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.

Cannabidiol: Barriers to Research and Potential Medical Benefits-June 24, 2015

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

  1. http://www.fda.gov/newsevents/testimony/ucm453989.htm
    Statement of Douglas C. Throckmorton, M.D.
    Deputy Director for Regulatory Programs
    Center for Drug Evaluation and Research
    Food and Drug Administration
    Department of Health and Human Services
    Before the
    Caucus on International Narcotics Control
    United States Senate
    June 24, 2015
    INTRODUCTION
    Chairman
    Grassley, Co-Chairman Feinstein, and Members of the Caucus, I am Dr.
    Douglas Throckmorton, Deputy Director for Regulatory Programs at the
    Center for Drug Evaluation and Research (CDER), Food and Drug
    Administration (FDA or the Agency), which is part of the Department of
    Health and Human Services (HHS). Thank you for the opportunity to be
    here today to discuss the important role that FDA plays in the
    regulation of researching marijuana, including its constituent
    compounds, such as cannabidiol, for potential medical uses. This is an
    important part of FDA's mission to protect and promote the public health
    by helping to ensure the safety, efficacy, and quality of medical
    products, including drugs. In addition, I will briefly discuss the
    regulatory oversight function of the Agency with respect to other
    products that may contain cannabidiol.
    FDA plays a critical role
    in regulating the development and potential use of cannabidiol and other
    constituents of marijuana as prescription drugs in the United States.
    FDA also, on its own and in partnership with other Federal agencies,
    supports the efficient and scientific assessment of cannabidiol and
    other constituents of marijuana in drug development. These activities
    are critical, if safe and effective drugs are to be developed from
    marijuana, including from its constituent components such as
    cannabidiol. FDA continues to believe that the drug approval process
    represents the best way to help ensure that safe and effective new
    medicines, including any such medicines derived from cannabidiol or
    other constituents of marijuana, are appropriately reviewed for safety
    and effectiveness, consistent with FDA's statutory requirements. It is
    important and appropriate to use the same scientific standards in the
    development and assessment of potential therapeutic uses of cannabidiol
    as with any unapproved drug that the Agency reviews.
    FDA's Role in Reviewing Cannabidiol as a Potential Prescription Drug
    The
    role of FDA in the regulation of cannabidiol as a potential
    prescription drug relates to our larger responsibility for the
    regulation of all drugs intended for human use. The Agency reviews drug
    product applications to determine whether drugs are safe and effective
    for their intended uses. Any product intended for use in the diagnosis,
    cure, mitigation, treatment, or prevention of disease is classified by
    FDA as a drug. This applies regardless of the product's form, the
    product's active or inactive ingredients, or the way in which the
    manufacturer chooses to market and label the product.
    In approving a drug for marketing, FDA reviews important information about the drug, including:
    The indication for which the drug has been studied, including specific uses in children or the elderly, if any
    Which patients may benefit from its use, including information about whether the drug has been tested in children
    What adverse effects have been reported for individuals taking the drug
    How the drug should be taken (e.g., orally, intravenously)
    The dose of the drug recommended in the intended patient populations
    How the drug is made (e.g., as a pill, liquid) and what is in the drug, including both active and inactive ingredients
    Getting
    a drug approved requires the collection and submission to FDA of
    clinical and non-clinical data about the proposed use of the drug for
    review as part of a New Drug Application (NDA) or Biologics License
    Application (BLA). Usually, the first step that a sponsor takes to
    obtain approval for a new drug is to test the drug in animals to
    determine drug toxicity. The sponsor uses those animal data, along with
    additional information about the drug's composition and manufacturing,
    to develop a plan for testing the drug in humans. The sponsor submits
    the animal data to FDA in the form of an Investigational New Drug (IND)
    application that includes protocols describing proposed human studies,
    the qualifications of the investigators who will conduct the clinical
    studies, and assurances of informed consent and protection of the
    rights, safety, and welfare of the human participants. FDA reviews the
    IND to confirm that the proposed studies, generally referred to as
    clinical trials, do not place human participants at unreasonable risk of
    harm. FDA also verifies that there are adequate assurances of informed
    consent and human subject protection. At that point, drug testing in
    humans can begin.1
    Typically, the initial clinical
    trials (Phase 1) assess how to safely administer and dose the drug when
    used in small numbers of healthy volunteers. If Phase 1 trials are
    successful, Phase 2 studies explore the effectiveness of the drug for a
    particular indication, over a range of doses, and determine short-term
    side effects. If Phase 2 studies are successful, pivotal Phase 3 studies
    are designed based on the information learned in the earlier studies to
    further study safety and assess the efficacy of the investigational
    drug for a particular indication in a defined patient population. Phase 3
    studies also can provide additional safety data, including long-term
    experience effects of the drug in certain patient groups, and efficacy
    of different doses of the drug. These later trials sometimes enroll
    several hundreds to thousands of participants-depending on the
    indication studied-to provide essential information about the
    investigational drug's safety and efficacy. Following the completion of
    these studies, the data might be submitted to FDA in an NDA or BLA for
    the Agency to review. Throughout the development process, FDA strongly
    encourages sponsors to work closely with the Agency to support efficient
    drug development.
    In addition to establishing the safety and
    efficacy of the investigational drug, manufacturers also must
    demonstrate that they are able to consistently manufacture a
    high-quality drug product. This is an essential part of drug development
    and presents special challenges when the drug is derived from a
    botanical source, such as marijuana. Botanicals include herbal products
    made from leaves, roots, stems, seeds, pollen, or any other part of a
    plant. Botanical products pose challenges that are unique to this class
    of product, including lot-to-lot consistency. These unpurified products,
    which may be either from a single plant source or from a combination of
    different plant substances, can have effects through unknown or
    undefined mechanisms, making it difficult to determine if the product is
    causing the change in a patient's condition, or the change is related
    to some other factor. For these reasons, a focus of drug development for
    botanicals is identification of a source that will provide the
    necessary assurance of consistent quality, lot to lot. To support
    development of drugs derived from botanical sources, FDA has released
    guidance providing information on the development and approval of such
    drugs that addresses these issues, as well as providing more general
    recommendations on studying botanicals.2
    Another
    important consideration is the need to identify a method to consistently
    provide a given dose of a drug. When the Institute of Medicine (IOM)
    reviewed the potential clinical use of marijuana, it identified the
    problems associated with obtaining consistent dosing using smoked
    products and recommended that clinical trials involving marijuana should
    be conducted with the goal of developing safe, alternative delivery
    systems:3
    If there is any future for marijuana
    as a medicine, it lies in its isolated components, the cannabinoids and
    their synthetic derivatives. Isolated cannabinoids will provide more
    reliable effects than crude plant mixtures. Therefore, the purpose of
    clinical trials of smoked marijuana would not be to develop marijuana as
    a licensed drug but rather to serve as a first step toward the
    development of nonsmoked rapid-onset cannabinoid delivery systems.Another
    consideration related to the regulation of cannabidiol as a potential
    medicine is its status as a constituent of a controlled substance, in
    this case marijuana. Under section 202 of the Controlled Substances Act
    (CSA), marijuana is currently listed as a Schedule I controlled
    substance.4 Schedule I includes those substances that have a
    high potential for abuse, have no currently accepted medical use in
    treatment in the United States, and lack accepted safety for use under
    medical supervision.5 Nevertheless, Schedule I substances,
    including drugs such as marijuana, can be and are the subject of
    clinical trials under the Federal Food, Drug and Cosmetic Act (FD&C
    Act), provided, among other factors, that FDA authorizes an IND
    application submitted by a sponsor. In addition, the CSA requires
    researchers to register with the Drug Enforcement Administration (DEA)
    before handling Schedule I controlled substances, including conducting
    clinical trials.6
    Through the drug development
    processes described above, FDA has approved two drugs for human use
    which contain synthetic cannabinoids: Marinol (Schedule III) and Cesamet
    (Schedule II). FDA approved Marinol capsules in 1985 for the treatment
    of nausea and vomiting associated with cancer chemotherapy in patients
    who had failed to respond adequately to existing antiemetic treatments.
    Marinol capsules include the active ingredient dronabinol, a synthetic
    delta-9-tetrahydrocannabinol, or THC, which is a psychoactive component
    of marijuana. Marinol capsules were also approved in 1992 for the
    treatment of anorexia associated with weight loss in patients with AIDS.
    FDA approved Cesamet capsules for the treatment of nausea and vomiting
    associated with chemotherapy in 1985. Cesamet capsules contain the
    synthetic cannabinoid nabilone as the active ingredient.
    These
    products have undergone FDA's rigorous approval process and have been
    determined to be safe and effective for their respective indications and
    dosing, and demonstrate the views of the IOM that the future of
    marijuana as a potential medicine lies in classical pharmacological drug
    development.7 As a result, patients who need medication can
    have confidence that any approved drug will be safe and effective for
    its indicated uses.
    FDA's Role in Supporting Development of New Therapies
    FDA
    also plays a role in supporting the development of new drugs. This role
    broadly affects all of drug development. Because of FDA's role as both a
    regulator and as a public health agency, FDA has a unique perspective
    on drug development, a perspective we use to identify and facilitate the
    development of new, innovative products to meet the needs of patients
    and the American public. We recognize that many scientific discoveries
    still need to be translated into treatments, even as patients are
    urgently waiting for new lifesaving therapies, and FDA is committed to
    helping bridge this gap.
    As a part of this activity to streamline
    drug development, FDA has been actively scrutinizing, strengthening, and
    streamlining our regulatory processes at various steps along the path
    from drug discovery to delivery-including the clinical development
    phase, the longest and most expensive period of drug development. We
    have developed and successfully used a number of flexible and innovative
    approaches to expedite the development and review of drugs-to the
    benefit of millions of American patients. For instance, in 2014 almost
    two-thirds (26 of 41, 63 percent) of new molecular entities approved by
    CDER were approved in the United States before any other country.8
    This is comparable to the previous year, 2013, in which almost three-
    quarters of CDER-approved new molecular entities (74 percent of 27) were
    approved first in the United States.9
    FDA has several
    programs that directly facilitate and expedite development and review of
    new drugs that address unmet medical needs in the treatment of serious
    or life-threatening conditions: Fast Track10, Accelerated Approval11, Priority Review12, and Breakthrough Designation13.
    A look at recent drug approvals in CDER suggests that these programs
    have played an important role in bringing innovative drugs to market.
    Nearly half of the 27 novel drugs approved by CDER in 2013 took
    advantage of at least one of these expedited drug development and review
    approaches. Utilization of these accelerated programs was even more
    pronounced in 2014, with two-thirds (27 of 41) of novel drugs approved
    by CDER using at least one of these programs.
    Development programs
    for drugs derived from marijuana and its constituents, including
    cannabidiol, may be eligible for these expedited review and development
    programs under appropriate circumstances and, in fact, some of these
    programs are currently being used in the development of such drugs. For
    example, in April 2014, GW Pharmaceuticals announced14 that
    FDA granted Fast-track designation to its investigational drug product
    Sativex®, composed primarily of two cannabinoids: cannabidiol and THC,
    administered as a metered-dose oromucosal spray, for the treatment of
    pain in patients with advanced cancer, who experience inadequate
    analgesia during optimized chronic opioid therapy. GW Pharmaceuticals
    has announced15 that Sativex is currently in Phase 3 clinical
    trials for this indication. In addition, on June 6, 2014, GW
    Pharmaceuticals announced16 that FDA granted Fast-track
    designation to its investigational cannabidiol product, Epidiolex®, in
    the treatment of Dravet syndrome, a rare and catastrophic
    treatment-resistant form of childhood epilepsy. In February 2015, Insys
    Therapeutics announced17 that FDA granted Fast-track designation to the cannabidiol formulation they are studying for the same condition.
    As
    it does in other new drug development processes, FDA is working with
    researchers who are conducting studies on the development of new drugs
    derived from marijuana, including from its constituents such as
    cannabidiol, meeting with them regularly as they plan and carry out the
    trials as a part of their INDs. Although marijuana is a Schedule I
    substance, it can be, and is being, used in clinical trials conducted
    under INDs. A number of government-funded research projects involving
    marijuana or its component compounds, including cannabidiol, have been
    completed or are currently in progress, many of which are listed on the www.ClinicalTrials.gov website.
    FDA
    also understands the interest in making investigational products
    available to patients while being studied for approval, and there are
    expanded access provisions in both FDA's statute and its regulations to
    make this possible, where appropriate and where the manufacturer chooses
    to participate. FDA's expanded access mechanisms are designed to
    facilitate the availability of investigational products to patients with
    serious diseases or conditions when there is no comparable or
    satisfactory alternative therapy available, either because the patients
    have exhausted treatment with, or are intolerant of, approved therapies,
    or because the patients are not eligible for an ongoing clinical trial.18
    FDA cannot mandate or require a drug company to provide an unapproved
    drug to patients, and the availability of an investigational product
    through expanded access depends on the agreement of the drug company to
    make the drug available for the expanded access use, either through the
    company's own expanded access program, or to a treating physician for
    administration to a patient or patients.
    As noted, Epidiolex, containing cannabidiol, is being developed for the treatment of certain seizure disorders in children.19
    GW Pharmaceuticals reports that 20 Epidiolex intermediate-size expanded
    access INDs have been authorized to treat approximately 420 children
    and that approximately 95 percent of Epidiolex expanded access INDs are
    for patients between 1 and 17 years of age.20
    FDA also
    has worked with investigators to provide clear information on how to
    conduct research in this area. To help address common basic questions
    about research with marijuana, FDA, the National Institute on Drug Abuse
    (NIDA), and DEA have posted materials online to help researchers.21
    We also know that a number of states are interested in allowing access
    to cannabidiol-containing oils to treat childhood epilepsy. FDA
    encourages and supports medical research into the safety and
    effectiveness of products containing cannabidiol and other marijuana
    constituents through adequate and well-controlled clinical trials
    conducted under an IND and consistent with DEA requirements for research
    on Schedule I substances. FDA has talked with representatives from
    several states considering support for medical research of marijuana and
    its derivatives, including cannabidiol, to provide scientific advice
    and to help ensure that their research is rigorous and appropriate.
    FDA's Role in Investigations and Enforcement Actions With Regard to Products Containing Cannabidiol
    FDA
    recognizes that DEA is the lead Federal Government agency for
    enforcement matters related to the diversion of controlled substances,
    including marijuana. Historically, FDA has deferred to DEA regarding the
    illegal sale and use of illicit drugs of abuse, such as Schedule 1
    drugs, which have no currently accepted medical use.
    However, in
    addition to regulating medical products, FDA maintains regulatory
    oversight over foods, including dietary supplements. For any product
    found to be in violation of the FD&C Act, FDA considers many factors
    in deciding whether or not to initiate an enforcement action. Those
    factors include, among others, Agency resources and the threat to the
    public health. FDA also may consult with its Federal and state partners
    in making decisions about whether to initiate a Federal enforcement
    action.
    In late February 2015, FDA issued several Warning Letters
    to firms that were marketing unapproved drugs for the diagnosis, cure,
    mitigation, treatment, or prevention of diseases. Some of these firms
    claimed that their products, which the firms made available nationwide
    via the Internet, contained cannabidiol which the firms claimed could
    help to address cancer, diabetes, multiple sclerosis, and other
    ailments. FDA tested those products and, in some, did not detect any
    cannabidiol.22 It is important to note that these products
    are not approved by FDA for the diagnosis, cure, mitigation, treatment,
    or prevention of any disease. Often they do not even contain the
    ingredients found on the label. Although FDA advised these firms that
    their products were in violation of the FD&C Act, and FDA is
    committed to keeping violative products off the market, consumers should
    be extremely cautious in purchasing and using such products. Marketing
    of products that are not what they claim to be, such as those purporting
    to contain cannabidiol but don't, does more than simply defraud
    consumers who anticipate that the products purchased will contain the
    ingredients expected. These products and marketing can create false hope
    in a population especially vulnerable: those seeking relief from
    serious medical conditions for themselves or their loved ones, including
    their children. Moreover, it might divert patients from products with
    demonstrated safety and effectiveness.
    The recent examples of the
    products being marketed to individuals seeking relief from serious
    medical conditions-products that FDA has not evaluated for safety and
    efficacy and that often do not even contain the ingredients they claim
    to contain-serves as another reminder of the important role FDA and the
    drug approval process play in this area.
    CONCLUSION
    FDA
    appreciates this opportunity to discuss the Agency's work in the
    regulation of cannabidiol for potential medical uses in the United
    States, which is a part of FDA's core mission to protect and promote the
    public health by helping to ensure the safety, efficacy, and quality of
    medical products, including drugs. There is considerable public
    interest in developing new therapies from marijuana and its
    constituents, especially cannabidiol. FDA will continue to play its role
    in ensuring that any such new therapies are safe, effective, and
    manufactured to a high quality, applying the drug development paradigm
    that continues to provide new medicines that meet these standards for
    patients. This paradigm, grounded in rigorous scientific research, is
    essential to determining any appropriate uses of marijuana and its
    constituents in the treatment of human disease. As a part of this
    important work, we are committed to collaborating with Federal and state
    agencies, researchers, and manufacturers also working on issues related
    to the use of cannabidiol and other constituents of marijuana in the
    United States. The drug approval process remains the best way to
    identify new treatments that are safe and effective for patients and to
    protect patients from products that are not what they purport to be.
    Thank you for your interest in this important topic, and I am happy to answer any questions.
    1 In
    the case of Schedule I controlled substances, such as marijuana from
    which cannabidiol is derived, the Controlled Substances Act requires
    researchers to register with the Drug Enforcement Administration (DEA)
    before handling the controlled substances, including proceeding with
    clinical trials using controlled substances. Registration requirements
    applicable to research involving Schedule I controlled substances differ
    from those for drugs controlled in Schedules II-V (21 U.S.C. 823(f)).
    2 FDA guidance document on the development of botanical drug products is posted at http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm070491.pdf.
    3 IOM Report, p. 11 (1999), Marijuana and Medicine: Assessing the Science Base.
    4 21 U.S.C. 812
    5 21 U.S.C. 812(b)(l)(A)-(C)
    6 21
    U.S.C. 823(f) (stating that registration applications by practitioners
    wishing to conduct Schedule I research shall be referred to the
    Secretary of HHS, who shall determine the qualifications and competency
    of each practitioner, as well as the merits of the research protocol);
    see also 21 CFR 1301.18 (outlining specific application procedures and
    information to be provided by Schedule I researcher applicants).
    7 Institute of Medicine, Marijuana and Medicine: Assessing the Science Base (IOM Report), p.193 (2003).
    8 CDER Report, “Novel new Drugs, 2014 Summary,” (1/14/15) is posted at http://www.fda.gov/downloads/Drugs/DevelopmentAppr...
    9 “President's
    Fiscal Year 2015 Budget Request for the FDA” Testimony of Commissioner
    Margaret Hamburg before the Senate Committee on Appropriations (April 3,
    2014) at http://www.fda.gov/newsevents/testimony/ucm392262.htm.
    10 Fast-track
    designation: Provides opportunities for frequent interactions with FDA
    to discuss the drug's development plan and ensure collection of
    appropriate data needed to support drug approval, including such things
    as the design of the proposed clinical trials and use of biomarkers.
    11 Accelerated
    Approval: Basing approval on an agreed-upon surrogate marker that is a
    measure, such as a blood test or urine marker, that is believed to be
    indicative of a disease state and treatment effect but not demonstrative
    of a direct health gain to the patient, or on an intermediate clinical
    endpoint likely to predict clinical benefit. Since its inception in
    1992, 89 original new drug or biologic applications have been approved
    in CDER under the Accelerated Approval pathway. It has long been
    successful in driving innovation in cancer and HIV therapies, but we are
    encouraging its broader application in other areas, helped by the 2012
    Food and Drug Administration Safety and Innovation Act (FDASIA), which
    clarified that FDA has the authority to consider epidemiologic,
    pharmacologic, or other evidence developed using biomarkers or other
    scientific methods or tools in determining whether an endpoint can
    support accelerated approval.
    12 Priority Review: Acting on drug applications within six months instead of 10 months for standard review
    13 Breakthrough
    Therapy designation: Providing all of the benefits of Fast-track
    designation plus intensive guidance on an efficient drug development
    program, beginning as early as Phase 1, and the commitment from FDA's
    review staff, including senior managers, to work closely together
    throughout the drug development and review process. FDA's new
    Breakthrough Therapy Designation was created as part of FDASIA. As of
    May 29, 2015, CDER received 258 requests for designation, and granted
    78. As of May 29, 2015, 24 drug development programs designated
    breakthrough have been approved by CDER. Of these 24, 14 were for
    original applications and 10 were for supplements (to expand or add a
    new indication to an already approved drug).
    14 http://www.gwpharm.com/GW Pharmaceuticals Anno...
    15 http://www.gwpharm.com/Third phase III Sativ...
    16 http://www.gwpharm.com/GW Pharmaceuticals Anno...
    17 http://www.insysrx.com/investors/recent-news/ (Link at February 26, 2015)
    18 http://www.fda.gov/ForPatients/Other/default.htm
    19 Epidiolex
    received orphan product designation for treatment of Dravet syndrome.
    http://www.accessdata.fda.gov/scripts/opdlisting/o...
    and more recently for the treatment of Lennox-Gastaut syndrome: http://www.accessdata.fda.gov/scripts/opdlisting/o...
    20 http://www.gwpharm.com/Epidiolex.aspx
    21 http://www.fda.gov/drugs/developmentapprovalprocess/howdrugsaredevelopedandapproved/approvalapplications/investigationalnewdrugindapplication/ucm362986.htm;
    http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM198650.pdf;
    http://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm070491.pdf;
    http://www.drugabuse.gov/drugs-abuse/marijuana;
    http://www.deadiversion.usdoj.gov/drugreg/faq.htm#...
    22 For information on these warning letters and the product testing, please see http://www.fda.gov/NewsEvents/PublicHealthFocus/uc...

     

Share This Page