Saaibi Solano Medicity S.A.S Bucaramanga; Colombia
4
Basingstoke and North Hampshire
Dr Emma Williams Basingstoke; Hampshire; UK
Hospital
Dr Emmanuel George Arrowe Park Hospital Wirral; Merseyside; UK
Dr Joseph Forstot RASF - Clinical Research Center Boca Raton; Florida; USA
Arizona Arthritis & Rheumatology
Dr Saima Chohan Phoenix; Arizona; USA
Associates, P.C
Dr Pooja Bhadbhade University of Kansas Hospital Kansas City; Kansas; USA
Dr Mary Howell Klein and Associates, M.D., P.A Hagerstown; Maryland; USA
Dr Elmer Thomas Arne Lovelace Scientific Resources, Inc Venice; Florida; USA
6
Dr Chandrakant Mehta Southland Arthritis - Hemet Hemet; California; USA
New England Research Associates
Dr Geoffrey Gladstein Bridgeport; Connecticut; USA
LLC
Wilmington; North Carolina
Dr Mark Harris Carolina Arthritis Associates
USA
The Center for Rheumatology and
Dr Herbert Baraf Wheaton; Maryland; USA
Bone Research
Greensboro; North Carolina
Dr Nilamadhab Mishra Medication Management, LLC
USA
Health Research of Oklahoma, Oklahoma City; Oklahoma
Dr Christine Codding
PLLC USA
Inland Rheumatology Clinical
Dr Eric Lee Upland; California; USA
Trials, Inc
Los Alamitos; California
Dr Rebekah Neal-Kraal Valerius Medical Group
USA
Dr Robert LaGrone Center for Inflammatory Disease Nashville; Tennessee; USA
Dr Jeanne-Elyse Cedeno Family Clinical Trials, LLC Pembroke Pines; Florida; USA
Marietta Rheumatology Associates,
Dr Roel Querubin Marietta; Georgia; USA
P.C
Dr Jitendra Vasandani West Texas Clinical Research Lubbock; Texas; USA
Dr David Snow Cape Fear Arthritis Care Leland; North Carolina; USA
Dr Nazanin Firooz Center for Rheumatology Research West Hills; California; USA
AA MRC LLC Ahmed Arif
Dr Ali Karrar Grand Blanc; Michigan; USA
Medical Research Center
Advanced Rheumatology of
Dr Tamar Brionez The Woodlands; Texas; USA
Houston
Dr Shaikh Arif Ali DM Clinical Research Tomball; Texas; USA
7
Precision Comprehensive Clinical
Dr Dhiman Basu Colleyville; Texas; USA
Research Solutions
Dr Tania Rivera Rheumatology Center of San Diego San Diego; California; USA
8
Dr Krystyna Kuc CERMED Bialystok; Poland
9
METHODS
After eligibility had been confirmed, subjects were randomized in a 2:2:2:1 ratio in 4 treatment groups
(olokizumab [OKZ] 64 mg once every 2 weeks [q2w], OKZ 64 mg once every 4 weeks [q4w],
adalimumab [ADA] 40 mg q2w and placebo [PBO]) by blinded study staff using the IWRS (an
automated web randomization system). The IWRS allocated the treatment group and assigned the study
treatment. This system also managed drug supply management and visit dispensation. Blinded study
staff requested study treatment assignments via the IWRS for all subsequent treatment study visits.
Cenduit Interactive Response Technology (C.I.R.T) system (Cenduit, LLC) was used in the study for
this purpose.
The treatment each subject received was not disclosed to the blinded study site staff, including the
Investigators, study coordinators, subjects, R-Pharm (Sponsor), or R-Pharm’s designee. Since the study
treatments were distinguishable, they were prepared by the unblinded pharmacist (or their unblinded
designee) and administered by a trained, unblinded study team member who was not involved in the
management of study subjects. An independent unblinded pharmacist (or designee) prepared OKZ and
PBO in syringes of 0.4 mL for subcutaneous (SC) injection, and ADA was supplied from a central
vendor in a prefilled, single-dose syringe of either 0.4 or 0.8 mL for SC injection. In order to maintain
the blind, OKZ and PBO were prepared in blinded SC syringes as per the instructions in the study-
specific manual prior to dispensing, in a manner that both treatments were identical in appearance.
ADA was also masked; however, as ADA was supplied in a larger SC syringe, the size of the masked
syringe was distinguishable from OKZ and PBO. A qualified unblinded study team member who was
not involved in the management of study subjects administered the assigned study treatment as a single
SC injection q2w or q4w at the relevant time points. Subjects were dosed individually and away from
other subjects to ensure that differences between OKZ, ADA, and PBO were not observed. To maintain
the blind, the unblinded study site staff was trained in methods that must be followed and documented
to prevent subjects from observing which SC injection they receive. Guidance on specific blinding
procedures were provided in the study-specific manual.
Access to randomization codes were restricted. The treatment codes were held by the IWRS. Only the
unblinded pharmacist (or their unblinded designee) or dedicated unblinded study staff who were not
directly involved in subject management were aware of the randomized drug assignment. The storage
and preparation of the study treatment were at a secured location that was not accessible to blinded
study staff. Additional measures to ensure that both Investigators and participants remained blinded to
study treatment included the following:
10
Joint assessments were made by an independent assessor, blinded to both the dosing
regimen and all other study assessments. Training video was provided to the assigned assessors
in the beginning of the study.
Laboratory results for C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR)
samples collected during the treatment period were not available to blinded study site staff. As
ESR was tested locally, the testing was performed, reviewed, registered and reported by
unblinded study site staff who was not responsible for managing subjects.
Certain efficacy assessments (American College of Rheumatology 20% improvement response
criteria ACR20, ACR50, ACR70, Disease Activity Score 28-joint count CRP (DAS28-CRP),
DAS28-ESR, Simplified Disease Activity Index (SDAI), and Clinical Disease Activity Index
(CDAI)) were not calculated by the Investigator during the course of the study, but were
computed in the statistical database for analysis purposes further.
All blinding procedures were respected.
Members of the independent Data Safety Monitoring Board (DSMB) reviewed separately safety data
during the study. In the event that ongoing safety monitoring had uncovered an issue needed to be
addressed by unblinding at the treatment group level, only members of the DSMB were permitted to
conduct additional analysis of the safety data.
The primary efficacy endpoint was the ACR20 response at Week 12, where a responder was defined
as any subject satisfying ACR20 criteria and remaining on randomized treatment and in the study at
Week 12. This endpoint served to demonstrate that the efficacy of OKZ was superior to PBO.
ACR20 response at Week 12, where a responder was defined as any subject satisfying ACR20
criteria and remaining on randomized treatment and in the study at Week 12; was used to
demonstrate that the efficacy of OKZ was non-inferior to ADA, provided that superiority of
ADA to PBO (assay sensitivity) was demonstrated concurrently based on the same endpoint.
11
Percentage of subjects achieving DAS28-CRP<3.2, and remaining on randomized treatment
and in the study at Week 12; served to demonstrate that the efficacy of OKZ was non-inferior
to ADA, provided that superiority of ADA to PBO (assay sensitivity) was demonstrated
concurrently based on the same endpoint.
Improvement of physical ability from baseline to Week 12, as measured by the Health
Assessment Questionnaire-Disability Index (HAQ-DI).
Percentage of subjects with CDAI ≤2.8 (remission) and remaining on randomized treatment
and in the study at Week 24.
Other efficacy (exploratory) endpoints included but were not limited to proportion of subjects
achieving an ACR20, ACR50, and ACR70 response and remaining on randomized treatment and in
the study, assessed at all other applicable time points; proportion of subjects with
SDAI ≤3.3 (remission) and remaining on randomized treatment and in the study, assessed at all other
applicable time points; proportion of subjects with CDAI ≤2.8 (remission) and remaining on
randomized treatment and in the study, assessed at all other applicable time points; proportion of
subjects with DAS28-CRP<3.2 and remaining on randomized treatment and in the study at all other
applicable time points.
Sample size requirements were calculated to provide sufficient disjunctive power to evaluate the
primary endpoint (superiority of OKZ relative to PBO with respect to the ACR20 response rate at
Week 12) as well as 3 secondary endpoints (non-inferiority of OKZ relative to ADA with respect to
the ACR20 response rate at Week 12, superiority of OKZ relative to PBO with respect to the proportion
of subjects with DAS28-CRP<3.2 at Week 12, and non-inferiority of OKZ relative to ADA with
respect to the proportion of subjects with DAS28-CRP<3.2 at Week 12).
Sample size was estimated taking into account a multiplicity control procedure and the resulting α
adjustment that was used to control an overall Type I error rate in the strong sense at a 1-sided α =
0.025 across the primary and secondary endpoints. More specifically, the α-control strategy was based
on using the Bonferroni adjustment for the tests related to each of the 2 OKZ dose regimens (ie, using
the 1-sided α = 0.0125 for each dose). A gate-keeping strategy was used for tests associated with the
primary and secondary endpoints for each OKZ dose regimen independently, with a fixed order of
hypothesis tests for the primary and secondary endpoints within each OKZ dose regimen.
12
Sample size was estimated based on simulation that implemented the above α adjustment and gate-
keeping procedure including the primary endpoint of ACR20 response rate at Week 12 (superiority of
OKZ to PBO) and the 3 secondary endpoints as detailed. Based on this simulation, an adequate sample
size was selected to provide sufficient disjunctive power for the primary endpoint as well as sufficient
disjunctive power for testing non-inferiority of OKZ relative to ADA with respect to the ACR20
response rate at Week 12, superiority of OKZ relative to PBO with respect to the proportion of subjects
with DAS28-CRP<3.2 at Week 12, and non-inferiority of OKZ relative to ADA with respect to the
proportion of subjects with DAS28-CRP<3.2 at Week 12. For each secondary endpoint, the disjunctive
power represents a probability of establishing a significant treatment effect for 1 or 2 OKZ dose
regimens for the primary endpoint and a significant treatment effect for 1 or 2 OKZ dose regimens for
a given secondary endpoint, conditional on the fact that each dose regimen tested for the secondary
endpoint must have null hypothesis previously rejected for all endpoints (primary and secondary) that
precede this endpoint in a fixed order of testing as per the gate-keeping procedure.
Based on the treatment effect assumptions and the methodology for estimating sample size via
simulation to ensure sufficient disjunctive power as described, a sample size of 1575 subjects
randomized in a 2:2:2:1 ratio (450, 450, 450, and 225 subjects per group, respectively) would yield
100% disjunctive power for testing the primary hypothesis (superiority of OKZ to PBO with respect
to ACR20 at Week 12), 92% disjunctive power for testing the secondary hypothesis of non-inferiority
of OKZ to ADA with respect to ACR20 rate at Week 12, 92% disjunctive power for the evaluation of
the secondary endpoint of superiority of OKZ to PBO with respect to DAS28-CRP<3.2 at Week 12,
and 88% disjunctive power for testing the secondary hypothesis of non-inferiority of OKZ to ADA
with respect to DAS28-CRP<3.2 at Week 12.
For selection of non-inferiority margin the following assumptions were used: in a meta-analysis of two
key pivotal clinical studies for registration of ADA,1,2 the difference between the ACR20 response rate
at Week 12 for ADA in combination with methotrexate (MTX) with N=280 and PBO in combination
with MTX with N=262 was equal to 35% (95% confidence interval: 27.3% to 42.6%). Therefore,
27.3% was estimated as the absolute largest non-inferiority margin that could be justified. Using an
assumption of 55% ACR20 response rate at Week 12 for ADA and a more conservative minimum
lower bound of 43% for OKZ was considered to be clinically meaningful. As a result, a non-inferiority
margin of 12% for the ACR20 response rate at Week 12 was used in this study.
Missing data
The main method for handling missing data resulting from premature withdrawal of a subject from the
study was based on a clinical approach where no treatment benefit was attributed to study treatment at
any time point post study withdrawal, if the subject was unable to complete the study for the planned
duration.
Therefore, for all binary primary and secondary endpoints (ACR20 at Week 12, DAS28-CRP32 at
Week 12, ACR50 at Week 24 and CDAI28 at Week 24), all subjects who discontinue prematurely,
regardless of reason, were considered as non-responders with respect to the definition of the
corresponding endpoint.
Similarly, for all endpoints that are continuous in nature (such as HAQ-DI at Week 12), a return to
baseline values was performed and was implemented using multiple imputation accounting for the
uncertainty of missing data according to the methodology of Rubin.5
Missing data could be also result from missed intermediate visits. For all binary endpoints, missing
intermediate responder status was imputed from the surrounding visits. If the status at visits both before
and after the missed visit was classified as responder, the subject was considered a responder at the
missed visit. Otherwise, the subject was considered a non-responder. If an assessment for a given
binary endpoint at the last visit was missing for a completing subject, data from the 2 previous visits
were used with the same logic for determining the response status.
For the ACR20/50/70 endpoints, if only some components of the response criteria were missing but
the available components allowed classifying the subject as a responder, the responder category was
used. Otherwise, the method based on surrounding visits as described above was applied.
Similarly, for all continuous endpoints, an average of values from the surrounding visits was imputed
for the missing visit. In the event that some visits remained missing, either due to sequential missing
visits, the preceding visit being baseline, or the missing visit being the last scheduled visit, then these
were imputed using the Markov chain Monte Carlo method.
Subjects were enrolled in the study only if they met all of the following criteria:
14
1. Male or female subjects ≥18 years of age.
2. Subjects willing and able to sign informed consent.
3. Subjects had to have a diagnosis of adult-onset RA classified by American College of
Rheumatology (ACR)/ European Alliance of Associations for Rheumatology (EULAR) 2010
revised classification criteria for rheumatoid arthritis (RA)6 for at least 12 weeks prior to Screening.
If the subject was diagnosed according to ACR 1987 criteria previously, the Investigator could
classify the subject per ACR 2010 retrospectively, using available source data.
4. Inadequate response to treatment with oral, SC, or intramuscular MTX for at least 12 weeks prior
to Screening at a dose of 15 to 25 mg/week (or ≥10 mg/week if intolerant to higher doses); a lower
dose of MTX (≥7.5 mg/week) was permitted for subjects with intolerant to higher doses enrolled
in the Republic of Korea, consistent with local clinical practice.
The dose and means of administering MTX had to have been stable for at least 6 weeks prior
to Screening.
5. Subjects had to be willing to take folic acid or equivalent throughout the study.
6. Subjects had to have moderately to severely active RA disease as defined by all of the following:
a) ≥6 tender joints (68-joint count) at Screening and baseline; and
b) ≥6 swollen joints (66-joint count) at Screening and baseline; and
c) CRP above upper limit of normal (ULN) at Screening based on the central laboratory results.
Subjects who met any of the following criteria were not eligible for the study:
1. Diagnosis of any other inflammatory arthritis or systemic rheumatic disease (eg, gout, psoriatic or
reactive arthritis, Crohn’s disease, Lyme disease, juvenile idiopathic arthritis, or systemic lupus
erythematosus)
Subjects could have secondary Sjogren’s syndrome or hypothyroidism.
2. Subjects who were Steinbrocker class IV functional capacity (incapacitated, largely or wholly
bed-ridden or confined to a wheelchair, with little or no self-care).
3. Prior exposure to any licensed or investigational compound directly or indirectly targeting IL-6 or
IL-6R (including tofacitinib or other Janus kinase(JAK) and spleen tyrosine kinase (SYK)
inhibitors)
4. Prior treatment with cell-depleting therapies, including anti-CD20 or investigational agents (eg,
Campath, anti-CD4, anti-CD5, anti-CD3, and anti-CD19).
5. Prior use of biological disease-modifying antirheumatic drugs (bDMARDs).
6. Use of parenteral and/or intra-articular (IA) glucocorticoids within 4 weeks prior to baseline.
15
7. Use of oral glucocorticoids greater than 10 mg/day prednisone (or equivalent) or change in dosage
within 2 weeks prior to baseline.
8. Prior documented history of no response to hydroxychloroquine and sulfasalazine.
9. Prior use of conventional DMARDs (cDMARDs) other than MTX within the following windows
prior to baseline (cDMARDs should not be discontinued to facilitate a subject’s participation in the
study, but should instead have been previously discontinued as part of a subject’s medical
management of RA):
Four weeks for sulfasalazine, azathioprine, cyclosporine, hydroxychloroquine, chloroquine,
gold, penicillamine, minocycline, or doxycycline
Twelve weeks for leflunomide, unless the subject had completed the following elimination
procedure at least 4 weeks prior to baseline: сholestyramine at a dosage of 8 g 3 times daily for
at least 24 hours, or activated charcoal at a dosage of 50 g 4 times daily for at least 24 hours
Twenty-four weeks for cyclophosphamide
10. Vaccination with live vaccines in the 6 weeks prior to baseline or planned vaccination with live
vaccines during the study.
11. Participation in any other study of investigational drug within 30 days or 5 times the terminal
half-life of the investigational drug, whichever was longer, prior to baseline.
12. Other treatments for RA (eg, Prosorba Device/Column) within 6 months prior to baseline
13. Use of IA hyaluronic acid injections within 4 weeks prior to baseline.
14. Use of non-steroidal anti-inflammatory drugs (NSAIDs) on unstable dose or switching of NSAIDs
within 2 weeks prior to baseline.
15. Previous participation in this study (randomized) or another study of OKZ.
16. Abnormal laboratory values, as defined below:
Creatinine level ≥1.5 mg/dL (132 µmol/L) for females or ≥2.0 mg/dL (177 µmol/L) for males
Alanine aminotransferase (ALT) or aspartate aminotransferase (AST) level ≥1.5× ULN
Platelets <100×109/L (<100,000/mm3)
White blood cell (WBC) count <3.5×109/L
Neutrophil count <2000×106/L (<2000/mm3)
Hemoglobin (Hb) level ≤80 g/L
Glycosylated Hb level ≥8%
17. Subjects with concurrent acute or chronic viral hepatitis B or C infection as detected by blood tests
at Screening (eg, positive for hepatitis B surface antigen [HBsAg], total hepatitis B core antibody
[anti-HBc], or hepatitis C virus antibody [HCV Ab])
Subjects who were positive for hepatitis B surface antibodies (anti-HBs), but negative for
HBsAg and anti-HBc, were eligible.
16
18. Subjects with human immunodeficiency virus (HIV) infection.
19. Subjects with:
Suspected or confirmed current active tuberculosis (TB) disease or a history of active TB
disease
Close contact (ie, sharing the same household or other enclosed environment, such as a social
gathering place, workplace, or facility, for extended periods during the day) with an individual
with active TB within 1.5 years prior to Screening.
History of untreated latent TB infection (LTBI), regardless of interferon-gamma release assay
(IGRA) result at Screening
Subjects with a history of untreated LTBI could be re-screened and enrolled if they fulfilled
all 3 of the following criteria:
Active TB was ruled out by a certified TB specialist or pulmonologist who was familiar
with diagnosing and treating TB (as acceptable per local practice);
The subject had completed at least 30 days of LTBI-appropriate prophylaxis prior to
baseline with agents recommended as preventative therapy for LTBI according to
country-specific/Centers for Disease Control and Prevention (CDC) guidelines
(treatment with isoniazid for 6 months was not an appropriate prophylactic regime for
this study and was not used); and
The subject was willing to complete the entire course of recommended LTBI therapy.
Positive IGRA result at Screening. If indeterminate, the IGRA could be repeated once during
the Screening Period. If there was a second indeterminate result, the subject was excluded.
Subjects with a positive IGRA result at Screening could be re-screened and enrolled if:
Active TB was ruled out by a certified TB specialist or pulmonologist who was familiar
with diagnosing and treating TB (as acceptable per local practice);
The subject had completed at least 30 days of LTBI-appropriate prophylaxis prior to
baseline with agents recommended as preventative therapy for LTBI according to
country-specific/CDC guidelines (treatment with isoniazid for 6 months was not an
appropriate prophylactic regime for this study and was not used); and
The subject was willing to complete the entire course of recommended LTBI therapy.
If a subject with a positive IGRA result at Screening had documented evidence of
completing treatment for LTBI with a treatment regime and treatment duration that were
appropriate for this study, the subject could be enrolled without further prophylaxis if
recommended by a certified TB specialist or pulmonologist who was familiar with
diagnosing and treating TB (as acceptable per local practice) and no new exposure in close
17
contact with an individual with active TB after completing the prophylactic treatment was
suspected.
20. Concurrent malignancy or a history of malignancy within the last 5 years (with the exception of
successfully treated carcinoma of the cervix in situ and successfully treated basal cell carcinoma
and squamous cell carcinoma not less than 1 year prior to Screening [and no more than 3 excised
skin cancers within the last 5 years prior to Screening]).
21. Subjects with any of the following cardiovascular (CV) conditions:
Uncompensated congestive heart failure, or class III or IV heart failure defined by the New
York Heart Association classification7
Untreated or resistant arterial hypertension Grade II-III (systolic blood pressure [BP]
>160 mm Hg and/or diastolic BP >100 mm Hg)
History or presence of concurrent severe and/or uncontrolled CV disorder (including but not
limited to acute coronary syndrome or stroke/transient ischemic attack in the previous 3 months
before Screening) that would, in the Investigator’s judgment, have contraindicated subject
participation in the clinical study, or clinically significant enough in the opinion of the
Investigator to have altered the disposition of the study treatment, or constituted a possible
confounding factor for assessment of efficacy or safety of the study treatment
22. Subjects with a history or presence of any concurrent severe and/or uncontrolled medical condition
(including but not limited to respiratory, hepatic, renal, gastrointestinal, endocrinological,
dermatological, neurological, psychiatric, hematological [including bleeding disorder], or
immunologic/immunodeficiency disorder[s]) that would, in the Investigator’s judgment, have
contraindicated subject participation in the clinical study, or clinically significant enough in the
opinion of the Investigator to have altered the disposition of the study treatment, or constituted a
possible confounding factor for assessment of efficacy or safety of the study treatment.
23. Uncontrolled diabetes mellitus.
24. Subjects with any infection requiring oral antibiotic or antiviral therapy in the 2 weeks prior to
Screening or at baseline, injectable anti-infective therapy in the last 4 weeks prior to baseline, or
serious or recurrent infection with history of hospitalization in the 6 months prior to baseline.
25. Subjects with evidence of disseminated herpes zoster infection, zoster encephalitis, meningitis, or
other non-self-limited herpes zoster infections in the 6 months prior to baseline.
26. Subjects with planned surgery during the study or surgery ≤4 weeks prior to Screening and from
which the subject had not fully recovered, as judged by the Investigator.
27. Subjects with diverticulitis or other symptomatic GI conditions that could have predisposed the
subject to perforations, including subjects with history of such predisposing conditions (eg,
diverticulitis, GI perforation, or ulcerative colitis).
18
28. Pre-existing central nervous system demyelinating disorders (eg, multiple sclerosis and optic
neuritis).
29. History of chronic alcohol or drug abuse as judged by the Investigator.
30. Female subjects who were pregnant, currently lactating, had lactated within the last 12 weeks, or
who were planning to become pregnant during the study or within 6 months of the last dose of
study treatment.
31. Female subjects of childbearing potential (unless permanent cessation of menstrual periods,
determined retrospectively after a woman had experienced 12 months of natural amenorrhea as
defined by the amenorrhea with underlying status [eg, correlative age] or 6 months of natural
amenorrhea with documented serum follicle-stimulating hormone levels >40 mIU/mL and
estradiol <20 pg/mL) who were not willing to use a highly effective method of contraception during
the study and for at least 6 months after the last administration of study treatment
OR
Male subjects with partners of childbearing potential not willing to use a highly effective method
of contraception during the study and for at least 3 months after the last administration of study
treatment
Highly effective contraception was defined as:
Female sterilization surgery: hysterectomy, surgical bilateral oophorectomy (with or without
hysterectomy), or tubal ligation at least 6 weeks prior to the first dose of study treatment
In the case of oophorectomy alone, only when the reproductive status of the woman had
been confirmed by documented follow-up hormone level assessment
Total abstinence if that was the preferred and constant lifestyle of the subject. Thus, periodic
abstinence such as ovulation, symptothermal, postovulation, calendar methods, and withdrawal
were not acceptable methods of contraception.
Male sterilization surgery: at least 6 months prior to Screening (with the appropriate
postvasectomy documentation of the absence of sperm in the ejaculate). For female subjects,
the vasectomized male had to be the only partner.
Placement of established intrauterine device (IUD): IUD copper or IUD with progesterone.
Barrier method (condom and intravaginal spermicide, cervical caps with spermicide,
diaphragm with spermicide) in combination with the following: established oral, injected, or
implanted hormone methods of contraception or contraceptive patch.
32. Subjects with a known hypersensitivity to any component of the OKZ drug product, ADA, or PBO.
33. Subjects with a known hypersensitivity or contraindication to any component of the rescue
medication.
19
34. History of severe allergic or anaphylactic reactions to human, humanized, or murine monoclonal
antibodies.
35. Subject's unwillingness or inability to follow the procedures outlined in the protocol.
36. Other medical or psychiatric conditions or laboratory abnormalities that could have increased
potential risk associated with study participation and administration of investigational products, or
that could have affected study results interpretation and, as per the Investigator's judgment, made
the subject ineligible.
All subjects were free to withdraw from participation in the study at any time, for any reason, specified
or unspecified, and without prejudice to further treatment.
If premature withdrawal occurred for any reason, the Investigator made every effort to determine the
primary reason for a subject’s premature withdrawal from the study and record this information on the
source documents and appropriate electronic сase report form (eCRF).
Subjects were completely withdrawn from study treatment and all assessments in the following cases:
Protocol listed the reasons for premature treatment discontinuation. Among them there were the
following circumstances:
Subject presented with any of the following elevated liver function tests (LFTs):
ALT or AST elevations >8× ULN at any time, regardless of total bilirubin or accompanying
symptoms;
ALT or AST >5× ULN for ≥2 weeks regardless of total bilirubin or accompanying
symptoms;
ALT or AST elevations >3× ULN and total bilirubin value >2× ULN;
ALT or AST elevations >3× ULN accompanied by symptoms consistent with hepatic
injury (fatigue, nausea, vomiting, right upper quadrant pain or tenderness, fever, or rash);
The subjects who discontinued randomized study treatment early (ie, prior to Week 24) adhered to the
established double-blind treatment period visit schedule outlined in the study Schedule of Events, as if
they were still receiving study treatment. In addition, all subjects who discontinued study treatment
early were required to attend the End of Treatment (EoT) Visit 2 weeks after the last dose of the study
treatment, as well as Safety Follow-Up assessments 4, 8, and 22 weeks after the last dose of study
treatment.
Concomitant treatment with MTX was detailed in the protocol. Specifically, at the discretion of the
Investigator, the dose of MTX could be reduced once during the study for safety reasons. Concomitant
treatment with folic acid ≥5 mg per week or equivalent was required for all subjects starting by
Visit 2 (Week 0). Specific treatments prohibited prior to and during the course of the study are
described in the Table below. Other medications and non-drug therapies not listed within the Table
that were considered necessary for the subject’s safety and well-being could be given at the discretion
of the Investigator.
Prior MTX and all other treatments for RA for the 6 months prior to the Screening Visit were recorded
in the eCRF. All medications/treatments received within 4 weeks prior to the Screening Visit were also
recorded in the eCRF. Doses, route of applications, duration of treatment, and reasons for prescription
were also recorded.
21
Prohibited medications
Treatment Restriction
bDMARDs/ kinase
Treatment with any licensed or investigational biologics directly or
inhibitors
indirectly targeting IL-6 or IL-6R (including tofacitinib or other JAK
or SYK inhibitors) was prohibited during the entire study and their
use prior to Screening was exclusionary.
Treatment with cell-depleting therapies, including anti-CD20 agents
or investigational agents (e.g., CAMPATH, anti-CD4, anti-CD5,
anti-CD3, and anti-CD19), was prohibited during the entire study
and their use prior to Screening was exclusionary.
Treatment with bDMARDs, including tumor necrosis factor
inhibitors, was prohibited during the entire study and their use prior
to Screening was exclusionary.
22
Treatment Restriction
Corticosteroids
Treatment with an oral glucocorticoid greater than 10 mg/day
prednisone, or equivalent, or a change in dosage within 2 weeks prior
to baseline was exclusionary.
Use of parenteral glucocorticoids within 4 weeks prior to baseline
was exclusionary. Use of parenteral glucocorticoids was strongly
discouraged during the entire study, but limited use was allowed in
the following circumstance:
– No more than 2 joints could be injected at or after Week 14 after
all study assessments for this time point were performed. The
injection must not exceed 40 mg methylprednisolone or
equivalent cumulative dose. Injected joints were rated as having
their pre-injection status for the remainder of the study.
NSAIDs
NSAIDs were prohibited during the entire study with the following
exceptions:
– Stable doses of NSAIDs were permitted during the study if the
subject had received stable doses for ≥2 weeks prior to baseline.
Doses of NSAIDs were kept constant throughout the entire study
unless the Investigator changes the dose for safety reasons.
Switching of NSAIDs was not allowed. However, if the subject
had an AE that requires discontinuation of the NSAID, an
alternative NSAID could be initiated per the local label (if not
contraindicated).
Aspirin used at daily doses up to 325 mg was permitted if indicated
for CV protection. At this dose, aspirin was not considered an
NSAID.
Analgesics
Analgesics, including opioids, were prohibited during the entire
study with the exception of paracetamol/acetaminophen (maximum
2000 mg per day (maximum 1000 mg per dose).
Paracetamol/acetaminophen were not to be taken within 24 hours
prior to joint assessment, including baseline assessment.
23
Treatment Restriction
Hyaluronic acid
IA hyaluronic acid was prohibited during the entire study and its use
within 4 weeks prior to baseline was exclusionary.
Vaccination
Live vaccinations were prohibited during the entire study and their
use within 6 weeks prior to baseline was exclusionary.
Abbreviations: AE, adverse event; bDMARD, biologic disease-modifying anti-rheumatic drug; cDMARD,
conventional disease-modifying anti-rheumatic drug; CV, cardiovascular; IA, intraarticular; IL-6,
interleukin-6; IL-6R, IL-6 receptor; JAK, Janus kinase; MTX, methotrexate; NSAID, non-steroidal
anti-inflammatory drug; RA, rheumatoid arthritis; SYK, spleen tyrosine kinase.
Allowed medications
The following medications were allowed (non-exhaustive list), including clarifications of the
prohibited medications listed previously:
Inhaled corticosteroids
Topical corticosteroids
Oral corticosteroids
Doses of ≤10 mg/day of prednisone or equivalent were permitted as long as the dose was
not changed within the 2 weeks prior to baseline; dose adjustments were not permitted
during the study unless the Investigator changed the dose for safety reasons.
IA corticosteroids
No more than 2 joints could be injected during the study at or after Week 14, after all study
assessments for this time point were performed. The cumulative dose for both injections did
not exceed 40 mg methylprednisolone or equivalent. Joints treated with IA corticosteroids
were rated with their pre-injection status for the remainder of the study and should be
omitted from all subsequent joint assessments.
NSAIDs
The subject was receiving a stable dose for ≥2 weeks prior to baseline, and the dose was
kept constant throughout the entire study unless the Investigator changed the dose for safety
reasons.
Subjects were classified in terms of their response to study treatment at Week 14 with nonresponders
defined as all subjects who did not improve by at least 20% in both swollen and tender (66-68 joint
assessment). Nonresponders in all groups were prescribed sulfasalazine and/or hydroxychloroquine
according to the local label of the prescribed drug(s) as a rescue medication starting at or as close as
possible to Week 14, in addition to the assigned study treatment without unblinding.
Nonresponders at Week 14 remained blinded to their assigned treatment. Rescue medication was
administered as open-label treatment. The choice of rescue medication (sulfasalazine,
hydroxychloroquine, or both) was made according to local practice, and the assigned rescue medication
regimen was maintained throughout the study.
For subjects who received rescue medication, periodic safety evaluations for toxicity resulting from
sulfasalazine and/or hydroxychloroquine were undertaken as per the drug label and local guidelines.
Sensitivity analyses
For the primary outcome sensitivity analyses were also conducted in which intermediate missing data
were multiply imputed within treatment, data after treatment discontinuation were included, and data
after discontinuing the study were multiply imputed based on the placebo group for all subjects in the
Intention-to-treat population (ITT) and in the Per protocol population (PP); in these analyses
differences in the response rates for the OKZ q2w group, the OKZ q4w group and the adalimumab
group compared with the placebo group were statistically significant (p<0.0001 for all comparisons).
In the tipping point analysis to demonstrate the robustness of the ACR20 response for OKZ q4w
compared with placebo, all points were statistically significant (p<0.0125). In the tipping point
analysis of the ACR20 response for OKZ q2w compared with placebo, all points were statistically
significant (p<0.0125). In the tipping point analysis of the ACR20 response for adalimumab compared
with placebo, all points were statistically significant (p<0.025).
25
For the secondary outcome: to further compare the efficacy of OKZ with that of adalimumab, analyses
were conducted in which data after treatment discontinuation were included, and intermediate missing
data and data after discontinuing the study were multiply imputed within treatment in the ITT
population and in the PP population; non-inferiority of OKZ relative to adalimumab was demonstrated.
In the tipping point analysis of the ACR20 response for OKZ q2w compared with adalimumab, all
points demonstrated non-inferiority of OKZ relative to adalimumab using a margin of -12%. In the
tipping point analysis of the ACR20 response for OKZ q4w compared with adalimumab, all points
demonstrated non-inferiority of OKZ relative to adalimumab using a margin of -12% .
Efficacy assessments
All efficacy assesments were computed in the statistical database for analysis purposes, icluding
DAS28-CRP that was kept blinded after Screening.
ACR20/50/70 Response Criteria
The calculations were based on a ≥20%, ≥50%, and ≥70% improvement from baseline in the swollen
joint count (SJC) assessed in 66 joints and in the tender joint count (TJC) assessed in 68 joints; and a
≥20%, ≥50%, and ≥70% improvement from baseline in at least 3 of the 5 remaining core set measures:
1) Patient Global Assessment of Disease Activity (VAS); 2) Patient Assessment of Pain (VAS); 3)
HAQ DI; 4) Physician Global Assessment (VAS) and 5) Level of acute phase reactant (level of CRP
in this study).
The DAS28-CRP was calculated using the SJC (28 joints), TJC (28 joints), CRP level, and the Patient
Global Assessment of Disease Activity (VAS) (in mm) according to the following formula:
The CDAI was calculated using the SJC (28 joints), TJC (28 joints), the Patient Global Assessment of
Disease Activity (VAS) (in cm), and the Physician Global Assessment (VAS) (in cm) according to the
following formula:
CDAI = SJC + TJC + Patient Global Assessment of Disease Activity (VAS) + Physician Global
Assessment (VAS).
26
Health Assessment Questionnaire – Disability Index
The HAQ-DI assessed the degree of difficulty experienced in 8 domains of daily living activities using
20 questions. The domains were dressing and grooming, arising, eating, walking, hygiene, reach, grip,
and common daily activities, and each domain (activity) consisted of 2 or 3 items. For each question,
the level of difficulty was scored from 0 to 3 where 0 = without any difficulty, 1 = with some difficulty,
2 = much difficulty, and 3 = unable to do. Each category was given a score by taking the maximum
score of each question (i.e., question in each category with the highest score for that category).
If the maximum score was 0 or 1, but a device related to that category was used, or help from another
person was provided for the category, then the category score was increased to 2. If the category score
was already a 2 (or above), the score in that category remained 2 with or without any aids or device
use. If the subject did not answer for any questions within a category, no score was provided for that
category. The HAQ DI was calculated by dividing the sum of the category scores by the number of
categories with at least 1 question answered. If fewer than 6 categories had responses, no disability
score was calculated. The HAQ DI was also include patient assessments of pain and health on a scale
of 0 to 100.
27
FIGURES AND TABLES
Figure S1. Gate-keeping strategy
Notes: pSup, q2w and pSup, q4w represent p-values from a one-sided test of superiority versus placebo for OKZ dose
regimens 64 mg q2w and q4w respectively; 97.5% CLL OKZ q2w - Adb and 97.5% CLL OKZ q4w - Adb represent the
lower confidence limit of the 97.5% 2-sided CI for the difference between proportions versus ADA for OKZ
dose regimens 64 mg q2w and q4w respectively.
Abbreviations: ACR20, American College of Rheumatology 20% improvement response criteria; ACR50,
American College of Rheumatology 50% improvement response criteria; ADA, adalimumab; CDAI, Clinical
Disease Activity Index; CI, confidence interval; DAS28-CRP, Disease Activity Score 28 (based on C-reactive
protein data); HAQ-DI, Health Assessment Questionnaire-Disability Index; OKZ, olokizumab; PBO, placebo;
q2w, every 2 weeks; q4w, every 4 weeks; Wk, week
28
Figure S2. Non-inferiority testing details
Abbreviations: ACR20, American College of Rheumatology 20% improvement response criteria; ADA,
adalimumab; CI, confidence interval; DAS28-CRP, Disease Activity Score 28 (based on C-reactive protein
data); NI, non-inferiority; OKZ, olokizumab; q2w, every 2 weeks; q4w, every 4 weeks; Wk, week
29
Figure S3. Core components of ACR response criteria during the double‐blind treatment period (ITT population)
30
Figure S4. CDAI low disease activity and DAS28-CRP remission during the double‐blind treatment period (ITT population)
60 60
50,2
50 50
47,0
39,6 46,8
40 40 35,1
37,6
37,3 31,9
30 30
24,3 25,6 25,1 28,6
20 17,7 20
20,8 10,7
7,0
10 10
0 0 0
0 4 8 12 16 20 24 0 4 8 12 16 20 24
Weeks since Baseline Weeks since Baseline
Abbreviations: ADA, adalimumab; CDAI, Clinical Disease Activity Index; DAS28 (CRP), Disease Activity Score 28 (based on C-reactive protein data); ITT, intention-
to-treat; OKZ, olokizumab; PBO, placebo; q2w, every 2 weeks; q4w, every 4 weeks
31
Figure S5. Mean changes in laboratory values during double-blind treatment
period (safety population)
400 4,62
350 4,56
300 4,5
250 4,44
200 4,38
150 4,32
7 150
Neutrophil count, 10*9/L
5 140
4 135
3 130
2 125
5,8
40,0
Total сholesterol level, mmol/L
5,6
30,0
ALT, U/L
5,4
20,0
5,2
10,0
5
0,0
4,8
1,8 4
1,7 3,5
LDL level, mmol/L
HDL level, mmol/L
1,6 3
1,5 2,5
1,4 2
1,3 1,5
0 4 8 12 16 20 24 0 4 8 12 16 20 24
Weeks since baseline Weeks since baseline
Abbreviations: ADA, adalimumab; ALT, alanine aminotransferase; HDL, high density lipoproteins; LDL, low
density lipoproteins; OKZ, olokizumab; PBO, placebo; q2w, every 2weeks; q4w, every 4 weeks
32
Table S1. The numbers of randomized patients from each country (ITT
population)
OKZ 64 mg OKZ 64 mg ADA 40mg
Placebo Total
q2wk q4wk q2wk
N=243 N=1648
N=464 N=479 N=462
Сountries, n (%)
Argentina 42 (9.1) 44 (9.2) 43 (9.3) 24 (9.9) 153 (9.3)
Bulgaria 20 (4.3) 18 (3.8) 19 (4.1) 8 (3.3) 65 (3.9)
Brazil 33 (7.1) 33 (6.9) 30 (6.5) 16 (6.6) 112 (6.8)
Colombia 17 (3.7) 18 (3.8) 16 (3.5) 8 (3.3) 59 (3.6)
Czech Republic 50 (10.8) 58 (12.1) 51 (11.0) 30 (12.3) 189 (11.5)
Germany 13 (2.8) 11 (2.3) 10 (2.2) 6 (2.5) 40 (2.4)
Estonia 3 (0.6) 4 (0.8) 5 (1.1) 1 (0.4) 13 (0.8)
United Kingdom 1 (0.2) 3 (0.6) 6 (1.3) 1 (0.4) 11 (0.7)
Hungary 13 (2.8) 15 (3.1) 13 (2.8) 10 (4.1) 51 (3.1)
South Korea 4 (0.9) 2 (0.4) 1 (0.2) 4 (1.6) 11 (0.7)
Lithuania 22 (4.7) 22 (4.6) 22 (4.8) 8 (3.3) 74 (4.5)
Latvia 3 (0.6) 1 (0.2) 0 0 4 (0.2)
Mexico 63 (13.6) 63 (13.2) 59 (12.8) 30 (12.3) 215 (13.0)
Poland 85 (18.3) 90 (18.8) 87 (18.8) 46 (18.9) 308 (18.7)
Romania 0 0 1 (0.2) 0 1 (<0.1)
Russia 21 (4.5) 20 (4.2) 25 (5.4) 11 (4.5) 77 (4.7)
Taiwan 3 (0.6) 2 (0.4) 2 (0.4) 1 (0.4) 8 (0.5)
United States 71 (15.3) 75 (15.7) 72 (15.6) 39 (16.0) 257 (15.6)
Regions, n (%)
Western 148 (31.9) 162 (33.8) 152 (32.9) 86 (35.4) 548 (33.3)
Latin America 155 (33.4) 158 (33.0) 148 (32.0) 78 (32.1) 539 (32.7)
Rest of the World 161 (34.7) 159 (33.2) 162 (35.1) 79 (32.5) 561 (34.0)
Note: Western region was defined as Czech Republic, Germany, Hungary, United Kingdom and United
States; Latin America: Argentina, Brazil, Columbia, Mexico, Rest of the World: Bulgaria, Estonia, Latvia,
Lithuania, Poland, Romania, Russia, South Korea and Taiwan.
Abbreviations: ADA, adalimumab; ITT, intention-to-treat; n, number of subjects; N, total number of subjects
in the population; OKZ, olokizumab; q2wk, every 2 weeks; q4wk, every 4 weeks; %, percentage of subjects
calculated relative to the total number of subjects in the population.
33
Table S2. Reasons for discontinuation at Week 12 (ITT population)
OKZ 64 mg q2wk OKZ 64 mg q4wk ADA 40mg q2wk Placebo
Reason for discontinuation, n (%)
N=464 N=479 N=462 N=243
Discontinued treatment 26 (5.6) 29 (6.1) 34 (7.4) 26 (10.7)
Primary reason for premature discontinuation from treatment
Lack of efficacy (investigator perspective) 0 0 1 (0.2) 3 (1.2)
Adverse event 17 (3.7) 19 (4.0) 17 (3.7) 6 (2.5)
Pregnancy 0 0 0 0
Protocol violation 1 (0.2) 2 (0.4) 7 (1.5) 3 (1.2)
Consistent non-compliance with study drug dosing 0 0 1 (0.2) 0
Consistent non-compliance with study procedures 0 0 0 0
Use of prohibited concomitant medications 0 0 2 (0.4) 0
Violation of eligibility criteria 1 (0.2) 2 (0.4) 4 (0.9) 3 (1.2)
Other 0 0 0 0
Subject refusal 6 (1.3) 5 (1.0) 5 (1.1) 13 (5.3)
Lack of efficacy (subject perspective) 1 (0.2) 0 2 (0.4) 5 (2.1)
Withdrawal of informed consent 5 (1.1) 2 (0.4) 3 (0.6) 7 (2.9)
Other 0 3 (0.6) 0 1 (0.4)
Lost to Follow-Up 2 (0.4) 0 1 (0.2) 1 (0.4)
Discontinuation by sponsor 0 1 (0.2) 1 (0.2) 0
Study terminated by sponsor or investigator 0 0 0 0
Other 0 2 (0.4) 2 (0.4) 0
Abbreviations: ADA, adalimumab; ITT, intention-to-treat; n, number of subjects; N, total number of subjects in the population; OKZ, olokizumab; q2wk, every 2 weeks;
q4wk, every 4 weeks; %, percentage of subjects calculated relative to the total number of subjects in the population.
34
Table S3. Representativeness of study participants7,8
Category Example
Sex and gender RA affects women more than men (ratio of 3:1).
Geography Age of patients is about the same among countries (Western, Latin and Rest of the World), prevalence of RA in North Africa,
Middle East and Asia relatively low.
Other considerations MTX is the standard of start-up therapy and 1/3 - 1/2 received GCS
Overall representativeness of this Age distribution was the same among countries (adult patients was investigated). The proportion of Asian patients who
trial underwent randomization overall was small (1.5%), Black or African American patients was 4,0%. Health status was the same
between gender and region.
Table S4. Rheumatoid arthritis reference values of disease activity scores as per disease severity9
Question, n (%) OKZ 64 mg q2wk OKZ 64 mg q4wk ADA 40mg q2wk Placebo
N=464 N=479 N=462 N=243
How many didn’t complete 12 weeks?
(Including patients who discontinued treatment at 26 (5.6) 29 (6.1) 34 (7.4) 26 (10.7)
week 12)
How many didn’t complete 12 weeks?
(Patients discontinued treatment before week 12 (not 24 (5.2) 21 (4.4) 31 (6.7) 23 (9.5)
including week 12)
How many are non-responders? 138 (29.7) 137 (28.6) 153 (33.1) 135 (55.6)
Abbreviations: ADA, adalimumab; ITT, intention-to-treat; n, number of subjects; N, total number of subjects in the population; OKZ, olokizumab; q2wk, every 2 weeks;
q4wk, every 4 weeks; %, percentage of subjects calculated relative to the total number of subjects in the population.
36
Table S6. ACR20 response rates by visit and treatment (ITT population)
Analysis (visit) OKZ 64 mg q4wk OKZ 64 mg q2wk ADA 40 mg q2wk Placebo
N = 479 N = 464 N=462 N=243
Week 8 (visit 7), Nx (%) 479 (100.0) 479 (100.0) 462 (100.0) 243 (100.0)
n (%) 317 (66.2) 318 (68.5) 302 (65.4) 93 (38.3)
Comparison vs. PBO risk difference (97.5% CI/95% CI)1 0.279 [0.192, 0.361] 0.303 [0.215, 0.384] 0.271 [0.194, 0.343]
Comparison vs ADA risk difference 97.5% CI 0.008 [-0.061, 0.077] 0.032 [-0.038, 0.100]
Week 12 (visit 9), Nx (%) 479 (100.0) 479 (100.0) 462 (100.0) 243 (100.0)
n (%) 342 (71.4) 326 (70.3) 309 (66.9) 108 (44.4)
Comparison vs. PBO risk difference (97.5% CI/95% CI)1 0.270 [0.183, 0.362] 0.268 [0.171, 0.341] 0.224 [0.148, 0.298]
P-value <0.0001 <0.0001 <0.0001
Comparison vs ADA 0.045 0.034
risk difference 97.5% CI -0.022, 0.112 -0.035, 0.102
Non-inferiority Achieved? Yes Yes
Note: Subjects discontinuing the treatment or the study are considered nonresponders; 197.5% CI was calculated for comparison of OKZ vs PBO, 95% CI was calculated for
comparison of ADA vs PBO.
Abbreviations: ACR20: American College of Rheumatology 20%; ADA, adalimumab; CI, confidence interval; ITT: Intent-to-Treat: OKZ: olokizumab; q2wk, every 2
weeks; q4wk, every 4 weeks.
Note: N = Number of subjects in the analysis population: Nx = Number of subjects contributing to the analysis at the corresponding time point; n = Number of responders;
% = For Nx the percentage of subjects is calculated relative to the total number of subjects in the population (N). For n the percentage is calculated relative to Nx.
Note: Intermediate missing data are imputed using surrounding visits.
Note: Response is calculated relative to baseline, the last available assessment prior to the first dose of the study treatment.
Note: Confidence Interval is calculated using Newcombe hybrid score method.
Note: P-values are 1-sided p-values from 2x2 chi-square test.
Note: Non-Inferiority for each OKZ dosing regimen versus Adalimumab is achieved if the lower limit of the 97.5% confidence interval is greater than the protocol defined
non-inferiority margin of -12%.
37
Table S7. ACR20 response rate at Week 12 by visit, treatment and region (ITT population)
38
Table S8. ACR20 response rate at Week 12 by anti-CCP and RF status (ITT population)
OKZ 64 mg q2wk OKZ 64 mg q4wk ADA 40mg q2wk PBO
N=464 N=479 N=462 N=243
RF+ (≥ 20 IU/ml)
Nx (%) 352 (75.9) 355 (74.1) 343 (74.2) 181 (74.5)
n (%) 254 (72.2) 260.(73.2) 236 68.8) 77(42.5)
Comparison vs. PBO risk difference (97.5% CI/95% CI)1 29.6 [19.6, 39.0] 30.7 [20.7, 40.0] 26.3 [17.4, 34.7]
p-value <0.001 <0.001 <0.001
Comparison vs. ADA risk difference 97.5% CI 3.4 [-4.4, 11.1] 4.4 [-3.3, 12.1]
Anti-CCP+ (> 10 IU/ml)
Nx (%) 355 (76.5) 361 (75.4) 324 (70.1) 188 (77.4)
n (%) 257 (72.4) 262 (72.6) 225 (69.4) 85 (45.2)
Comparison vs. PBO risk difference (97.5% CI/95% CI)1 27.2 [17.3, 36.5] 27.4 [17.5, 36.7] 24.2 [15.4, 32.7]
p-value <0.001 <0.001 <0.001
Comparison vs ADA risk difference 97.5% CI 2.9 [-4.8, 10.7] 3.1 [-4.6, 10.9]
Either RF+ and/or Anti-CCP+
Nx (%) 383 (82.5) 391 (81.6) 367 (79.4) 203 (83.5)
n (%) 277 (72.3) 286 (73.1) 251 (68.4) 90 (44.3)
Comparison vs. PBO risk difference (97.5% CI/95% CI)1 28.0 [18.5, 37.0] 28.8 [19.3, 37.7] 24.1 [15.6, 32.1]
p-value <0.001 <0.001 <0.001
Comparison vs ADA risk difference 97.5% CI 3.9 [-3.5, 11.4] 4.8 [-2.6, 12.1]
Both RF+ and Anti-CCP+
Nx (%) 324 (69.8) 325 (67.8) 300 (64.9) 166 (68.3)
n (%) 234 (72.2) 236 (72.6) 210 (70.0) 72 (43.4)
Comparison vs. PBO risk difference (97.5% CI/95% CI)1 28.8 [18.4, 38.6] 29.2 [18.8, 39.0] 26.6 [17.3, 35.4]
p-value <0.001 <0.001 <0.001
Comparison vs ADA risk difference 97.5% CI 2.2 [-5.9, 10.3] 2.6 [-5.5, 10.7]
Seronegative
Nx (%) 79 (17.0) 87 (18.2) 95 (20.6) 40 (16.5)
n (%) 49 (62.0) 56 (64.4) 58 (61.1) 18 (45.0)
39
OKZ 64 mg q2wk OKZ 64 mg q4wk ADA 40mg q2wk PBO
N=464 N=479 N=462 N=243
Comparison vs. PBO risk difference [97.5% CI/95% CI]* 17.0 [-4.3,36.6] 19.4 [-1.6, 38.5] 16.1 [-2.1, 33.0]
p-value 0.038 0.020 0.043
Comparison vs ADA risk difference 97.5% CI 1.0 [-15.4, 17.0] 3.3 [-12.6, 18.0]
Notes: 197.5% CI was calculated for comparison of OKZ vs PBO, 95% CI was calculated for comparison of ADA vs PBO. Percentage for Nx is calculated relative to
the total number of subjects in the population, percentage for n is calculated relative to Nx, p-values are 1-sided p-values from 2x2 chi-square test.
Abbreviations: ACR20, American College of Rheumatology 20% improvement response criteria; ADA, adalimumab; Anti-CCP, anti-cyclic citrullinated peptide; CI,
confidence interval; ITT, intent-to-treat; N, number of subjects in the analysis population; Nx, number of subjects contributing to the analysis at the corresponding time
point; n, number of responders; OKZ, olokizumab; PBO, placebo; q2wk, every 2 weeks; q4wk, every 4 weeks; RF, rheumatoid factor.
40
Table S9. ACR20 response rate at Week 12 by time since diagnosis (ITT population)
OKZ 64 mg q2wk OKZ 64 mg q4wk ADA 40mg q2wk Placebo
N=464 N=479 N=462 N=243
0 to < 2 Years
Nx (%) 127 (27.4) 123 (25.7) 130 (28.1) 70 (28.8)
n (%) 90 (70.9) 90 (73.2) 83 (63.8) 34 (48.6)
Comparison vs. PBO risk difference (97.5% CI/95% CI)1 22.3 [6.1, 37.5] 24.6 [8.3, 39.7] 15.3 [1.0, 29.0]
p-value <0.001 <0.001 0.018
Comparison vs ADA risk difference 97.5% CI 7.0 [-6.0, 19.7] 9.3 [-3.8, 21.9]
≥ 2 to < 5 Years
Nx (%) 111 (23.9) 116 (24.2) 112 (24.2) 57 (23.5)
n (%) 83 (74.8) 76 (65.5) 82 (73.2) 29 (50.9)
Comparison vs. PBO risk difference (97.5% CI/95% CI)1 23.9 [6.5, 40.3] 14.6 [-2.9, 31.6] 22.3 [7.0, 36.9]
p-value <0.001 0.032 0.002
1.6 -7.7
Comparison vs ADA risk difference 97.5% CI
[-11.5, 14.6] [-20.9, 6.0]
≥ 5 Years
Nx (%) 224 (48.3) 240 (50.1) 220 (47.6) 116 (47.7)
n (%) 153 (68.3) 176 (73.3) 144 (65.5) 45 (38.8)
Comparison vs. PBO risk difference (97.5% CI/95% CI)1 29.5 [16.8, 41.0] 34.5 [22.1, 45.7] 26.7 [15.5, 36.9]
p-value <0.001 <0.001 <0.001
Comparison vs ADA difference 97.5% CI 2.8 [-7.1, 12.7] 7.9 [-1.7, 17.4]
Notes: 197.5% CI was calculated for comparison of OKZ vs PBO, 95% CI was calculated for comparison of ADA vs PBO. Percentage for Nx is calculated relative
to the total number of subjects in the population, percentage for n is calculated relative to Nx, p-values are 1-sided p-values from 2x2 chi-square test.
Abbreviations: ACR20, American College of Rheumatology 20% improvement response criteria; ADA, adalimumab; CI, confidence interval; ITT, intent-to-treat;
N, number of subjects in the analysis population; Nx, number of subjects contributing to the analysis at the corresponding time point; n, number of responders; OKZ,
olokizumab; PBO, placebo; q2wk, every 2 weeks; q4wk, every 4 weeks.
41
Table S10. Main efficacy results for other endpoints (ITT population)
OKZ 64 mg q2wk OKZ 64 mg q4wk ADA 40mg q2wk PBO
N=464 N=479 N=462 N=243
DAS28 (CRP) <2.6†week 24, n (%) 148 (31.9) 168 (35.1) 132 (28.6) 26 (10.7)
Comparison vs. PBO risk difference (97.5% CI/95% CI)1 21.2 [14.2; 27.4]# 24.4 [17.3; 30.6]# 17.9 [11.9; 23.3]#
CDAI <10, n (%) week 242 211 (45.5) 224 (46.8) 227 (49.1) 59 (24.3)
Comparison vs. PBO risk difference (97.5% CI/95% CI)1 21.2 [12.8; 28.8]# 22.5 [14.1; 30.1]# 24.8 [17.5; 31.6]#
ACR70 response, n (%) week 24 133 (28.7) 129 (26.9) 119 (25.8) 27 (11.1)
Comparison vs. PBO risk difference (97.5% CI/95% CI)1 17.6 [10.6; 23.7]# 15.8 [9.0; 21.9]# 14.7 [8.7; 20.0]#
Comparison vs. PBO risk difference (97.5% CI/95% CI) 1 5.6 [-0.3; 10.9]* 3.5 [-3.1; 9.4] 4.1 [-1.6; 9.4]
HAQ-DI improvement of ≥0.22, n (%) week 12 322 (69.4) 340 (71.0) 313 (67.7) 130 (53.5)
Comparison vs. PBO risk difference (97.5% CI/95% CI)1 15.9 [7.3; 24.4]# 17.5 [8.9; 25.9]# 14.2 [6.5; 21.6]#
Footnotes: 1Risk difference in percentage points; 97.5% CI was calculated for comparison of OKZ vs. PBO; 95% CI was calculated for comparison of ADA vs. PBO;
†p≤0.05, ⱡp<0.001; #p<0.0001 compared with placebo.
Abbreviations: ACR, American College of Rheumatology response; ADA, adalimumab; CDAI, Clinical Disease Activity Index; CI, confidence interval; CRP, C-
reactive protein; DAS28-CRP, Disease Activity Score 28 based on CRP; HAQ-DI, Health Assessment Questionnaire Disability Index; NRI, non-responder imputation;
N, number of subjects; n, number of responders; OKZ, olokizumab; PBO, placebo; q2wk, every 2 weeks; q4wk, every 4 weeks; SE, standard error; Wk, week
42
Table S11. Treatment-emergent adverse events (safety population)
System Organ Class OKZ 64 mg q2wk OKZ 64 mg q4wk ADA 40 mg q2wk Placebo
Preferred Term (more than 4% in any group), n (%) N=463 N=477 N=462 N=243
Subjects with at Least One TEAE 324 (70.0) 338(70.9) 302(65.4) 154(63.4)
Infections and infestations 140 (30.2) 162 (34.0) 148 (32.0) 84 (34.6)
Nasopharyngitis 29 (6.3) 26 (5.5) 29 (6.3) 18 (7.4)
Upper respiratory tract infection 28 (6.0) 29 (6.1) 26 (5.6) 16 (6.6)
Urinary tract infection 7 (1.5) 14 (2.9) 20 (4.3) 9 (3.7)
Bronchitis 12 (2.6) 10 (2.1) 12 (2.6) 11 (4.5)
Investigations 108 (23.3) 127 (26.6) 53 (11.5) 26 (10.7)
ALT increased 42 (9.1) 53 (11.1) 9 (1.9) 4 (1.6)
AST increased 23 (5.0) 24 (5.0) 8 (1.7) 2 (0.8)
Metabolism and nutrition disorders 93 (20.1) 62 (13.0) 38 (8.2) 17 (7.0)
Hypercholesterolaemia 29 (6.3) 20 (4.2) 6 (1.3) 3 (1.2)
Dyslipidaemia 25 (5.4) 15 (3.1) 4 (0.9) 4 (1.6)
Hyperlipidaemia 22 (4.8) 10 (2.1) 5 (1.1) 2 (0.8)
43
System Organ Class OKZ 64 mg q2wk OKZ 64 mg q4wk ADA 40 mg q2wk Placebo
Preferred Term (more than 4% in any group), n (%) N=463 N=477 N=462 N=243
Skin and subcutaneous tissue disorders 44 (9.5) 37 (7.8) 22 (4.8) 15 (6.2)
Nervous system disorders 34 (7.3) 22 (4.6) 28 (6.1) 19 (7.8)
Headache 10 (2.2) 11 (2.3) 14 (3.0) 12 (4.9)
Vascular disorders 30 (6.5) 43 (9.0) 20 (4.3) 10 (4.1)
Hypertension 25 (5.4) 28 (5.9) 13 (2.8) 8 (3.3)
Respiratory, thoracic and mediastinal disorders 27 (5.8) 28 (5.9) 34 (7.4) 11 (4.5)
Notes: AEs were collected from the signature of the informed consent form until the last visit of the subject in the study (up to 22 weeks after the last dose of study
treatment, thus up to approximately 44 weeks). A TEAE was defined as an AE that first occurred or worsened in severity after the first dose of the study treatment;
%, percentage of subjects calculated relative to the total number of subjects in the population.
MedDRA, Medical Dictionary for Regulatory Activities (Version 21.1) was used to code AEs.
Abbreviations: ADA, adalimumab; AE, adverse event; ALT, alanine aminotransferase; AST, aspartate aminotransferase; MACE, Major Adverse Cardiac Events;
MedDRA, Medical Dictionary for Regulatory Activities; N, number of subjects; n, number of subjects with events; OKZ, olokizumab; PBO, placebo; q2wk, every 2
weeks; q4wk, every 4 weeks; TEAE, treatment-tmergent adverse event.
44
Table S12. Key serious treatment-emergent adverse events (safety population)
OKZ 64 mg q2wk OKZ 64 mg q4wk ADA 40 mg q2wk Placebo
Number of subjects (%)
N=463 N=477 N=462 N=243
Subjects with at least one key TESAE 22 (4.8) 20 (4.2) 26 (5.6) 12 (4.9)
Serious Infections 6 (1.3) 7 (1.5) 16 (3.5) 4 (1.6)
Pneumonia 1 (0.2) 1 (0.2) 6 (1.3) 0
Sepsis 1 (0.2) 1 (0.2) 2 (0.4) 0
Urosepsis 0 0 2 (0.4) 2 (0.8)
Cellulitis 0 2 (0.4) 0 0
Urinary tract infection 0 0 2 (0.4) 0
Herpes zoster 0 0 1 (0.2) 0
Pulmonary tuberculosis 0 1 (0.2) 0 0
Neoplasms benign, malignant and unspecified (incl. cysts and
3 (0.6) 2 (0.4) 2 (0.4) 3 (1.2)
polyps)
Investigations 3 (0.6) 2 (0.4) 1 (0.2) 2 (0.8)
ALT increased 1 (0.2) 2 (0.4) 1 (0.2) 0
Transaminases increased 2 (0.4) 0 0 1 (0.4)
General disorders and administration site conditions 2 (0.4) 1 (0.2) 1 (0.2) 1 (0.4)
45
OKZ 64 mg q2wk OKZ 64 mg q4wk ADA 40 mg q2wk Placebo
Number of subjects (%)
N=463 N=477 N=462 N=243
Cardiac disorders 0 2 (0.4) 2 (0.4) 0
Respiratory, thoracic and mediastinal disorders 1 (0.2) 0 3 (0.6) 0
Interstitial lung disease 0 0 2 (0.4) 0
Gastrointestinal disorders 1 (0.2) 0 2 (0.4) 0
Musculoskeletal and connective tissue disorders 1 (0.2) 0 2 (0.4) 0
A TEAE Leading to death** 3 (0.6) 2 (0.4) 1 (0.2) 1 (0.4)
Subjects with at least one MACE*** 4 (0.9) 2 (0.4) 2 (0.4) 1 (0.4)
Notes: *One cholecystitis (0.2%) and one cholelytiasis (0.2%) in OKZ q2w; one hepatotoxicity (0.2%) and one hepatic steatosis (0.2%) in OKZ q4w; one
hepatotoxicity (0.4%) in PBO.
**Cerebrovascular accident (stroke) 1(0.2%), infections and infestations (sepsis 1, septic shock 1) 2 (0.4%) in OKZ q2w; infections and infestations (sepsis) 1(0.2%),
cardiac disorders (MI) 1 (0.2%) in OKZ q4w; infections and infestations (sepsis) 1 (0.2%) in ADA and sudden death 1 (0.4%) in PBO.
***One fatal cerebrovascular accident (stroke) (0.2%), one death due to undetermined cause (0.2%), two non-fatal strokes/ TIAs (0.6%) in OKZ q2w; one
cardiovascular death (0.2%) and non-fatal MI (0.2%) in OKZ q4w; two non-fatal strokes/ TIAs (0.4%) in ADA; one cardiovascular death (0.4%) in PBO.
AEs were collected from the signature of the informed consent form until the last visit of the subject in the study (up to 22 weeks after the last dose of study treatment,
thus up to approximately 44 weeks). A TEAE was defined as an AE that first occurred or worsened in severity after the first dose of the study treatment; %, percentage
of subjects calculated relative to the total number of subjects in the population.
Abbrviations: ADA, adalimumab; AE, adverse event; ALT, alanine aminotransferase; MACE, major adverse cardiac events; MI, myocardial infarction; N, number
of subjects; n, number of subjects with events; OKZ, olokizumab; PBO, placebo; q2wk, every 2 weeks; q4wk, every 4 weeks; TEAE, treatment-emergent adverse
event; TESAE, treatment-emergent serious adverse event; TIA, transient ischemic attack.
Definition: MACEs were defined as cardiovascular death, death from undetermined cause, non-fatal myocardial infarction, non-fatal stroke or transient ischemic
attack, hospitalization for unstable angina requiring unplanned revascularization, and coronary revascularization procedures.
46
Table S13. Selected hematology assessments outside of the normal ranges (safety population)
Number of patients, n (%) OKZ 64 mg q2wk OKZ 64 mg q4wk ADA 40 mg q2wk Placebo
N=463 N=477 N=462 N=243
WBC count < 4000/mm3 Nх (%) 463 (100.0) 475 (99.6) 462 (100.0) 243 (100.0)
Baseline
(or < 4 ·109/L) n (%) 3 (0.6) 4 (0.8) 4 (0.9) 5 (2.1)
Nx (%) 427 (92.2) 437 (91.6) 418 (90.5) 213 (87.7)
Wk 12
n (%) 42 (9.1) 36 (7.5) 7 (1.5) 3 (1.2)
Nх (%) 407 (87.9) 411 (86.2) 389 (84.2) 198 (81.5)
Wk 24
n (%) 39 (8.4) 40 (8.4) 7 (1.5) 2 (0.8)
ANC count < 1500/mm3 Nх (%) 463 (100.0) 475 (99.6) 462 (100.0) 243 (100.0)
Baseline
(or < 1.5 ·109/L) n (%) 0 (0.0) 0 (0.0) 0 (0.0) 1 (0.4)
Nх(%) 426 (92.0) 433 (90.8) 416 (90.0) 213 (87.7)
Wk 12
n(%) 16 (3.5) 11 (2.3) 2 (0.4) 1 (0.4)
Nх (%) 406 (87.7) 411 (86.2) 387 (83.8) 198 (81.5)
Wk 24
n (%) 10 (2.2) 13 (2.7) 1 (0.2) 0 (0.0)
ANC count < 1000/mm 3
Nх (%) 463 (100.0) 475 (99.6) 462 (100.0) 243 (100.0)
Baseline
(or < 1.0 ·109/L) n (%) 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0)
Nх (%) 426 (92.0) 433 (90.8) 416 (90.0) 213 (87.7)
Wk 12
n (%) 1 (0.2) 0 (0.0) 0 (0.0) 0 (0.0)
Nх (%) 406 (87.7) 411 (86.2) 387 (83.8) 198 (81.5)
Wk 24
n (%) 1 (0.2) 1 (0.2) 0 (0.0) 0 (0.0)
ALC count < 500/mm3 Nх (%) 463 (100.0) 475 (99.6) 462 (100.0) 243 (100.0)
Baseline
(or < 0.5 ·10 /L)
9
n (%) 0 (0.0) 0 (0.0) 2 (0.4) 1 (0.4)
Wk 12 Nх (%) 426 (92.0) 433 (90.8) 416 (90.0) 213 (87.7)
47
Number of patients, n (%) OKZ 64 mg q2wk OKZ 64 mg q4wk ADA 40 mg q2wk Placebo
N=463 N=477 N=462 N=243
n (%) 1 (0.2) 1 (0.2) 1 (0.2) 1 (0.4)
Nх (%) 406 (87.7) 411 (86.2) 387 (83.8) 198 (81.5)
Wk 24
n (%) 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0)
Hb ≤80 g/L Nх (%) 463 (100.0) 476 (99.8) 462 (100.0) 243 (100.0)
Baseline
n (%) 1 (0.2) 0 1 (0.2) 0
Nх (%) 428 (92.4) 440 (92.2) 422 (91.3) 215 (88.5)
Wk 12
n (%) 0 0 0 1 (0.4)
Nх (%) 409 (88.3) 411 (86.2) 391 (84.6) 200 (82.3)
Wk 24
n (%) 0 0 0 1(0.4)
Notes: percentage of subjects with non-missing results was calculated relative to the total number of subjects in the population. Percentage of subjects with abnormal
parameters was calculated relative to the number of subjects with non-missing results at a given visit.
Abbreviations: ADA, adalimumab; ALC, absolute lymphocyte count; ANC, absolute neutrophil count; Hb, hemoglobin; N, number of subjects in the arm; Nx(%),
number and percent of the subjects with non-missing results; n(%), number and percent of the subjects with abnormal parameters; OKZ, olokizumab; PBO, placebo;
q2wk, every 2 weeks; q4wk, every 4 weeks; WBC, white blood cells; Wk, week.
48
Table S14. Selected chemistry assessments outside of the normal ranges (safety population)
OKZ 64 mg q2wk OKZ 64 mg q4wk ADA 40 mg q2wk Placebo
Number of
N=463 N=477 N=462 N=243
Nx 463 (100.0) 477 (100.0) 462 (100.0) 243 (100.0)
ALT >1x ULN to ≤ 3x ULN 43 (9.3) 58 (12.2) 37 (8.0) 21 (8.6)
ALT, n (%) BL
> 3x ULN to ≤ 5x ULN 1 (0.2) 0 (0.0) 2 (0.4) 1 (0.4)
> 5x ULN 1 (0.2) 0 (0.0) 0 (0.0) 0 (0.0)
Nx 463 (100.0) 477 (100.0) 461 (99.8) 243 (100.0)
ALT, n (%) ALT >1x ULN to ≤ 3x ULN 247 (53.3) 250 (52.4) 153 (33.2) 75 (30.9)
post-BL worst case > 3x ULN to ≤ 5x ULN 34 (7.3) 30 (6.3) 11 (2.4) 2 (0.8)
> 5x ULN 8 (1.7) 10 (2.1) 3 (0.7) 3 (1.2)
Nx 463 (100.0) 477 (100.0) 462 (100.0) 243 (100.0)
Total bilirubin, n (%) BL > 2x ULN 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0)
> 3x ULN 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0)
Total bilirubin, n (%) Nx 463 (100.0) 477 (100.0) 461 (99.8) 243 (100.0)
post-BL worst case > 2x ULN 4 (0.9) 4 (0.8) 0 (0.0) 1 (0.4)
Nx 459 (99.1) 473 (99.2) 458 (99.1) 237 (97.5)
Cholesterol, n (%) BL
High 174 (37.9) 177 (37.4) 191 (41.7) 92 (38.8)
Cholesterol, n (%) Post-BL Nx 459 (99.1) 474 (99.4) 459 (99.4) 243 (100.0)
High 346 (75.4) 357 (75.3) 295 (64.3) 129 (53.1)
Wk 12, n (%) 126 ( 29.2) 126 ( 28.8) 64 ( 15.2) 17 ( 8.1)
Cholesterol shifts from normal
Mean change, mmol/L, (SD) 1.31 (0.66) 1.40 (0.87) 0.90 (0.56) 0.67 (0.50)
to high
Wk 24, n (%) 122 ( 30.3) 109 ( 26.6) 66 ( 16.8) 21 ( 10.6)
49
OKZ 64 mg q2wk OKZ 64 mg q4wk ADA 40 mg q2wk Placebo
Number of
N=463 N=477 N=462 N=243
Mean change, mmol/L, (SD) 1.39 (0.73) 1.32 (0.69) 0.98 (0.57) 0.81 (0.74)
Nx 459 ( 99.1) 473 ( 99.2) 458 ( 99.1) 237 ( 97.5)
LDL Cholesterol, n (%) BL
High 101 ( 22.0) 119 ( 25.2) 113 ( 24.7) 50 ( 21.1)
LDL Cholesterol, n (%) Post- Nx 459 ( 99.1) 474 ( 99.4) 459 ( 99.4) 243 (100.0)
BL High 259 ( 56.4) 272 ( 57.4) 200 ( 43.6) 81 ( 33.3)
Wk 12, n (%) 97 ( 22.6) 99 ( 22.7) 46 ( 11.0) 13 ( 6.2)
LDL Cholesterol shifts from Mean change, mmol/L, (SD) 1.06 (0.58) 1.15 (0.85) 0.88 (0.49) 0.60 (0.26)
normal to high Wk 24, n (%) 96 ( 23.9) 87 ( 21.3) 51 ( 13.0) 19 ( 9.6)
Mean change, mmol/L,(SD) 1.18 (0.60) 1.08 (0.53) 0.91 (0.50) 0.77 (0.61)
Notes: there were no subjects with concomitant elevations of ALT or AST ≥3xULN and bilirubin ≥2xULN. Percentage of subjects with non-missing results (Nx) was
calculated relative to the total number of subjects in the population. All other percentages of subjects were calculated relative to the number of subjects with non-missing
results at a given visit.
Abbreviations: ADA, adalimumab; ALT, alanine aminotransferase; BL, baseline; LDL, low density lipoproteins; N, number of subjects in the population; Nx, number
of the subjects with non-missing results; n(%), number and percent of the subjects with abnormal parameters; OKZ, olokizumab; PBO, placebo; q2wk, every 2 weeks;
q4wk, every 4 weeks; SD, standard deviation; ULN, upper limit of normal; Wk, week.
50
Contributorship
CJSC R-Pharm was involved in the study design, collection, analysis, interpretation of data, and
validation of information provided in the manuscript. EN, MS, SF, and EK were involved in study
conceptualization and data analysis. All authors had unrestricted access to the study data, contributed
to the interpretation of the results and participated in the development of this manuscript. The authors
did not receive honoraria related to the development of this manuscript. All authors were responsible
for all the content and editorial decisions. The authors meet the authorship criteria recommended by
the International Committee of Medical Journal Editors (ICMJE).
Medical writing assistance, under the direction of the authors, and editorial support, including
development of the first draft, were provided by Sofia Kuzkina according to CONSORT 2010
Statement: updated guidelines for reporting parallel-group randomized trials
(https://www.bmj.com/content/340/bmj.c332) and Good Publication Practice guidelines.
REFERENCES:
1. Keystone E, Kavanaugh A, Sharp J, et al. Radiographic, clinical, and functional outcomes of treatment
with adalimumab (a human anti-tumor necrosis factor monoclonal antibody) in patients with active
rheumatoid arthritis receiving concomitant methotrexate therapy: a randomized, placebo-controlled.
Arthritis and Rheumatism 2004;50(5):1400–11.
2. Weinblatt M, Keystone E, Furst D, et al. Adalimumab, a fully human anti-tumor necrosis factor alpha
monoclonal antibody, for the treatment of rheumatoid arthritis in patients taking concomitant
methotrexate: the ARMADA trial. Arthritis and Rheumatism 2003;48(1):35–45.
3. Keystone E, van der Heijde D, Kavanaugh A, et al. Clinical, functional, and radiographic benefits of
longterm adalimumab plus methotrexate: final 10-year data in longstanding rheumatoid arthritis. The
Journal of Rheumatology 2013;40(9):1487–97.
4. Taylor P, Keystone E, van der Heijde D, et al. Baricitinib versus Placebo or Adalimumab in Rheumatoid
Arthritis. The New England journal of medicine 2017;376(7):652–62.
5. Rubin DB. Multiple Imputation for Nonresponse in Surveys. Hoboken, NJ, USA: John Wiley & Sons,
Inc.; 1987.
6. Aletaha D, Neogi T, Silman AJ, et al. 2010 Rheumatoid arthritis classification criteria: An American
College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis &
Rheumatism 2010;62(9):2569–81.
8. Yip K, Navarro-Millán I. Racial, ethnic, and healthcare disparities in rheumatoid arthritis. Current
Opinion in Rheumatology 2021;33(2):117–21.
9. Aletaha D, Smolen JS. The Definition and Measurement of Disease Modification in Inflammatory
Rheumatic Diseases. Rheumatic Disease Clinics of North America 2006;32(1):9–44.
51