1_ajpr_1_1_2011

Upload: vundru-vikash

Post on 05-Apr-2018

215 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/2/2019 1_AJPR_1_1_2011

    1/6

    Asian J. Pharm. Res. 2011; Vol. 1: Issue 1, Pg 01-06 [AJPRes.]

    1

    ISSN- 22315683 (Print) www.asianpharmaonline.orgISSN- 22315691 (Online) 0974-3618

    REVIEW ARTICLE

    Regulatory Requirements for Drug Development and Approval in United

    States: A Review

    Swati Rawat1* and Akhilesh Gupta

    2

    1Shri Bhagwan College of Pharmacy Aurangabad (M.S.)

    2Kunwar Haribansh Singh College of Pharmacy Jaunpur (U.P.)

    *Corresponding Author E-mail: [email protected]

    ABSTRACT:In this paper a brief history and an overview of the regulatory process for drug approval in the United States through

    illustrations of Investigational New Drug (INDs) Applications and New Drug Applications (NDAs), abbreviated newdrug applications (ANDAs) and supplemental new drug applications (SNDAs) are provided. For INDs, the regulatory

    requirements for a well-designed protocol, the role and responsibility of institutional review boards, and the

    applicability of treatment INDs are discussed. For NDAs, issues regarding the application of expanded access, the

    submission of abbreviated NDAs for a generic drug, the submission of supplemental NDAs for labeling changes, and

    the role and responsibility of advisory committees are addressed. Along with this a brief description of review steps

    taken by FDA is provided.

    KEYWORDS: Investigational New Drug Application; New Drug Application; International Conference onHarmonization.

    INTRODUCTION:HISTORY:

    The research, development, and approval of a drug productis a continuous but lengthy process involving drug

    discovery, laboratory development, animal studies, clinical

    trials, and regulatory registration. This lengthy process is

    necessary to assure the effectiveness and safety of the drug

    product. In the United States, however, no regulations were

    put forth until the Pure Food and Drug Act was passed by

    Congress in 1906. The purpose of this act is to prevent

    misbranding and adulteration of food and drugs yet it does

    not give the government any authority to inspect food and

    drugs. The act was amended in 1912 (the Sherley

    Amendment) to prohibit labeling medicines with false and

    fraudulent claims. In 1931, the United States Food and

    Drug Administration (FDA) was formed. The provisions of

    the FDA are intended to ensure that:1. Food is safe and wholesome,

    2. Drugs, biological products, and medical devices are safe

    and effective,

    3. Cosmetics are unadulterated,

    4. The use of radiological products does not result in

    unnecessary exposure to radiation, and

    5. All of these products are honestly and informatively

    labeled (1).

    Received on 21.02.2011 Accepted on 15.03.2011

    Asian Pharma Press All Right ReservedAsian J. Pharm. Res. 1(1): Jan.-Mar. 2011; Page 01-06

    Late 1930s, when the Elixir Sulfanilamide disaster

    occurred. The disaster was a safety concern of a liquid

    formulation of a sulfa drug which caused more than 100

    deaths. This drug had never been tested in humans before it

    was marketed. This safety concern led to the passage of the

    Federal Food, Drug and Cosmetic Act (FDandC Act) in

    1938. The FDandC Act extended its coverage to cosmetics

    and therapeutic devices. More importantly, the FDandC Act

    requires pharmaceutical companies to submit full reports of

    investigations regarding the safety of new drugs. In 1962,

    the significant Kefauver-Harris Drug Amendments to the

    FDandC Act were passed which not only strengthened the

    safety requirements for new drugs but also established an

    efficacy requirement for new drugs for the first time. In

    1984, Congress passed the Price competition and Patent

    Term Restoration Act to provide for increased patent

    protection to compensate for patent life lost during the

    approval process. Based on this act, the FDA was

    authorized to approve generic drugs through the evaluation

    of bioequivalency on healthy male subjects. In addition, the

    FDA also has the authority to designate prescription drugs

    or over-the-counter (OTC) drugs1.

    REGULATORY PROCESS:

    For approval of drug products, each country and/or region

    such as the European Community (EC), Japan, and the

    United States has similar but slightly different regulatory

    processes and requirements for the conduct of clinical trials

  • 8/2/2019 1_AJPR_1_1_2011

    2/6

    Asian J. Pharm. Res. 2011; Vol. 1: Issue 1, Pg 01-06 [AJPRes.]

    2

    and the submission, review, and approval of clinical results.

    This section, for illustration purpose, will focus on the

    regulatory process and requirements adopted in the United

    States.

    For evaluation and approval of drugs, sponsors are requiredto submit to the FDA substantial evidence of effectiveness

    and safety accumulated from adequate and well controlled

    clinical trials. The current regulations for conducting

    clinical trials and the submission, review, and approval of

    clinical results for pharmaceutical compounds in the United

    States can be found in the Code of Federal Regulations

    (CFR) (see, eg, 21 CFR Parts 50, 56, 312, and 314).

    The FDA has jurisdiction over administration of regulation

    and approval of drug products. These regulations cover

    Investigational New Drug Applications, and New Drug

    Applications for new drugs, orphan drugs, and over-the-

    counter human drugs, Abbreviated New Drug Applications

    for generic drugs, Establishment License Applications orProduct License Applications for biological products, and

    Investigational Device Exemptions, and Premarket

    Approval of Medical Devices for medical devices.

    A treatment consisting of a combination of drugs, biological

    products, and/or medical devices is usually referred to as

    combined therapy. If a treatment consists of a combination

    of drugs, biologics, and/or devices, such as a drug with a

    device, a biologic with a device, a drug with a biologic, or a

    drug with a biologic in conjunction with a device, then it is

    defined as a combined product. For a combined product

    consisting of different pharmaceutical entities, FDA

    requires that each of the entities be reviewed separately by

    appropriate centers at the FDA. In order to avoid confusionof jurisdiction over a combination product and to improve

    the efficiency of the approval process, the principle of

    primary mode of action of a combination product was

    established in the Safe Medical Devices Act (SMDA) in

    1990 (21 U.S.C. 353). In 1992, based on this principle,

    three intercenter agreements were signed between the

    Center for Drug Evaluation and Research (CDER) and

    Center for Biologics Evaluation and Research (CBER),

    between CDER and Center for Devices and Radiological

    Health (CDRH) and between CBER and CDRH to establish

    the ground rules for assignment of a combined product and

    intercenter consultation (2).

    As described earlier, different regulations exist for differentproducts. The spirit and principles for the conduct,

    submission, review, and approval of clinical trials, however,

    are the same. Therefore, for the purpose of illustration, a

    detailed discussion will be provided only on INDs and

    NDAs for drug products2.

    INVESTIGATIONAL NEW DRUG APPLICATION

    (IND):

    The FDA first enters the picture when a drug sponsor

    submits an IND to the agency. Sponsors--companies,

    research institutions, and other organizations that take

    responsibility for marketing a drug--must show the FDA

    results of pre-clinical testing they've done in laboratory

    animals and what they propose to do for human testing. At

    this stage, the FDA decides whether it is reasonably safe to

    move forward with testing the drug on humans.

    Before a drug can be studied in humans, its sponsor must

    submit an IND to the FDA. Unless otherwise notified, the

    sponsor may begin to investigate the drug 30 days after the

    FDA has received the application. The IND requirements

    extend throughout the period during which a drug is under

    study. As mentioned in Section 312.1 and 312.3 of 21 CFR,

    an IND is synonymous with the Notice of Claimed

    Investigational Exemption for a New Drug. Therefore, an

    IND is, legally speaking, an exemption to the law that

    prevents the shipment of a new drug for interstate

    commerce. Consequently, the drug companies which file an

    IND have the flexibility of conducting clinical

    investigations of products across the United States.

    Kessler indicated that there are two types of INDs:

    commercial and noncommercial (3). A commercial IND

    permits the sponsor to gather the data on clinical safety and

    effectiveness that are needed for an NDA. If the drug is

    approved by the FDA, the sponsor is allowed to market the

    drug for specific uses. On the other hand, a noncommercial

    IND allows the sponsor to use the drug in research or early

    clinical investigation to obtain advanced scientific

    knowledge of the drug. Note that the FDA itself does not

    investigate new drugs or conduct clinical trials.

    Pharmaceutical manufacturers, physicians, and other

    research organizations such as the National Institutes of

    Health (NIH) may sponsor INDs. If a commercial IND

    proves successful, the sponsor ordinarily submits an NDA.During this period, the sponsor and the FDA usually

    negotiate over the adequacy of the clinical data and the

    wording proposed for the label accompanying the drug

    which sets out description, clinical pharmacology,

    indications and usage, contraindications, warnings,

    precautions, adverse reactions, and dosage and

    administration.

    By the time an IND is filed, the sponsor should have

    enough information about chemistry, manufacturing, and

    controls (CMC) of the drug substance and drug product to

    assure the identity, strength, quality, and purity of the

    investigational drug covered by the IND. In addition, the

    sponsor should provide adequate information aboutpharmacological studies for absorption, distribution,

    metabolism, and excretion (ADME) and acute, subacute,

    and chronic toxicological studies and reproductive tests in

    various animal species to support that the investigational

    drug is reasonably safe to be evaluated in clinical trials of

    various durations in humans.

    The central focus of the initial IND submission should be

    on the general investigational plan and protocols for

    specific human studies. Therefore, a copy of protocol(s)

    which include study objectives, investigators, criteria for

  • 8/2/2019 1_AJPR_1_1_2011

    3/6

    Asian J. Pharm. Res. 2011; Vol. 1: Issue 1, Pg 01-06 [AJPRes.]

    3

    inclusion and exclusion, study design, dosing schedule,

    endpoint measurements, and clinical procedure should be

    submitted along with the investigational plan and other

    information such as chemistry, manufacturing, and controls,

    pharmacology and toxicology, previous human experiences

    with the investigational drug, and any additional andrelevant information related to the investigational drug.

    Note that the FDA requires that all sponsors submit an

    original and two copies of all submissions to the IND file,

    including the original submission and all amendments and

    reports3.

    TREATMENT INDs:

    During the clinical investigation of the drug under an IND,

    it may be necessary and ethical to make the drug available

    to those patients who are not enrolled in clinical trials.

    Since 1987, the FDA permits an investigational drug to be

    used for treatment under a treatment protocol or treatment

    IND if the drug is intended to treat a serious or immediately

    life-threatening disease, especially when there is nocomparable or satisfactory alternative drug or other therapy

    available to treat that stage of the disease in the intended

    patient population. The FDA, however, may deny a request

    for treatment use of an investigational drug under a

    treatment protocol or treatment IND if the sponsor fails to

    show that the drug may be effective for its intended use in

    its intended patient population or the drug may expose the

    patients to an unreasonable and significant additional risk of

    illness or injury.

    At any time a sponsor may withdraw an effective IND

    without prejudice. If an IND is withdrawn, however, FDA

    shall be notified and all clinical investigations conducted

    under the IND shall be stopped. If an IND is withdrawnbecause of a safety reason, the sponsor shall promptly

    inform the FDA, all investigators, and all reviewing IRBs

    with the reasons for the withdrawal.

    If there are any deficiencies in an IND or in the conduct of

    an investigation under an IND, the FDA may terminate the

    IND. If an IND is terminated, the sponsor shall end all

    clinical investigations conducted under the IND and recall

    or dispose of all unused supplies of the drug. Some

    examples of deficiencies in an IND are discussed under 21

    CFR 312.44. For example, the FDA may propose

    terminating the IND if it finds that human subjects would

    be exposed to an unreasonable and significant risk of illness

    or injury. In such a case, the FDA will notify the sponsor inwriting and invite correction or explanation within a period

    of 30 days. A terminated IND is subject to reinstatement

    based on additional submissions that eliminate such risk. In

    this case, a regulatory hearing on the question of whether

    the IND should be reinstated will be held.

    COMMUNICATION WITH THE FDA:

    FDA encourages open communication regarding any

    scientific or medical question that may arise during the

    clinical investigation. Basically, it is suggested that such

    communication be arranged at the end of the Phase II study

    and prior to a marketing application. The purpose of an end-

    of-Phase II meeting is to review the safety of a drug

    proceeding to Phase III. This meeting is helpful not only

    because it evaluates the Phase III plan and protocols but it

    also identifies any additional information necessary to

    support a marketing application for the uses underinvestigation. Note that a similar meeting may be held at the

    end of Phase I to review results of tolerance/safety studies

    and to review the adequacy of the remaining development

    program. At the end of Phase I a meeting would be

    requested by a sponsor when a drug or biologic product is

    being developed for a life-threatening disease and the

    sponsor wishes to file under the expedited registration

    regulations. The purpose of pre-NDA meetings is not only

    to uncover any major unresolved problems but also to

    identify those studies that are needed for establishment of

    drug effectiveness. In addition, the communication enables

    the sponsor to acquaint FDA reviewers with the general

    information to be submitted in the marketing application.

    More importantly, the communication provides theopportunity to discuss:

    1. Appropriate methods for statistical analysis of the data,and

    2. The best approach to the presentation and formatting of

    the data4.

    CLINICAL TRIALS:Drug studies in humans can begin only after an IND is

    reviewed by the FDA and a local institutional review board

    (IRB). The board is a panel of scientists and non-scientists

    in hospitals and research institutions that oversees clinical

    research.

    IRBs approve the clinical trial protocols, which describe thetype of people who may participate in the clinical trial, the

    schedule of tests and procedures, the medications and

    dosages to be studied, the length of the study, the study's

    objectives, and other details. IRBs make sure the study is

    acceptable, that participants have given consent and are

    fully informed of their risks, and that researchers take

    appropriate steps to protect patients from harm.

    Phase 1 studies are usually conducted in healthy

    volunteers. The goal here is to determine what the drug's

    most frequent side effects are and, often, how the drug is

    metabolized and excreted. The number of subjects typically

    ranges from 20 to 80.

    Phase 2 studies begin if Phase 1 studies don't reveal

    unacceptable toxicity. While the emphasis in Phase 1 is on

    safety, the emphasis in Phase 2 is on effectiveness. This

    phase aims to obtain preliminary data on whether the drug

    works in people who have a certain disease or condition.

    For controlled trials, patients receiving the drug are

    compared with similar patients receiving a different

    treatment--usually a placebo or a different drug. Safety

    continues to be evaluated, and short-term side effects are

    studied. Typically, the number of subjects in Phase 2

    studies ranges from a few dozen to about 300.

  • 8/2/2019 1_AJPR_1_1_2011

    4/6

    Asian J. Pharm. Res. 2011; Vol. 1: Issue 1, Pg 01-06 [AJPRes.]

    4

    Phase 3 studies begin if evidence of effectiveness is shown

    in Phase 2. These studies gather more information about

    safety and effectiveness, studying different populations and

    different dosages and using the drug in combination with

    other drugs. The number of subjects usually ranges from

    several hundred to about 3,000 people.

    Phase 4 studies occur after a drug is approved. They may

    explore such areas as new uses or new populations, long-

    term effects, and how participants respond to different

    dosages.

    NEW DRUG APPLICATION (NDA):This is the formal step a drug sponsor takes to ask that the

    FDA consider approving a new drug for marketing in the

    United States. An NDA includes all animal and human data

    and analyses of the data, as well as information about how

    the drug behaves in the body and how it is manufactured.

    When an NDA comes in, the FDA has 60 days to decidewhether to file it so that it can be reviewed. The FDA can

    refuse to file an application that is incomplete. For example,

    some required studies may be missing. In accordance with

    the Prescription Drug User Fee Act (PDUFA), the FDA's

    Center for Drug Evaluation and Research (CDER) expects

    to review and act on at least 90 percent of NDAs for

    standard drugs no later than 10 months after the

    applications were received. The review goal is six months

    for priority drugs. The Tufts Center for the Study of Drug

    Development in Boston estimates that about 1 in 5 drugs

    that enter clinical testing ultimately are approved by the

    FDA.

    How often the FDA meets with a drug sponsor varies, butthe two most common meeting points are at the end of

    Phase 2 clinical trials and pre-NDA--right before a new

    drug application is submitted.

    At the end of Phase 2, the FDA and sponsors try to come to

    an agreement on how the large-scale studies in Phase 3

    should be done. The pre-NDA meeting is for discussing

    what the FDA expects to see in the application.

    There is also continuous interaction throughout the review

    process5.

    REVIEWING APPLICATIONS:

    Though FDA reviewers are involved with a drug's

    development throughout the IND stage, the official review

    time is the length of time it takes to review a new drugapplication and issue an action letter, an official statement

    informing a drug sponsor of the agency's decision.

    Once a new drug application is filed, an FDA review team--

    medical doctors, chemists, statisticians, microbiologists,

    pharmacologists and other experts--evaluates whether the

    studies the sponsor submitted show that the drug is safe and

    effective for its proposed use. No drug is absolutely safe; all

    drugs have side effects. "Safe" in this sense means that the

    benefits of the drug appear to outweigh the risks.

    The review team analyzes study results and looks for

    possible problems with the application, such as weaknesses

    of the study design or analyses. Reviewers determine

    whether they agree with the sponsor's results and

    conclusions, or whether they need any additional

    information to make a decision.Each reviewer prepares a written evaluation containing

    conclusions and recommendations about the application.

    These evaluations are then considered by team leaders,

    division directors, and office directors, depending on the

    type of application.

    Reviewers receive training that fosters consistency in drug

    reviews, and good review practices remain a high priority

    for the agency. For example, CDER recently held a two-day

    retreat in which clinical reviewers discussed review

    priorities, including improved communication between drug

    review divisions in CDER regarding drugs being reviewed

    for more than one indication.

    Sometimes the FDA calls on advisory committees made upof outside experts who help the agency decide on drug

    applications. Whether an advisory committee is needed

    depends on many things.

    ACCELERATED APPROVAL:

    Traditional approval requires that clinical benefit be shown

    before approval can be granted. Accelerated approval is

    given to some new drugs for serious and life-threatening

    illnesses that lack satisfactory treatments. This allows an

    NDA to be approved before measures of effectiveness that

    would usually be required for approval are available.

    Instead, less traditional measures called "surrogate

    endpoints" are used to evaluate effectiveness. These arelaboratory findings or signs that may not be a direct

    measurement of how a patient feels, functions, or survives,

    but are considered likely to predict benefit. For example, a

    surrogate endpoint could be the lowering of HIV blood

    levels for short periods of time with anti-retroviral drugs.

    Most drugs to treat HIV have been approved under

    accelerated approval provisions, with the company required

    to continue its studies after the drug is on the market to

    confirm that its effects on virus levels are maintained and

    that it ultimately benefits the patient. Under accelerated

    approval rules, if studies don't confirm the initial results, the

    FDA can withdraw the approval.

    Because premarket review can't catch all potential problems

    with a drug, the FDA continues to track approved drugs for

    adverse events through a postmarketing surveillance

    program6.

    "APPROVABLE" OR "NOT APPROVABLE"

    If the FDA decides that the benefits of a drug outweigh the

    risks, the drug will receive approval and can be marketed in

    the United States. But if there are problems with an NDA,

    the FDA may decide that a drug is "approvable" or "not

    approvable."

  • 8/2/2019 1_AJPR_1_1_2011

    5/6

    Asian J. Pharm. Res. 2011; Vol. 1: Issue 1, Pg 01-06 [AJPRes.]

    5

    A designation of approvable means that the drug can

    probably be approved, provided that some issues are

    resolved first. This might involve the sponsor and the FDA

    coming to a final agreement on what should go on the

    drug's label, for example. It could also involve more

    difficult issues, such as the adequacy of information on howpeople respond to various dosages of the drug.

    A designation of "not approvable" describes deficiencies

    significant enough that it is not clear that approval can be

    obtained in the future, at least not without substantial

    additional data.

    Common problems include unexpected safety issues that

    crop up or failure to demonstrate a drug's effectiveness. A

    sponsor may need to conduct additional studies--perhaps

    studies of more people, different types of people, or for a

    longer period of time.

    Manufacturing issues are also among the reasons that

    approval may be delayed or denied. Drugs must bemanufactured in accordance with standards called good

    manufacturing practices, and the FDA inspects

    manufacturing facilities before a drug can be approved. If a

    facility isn't ready for inspection, approval can be delayed.

    Any manufacturing deficiencies found would need to be

    corrected before approval.

    The FDA outlines the justification for its decision in an

    action letter to the drug sponsor. When the action is either

    approvable or not approvable, CDER gives the sponsor a

    chance to meet with agency officials to discuss the

    deficiencies. At that point, the sponsor can choose to ask for

    a hearing or correct any deficiencies and submit new

    information.

    ABBREVIATED NEW DRUG APPLICATIONS:

    An abbreviated NDA (ANDA) is usually reserved for drug

    products (eg, generics) which duplicate products previously

    approved under a full NDA. For an ANDA, reports of

    nonclinical laboratory studies and clinical investigations

    except for those pertaining to in vivo bioavailability of the

    drug product are not required. The information may be

    omitted when the FDA has determined that the information

    already available is adequate to establish that a particular

    dosage form of a drug meets the statutory standards for

    safety and effectiveness. The duplicate products are usually

    referred to as products with the same active ingredient(s),

    route of administration, dosage form, strength, or conditionof use which may be made by different manufacturers.

    As mentioned earlier, under the Drug Price Competition

    and Patent Term Restoration Act passed in 1984, the FDA

    may approve generic drug products if the generic drug

    companies can provide evidence that the rates and extents

    of absorption of their drug products do not show significant

    differences from those of the innovator drug products when

    administered at the same molar dose of the therapeutic

    moiety under similar experimental conditions (21 CFR

    320). The Drug Price Competition and Patent Term

    Restoration Act states that FDA has authority for all generic

    drug approvals through an ANDA submission for

    bioequivalence review. An ANDA submission should

    include product information, pharmacokinetic data and

    analysis, statistical analysis, analytical methodology and

    validation, and clinical data.To ensure the validity of bioequivalence assessment, the

    Division of Bioequivalence's Office of Generic Drugs of

    CDER issued a "Guidance on Statistical Procedures for

    Bioequivalence Studies Using a Standard TwoTreatment

    Crossover Design" in July 1992. The guidance sets forth

    regulations for valid statistical analysis for bioequivalence

    assessment. In addition, any relevant clinical findings,

    adverse reactions, and deviations from the protocol also

    need to be included in the ANDA submission. Recently, to

    assure drug interchangeability, a draft guidance on "In Vivo

    Bioequivalence Studies Based on Population and Individual

    Bioequivalence Approaches" was developed by the FDA

    and is currently being circulated for public comments. The

    draft guidance is intended to replace the 1992 guidance forassessment of bioequivalence for regulatory approval of

    new dosage forms of an innovator drug and its generic

    copies after it is finalized7.

    SUPPLEMENTAL NEW DRUG APPLICATIONS:

    A supplemental NDA (SNDA) is referred to as

    documentation submitted to the FDA on a drug substance or

    product which is already the subject of an approved NDA.

    Supplements may be submitted for a variety of reasons such

    as labeling changes, a new or expanded clinical indication,

    or a new dosage form. For example, for labeling changes,

    the sponsor may want to add a new specification or test

    method or changes in the methods, facility, or controls to

    provide increased assurance that the drug will have thecharacteristics of identity, strength, quality, and purity that

    it purports to possess. For drug substance and/or drug

    product, the sponsor may want to relax the limits for a

    specification, establish a new regulatory analytical method,

    or delete a specification or regulatory analytical method. In

    addition, the sponsor may want to extend the expiration

    date of the drug product based on data obtained under a new

    or revised stability testing protocol that has not been

    approved in the application or to establish a new procedure

    for reprocessing a batch of the drug product that fails to

    meet specification. It should be noted, however, that in an

    SNDA, the sponsor is required to fully describe the change

    in each condition established in an approved application

    beyond the variation already provided for in theapplication

    8.

    ADVISORY COMMITTEES:

    The FDA has established advisory committees which

    consist of clinical, pharmacological, and statistical experts

    and one consumer advocate (not employed by the FDA) in

    designated drug classes and subspecialities. The

    responsibilities of the committees are to review data

    presented in NDAs and to advise the FDA as to whether or

    not substantial evidence of safety and effectiveness exists

    based on adequate and well controlled clinical studies. In

  • 8/2/2019 1_AJPR_1_1_2011

    6/6

    Asian J. Pharm. Res. 2011; Vol. 1: Issue 1, Pg 01-06 [AJPRes.]

    6

    addition, the committee may also be asked at times to

    review certain INDs, protocols, or important issues relating

    to marketed drugs and biologics. The advisory committees

    not only supplement the FDA's expertise but also allow an

    independent peer review during the regulatory process9.

    Note that the FDA usually prepares a set of questions forthe advisory committee to address at the meeting. For

    example, the following is a list of some typical questions:

    1. Are there two or more adequate and wellcontrolled trials?

    2. Have the patient populations been well enough

    characterized?

    3. Has the dose-response relationship been sufficiently

    characterized? and

    4. Do you recommend the use of the drug for the indication

    sought by the sponsor for the intended patient population?

    DRUG REVIEW STEPS:

    The review steps taken by FDA are as described in brief10

    :

    1 Pre-clinical (animal) testing.2. An investigational new drug application (IND) outlineswhat the sponsor of a new drug proposes for human testing

    in clinical trials.

    3. Phase 1 studies (typically involve 20 to 80 people).4. Phase 2 studies (typically involve a few dozen to about300 people).

    5. Phase 3 studies (typically involve several hundred toabout 3,000 people).

    6. The pre-NDA period, just before a new drugapplication (NDA) is submitted.

    A common time for the FDA and drug sponsors to meet.

    7. Submission of a new drug application is the formalstep asking the FDA to consider a drug for marketing

    approval.

    8. After an NDA is received, the FDA has 60 days todecide whether to file it so it can be reviewed.

    9. If the FDA files the NDA, an FDA review team isassigned to evaluate the sponsor's research on the drug's

    safety and effectiveness.

    10. The FDA reviews information that goes on a drug'sprofessional labeling, guidance on how to use the drug.

    11. The FDA inspects the facilities where the drug will bemanufactured as part of the approval process.

    12. FDA reviewers will approve the drug or find it either"approvable" or "not approvable."

    CONCLUSION:This paper provides a brief history and an overview of the

    regulatory approval process in drug development currently

    adopted in the United States. For drug sponsors and others

    who want a basic understanding of the drug development

    process, the FDA has published a series of articles in a

    special report called From Test Tube to Patient: Improving

    Health Through Human Drugs" These articles provide

    background information on a broad range of topics from

    laboratory and animal studies, to reporting unsafe medical

    products already on the market. Another resource for drug

    development information is an interactive chart which

    graphically displays the process with an emphasis on

    preclinical (animal) research and clinical (human) studies or

    trials conducted by the drug's sponsor. It should be noted

    that most regulatory authorities in different countries have

    similar but slightly different requirements for approval of

    drug products. In practice, similar studies may be

    repeatedly conducted in order to fulfill regulatory

    requirements in different marketplaces. Therefore, there is astrong desire to establish international standards for

    regulatory requirements so that the information generated

    from a specific region will be mutually accepted by other

    regions. The necessity to standardize regulatory

    requirements has been recognized by both regulatory

    authories and the pharmaceutical industry. As a result, the

    International Conference on Harmonization (ICH) which

    consists of the European Community, the United States, and

    Japan was formed to evaluate and develop technical

    requirements for the registration of pharmaceuticals for

    human use. A number of guidance and draft guidelines for

    good pharmaceutical practices have been developed to

    assist pharmaceutical companies in drug research and

    development.

    REFERENCES:1. Fairweather WR. Statisticians, the FDA and a time of transition.

    Presented at Pharmaceutical Manufacturers Association

    Education and Research Institute Training Course in Non-

    Clinical Statistics, Georgetown University Conference Center,February 6-8, 1994, Washington, DC.

    2. Margolies ME Regulations of combination products. App ClinTrials. 1994; 3:50-65.

    3. Kessler DA. The regulation of investigational drugs. New Eng JMed 1989; 320:281-288.

    4. Petricciani JC. An overview of FDA, IRBs and regulations. IRB.1981; 3:1.

    5. Barry MJ, Fowler Jr. FJ, O'Leary MP, Bruskewitz RC, HoltgreweHL, Mebust WK, Cockett AT. The American Urological

    Association Symptom Index for Benign Prostatic Hyperplasia. JUrol. 1992; 148: 1549-1557.

    6. Chow SC, Liu JP. Design and Analysis of Clinical Trials. NewYork, New York: John Wiley and Sons; 1998.

    7. Kessler DA, Feiden KL. Faster evaluation of vital drugs.Scientific Am. 1995; 272:48-54.

    8. Tessman DK, Gipson B, Levins M. Cooperative fasttrackdevelopment: the fludara story. App Clin Trials. 1994; 3:55-62.

    9. Food and drug administration publications; Publication No.(FDA) 02-3242.

    10. http://www.fda.gov, accessed on 21/12/2007.