Drug Evaluation & Regulation. CONCEPTS A.Safety and Efficacy: Because society expects prescription drugs to be safe and effective, governments have regulated.

Slides:



Advertisements
Similar presentations
© 2004 by Thomson Delmar Learning, a part of the Thomson Corporation. Fundamentals of Pharmacology for Veterinary Technicians Chapter 2 Veterinary Drug.
Advertisements

The Drug Discovery Process
Drug design and testing,. Drug Names Chemical name- describes its molecular structure and distinguishes it from other drugs.
Drug Research and Development (R&D) Karol Godwin DVM.
Drugs Used During Pregnancy & Lactation
1 Pharmacology/Toxicology information to submit an IND for an anticancer drug.
Clinical Trials Importance in future therapies. What are the Requirements to Produce New Drugs? Drug must work significantly better than a control treatment.
Clinical Trials — A Closer Look. The Food and Drug Administration (FDA) is the main consumer watchdog for numerous products: Drugs and biologics (prescription.
Clinical Trials Medical Interventions
JMV 1843 pharmacological profile
U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES NATIONAL INSTITUTES OF HEALTH Working with FDA: Biological Products and Clinical Development Critical Path.
The Application of the Scientific Method: Preclinical Trials Copyright PEER.tamu.edu.
What Do Toxicologists Do?
Pharmaceutical Development and Review Process Rev. 10/21/2014 APGO Interaction with Industry: A Medical Student Guide.
+ Drug Development and Review Process. + Objectives Learn the processes involved in drug discovery and development Define the phases involved in FDA drug.
Drug Discovery Process
Biomedical research methods. What are biomedical research methods? An integrated approach using chemical, mathematical and computer simulations, in vitro.
Food and Drug Administration Preclinical safety data for “first in human” (FIH) clinical trials in healthy volunteer subjects Oncology Drug Advisory Committee.
Stages of drug development
The Drug Development Process
Eureka Pre-Clinical Investigation Animal toxicology Animal pharmacokinetics/ pharmacodynamics Clinical Investigation Phase I Safety and pharmacology Phase.
Development of a drug Based on: a known active drug known receptor known endogenous ligand Of natural origin: microorganisms Plants Animals.
Medicines and drugs Option D Part 1.
Clinical Trials — A Closer Look. The Food and Drug Administration (FDA) is the main consumer watchdog for numerous products: Drugs and biologics (prescription.
Clinical Trials Amir Zarrinhaghighi
Dr. Dipa Brahmbhatt VMD MpH DRUG DEVELOPMENT AND CONTROL Chapter 2 Dr. Dipa Brahmbhatt VMD MpH
A substance used in the diagnosis, treatment, or prevention of a disease or as a component of a medication A substance used in the diagnosis, treatment,
ADVERSE EFFECTS OF DRUGS Phase II May Adverse Drug Reaction An adverse reaction to a drug is a harmful or unintended response. ADRs are claimed.
PHARMACOVIGILANCE AND CLINICAL TRIALS DIVISION 20 August 2015 Victoria Falls Protecting Your Right to Quality Medicines and Medical Devices.
From the Lab to Market Unit 3.04 Understanding Biotechnology research & Development.
Nonclinical Perspective on Initiating Phase 1 Studies for Small Molecular Weight Compounds John K. Leighton, PH.D., DABT Supervisory Pharmacologist Division.
Biomedical Research Objective 2 Biomedical Research Methods.
Biomedical Research.
CHEE DRUG PRODUCT DEVELOPMENT u Drug ä agent intended for use in the diagnosis, mitigation, treatment, cure, or prevention of disease in man or animals.
Investigational Drugs in the hospital. + What is Investigational Drug? Investigational or experimental drugs are new drugs that have not yet been approved.
AN INTRODUCTION TO CLINICAL PHARMACOLOGY. BEFORE STARTING DRUG THERAPY, DOCTORS CONSIDER  Whether intervention needed ?  Objective of treatment ? 
Joint Dermatologic and Ophthalmic Drugs & Drug Safety and Risk Management Advisory Committee February 26 & 27, 2004 RISK MANAGEMENT OPTIONS FOR PREGNANCY.
FDA Case Studies Pediatric Oncology Subcommittee March 4, 2003.
MAIN TOXICITY TESTING. TESTING STRATEGIES A number of different types of data are used in order to establish the safety of chemical substances for use.
The New Drug Development Process (www. fda. gov/cder/handbook/develop
The FDA: Basic Facts It takes 12 to 15 years to develop a single drug Only 1 in 10,000 potential medications makes it completely through the process Only.
History of Pediatric Labeling
Chapter 2 Veterinary Drug Development and Control
C LINICAL R ESEARCH. CONTENTS Drug Development Process Pre – Clinical Studies Clinical Trials Phase I Phase II Phase III Phase IV 2.
“Journey of a Drug” From Test Tube TO Prescribing Physician.
European Patients’ Academy on Therapeutic Innovation Special Populations.
Clinical Trials - PHASE II. Introduction  Important part of drug discovery process  Why important??  Therapeutic exploratory trial  First time in.
E-Clinical
Pharmacodynamics Department Of Pharmacology. Tolerance: Decreased response to a drug following repeated administration though the dose is constant. Acute.
Clinical trials are a set of procedures in medical research conducted to allow safety and efficacy data to be collected for health interventions. The.
A substance used in the diagnosis, treatment, or prevention of a disease or as a component of a medication recognized or defined by the U.S. Food, Drug,
Section I General principle of Pharmacology. Where can you get information about general principle of Pharmacology?  Text books:  Katzung, Basic and.
The process of drug development. Drug development 0,8 – 1 mld. USD.
Clinical Trials.
FDA DRUG APPROVAL FDA’s Lengthy Drug Approval Process in Twelve Steps Overview of the FDA Drug Approval Process Drug Developed June 13, 2016 | Emilia Varrone.
Pharmacology Science that studies interactions of drugs with organism on different levels (subcellular, cellular, organ, systemic) Studies: - relationship.
Drug Development Process Stages involved in Regulating Drugs
The Stages of a Clinical Trial
Drug Synthesis Noadswood Science, 2016.
Drug Discovery &Development
Regulatory– Terms & Definitions רגולציה - מונחים והגדרות
Schedule 2 Drugs Henderson.
Basic & Clinical Evaluation of New Drugs
Clinical Trials — A Closer Look
Prof. Dr. Basavaraj K. Nanjwade
Drug and Drug Products Quality & Testing
Drug Design and Drug Discovery
By Amany Helmy Hasanin Assistant Professor of Clinical Pharmacology
Objective 2 Biomedical Research Methods
Design Issues in Human studies of Psychopharmacology placebo controls double blind studies.
Presentation transcript:

Drug Evaluation & Regulation

CONCEPTS A.Safety and Efficacy: Because society expects prescription drugs to be safe and effective, governments have regulated the development and marketing of new drugs. The Food & Drug Administration (FDA) is the regulatory body in the USA that proposes and administers these regulations.

The FDA requires evidence of: –relative safety (derived from acute and subacute toxicity testing in animals) –probable therapeutic action (from the pharmacologic profile in animals) before human testing is permitted. –Some information about the pharmacokinetics of a compound. The cost of development of a new drug, including false starts and discarded molecules, is currently several hundred million dollars.

B.Animal Testing: The amount of animal testing required before human studies begin is a function of the proposed use and the urgency of the application. Thus, a drug proposed for occasional nonsystemic use requires less extensive testing than one destined for chronic systemic administration. Anticancer drugs and drugs proposed for use in AIDS, because of the urgent need for new agents, require less evidence of safety than do drugs used in treatment of less threatening diseases and are often investigated and approved on an accelerated schedule.

1.Acute toxicity: –Required for all drugs. –Involve administration of single doses of the agent up to the lethal level in at least two species, e.g., one rodent and one nonrodent.

The aim of these studies is to: identify the dose of the drug which causes major adverse effects to help set doses for further preclinical studies to predict the effects of overdose in humans.

2. Subacute and chronic toxicity: –Required for most agents, especially those intended for chronic use. –Tests are usually conducted for at least the amount of time proposed for human application, 2-4 weeks (subacute) or 6-24 months (chronic), –in at least two species.

C.Types of Animal Tests: Tests done with animals often include: –general screening tests for pharmacologic effects –hepatic and renal function monitoring –blood and urine tests –gross and histopathologic examination of tissues –tests of reproductive effects and carcinogenicity.

1.Pharmacologic profile: A description of all the pharmacologic effects of a drug (e.g., effects on blood pressure, gastrointestinal activity, respiration, renal function, endocrine function, the central nervous system). 2.Reproductive toxicity: involves the study of the fertility effects of the candidate drug and its teratogenic and mutagenic effects.

 Teratogenesis: induction of developmental defects in the fetus (by exposure of the fetus to a drug, radiation, etc). Teratogenesis is studied by treating pregnant female animals of at least two species at selected times during early pregnancy when organogenesis is known to take place and later examining the fetuses or neonates for abnormalities. pregnant female

Examples of teratogenic drugs: –thalidomide, ethanol, glucocorticoids, valproic acid, isotretinoin, warfarin, lithium, and androgens.  Mutagenesis is induction of changes in the genetic material of animals of any age and therefore induction of heritable abnormalities.

The Ames test, the standard in vitro test for mutagenicity, uses a special strain of salmonella bacteria that naturally depends on specific nutrients in the culture medium. Loss of this dependence during exposure to the test drug signals a mutation. in the absence of histidine, the bacteria are unable to grow on minimal medium

The dominant lethal test is an in vivo mutagenicity test carried out in mice. Male animals are exposed to the test substance before mating. Abnormalities in the results of subsequent mating (loss of embryos, deformed fetuses, etc) signal a mutation in the male's germ cells. Many carcinogens (e.g., aflatoxin, cancer chemotherapeutic drugs, and other agents that bind to DNA) have mutagenic effects.

3.Carcinogenesis: Carcinogenesis is the induction of malignant characteristics in cells. Agents with known carcinogenic effects include coal tar, aflatoxin, dimethylnitrosamine and other nitrosamines, urethane, vinyl chloride, and the polycyclic aromatic hydrocarbons in tobacco smoke, e.g., benzo[a]pyrene.

D.Clinical Trials: Human testing in the USA requires the prior approval of an lnvestigational New Drug Exemption application (IND), which has been submitted by the manufacturer to the FDA. The major clinical testing process is informally divided into three phases before a New Drug Application (NDA) can be submitted. The NDA constitutes the request for approval of general marketing of the new agent for prescription use. A fourth phase of study follows NDA approval.

1.Phase I: Consists of careful evaluation of the dose-response relationship in a small number of normal human volunteers (e.g., 20-30). An exception is in phase I trials of cancer chemotherapeutic agents; these are carried out by administering the agents to patients with cancer. In phase 1 studies, the acute effects of the agent are studied over a broad range of dosages, stalling with one that produces no detectable effect and progressing to one that produces either a significant physiologic response or a very minor toxic effect.

2. Phase II: involves evaluation of a drug in a moderate number of patients (eg, ) with the target disease. A placebo or positive control drug is included in a single- blind or double-blind design. The study is carried out under very carefully controlled conditions and patients are very closely monitored, often in a hospital research ward. The goal is to determine whether the agent has the desired therapeutic effects at doses that are tolerated by sick patients.

3. Phase III: Consists of a large design involving many patients (e.g., or more in many centers) and many clinicians who are using the drug in the manner proposed for its ultimate general use, e.g., in outpatients. Such studies usually include placebo and positive controls in a double-blind crossover design. The goal is to explore further the spectrum of beneficial actions of the new drug, to compare it with older therapies, and to discover toxicities, if any, that occur so infrequently as to be undetectable in phase II studies.

4. Phase IV: Phase IV represents the postmarketing surveillance phase of evaluation, in which it is hoped that toxicities that occur very infrequently will be detected and reported early enough to prevent major therapeutic disasters. Unlike the first three phases, phase IV is not rigidly regulated by the FDA.

 Orphan Drugs:  An orphan drug is a drug for a rare disease (one affecting fewer than 200,000 people).  The study of such agents has often been neglected because the sales of an effective agent for an uncommon ailment might not pay the costs of development.  In the USA, current legislation provides for tax relief and other incentives designed to encourage the development of orphan drugs.

 Adverse Drug Reactions (ADRs) An adverse reaction to a drug is a harmful or unintended response. Some adverse reactions, such as: –Overdose –excessive effects –drug interactions.