Food and Drug Administration (FDA) Disclosure:

The statements in this forum have not been evaluated by the Food and Drug Administration and are generated by non-professional writers. Any products described are not intended to diagnose, treat, cure, or prevent any disease.

Website Disclosure:

This forum contains general information about diet, health and nutrition. The information is not advice and is not a substitute for advice from a healthcare professional.

FDA Testimony: Potential Merits of Cannabinoids for Medical Uses April 1, 2004

Discussion in 'Medical Marijuana Usage and Applications' started by jainaG, Sep 28, 2015.

  1. http://www.fda.gov/newsevents/testimony/ucm114741.htm


    Statement of:
    Robert J. Meyer, M.D.

    Director

    Office of Drug Evaluation II

    Center for Drug Evaluation and Research

    Food and Drug Administration

    U.S. Department of Health and Human Services before the Subcommittee on Criminal Justice, Drug Policy, and Human Resources
    House Committee on Government Reform
    http://www.fda.gov/newsevents/testimony/ucm114741....


    April 1, 2004

    INTRODUCTION

    Good afternoon, Mr. Chairman and Members of the Subcommittee. I am
    Dr. Robert Meyer, Director of the Office of Drug Evaluation II at the
    Food and Drug Administration's (FDA or the Agency), Center for Drug
    Evaluation and Research (CDER). I am pleased to be here today with my
    colleague, Dr. Nora Volkow, Director of the National Institute on Drug
    Abuse (NIDA). FDA appreciates the opportunity to discuss the need for a
    science-based approach to evaluating the merits of marijuana for
    medicinal purposes.


    In my testimony today, I will first describe the FDA drug approval
    process. Second, I will clarify FDA's role in facilitating the objective
    evaluation of the potential merits of cannabinoids for medical uses as
    well as FDA's role with respect to enforcement efforts relating to
    Schedule I Controlled Substances such as marijuana.


    FDA APPROVAL PROCESS


    FDA's primary mission for over 90 years has been to promote and
    protect the public health, under the authority of the Federal Food,
    Drug, and Cosmetic (FD&C) Act and the Public Health Service Act.
    These statutes were enacted and amended, in part, in response to public
    health tragedies resulting from the sale to, and use by, an unsuspecting
    public of unsafe and ineffective products sold as medicines and medical
    devices. The FD&C Act requires that new drugs be shown to be safe
    and effective before being marketed in this country.


    The single most important public health provision in these statutes
    is the requirement that a person wishing to sell to the public a product
    to prevent, cure or mitigate illness or injury must first prove that
    such product is safe, and actually does what the vendor claims it does.
    This statutory provision affords patients the most effective protection
    against untested and unproven products.


    A new drug or biologic (referred to in this statement as a drug) may
    not be distributed in interstate commerce (except for clinical studies
    under an investigational new drug application) until a sponsor, usually
    the drug manufacturer, has submitted and FDA has approved a new drug
    application (NDA) or a biologics license application (BLA) for the
    product. For approval, an NDA or BLA must contain sufficient scientific
    evidence demonstrating the safety and effectiveness of the drug for its
    intended uses.


    The evidence of safety and effectiveness usually is obtained through
    controlled clinical trials. The disciplined, systematic, scientific
    conduct of such trials is the most effective and certain means of
    obtaining the data that document safety and efficacy of a drug and how
    to use the new product so that it will have the most beneficial effect.


    INVESTIGATIONAL NEW DRUG APPLICATION PROCESS

    The first step a sponsor usually must take to obtain approval for a new
    drug is to test the drug in animals for toxicity. The sponsor then takes
    that animal testing data, along with additional information about the
    drug's composition and manufacturing, and develops a plan for testing
    the drug in humans. The sponsor submits these data, along with proposed
    studies, the qualifications of the investigators who will conduct the
    clinical studies, and assurances of informed consent and protection of
    the rights and safety of the human subjects, to FDA in the form of an
    investigational new drug application (IND).


    FDA reviews the IND for assurance that the proposed studies,
    generally referred to as clinical trials, do not place human subjects at
    unreasonable risk of harm. FDA also verifies that there are adequate
    assurances of informed consent and human subject protection. At that
    point the first of three phases of study in humans can begin. Phase I
    studies primarily focus on the safety of the drug in humans. Phase I
    studies carefully assess how to safely administer and dose the drug with
    an emphasis on evaluation of the toxic manifestations of the therapy,
    how the body distributes and degrades the drug, and how side effects
    relate to dose. Phase I studies typically include fewer than 100 healthy
    volunteers or subjects.


    Phase II studies are clinical studies to explore the effectiveness of
    the drug for a particular indication over a range of doses and to
    determine common short-term side effects. Phase II studies typically
    involve a few hundred subjects. Once Phase II studies are successfully
    completed, the drug's sponsor has learned much about the drug's
    appropriate dosing and its apparent safety and effectiveness. The next
    step is to conduct Phase III studies involving up to several thousand
    subjects. These studies establish efficacy for a particular indication,
    examine additional uses, may provide further safety data including
    long-term experience, and consider additional population subsets, dose
    response, etc. FDA strongly encourages sponsors to work closely with the
    Agency in planning definitive Phase III clinical trials to help assure
    that the trials are designed to have the greatest likelihood of
    producing results sufficient to provide adequate data and permit the
    Agency to make appropriate decisions about the safety and efficacy of
    the product.


    Once Phase III trials are completed, the sponsor submits the results
    of all the relevant testing to FDA in the form of an NDA. FDA's medical
    officers, chemists, statisticians, and pharmacologists review the
    application to determine if the sponsor's data in fact show that the
    drug is both safe and effective. The drug's manufacturing process is
    evaluated to confirm that the product can be produced consistently with
    high quality. It is common to allow subjects in Phase II and III studies
    to continue on a therapy if it seems to be providing benefit. This
    practice provides long-term safety information at an early stage in this
    process. At present, there are literally thousands of clinical trials
    ongoing, involving hundreds of thousands of subjects. There are over
    15,000 active INDs for drugs, therapeutic biologics, and biologics filed
    with the Agency.


    Results of controlled clinical trials are the basis of evidence-based
    medicine. These allow physicians and patients to use therapies with a
    clear understanding of their benefits and risks and, in some cases, a
    basis for strong public health recommendations for treatments.


    Clinical trials also have saved us from unwanted public health
    consequences. For example, when azidothymidine (AZT) was the only
    approved AIDS treatment, dideoxycytidine (ddC) was made available under
    treatment-IND for the several years while clinical trials were underway.
    These trials were to assess whether ddC was superior to AZT or if it
    was effective for patients intolerant of AZT. Although the product, ddC,
    could cause permanent, sometimes severe nerve damage, there was great
    demand for early access to the product. It was even manufactured by
    sources other than the company (probably by amateur chemists) and this
    “bathtub” ddC was made available through buyers clubs when the demand
    exceeded the sponsor's supply. FDA acted with the sponsor, the buyers
    clubs, patient advocates, and investigators to make more of the drug
    available and get the illicit, poorly manufactured product off the
    market.


    What did the ddC clinical trials show? In a head-to-head comparison
    versus AZT as initial therapy, an independent data safety monitoring
    board stopped the trial early because the death rate in the ddC group
    was at least twice higher than in the AZT group. For patients intolerant
    to AZT, a clinical trial compared switching to ddC versus
    dideoxyinosine (ddI). In this study the trend was that ddC had superior
    survival to ddI. Later studies showed that ddC in combination with AZT
    had superior survival to AZT alone. Each of these studies involved
    hundreds of patients and was essential to determining where ddC improved
    survival and where it did not. Although some of the early access uses
    were later found to be poor choices, physicians considered it reasonable
    at the time to provide the drug while the question was still being
    answered. The important point is that patients are only well served by
    early access when the controlled clinical trials proceed in parallel
    with early access.


    A second example that illustrates the importance of conducting
    clinical trials is the recently announced results of the Women's Health
    Initiative (WHI) study of estrogen and progesterone in treating
    post-menopausal women conducted by the National Institutes of Health.
    This large (more than 16,000 women), scientifically rigorous clinical
    trial was done to confirm the widely held belief that
    estrogen/progesterone therapy in post-menopausal women would
    significantly reduce the risk of cardiovascular events, such as heart
    attacks and strokes. There was also some hope that this post-menopausal
    therapy might lessen the onset of Alzheimer's disease. These widely held
    beliefs were based on scientific evidence that was not from clinical
    trials, such as epidemiology. On the strengths of these beliefs,
    post-menopausal hormone therapy was very widely used and growing in
    popularity.


    The WHI trial or post-menopausal estrogen/progesterone preceded but
    was stopped early due to an excess of harm in women taking these drugs
    compared to placebo. Surprisingly and importantly, women given the
    active drugs were more likely to suffer heart attacks and strokes and
    appeared to be more likely to develop dementia. This study not only
    failed to prove the widely held notion that this therapy was good for
    preventing these types of occurrences, but actually confirmed harm.
    These important results have led to significant changes in the use of
    post-menopausal hormones.


    FDA sometimes uncovers individuals who do not comply with statutory
    and regulatory drug approval requirements. This puts patients at risk of
    using unproven products and also denies to all patients the knowledge
    of whether the untested therapies may actually work. Distribution of
    unproven products and subsequent widespread use combined with little
    accountability or liability reduces the incentive for manufacturers and
    health care practitioners to conduct studies of safety and
    effectiveness. We constantly work to find ways to make safe and
    effective products available to patients as quickly and efficiently as
    possible, consistent with the protections established in the law. It is
    essential to preserve the system of controlled clinical trials that
    provides the information necessary to make the final determination on
    the safety and effectiveness of unapproved products. The two concepts,
    the protection of public health and making available treatments for
    individuals, can and must co-exist.


    HUMAN SUBJECT PROTECTION

    The FD&C Act and its implementing regulations are one part of a
    complex system of safeguards designed to protect human subjects. Each
    participant in a research effort --the company that sponsors the
    research, the clinical investigator who conducts the research, and the
    Institutional Review Board (IRB) is obliged to protect the interests of
    the people who are taking part in the experiments. FDA's responsibility
    is to see that the safeguards are met. FDA monitors the activities of
    research sponsors, researchers, IRBs and others involved in the trial.
    We take very seriously our role to protect people enrolled in clinical
    trials.


    The sponsors of research --usually, manufacturers or academic bodies,
    but sometimes individual physicians --must select well?qualified
    clinical investigators, design scientifically-sound protocols, make sure
    that the research is properly conducted, and make certain that the
    clinical investigators conduct the research in compliance with all
    pertinent regulations, including requirements for obtaining informed
    consent and review by an IRB. The primary regulatory obligations of the
    clinical investigator are to: 1) conduct or supervise the study; 2)
    conduct the study according to the approved protocol or research plan;
    3) ensure that the study is reviewed and approved by an IRB that is
    constituted and functioning according to FDA and other Federal
    requirements; 4) obtain informed consent; 5) maintain adequate and
    accurate records of study observations (including adverse reactions); 6)
    administer the drug only to subjects under the investigator's personal
    supervision or under the supervision of a sub-investigator responsible
    to the investigator; 7) report to the sponsor adverse experiences that
    occur in the course of the investigation; and 8) promptly report to the
    IRB all unanticipated problems involving risks to humans or others.


    The core of FDA's informed consent regulations, Title 21, Code of
    Federal Regulations (CFR) Part 50, is that the clinical investigator
    must generally obtain the informed consent of a human subject or his/her
    legally authorized representative before any FDA?regulated research can
    be conducted. The researcher has to make sure that, whenever possible,
    the study participants fully understand the potential risks and benefits
    of the experiment before the experiment begins. The information
    provided must be in a language understandable to the subject, and must
    not require the subject to waive any legal rights, or release those
    conducting the study from liability for negligence. The clinical
    investigator must tell the human subjects important information about
    the study and its potential consequences so that the person can decide
    whether to be in the experiment. The entire informed consent process
    involves giving the subject all the information concerning the study
    that he or she would reasonably want to know, ensuring that the subject
    has comprehended this information, and obtaining the subject's written
    consent to participate.


    An IRB is a group (consisting of experts and lay persons) formally
    designated to review, approve the initiation of, and periodically review
    the progress of, research involving human subjects. The primary
    function of IRBs is to protect the rights and welfare of the people who
    are in trials. FDA's regulations, 21 CFR Part 56, contain the general
    standards for the composition, operation, and responsibility of an IRB
    that reviews clinical investigations submitted to FDA under sections
    505(i), and 520(g) of the FD&C Act. IRBs must scrutinize and approve
    each of the clinical trials that are conducted on FDA-regulated
    products in this country each year. IRBs must develop and follow
    procedures for their initial and continuing review of the trials. Among
    other requirements, IRBs must make sure that the risks to subjects are
    minimized and do not outweigh the anticipated study benefits, that the
    selection of participants is equitable, that there are adequate plans to
    monitor data gathered in the trial and provisions to protect the
    privacy of subjects and the confidentiality of data. The IRB has the
    authority to approve, modify, or disapprove a clinical trial. The IRB
    must approve the informed consent form that will be used. If the
    researchers fail to adhere to IRB requirements, the IRB has the
    authority and the responsibility to take appropriate steps, which may
    include termination of the trial. The IRB is required to conduct
    continuing review of ongoing research at intervals appropriate to the
    degree of risk, but not less than once per year. It also has the
    authority to observe or have a third party observe the consent process
    and the research.


    IRBs are currently not required to register with FDA nor inform FDA
    when they begin reviewing studies. However, FDA performs on-site
    inspections of IRBs that review research involving products that FDA
    regulates, including IRBs in academic institutions and hospitals as well
    as those independent from where the research will be conducted. The
    primary focus of FDA's IRB Program is the protection of the rights and
    welfare of research subjects, rather than validating the data obtained
    from research.


    Marijuana


    FDA has not approved marijuana for medical use in the United States.
    Despite its status as an unapproved new drug, there has been
    considerable interest in its use for the treatment of a number of
    conditions, including glaucoma, AIDS wasting, neuropathic pain,
    treatment of spasticity associated with multiple sclerosis, and
    chemotherapy-induced nausea. Under the Controlled Substances Act (CSA)
    Congress listed marijuana in Schedule I. Schedule I substances have a
    very high potential for abuse, no accepted medical use in the United
    States, and lack accepted safety data for use under medical supervision.
    Schedule I substances can still be the subject of an IND; however, the
    conditions for its use are more restrictive.


    Pursuant to the FD&C Act, FDA is responsible for the approval and
    marketing of drugs for medical use, including controlled substances.
    DEA is the lead Federal agency responsible for regulating controlled
    substances and enforcing the CSA. The CSA separates controlled
    substances into five schedules, depending upon their approved medical
    use and abuse potential. Unlike Schedule I controlled substances,
    Schedule II substances are approved for medical use, although they also
    have a very high potential for abuse. Schedules III, IV, and V include
    those controlled substances that have been approved for medical use, but
    whose potential for abuse is diminished.


    FDA's Office of Criminal Investigations (OCI) is responsible for
    managing and conducting the Agency's criminal investigations. As a part
    of its duties, OCI has worked closely with DEA on a number of criminal
    investigations involving the illegal sale, use, and diversion of
    controlled substances including controlled substances sold over the
    Internet. OCI's close working relationship with DEA and local law
    enforcement agencies has led to many successful criminal cases involving
    controlled substances. FDA cooperates with DEA and other state and
    Federal agencies. OCI is often requested by these entities to provide
    assistance. Both OCI and DEA have worked together in the past to utilize
    the full range of regulatory and administrative tools available to them
    to pursue cases involving controlled substances. However, the primary
    responsibility for enforcing the CSA resides with DEA, and, FDA
    generally defers to DEA on criminal enforcement efforts related to
    Schedule I controlled substances. The criminal penalties related to
    Schedule I controlled substances are far greater under the CSA than
    those available under the FD&C Act for the distribution of an
    unapproved new drug.


    The Department of Health and Human Services (HHS) and FDA support the
    medical research community who intend to study marijuana in
    scientifically valid investigations and well-controlled clinical trials,
    in-line with the FDA's drug approval process. HHS and FDA recognize the
    need for objective evaluations of the potential merits of cannabinoids
    for medical uses. If the scientific community discovers a positive
    benefit, HHS also recognizes the need to stimulate development of
    alternative, safer dosage forms. In February 1997, an NIH-sponsored
    workshop analyzed available scientific information and concluded that
    “in order to evaluate various hypotheses concerning the potential
    utility of marijuana in various therapeutic areas, more and better
    studies would be needed.”


    In March 1999, the Institute of Medicine (IOM) issued a detailed
    report that supports the absolute need for evidence-based research into
    the effects of marijuana and cannabinoid components of marijuana, for
    patients with specific disease conditions. The IOM report also
    emphasized that smoked marijuana is a crude drug delivery system that
    exposes patients to a significant number of harmful substances and that
    “if there is any future of marijuana as a medicine, it lies in its
    isolated components, the cannabinoids and their synthetic derivatives.”
    As such, the IOM recommended that clinical trials should be conducted
    with the goal of developing safe delivery systems.


    In May 1999, HHS released “Guidance on Procedures for the Provision of Marijuana for Medical Research,”
    a document intended to provide the medical research community who
    intend to study marijuana in scientifically valid investigations and
    well-controlled clinical trials on HHS procedures for providing
    research-grade marijuana to sponsors. The HHS guidance is intended to
    facilitate the research needed to evaluate pending public health
    questions regarding marijuana by making research-grade marijuana
    available for well-designed studies on a cost-reimbursable basis. The
    focus of this HHS program is the support of quality research for the
    development of clinically meaningful data regarding marijuana. An
    appropriate scientific study of a drug requires, among other things,
    that the drug used in the research must have a consistent and
    predictable potency, must be free of contamination, and must be
    available in sufficient amounts to support the needs of the study. NIDA
    allocates resources to cultivate a grade of marijuana that is suitable
    for research purposes. The HHS Guidance outlines the procedures for
    obtaining research-grade marijuana including: 1) the researcher must
    make an inquiry to NIDA to determine the availability and costs of
    marijuana, and NIDA has to determine that marijuana is available to
    support the study; 2) researchers who propose to conduct investigations
    in humans must proceed through the FDA process for filing an IND
    application: and 3) all researchers must obtain from DEA registration to
    conduct research using a Schedule I controlled substance.


    FDA regulates smoked marijuana, a botanical product, when it is being
    investigated for use in the diagnosis, cure, mitigation, treatment or
    prevention of disease in man or other animals, as a drug, under the
    FD&C Act. Botanicals include herbal products made from leaves, as
    well as products made from roots, stems, seeds, pollen or any other part
    of a plant. Botanical products pose some issues that are unique to this
    class of product, including the problem of lot-to-lot consistency.
    These unpurified products, which may be either from a single plant
    source or from a combination of different plant substances, often exert
    their reported effects through mechanisms that are either unknown or
    undefined. For these reasons, the exact chemical nature of these
    products may not be known. In addition, issues of strength, potency,
    shelf life, dosing and toxicity monitoring need to be addressed. If a
    product varies greatly, as can occur with botanicals, it is critical to
    obtain lot-to-lot product consistency. Without this it is difficult to
    determine if the product is causing the change in a patient's condition,
    or the change is related to some other factor. Because of the problems
    associated with obtaining lot-to-lot consistency with botanical
    marijuana, it is not surprising that IOM recommended that clinical
    trials should be conducted with the goal of developing safe delivery
    systems.


    HHS performed a scientific and medical evaluation of marijuana in
    2001 and concluded with a recommendation to DEA that marijuana should
    remain in Schedule I pursuant to section 201(b) of the CSA. HHS's
    scientific and medical evaluation and scheduling recommendation can be
    found at Volume 66, Federal Register page 20038 (April 18,
    2001). After receiving an HHS evaluation and recommendation, DEA is
    responsible for scheduling substances and as noted previously, has
    primary responsibility for the regulation and distribution of Schedule I
    substances.


    FDA Approval of Safer Dosage Forms of Cannabinoids

    FDA has approved two drugs, Marinol and Cesamet, for therapeutic uses in
    the U.S., which contain active ingredients that are present in
    botanical marijuana. On May 31, 1985, FDA approved Marinol Capsules,
    manufactured by Unimed, for nausea and vomiting associated with cancer
    chemotherapy inpatients that had failed to respond adequately to
    conventional antiemetic treatments. Marinol Capsules include the active
    ingredient dronabinol, a synthetic delta-9- tetrahydrocannabinol or THC,
    which is considered the psychoactive component of marijuana. On
    December 22, 1992, FDA approved Marinol Capsules for the treatment of
    anorexia associated with weight loss in patients with AIDS. Although FDA
    approved Cesamet Capsules for the treatment of nausea and vomiting
    associated with chemotherapy on December 26, 1985, this product was
    never marketed in the U.S. Cesamet Capsules contain nabilone as the
    active ingredient, a synthetic cannabinoid. Nabilone is not naturally
    occurring and not derived from marijuana, as is THC.


    These products have been through FDA's rigorous approval process and
    have been determined to be safe and effective for their respective
    indications. It is only through the FDA drug approval process that solid
    clinical data can be obtained and a scientifically based assessment of
    the risks and benefits of an investigational drug is made. Upon FDA
    approval for marketing, consumers who need the medication can have
    confidence that the approved medication will be safe and effective.


    CONCLUSION


    Having access to a drug or medical treatment, without knowing how to
    use it or even if it is effective, does not benefit anyone. Simply
    having access, without having safety, efficacy, and adequate use
    information does not help patients. FDA has and will continue to use its
    IND and other expanded access programs to provide patients freedom to
    choose investigational medical treatments while reasonably ensuring
    safety, informed choice, and systematic data collection that allows us
    to review drug applications.


    FDA will continue to be receptive to sound, scientifically based
    research into the medicinal uses of botanical marijuana and other
    cannabinoids. FDA will continue to facilitate the work of manufacturers
    interested in bringing to the market safe and effective products.


    I would like to thank the Subcommittee again for the opportunity to
    testify today on this important issue. I would be happy, at this time,
    to answer any questions Members of the Subcommittee may have.




     

Share This Page