+ All Categories
Home > Documents > Drug Development

Drug Development

Date post: 16-Nov-2014
Category:
Upload: api-3750955
View: 193 times
Download: 2 times
Share this document with a friend
Popular Tags:
33
DRUG DEVELOPMENT
Transcript
Page 1: Drug Development

DRUG DEVELOPMENT

Page 2: Drug Development

The number of new chemical entities (NCEs) gaining market approval has decreased over the last decade down to 20 per year.

The estimated average of new NCEs needed for the pharmaceutical and biotech industry to sustain a 5 percent growth rate is 50.

Page 3: Drug Development

STANDARDS

•GLP-Good Laboratory Practice refers to nonclinical laboratory studies that support or are intended to support applications for research or marketing permits;

•GMP-Good Manufacturing Practice, also known as cGMP ("current" GMP), is a set of regulations requiring that quality, safety, and effectiveness be built into foods, drugs, medical devices, and biological products.

•21 CFR-describing the code of regulations for food and drugs. Particularly relevant is code describing the standards and regulations on electronic data and electronic signatures.

Page 4: Drug Development

Pre-clinical Data

Under FDA requirements, a sponsor must first submit data showing that the drug is reasonably safe for use in initial, small-scale clinical studies.

Depending on whether the compound has been studied or marketed previously, the sponsor may have several options for fulfilling this requirement: (1) compiling existing nonclinical data from past in-vitro laboratory or animal studies of the compound;

(2) compiling data from previous clinical testing or marketing of the drug in the United States or another country whose population is comparable to the U.S. population; or

(3) undertaking new preclinical studies designed to provide the evidence

Page 5: Drug Development

o Genotoxicity screening is performed, as well as investigations on drug absorption and metabolism, the toxicity of the drug's metabolites, and the speed with which the drug and its metabolites are excreted from the body.

oAt the preclinical stage, the FDA will generally ask, at a minimum, that sponsors:

(1) develop a pharmacological profile of the drug;

(2) determine the acute toxicity of the drug in at least two species of animals, and

(3) conduct short-term toxicity studies ranging from two weeks to three months, depending on the proposed duration of use of the substance in the proposed clinical studies.

Page 6: Drug Development

•Process Development/CMC/API

oUpon nomination of a development candidate, it becomes imperative that the highest-quality compound can be provided for preclinical and clinical development repeatedly and consistently at reasonable cost and in a timely manner.

The initial synthetic route will be revised and optimized to achieve:

oAccessibility of readily available and cost-effective starting material

oMinimization of synthetic and purification steps

oFeasibility of scale-up from microgram to gram, and possibly to kilogram scale.

oReduced cost of goods (COGS)

Page 7: Drug Development

The FDA will require a Chemistry, Manufacturing and Controls (CMC) documentation package for any drug entering clinical trials.

CMC:

oActive Pharmaceutical Ingredients (API)

oDescription and characterization

oManufacturer

oSynthesis/method of manufacture

oProcess controls

oSpecifications (list of tests, methods and acceptance criteria)

oPurity profiles

oContainer/closure system for drug substance (DS) storage

oContainer/closure system for drug product (DP) shelf life

oStability

Page 8: Drug Development

• IND Application

o Investigational new drug (IND) application is the result of a successful preclinical development program. The IND is also the vehicle through which a sponsor advances to the next stage of drug development known as clinical trials (human trials).

o Generally, this includes data and information in three broad areas:

1. Animal Pharmacology and Toxicology Studies

2. Manufacturing Information: This information is assessed to ensure the company can adequately produce and supply consistent batches of the drug.

3. Clinical Protocols and Investigator Information

Page 9: Drug Development

oTypes of INDs:

o"Commercial INDs" are applications that are submitted primarily by companies whose ultimate goal is to obtain marketing approval for a new product.

o“Non-commercial" INDs, which, in fact, account for the vast majority of INDs filed. Submitted by NIH and other sponsors, these INDs include "Investigator INDs," "Emergency Use INDs," and "Treatment INDs."

Page 10: Drug Development

CLINICAL TRIALS

Forty percent of compounds fail in Phase 1, 62 percent of successful Phase 1 compounds fail in phase 2, 40 percent of successful Phase 2 compounds fail in Phase 3, and a surprising 23 percent of successful Phase 3 compounds fail at the registration stage, when the FDA denies approval for a completed New Drug Application (NDA.)

In 1991, the main reason for failure was problems in PK/bioavailability (40 percent) followed by lack of efficacy (30 percent) and toxicology (12 percent). In 2000, the main reason for failure was lack of efficacy (27 percent), followed by commercial and market reasons (21 percent) and toxicology (20 percent).

All current clinical trials registered with the FDA are listed at this website: http://clinicaltrials.gov/.

Page 11: Drug Development

Phase 1 includes the initial introduction of an investigational new drug into humans.

These studies are usually conducted in healthy volunteer subjects.

These studies are designed to determine the metabolic and pharmacologic actions of the drug in humans, the side effects associated with increasing doses, and, if possible, to gain early evidence on efficacy.

During Phase 1, sufficient information about the drug's pharmacokinetics and pharmacological effects should be obtained to permit the design of well-controlled, scientifically valid Phase 2 studies.

Page 12: Drug Development

oPhase 1 studies also evaluate drug metabolism, structure-activity relationships (SAR), and the mechanism of action (MOA) in humans

o. The total number of subjects included in Phase 1 studies varies with the drug, but is generally in the range of 20 to 80.

•In Phase 1 studies, CDER (Center for Drug Evaluation and Research) can impose a clinical hold (i.e., prohibit the study from proceeding or stop a trial that has started) for reasons of safety, or because of a sponsor's failure to accurately disclose the risk of study to investigators.

Page 13: Drug Development

•Phase II

oPhase 2 includes early controlled clinical studies conducted to obtain some preliminary data on the efficacy of the drug for a particular indication (or indications) in patients with the disease. This testing phase also helps determine common short-term side effects and risks associated with the drug.

oDecisive or pivotal trials are usually run as randomized controlled trials (RCT). Randomization introduces a deliberate element of chance into the assignment of treatments to trial patients.

Page 14: Drug Development

oPhase 2a: Pilot trials to evaluate efficacy and safety in selected populations of about 100 to 300 patients who have the condition to be treated, diagnosed, or prevented. They often involve hospitalized patients who can be closely monitored. Objectives may focus on dose-response, type of patient, frequency of dosing, or any of a number of other issues involved in safety and efficacy.

oPhase 2b: Well-controlled trials to evaluate safety and efficacy in patients who have the condition to be treated, diagnosed, or prevented. These trials usually represent the most rigorous demonstration of a medicine's efficacy.

Page 15: Drug Development

•Phase III

oPhase 3 studies are expanded, controlled trials. They are performed after preliminary evidence of effectiveness has been obtained in Phase 2, and are intended to gather the additional information about safety and effectiveness needed to evaluate the overall benefit-risk relationship of the drug.

oPhase 3 trials should provide an adequate basis for extrapolating the results to the general population and conveying that information in the physician labeling. These studies usually include several hundred to several thousand people.

Page 16: Drug Development

At the completion of Phase III an application for licensure is submitted to the FDA, employing either a new drug application (NDA) or a biologics license application (BLA). The FDA will evaluate the applications and decide:

•Whether the drug is safe and effective as indicated, while weighing the risks against the benefits;

•If the drug product is labelled correctly;

•If the drug has been characterized adequately to validate the drug's identity, strength, quality, potency and purity attributes; and

•Whether the drug substance and drug product have been manufactured following current Good Manufacturing Practices (cGMPs.)

Page 17: Drug Development

In both Phase 2 and 3, the Center for Drug Evaluation and Research (CDER), a branch of the FDA, can impose a clinical hold if a study is unsafe or if the protocol design is deficient in meeting its stated objectives.

FDA approval/disapproval decisions are based on the results of pivotal studies. To be considered pivotal, a study must meet at least these 4 criteria:

oBe controlled using placebo or a standard therapy.

oHave a double-blinded design when such a design is practical and ethical.

oBe randomized.

oBe of adequate size.

Page 18: Drug Development

NDA

The components of any NDA are, in part, a function of the nature of the subject drug and the information available to the applicant at the time of submission. As outlined in Form FDA-356h (Application to Market a New Drug for Human Use Or As An Antibiotic Drug For Human Use) NDAs can consist of as many as 15 different sections:

oIndex;

oSummary;

oChemistry, Manufacturing, and Control (CMC);

oSamples, Methods Validation Package, and Labeling;

oNonclinical Pharmacology and Toxicology;

oHuman Pharmacokinetics and Bioavailability;

Page 19: Drug Development

oMicrobiology (for anti-microbial drugs only);

oClinical Data;

oSafety Update Report (typically submitted 120 days after the NDA's submission);

oStatistical;

oCase Report Tabulations;

oCase Report Forms;

oPatent Information;

oPatent Certification; and

oOther Information.

oReviewers attempt to confirm and validate the sponsor's conclusion that a drug is safe and effective for its proposed use. The review is likely to involve a reanalysis or an extension of the analyses presented in the NDA

Page 20: Drug Development

oWhen the technical reviews are complete, each reviewer develops a written evaluation of the NDA that presents their conclusions and their recommendations on the application.

oThe division director or office director then evaluates the reviews and recommendations and decides the action that the division will take on the application.

oThe result is an action letter that provides an approval, approvable, or non-approvable decision and a justification for that recommendation.

Page 21: Drug Development

•Phase IV

Phase 4 trials are done after a drug has received a market approval. These trials are monitoring drugs that are available for doctors to prescribe, rather than experimental drugs that are still being developed. Pharmaceutical companies run Phase 4 trials to find out:

oMore about safety and side effects of the drug.

oWhat the long-term risks and benefits are?

oHow well the drug works when it is used more widely than in clinical trials?

Page 22: Drug Development
Page 23: Drug Development

Moving from Research into Development

Market potential and health benefits.

•In practice, the best policy is to have no more drugs in development than are sustainable, with preference for resources given to those that have the best chance of being first in class and/or first to market. Factors of most importance include:

1.Identifying drug candidates with poor physicochemical properties, such as poor aqueous solubility and/or poor dissolution characteristics.

2.Identifying poorly adsorbed or metabolized compounds with suboptimal pharmokinetics and/or toxicological properties.

Page 24: Drug Development

Hydrophobic drugs are examples of molecules that exhibit several of these problems. Physicochemical parameters such as solubility, lipophilicity and stability should be evaluated in any early development program, providing drug molecules with the most preferred physicochemical properties for formulation and dosage form development.

With respect to issues related to toxicological problems, gene transcriptional profiling has the potential to predict drug candidate toxicity and with the development of transgenic and knockout animal models, preclinical toxicogenomic studies are greatly aided.

Page 25: Drug Development

There are alternative strategies that seek shortcuts in the regulatory pathways, such as re-profiling marketed drugs for alternative indications. With this approach, costs are lowered and risks minimized as often many of the more costly and time-consuming regulatory requirements have already been identified and completed. Several famous drugs were found to have their greatest therapeutic potential in other unanticipated indications in this way. Examples include Viagra (erectile dysfunction), Evista (osteoporosis) and Hytrin (benign prostatic hyperplasia). Furthermore, subsequent to approval, nearly 90% of marketed drugs find important new clinical indications.

Page 26: Drug Development
Page 27: Drug Development
Page 28: Drug Development
Page 29: Drug Development
Page 30: Drug Development
Page 31: Drug Development
Page 32: Drug Development
Page 33: Drug Development

Recommended