Docsity
Docsity

Prepare for your exams
Prepare for your exams

Study with the several resources on Docsity


Earn points to download
Earn points to download

Earn points by helping other students or get them with a premium plan


Guidelines and tips
Guidelines and tips

Drug Discovery and Development Process, Lecture notes of Pharmacy

An overview of the drug discovery and development process, including the discovery of anti-HIV and cancer drugs, target discovery, lead discovery and optimization, pre-clinical studies, clinical trials, and post-marketing surveillance. It also discusses the role of natural products in drug discovery and the advantages and disadvantages of using them. The document further explains the preclinical phase and the different types of toxicity testing involved in it.

Typology: Lecture notes

2022/2023

Available from 01/04/2024

juju-43
juju-43 🇵🇭

9 documents

1 / 7

Toggle sidebar

This page cannot be seen from the preview

Don't miss anything!

bg1
DRUG DISCOVERY AND DEVELOPMENT PRPM150
TRANSCRIBED BY: JULIA MARIE BABAYEN-ON
M1: INTRODUCTION TO DRUG DISCOVERY AND
DEVELOPMENT
Diseases and conditions that were once deemed incurable
or fatal have been conquered with therapeutic agents
designed to extend and improve quality of life.
DISCOVERY OF ANTI-HIV DRUG
Human immunodeficiency virus (HIV)
Causative agent of AIDS
AZT (Retrovir®, azidothymidine)
First RT inhibitor approved for tx of AIDS
Highly active antiretroviral therapy (HAART)
Multidrug cocktail treatment regimens
CANCER DRUG DEVELOPMENT
Survival rates for many types of cancer have dramatically
improved as a result of the discovery and development of
novel therapeutic agents.
Overall cancer death rates in the United States have
declined by 27% between 1991 and 2016
THE PROCESS
A multidimensional one and as such requires the
coordinated effort of individuals with a wide array of
expertise
A. The cost associated with the identification of useful and
marketable therapeutic entities is staggering
B. As of 2011, it is estimated that a single new drug costs
over $1.75 Billion to discover and develop.
Identification of a single marketed drug can require an
initial examination of over 100,000 candidate
compounds, hundreds of preclinical animal studies, and
numerous clinical trials involving thousands of patients.
Success rate: <0.001%
DRUG DISCOVERY
TARGET DISCOVERY
The choice of a disease area and identification of a
biological target
LEAD DISCOVERY
Structurally related compounds with the desired biological
activity are identified (HIT and LEAD compounds) through
biological screening of large numbers of compounds.
LEAD OPTIMIZATION
Lead compounds are modified and studied to identify
potential candidates for drug development.
DRUG DEVELOPMENT
PRE-CLINICAL STUDIES
Decides whether a drug is ready for clinical trials and
involves extensive studies (e.g. animal models) that yield
preliminary information on;
efficacy,
Toxicity
pharmacokinetic
safety
SUBMISSION OF AN INVESTIGATIONAL NEW DRUG
APPLICATION (INDA)
INDA requests permission to move a clinical candidate into
human study.
pf3
pf4
pf5

Partial preview of the text

Download Drug Discovery and Development Process and more Lecture notes Pharmacy in PDF only on Docsity!

TRANSCRIBED BY : JULIA MARIE BABAYEN-ON

M1: INTRODUCTION TO DRUG DISCOVERY AND

DEVELOPMENT

● Diseases and conditions that were once deemed incurable or fatal have been conquered with therapeutic agents designed to extend and improve quality of life. DISCOVERY OF ANTI-HIV DRUG ● Human immunodeficiency virus (HIV) ○ Causative agent of AIDS ● AZT (Retrovir®, azidothymidine) ○ First RT inhibitor approved for tx of AIDS ● Highly active antiretroviral therapy (HAART) ○ Multidrug cocktail treatment regimens CANCER DRUG DEVELOPMENT ● Survival rates for many types of cancer have dramatically improved as a result of the discovery and development of novel therapeutic agents. ● Overall cancer death rates in the United States have declined by 27% between 1991 and 2016 THE PROCESS ● A multidimensional one and as such requires the coordinated effort of individuals with a wide array of expertise A. The cost associated with the identification of useful and marketable therapeutic entities is staggering B. As of 2011, it is estimated that a single new drug costs over $1.75 Billion to discover and develop. ● Identification of a single marketed drug can require an initial examination of over 100,000 candidate compounds, hundreds of preclinical animal studies, and numerous clinical trials involving thousands of patients. ● Success rate: <0.001%

DRUG DISCOVERY

TARGET DISCOVERY

● The choice of a disease area and identification of a biological target LEAD DISCOVERY ● Structurally related compounds with the desired biological activity are identified (HIT and LEAD compounds) through biological screening of large numbers of compounds. LEAD OPTIMIZATION ● Lead compounds are modified and studied to identify potential candidates for drug development.

DRUG DEVELOPMENT

PRE-CLINICAL STUDIES

● Decides whether a drug is ready for clinical trials and involves extensive studies (e.g. animal models) that yield preliminary information on; ○ efficacy, ○ Toxicity ○ pharmacokinetic ○ safety SUBMISSION OF AN INVESTIGATIONAL NEW DRUG APPLICATION (INDA) ● INDA requests permission to move a clinical candidate into human study.

TRANSCRIBED BY : JULIA MARIE BABAYEN-ON

● INDA provides regulatory agencies with detailed preclinical data and detailed clinical protocols that describe how the compounds will be studied in human populations. CLINICAL TRIALS (PHASE I, II, AND III) ● PHASE I Safety and tolerability of an IND are examined in a small number of healthy individuals, typically 20 to 100 people. ● PHASE II - Safety and efficacy of IND are determined in around 100 to 300 patients. ● PHASE III - Efficacy of IND in larger population including comparison with standard therapies. SUBMISSION OF NEW DRUG APPLICATION (NDA) TO REGULATORY BODY ● NDA typically contains comprehensive details of both animal and human studies, ○ Safety findings ○ manufacturing procedures ○ dosing methods ○ storage conditions CLINICAL TRIAL (PHASE IV)/POSTMARKETING SURVEILLANCE (PMS ● PMS detects rare and long-term adverse effects across a much larger population of patients than could be supported in Phase III. ● Impact of PMS can include; ○ alterations to labeling based on safety results, ○ contraindication for use of the new drug in combination with other medications, or withdrawal of marketing approval if the findings are severe enough. ● Companies often use the data in PMS to identify competitive advantages, new markets, and new indications for the products.

ETHNOPHARMACOLOGY AND NATURAL PRODUCT

RESEARCH

● A glance at the pharmacopeia will show that many therapeutic agents, particularly anti infective and antineoplastic drugs, originate from natural products, rather than synthetic molecules. ● Examples: ○ Antibiotics - derived from fungal metabolites. ○ Paclitaxel - anticancer drug from yew tree ○ Vinca alkaloids - anticancer drugs produced by plants of the periwinkle family ○ Opiates - analgesic compounds from poppies ○ Artemether - antimalarial drug, semisynthetic derivative of artemisinin, produced by Chinese herb ● ADVANTAGES: ○ Such ready-made, highly evolved biomolecules stand a better chance of interacting with selected drug targets than do random synthetic molecules, and the pool from which they come is huge and largely untapped. ● DISADVANTAGES: ○ Accessibility due to geographical, cultural and political reasons ○ Continuing availability of the active compound may be uncertain. ○ Purification and structure determination of natural products is often difficult and time consuming "Exploiting such a ready-made compound library from natural products is seen as an attractive strategy for important therapeutic breakthroughs.

M2 : DRUG DISCOVERY AND DEVELOPMENT

PROCESS

TARGET IDENTIFICATION AND VALIDATION

● Drugs were mostly found by identifying API by: ○ Traditional medicines with folkloric use [ETHNOPHARMACOLOGY] ○ Serendipity ○ Classical methods in pharmacology ● Drug discovery and development involves ○ screening hit compounds, ○ medicinal chemistry, and ○ optimization of those target hits ■ For safety ■ efficacy ■ metabolic stability ■ and oral bioavailability Aims to identify the key molecular drivers of disease called TARGETS. TARGETS –naturally existing cellular or molecular structure involved in the pathophysiology of interest.

CLASSIFICATION

● Carbohydrates, lipids and PROTEINS ● “DRUG BANK” database most comprehensive databases of existing drug and targets RECEPTORS ● G-PROTEIN COUPLED RECEPTORS (GPCRs) ○ Most common; activate internal signal transduction pathways, and ultimately, cellular responses.

TRANSCRIBED BY : JULIA MARIE BABAYEN-ON

● The number of hits needs to be reduced, or filtered, to reach a more manageable figure. ● Pharmacodynamics ○ Activity in a biochemical assay is strictly a measure of how a molecule interacts with a target ● Pharmacokinetics ○ Most commercially successful drugs are administered orally, ○ Diffusion across Caco-2 (colon carcinoma) cell membranes (most common membrane permeability assay) ○ Ability for cytochrome P-450 enzymes to metabolize a hit is tested with liver microsome ● Final Concerns for Promotion of a Hit to a Lead ○ The surviving hits, sometimes called compounds of interest or similar, receive additional scrutiny. ○ Each remaining hit undergoes a handful of structural modifications.

SPECIAL CASES

SERENDIPITY

● Some scientific discoveries, including drugs, are labeled as serendipitous. ● Whether serendipitous drug discoveries are accidental is arguable, but the discoveries are unexpected. ● The sagacity required for such discoveries must be credited in part to the powers of observation of the researchers CLINICAL OBSERVATIONS ● Selective optimization of side activities (SOSA) ○ a drug with poor target specificity can be modified to enhance the side effect and decrease the original effect

LEAD OPTIMIZATION

● Aim: Identification of a compound which possesses the required properties (physicochemical, biological and pharmacological) of the targeted preclinical development candidate ● Focus: ADME parameter THE FRAMEWORK FOR SUCCESSFUL LEAD OPTIMIZATION

PRE-FORMULATION STUDIES

● Objectives: ○ To lay down FOUNDATION for developing a new drug entity into a pharmaceutical formulation ○ To provide STABILITY to the formulation OPTIMIZATION OF NEW DRUG ENTITY ● To improve stability and bioavailability through developing the molecule into: ○ Salt - Most widely used approach ○ Prodrug - Chemically modified inactive derivative of the active form DETERMINATION OF CHEMICAL PROPERTIES

  1. Partition coefficient - Drug’s ability to cross the phospholipid cell membrane
  2. Dissociation constant - Solubility in pH-dependent environment and extent of ionization
  3. Chirality - Ability of molecular entities to exist in different racemic forms
  4. Stability - Conditions in which the molecule is susceptible to degradation DETERMINATION OF PHYSICAL PROPERTIES
  5. Solubility - Most widely studied techniques during preformulation analysis
  6. Crystalline vs Amorphous form - Crystalline form is more stable but less water soluble
  7. Polymorphism - Ability of a compound to exist in different crystalline forms with different internal lattices
  8. Deliquescence and Hygroscopicity - Moisture level
  9. Particle size - Affects dissolution rate, solubility, bioavailability, content uniformity
  10. Particle shape - Affects surface area, flow properties, and compaction force
  11. Density - Dependent on particle size distribution and shape
  12. Flow property - Measured using Hausner’s ratio, Carr’s index, and angle of repose DRUG EXCIPIENT COMPATIBILITY
  13. Physical incompatibility a. No formation of new compounds; may involve organoleptic changes
  14. Chemical incompatibility a. Production of new unstable chemical entity
  15. Therapeutic incompatibility

TRANSCRIBED BY : JULIA MARIE BABAYEN-ON

a. aka: BIOPHARMACEUTICAL INTERACTION b. Associated with alteration in drug absorption

M3 : PRECLINICAL PHASE

PRECLINICAL TRIALS

● “Preclinical trials or nonclinical trials” ● Laboratory test of a new drug substance or medical devices, usually done on animal subjects ○ Determines a products’ ultimate safety profile ○ Develop adequate safety data before human trial ○ Part of INDA

TYPES OF TOXICITY TESTING

ACUTE TOXICITY TESTING

● Single high doses are given to small groups of animals ● Observed for mortality for 1-3 days ● Organ toxicity is examined by histopathology ● Lethal dose (LD50) ○ dose which kills 50% animals ○ Involves large numbers of animals, 14 days observation

REPEATED DOSE TOXICITY TESTING

● Carried out for a minimum of 28 days ● The test substance is administered regularly at a specific time SUB-CHRONIC ORAL TOXICITY TESTING ● The test substance is administered orally for 90 days. ● The test substance is administered regularly at a specific time. CHRONIC ORAL TOXICITY TESTING ● The test compound is administered over more than 90 days, and the animals are observed periodically. ● Provides inferences about the long-term effect of a test substance ● Essential for new drug entities NEUROTOXICITY STUDIES ● Studies the effects of a test substance on the CNS CARCINOGENICITY TESTING ● Observed for signs of toxicity and development of tumors. ● 18 months for mice and hamsters; 24 months for rats MUTAGENICITY TESTING ● Assess submicroscopic changes in the base sequence of DNA ○ Mutagenicity: permanent transmissible variations that can increase the frequency of mutations GENOTOXICITY TESTING ● Used to identify gene mutations, chromosome changes, and alterations in the DNA sequencing ○ Genotoxicity: ability of harmful substances to damage genetic information ONE-GENERATION REPRODUCTION TOXICITY TESTING ● Rodents are preferred (M and F) ● Observed for 1 spermatogenic cycle and 2 estrous cycles Animals are allowed to mate and parturition is observed. DEVELOPMENT TOXICITY/EMBRYOTOXICITY ● The compound is administered between the 8th and 14th day of pregnancy, and embryo lethal effects are observed. ● At the end of the study or on the 21st day of the study, a cesarean section is performed and parameters such as fetal deformation and mortality are observed. TOXICOKINETICS ● Deals with the kinetic patterns of higher doses of chemicals/toxins/xenobiotics

TRANSCRIBED BY : JULIA MARIE BABAYEN-ON

FACTORS TO CONSIDER WHEN USING IN VITRO

METHODS

● Know whether in vitro methods are accepted by regulatory authorities in your jurisdiction or not. ● Consider animal welfare issues. ● Know the limitations of in vitro methods

GUIDELINES FOR ETHICAL CONDUCT IN THE CARE

AND USE OF ANIMALS FOR PRECLINICAL TRIALS

IN DRUG TESTING

● Department of Agriculture’s (DA) Bureau of Animal Industry (BAI) ● Animal Welfare Act of 1998 (Republic Act No. 8485

ADMINISTRATIVE ORDER (AO) NO.

● The requirements for the authorization to conduct research using animals are the following:

  1. Description of the Animal Care and Use Program (ACUP) signed by a duly licensed veterinarian representing the entity,
  2. Animal Care and Use Program Accreditation Certificate issued by a duly recognized body or association,
  3. Animal Technician Training Program on Laboratory animal care and use, and
  4. Certification of Assurance that an Institutional Animal Care and Use Committee (IACUC) is in existence in the establishment. ETHICAL CONSIDERATION IN ANIMAL EXPERIMENTATION – 3R’S ● Replacement ● Reduction ● Refinement