Role of Toxicopathologist in Drug Discovery and Development

Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Role of Toxicopathologist in Drug Discovery and Development

Dr. Pavan Patil, MVSc, DABT, DIBTP


Sr. Scientist, Toxicology/ Clinical Safety
Johnson & Johnson Ltd, Mumbai, India

1
Outline

➢ Drug Research and Development Process

• Target Identification
• Target Validation
• Lead Optimization
• Candidate Selection
• Preclinical Research
• Clinical Development
• Regulatory Review, Approval
• Post-Marketing Safety Surveillance

• Role of Toxicopathologist in Drug Discovery and Development

2
The Drug Research and Development Process

(Ashburn 2004)
3
Target Identification

• Target identification starts with isolating the function of a possible therapeutic target (gene/nucleic acid/protein)
and its role in the disease.

• Identification of the target is followed by characterization of the molecular mechanisms addressed by the target

• Targets has to be
➢ Efficacious and Safe
➢ Usable as drugs,
➢ Capable of meeting clinical and commercial requirements.

• Various Approaches for Target Identification


➢ Data mining using bioinformatics
— identifying, selecting potential disease targets
➢ Genetic association
— genetic polymorphism and connection with the disease
➢ Expression profile
— changes in mRNA/protein levels
— In vitro cell-based mechanistic studies
➢ Functional screening
— knockdown, knockout or using target specific tool
4
Target Validation

• Target validation is the process by which the expected molecular target – gene/nucleic acid/protein is
certified.

• Conducting careful and precise target validation experiments is essential for the success of drug
development.

• Target validation includes


➢ Determining the structure activity relationship (SAR) of analogs
➢ Generating a drug-resistant mutant of the presumed target
➢ Knockdown or over expression of the presumed target
➢ Monitoring the known signaling systems downstream of the presumed target

• Whilst the validation of a drug‘s efficacy and toxicity in numerous disease-relevant cell models and
animal models is extremely valuable

5
Lead Optimization

• Lead optimization is the process by which a drug candidate is designed


after an initial lead compound is identified.

• The purpose of lead optimization is to maintain favorable properties,


specificity and selectivity in lead compounds, while improving on
deficiencies in lead structure.

• The process involves series of synthesis and characterization of a potential


drug

• In initial drug discovery, the resulting leads from hit-to-lead high


throughput screening tests undergo lead optimization, to identify
promising compounds.

• Pharmacodynamic and pharmacokinetic parameters and toxicological


properties are also evaluated.

6
Candidate Selection

• Out of the several promising “high-quality” leads, one is taken forward as a clinical candidate.

• For a drug candidate to be deemed suitable for preclinical and clinical testing it should;
➢ Bind selectively to the target,
➢ Elicit the desired functional response when interacting with the target molecule
➢ Must have adequate bioavailability and biodistribution to elicit the desired response
➢ It must have a toxicity profile

• Additionally, following factors are also considered;


➢ future manufacturing suitability and scale-up
➢ commercial viability and cost-effectiveness

• When the candidate molecule shows promise as a therapeutic, it must be characterized—


the molecule’s size, shape, strengths and weaknesses, preferred conditions for maintaining function, bioactivity,
bioavailability and toxicity

• Characterization studies will undergo analytical method development and validation.

• Early stage pharmacology studies help to characterize the underlying mechanism of action of the compound.
7
Formulation and Development

• Physicochemical properties of clinical candidates are characterized to


produce a bioavailable, stable and optimal dosage form for a specific
administration route.

• During preformulation studies the following parameters are evaluated:

➢ Solubility in different media and solvents


➢ Dissolution
➢ Accelerated Stability Services under various conditions
➢ Solid state properties (polymorphs, particle size/ shape)
➢ Optimization of existing formulations
➢ Process development for selected dosage forms
➢ Novel formulations for improved delivery of existing dosage
forms

8
Preclinical Research

• Preclinical testing analyzes the bioactivity, safety, and efficacy of the


formulated drug product through laboratory and animal testing

• In Vivo, In Vitro & Ex Vivo Assays

➢ In vivo, is research conducted using mice, rat and dog models.


➢ In vitro is research conducted in a laboratory.
➢ Ex vivo uses animal cells or tissues from a non-living animal.
o a cell is used as the basis for small explant cultures that
provide a dynamic, controlled, and sterile environment.

• In Silico Assays

➢ In silico assays are test systems or biological experiments


performed on a computer or via computer simulation.

9
Preclinical Research

Acute Studies
• One or more doses administered over a period of up to 24 hours to determine toxic dose levels and observe
clinical indications of toxicity.
• Acute tox studies helps determine doses for repeated dose studies in animals and Phase I studies in humans.

Repeated Dose Studies


• Repeated dose studies may be referred to as subacute, subchronic, or chronic.
• The specific duration should anticipate the length of the clinical trial that will be conducted on the new drug.

Genetic Toxicity Studies


• These studies assess the likelihood that the drug compound is mutagenic or carcinogenic.
• Procedures such as the Ames test (conducted in bacteria) detect genetic changes.
• DNA damage is assessed in tests using mammalian cells such as the Mouse Micronucleus Test.
• The Chromosomal Aberration Test and similar procedures detect damage at the chromosomal level.

10
Preclinical Research

Reproductive Toxicity Studies


• Segment I reproductive tox studies detect effects of the drug on fertility.
• Segment II and III studies detect effects on embryonic and post-natal development.
• Reproductive tox studies must be completed before a drug can be administered to women of child-bearing
age.

Carcinogenicity Studies
• Carcinogenicity studies are usually needed only for drugs intended for chronic or recurring conditions.
• They are time consuming and expensive and must be planned for early in the preclinical testing process.

Toxicokinetic Studies
• These are typically similar in design to PK/ADME studies except that they use much higher dose levels.
• They examine the effects of toxic doses of the drug and help estimate the clinical margin of safety.

There are numerous FDA and ICH guidelines that give a wealth of detail on the different types of preclinical
toxicology studies and the appropriate timing for conduct of studies

11
Clinical Development

Phase I – Healthy Volunteer Study

• This phase is the first time the drug is tested on


humans; less than 100 volunteers
• These studies are used to evaluate pharmacokinetic
parameters and tolerance
• These studies include initial single-dose studies, dose
escalation and short-term repeated-dose studies.

Phase II – Studies in Patient Population

• Phase II assesses drug safety and efficacy in an


additional 100-500 patients, who may receive a placebo
or standard drug previously used as treatment.
• Analysis of optimal dose strength helps create
schedules while adverse events and risks are recorded.

12
Clinical Development

Phase III – Studies in Patient Population

• Phase III enrolls 1,000-5,000 patients, enabling


medication labeling and instructions for proper drug use.
• Phase III trials require extensive collaboration,
organization, coordination and regulation in anticipation
of full-scale production following drug approval.

Complexity of Study Design, Associated Cost &


Implementation Issues

• Trials must be safe and efficacious and be completed


under the drug development budget, using a
methodology to ensure the drug works as well as
possible for its intended purpose.
• This rigorous process must be set up correctly and enroll
many volunteers to be effective.

13
Regulatory Review and Approval

The data submitted to the regulatory authorities should give confidence


• The drug is safe and effective as a treatment for the condition it has been developed for
• The drug’s therapeutic benefits outweigh the risks
• The drug’s labeling is fit-for-purpose and whether all required details are included
• The methods used to manufacture the drug and measures to ensure the drug's qualities are satisfactory

14
Product Launch

Once the drug receives approval from the relevant regulatory authority, numerous activities are initiated to prepare
for the launch of the product.

These include:
• Manufacturing scale-up
• Printing of final product label information, packaging and artwork
• Product storage, shipping and distribution arrangements
• Production staff and quality team availability

Post-marketing Safety Surveillance

• Post-marketing safety surveillance is the term used for the monitoring of a drug after it has received approval
and has reached the market.
• It is designed to evaluate the long-term safety and efficacy of a drug, potential “real-world” problems with
formulation, and use for unapproved conditions or “off-label”

15
Role of Toxicopathologist in Drug Development

• Select relevant species for nonclinical studies and Genetically


modified animal models such as transgenic or knockout mice for
discovery, nonclinical toxicity and pharmacokinetic studies.

• Use their expertise to identify and describe findings in nonclinical


studies taking into account species and age.

• Develop critical interpretative expertise for differentiating test


compound effects from background findings known to occur in a
particular species.

• The toxicologic pathologist working in discovery is uniquely


equipped to provide a ‘‘whole animal’’ perspective within a team of
chemists, biologists and pharmacologists

16
Role of Toxicopathologist in Drug Development

• Ensure the selected models accurately reflect


human disease states and expected
pharmacology

17
Role of Toxicopathologist in Drug Development

• The toxicologic pathologist assists in identifying suitable biomarkers.

• Correlate the results from specialized techniques such as


immunohistochemistry, immunofluorescence, in situ hybridization,
and reverse transcription-polymerase chain reaction (RT-PCR), with
observations made from tissue sections or cell preparations.

• Additionally, test compound effects are evaluated by looking at


clinical pathology parameters

➢ Alterations in numbers and types of bone marrow or circulating


blood cells;
➢ Levels of proteins, electrolytes, and tissue-specific enzymes in
blood and urine that reflect specific organ status;
➢ Functional parameters such as blood clotting times.

18
Role of Toxicopathologist in Drug Development

• Identify target organ toxicity by evaluating macroscopic, and


microscopic changes, and to interpret these findings within the
context of overall study results, including toxicokinetics and in-life
observations.

• Evaluate data from a series of nonclinical studies of increasing


duration, beginning with acute, single-dose studies, then repeated
dose range-finding studies (often 2 weeks in duration), followed by
subacute (4 and 13 weeks) and chronic (26 and 52 weeks)
repeated dose studies.

• This assessment results in the establishment of a no-observed-


adverse-effect-level (NOAEL), which is used to calculate safe
starting doses of the compound in clinical trials

19
20
21
References
• Deore, AB, Dhumane JR, Wagh HV, Sonawane RB, The Stages of Drug Discovery and Development Process. Asian
Journal of Pharmaceutical Research and Development. 2019; 7(6):62-67
• Shayne CG. Introduction: drug Discovery in the 21stCentury. Drug Discovery Handbook, Wiley Press, 2005; 1-10.
• Smith GC, OíDonnel JT. The Process of New Drug Discovery and Development, Eds., 2nd edition, Informa Healthcare,
New York 2006
• Raquel Maeso-Díaz et al, New Rat Model of Advanced NASH Mimicking Pathophysiological Features and
Transcriptomic Signature of The Human Disease, Cells 2019, 8, 1062
• Lindsay MA. Target discovery. Nature Reviews Drug Discovery, 2003; 2:831–838.
• Robert A. Ettlin, Toxicologic Pathology in the 21st Century, Toxicologic Pathology, 41: 689-708 (2013)
• Susan van Tongeren, Jane A. Fagerland et al, The Role of the Toxicologic Pathologist in the Biopharmaceutical
Industry, International Journal of Toxicology 30(5) 568-582 (2011)
• Yvonne Oligschlaeger and Ronit Shiri-Sverdlov, NAFLD Preclinical Models: More than a Handful, Less of a Concern?
Biomedicines 2020, 8(2), 28
• Ashburn, T. T., & Thor, K. B. (2004). Drug repositioning: identifying and developing new uses for existing drugs.
Nature Reviews Drug Discovery, 3(8), 673–683.

22

You might also like