• where experts go to learn about FDA
  • New Citizen Petition Asks FDA to Require Homeopathic Drugs to be Tested for Effectiveness or Include a Disclaimer

    By Susan J. Matthees

    The Center for Inquiry, a non-profit organization with a mission to “promote scientific inquiry,” recently submitted a Citizen Petition to FDA requesting that the Agency initiate rulemaking for a proposed rule that would require all homeopathic drugs to be tested for effectiveness and, until the drugs are tested, require all advertisements for the products carry to state “WARNING:  The FDA has not determined that this product is safe, effective, and not misbranded for its intended use.” 

    FDC Act § 505 prohibits a new drug from being introduced into interstate commerce without an application approved by FDA based, in pertinent part, on substantial evidence of effectiveness.  Although homeopathic drugs are not exempt from FDC Act § 505, most are marketed under FDA’s Compliance Policy Guide (“CPG”) “Conditions Under Which Homeopathic Drugs May be Marketed” and are not approved by FDA.  Pursuant to the CPG, marketed homeopathic drugs must meet the standards for strength, quality, and purity set forth in the Homeopathic Pharmacopeia of the United States, must have indications included in a Materia Medica, and must be labeled in accordance with the FDC Act and FDA’s regulations. 

    The Center for Inquiry does not explicitly ask FDA to revoke the CPG, remove homeopathic drugs from the market, or approve homeopathic drugs; the only explicit requests are that FDA use its authority to require efficacy testing for homeopathic drugs and a warning label for homeopathic drugs that are not tested.  The petition is short on legal analysis, and instead is dedicated to explaining that scientific evidence demonstrates that homeopathic drugs are not effective.  In particular, the Center for Inquiry mentions a product manufactured by Boiron as a product that allegedly has no credible scientific evidence of effectiveness.  As you may recall, a federal court in California recently decided to allow a case against Boiron to proceed.  The plaintiffs in that case allege that Boiron violated the California Legal Remedies Act and the California Unfair Competition Law and committed common law fraud by marketing homeopathic drugs that are not effective.  The Center for Inquiry does not mention the California case. 

    Medical Devices Law and Regulation Answer Book 2011–12

    The Practising Law Institute ("PLI") has released a new Medical Devices Law and Regulation Answer Book 2011–12, edited by Suzan Onel (K&L Gates LLP) and Karen M. Becker (Becker & Associates Consultants).  This Q&A guide is intended to orient and guide those new to the industry, and to be a useful annotated resource for the more experienced regulatory professional.  It has 19 chapters covering a wide range of topics in the life cycle of a medical device, including:

    • Device Premarket Submissions 
    • Post Market Considerations 
    • In Vitro Diagnostic ("IVD") Devices 
    • Interacting with FDA 
    • International Considerations 
    • HIPAA’s Impact on Manufacturers

    The book has more than 30 contributors from leading law firms and consulting firms specializing in medical device work.  In the interest of full disclosure, we (proudly) note that Jeff Shapiro, a Director of our firm, contributed Chapter 3 on Device Premarket Submissions.  We also received a free review copy.

    The single volume Medical Devices Law and Regulation Answer Book 2011–12 is $235 and is available for a 30-day free examination.

    The table of contents is unusually detailed because it shows the questions that the book answers in each chapter.  The table of contents (and other information about the book and the contributing authors) can be found here.  The FDA Law Blog is pleased to offer its readers a 25% discount off the purchase price.  When ordering, contact Andrew Given at 212-824-5887 or agiven@pli.edu and let him know you would like the FDA Law Blog discount.

    Categories: Medical Devices

    FDA and FSIS Request Information on Sodium Reduction

    By Ricardo Carvajal

    FDA and FSIS have published a notice seeking information on issues related to reduction of dietary sodium.  Last year, the CDC determined that the government’s recommendation to limit sodium to no more than 1,500 mg/day applied to nearly 70% of adults (those deemed to be at greater risk for hypertension), but even the higher recommended limit of 2,300 mg/day has proven difficult to meet.  Average intake among those over 2 years of age is nearly 3,500 mg/day.  According to CDC, 40 percent of daily sodium intake comes from grain-based products (regulated by FDA) and 30 percent comes from processed meat products (regulated by FSIS).  Given the government’s estimate that 75% of dietary sodium is added during food manufacture and restaurant preparation, the topic of sodium reduction is of obvious interest to industry.

    The notice catalogues 25 years’ worth of government and industry efforts to help consumers reduce sodium intake.  Given little progress on the issue, the two agencies are now “considering potential ways to promote gradual, achievable and sustainable reduction of sodium intake over time,” and acknowledge the need for “research on a variety of issues, including the development of possible targets” for sodium reduction.  The notice acknowledges that a number of factors “may inform judgments about appropriate opportunities for sodium reduction,” including the role of sodium in food safety, its impact on food processing, and the fact that consumers prefer salty foods.  The notice seeks information on a number of sodium reduction issues, including current industry initiatives, potential effective strategies, incentives for innovation in reformulation, establishment of targets, potential unintended consequences, and economic impacts.

    Those wishing to submit information should note that any information not marked confidential “will be included in the public version of the official record without prior notice.” 

    FDA Public Meeting on Mobile Medical Apps and Stand-Alone Clinical Decision Support Software

    By Carmelina G. Allis

    Below is a summary of some of the key issues discussed during FDA’s September 12 and 13, 2011, public meeting on the recently issued draft guidance document for mobile med apps.  During the meeting, FDA also requested input on how to regulate stand-alone software that provides clinical decision support, even though those support systems are not specifically addressed in the draft guidance.

    In general, FDA received positive feedback regarding the draft guidance document on mobile apps.  However, it is clear that the agency still has some work to do, in particular, it needs to define the regulatory landscape in the area of stand-alone clinical decision support software.  There are some unanswered questions in both areas, as you will see from the discussion below:

    Mobile Medical Apps

    • There were three main proposals regarding mobile medical apps: (1) FDA should classify apps based on the level of risk to patient health; (2) FDA should issue guidance to explain what kinds of apps and/or claims the agency will not regulate; and (3) there should be a way to ensure that apps do not adversely affect the main device they connect to (and vice versa), such as requiring good software practices.
    • Because many app “manufacturers” are not FDA-regulated entities, the draft guidance document needs clarity and information in order to help those that are having to learn the regulatory landscape.  The discussion on mobile medical apps centered around what does “intended use” mean and how to apply it.  A simple roadmap that helps entities/individuals to determine “how do I know that I am a mobile med app manufacturer” was suggested.
    • The discussion on mobile med apps as “accessories” centered around proposals that FDA classify accessories based on intended use and functionality rather than specific characteristics or technology.
    • There was some discussion that the draft guidance goes too far – for example, could general-purpose websites that have dosage calculators fall within the definition of a mobile medical app if that website (and, thus, the dosage calculator) can be accessed via a mobile phone?
    • Another issue discussed was that of “interoperability” – there are apps that get information from many different sources/devices, and then an action is triggered.  How does FDA intend to regulate those apps?  Some suggested a risk-based approach.  However, the problem with the risk-based approach is how to define it: the risk if something goes wrong with the app vs. the risk that the use of the app poses?  FDA’s Bakul Patel noted that the agency discusses those issues internally all the time, but that at the end of the day, the issue is about patient safety.  Another issue regarding interoperatibility relates to who has the regulatory burden to ensure safety and effectiveness.  Is it the app manufacturer only?  But how do you ensure that the hardware (e.g., the iPad or Android platform, which FDA has said will not regulate) is safe for a particular app?
    • It was suggested that FDA not discourage people from being able to make claims about the platform because of fear of becoming a medical device.  This issue came up because of comments regarding the Ford car that will supposedly provide access to apps specifically targeted to diabetics.  Does the car become an accessory to the app?  Does it all depend on the claims made by Ford?  Or could FDA allow certain claims and not regulate the car?

    Stand-Alone Clinical Decision Support Systems

    • FDA said that the factors it generally considers on how to regulate stand-alone clinical decision support systems involve: (1) the level of impact on subject health/condition; (2) the degree of acceptance in the clinical practice; and (3) the ability to identify erroneous output (due to incorrect output or clinically wrong information).
    • There appears to be a strong concensus among some that national/international standards should be imposed/required to ensure that clinical decision support systems adhere to good software practices.
    • Others suggested that regulatory requirements for clinical decision support systems should be based on intended use and risk.  Two general types of support software systems were outlined: (1) those that provide generic decision support, such as providing summaries of clinical articles or anecdotes, which FDA has said it does not intend to regulate, and (2) those that provide patient-specific support.  Among the latter, three main types were discussed:

    – systems that provide or assist with simple functions;
    – systems that provide assistance with therapeutics; and
    – systems that assist with diagnoses.

    No particular regulatory approach was proposed, as the landscape of clinical decision support systems varies immensely on intended use, intended population, functionality, and technological characteristics.  For example, while some clinical decision support systems are based on simple rules, like medication reminder software, others are based on more complex algorithms, such as assisting medical practitioners with diagnoses assessments or computing chemotherapy doses.  One possible approach not publicly discussed at the meeting would be for FDA to create a classification regulation requiring 510(k)s with special controls that applies to a well defined, but broad group of stand-alone clinical decision software (e.g., a classification regulation for software intended to assist medical practitioners with therapy determinations).

    At this time it is difficult to advocate a particular regulatory pathway to clients for these device types, because there are no classification regulations that apply to many of these stand-alone clinical decision systems.  In addition, FDA has not defined what system types it intends and does not intend to regulate.  And because the 510(k)/de novo pathway is unpredictable and inefficient, manufacturers are unwilling to pursue any specific pathway to market until FDA further defines the regulatory landscape.  FDA does not want to be inundated with de novos or PMAs (which would be the default for lack of a predicate device), both of which are time-consuming and resource-intensive regulatory processes.  And that is why the suggested approach of creating a regulation with special controls for a defined, but general-scope stand-alone clinical decision support system looks appealing.

    Categories: Medical Devices

    USTR Embraces “TEAM” Approach in TPP Talks; Senators Back 12-Year Exclusivity for Biologics

    By Kurt R. Karst –      

    September 12th marked the beginning of the eighth round of Trans-Pacific Partnership (“TPP”) negotiations, which are taking place in Chicago, Illinois.  The TPP is an Asia-Pacific regional trade agreement being hammered out among the United States and eight other partners.  The TPP would cover trade in goods and services and also includes a proposed chapter on intellectual property.  As we previously reported, the chapter on intellectual property is where the latest battle over biologics exclusivity is happening, and which was the subject of several letters from Members of Congress sent earlier this year. 

    In a new round of letters reported on by Patent Docs, several U.S. Senators urge U.S. Trade Representative (“USTR”) Ron Kirk to support a 12-year period of exclusivity for biological products.  Colorado Senators Mark Udall (D) and Michael Bennett (D) state in their letter that “[b]eginning the biologics negotiations on an intellectual property standard consistent with U.S. law will make sure that Coloradans can continue to lead the world in the innovation of biologics while also assuring a reasonable pathway for biosimilar products.”  Meanwhile, another letter signed by a bipartisal group of 37 U.S. Senators says that they are united in urging the USTR to “propose a strong minimum term of regulatory data protection for biologics consistent with U.S. law.”

    On the same day the letters were sent to USTR Kirk, the USTR issued a white paper outlining a new strategic initiative, titled “Trade Enhancing Access to Medicines” (or “TEAM”).  The TEAM strategy, which is supported by the U.S. Agency for International Development, is “designed to deploy the tools of trade policy to promote trade in, and reduce obstacles to, access to both innovative and generic medicines, while supporting the innovation and intellectual property protection that is vital to developing new medicines and achieving other medical breakthroughs,” according to the white paper.  “The TEAM initiative reflects fresh thinking about trade and access to medicines.  It is about more than allowing access to medicines.  It is about working with trading partners to develop strong and common standards to help drive access – propelling the TPP countries to the front of the line for important innovative medicines and for generic competition, while promoting U.S. jobs and exports.”

    The USTR’s white paper identifies several goals, including:

    Expedite access to innovative and generic medicines through a “TPP access window”: Promote the availability of life-saving and life-enhancing medicines in TPP markets and simultaneously establish a pathway for generics to enter those markets as quickly as possible by conditioning obligations to apply certain pharmaceutical-specific intellectual property protections on the requirement that innovators bring medicines to TPP markets within an agreed window of time.

    The white paper is silent on the term of any biologics exclusivity period; however, we note that President Obama’s Budget for Fiscal Year 2012 proposed that “beginning in 2012, innovator brand biologic manufacturers would have 7 years of exclusivity. . . .”  A letter sent to President Obama this past summer concerning TPP negotiations stated that “[w]ere the TPP ultimately to contain a 12 year biologics exclusivity provision, it would impede the ability of Congress to achieve the Administration’s proposed 7 year change without running afoul of U.S. trade obligations.”

    Another Reminder: Nutrition Labeling of Single Meat and Poultry Products is Coming Soon

    By Riëtte van Laack

    As we previously reported, on December 29, 2010, the Food Safety Inspection Service ("FSIS") published its final rule on nutrition labeling of single ingredient meat and poultry products and ground or chopped meat and poultry products. The new rule requires nutrition labeling on the major cuts of single-ingredient raw meat and poultry products and on all ground or chopped meat and poultry products, with or without added seasonings, unless an exemption applies. The new rule also provides that a lean percentage statement of the fat percentage may be included on ground or chopped product that does not meet the regulatory criteria for a “low fat” label as long as the label meets certain specified criteria.  The rule takes effect on January 1, 2012. 

    In August, FSIS issued a notice, informing Inspection Program Personnel of the upcoming implementation date of this rule and instructing the Inspection Personnel to make plant management aware of the final rule and the implementation date of January 1, 2012.  In further efforts to prepare industry for the implementation of this rule, FSIS announced a series of webinars regarding the implementation of the rule. 

    FSIS’s website includes a presentation that gives an overview of the nutrition labeling final rule and its requirements, and answers to questions that were submitted via askFSIS. 

    The Feds Challenge Mobile Medical Apps

    By Carmelina G. Allis

    If you thought you’ve had enough trying to figure out whether FDA regulates your mobile app, watch out for the FTC.  The FTC has brought its first case targeting health claims related to mobile medical apps.

    The mobile apps, sold in Apple’s iTunes Store and Google’s Android Marketplace under the names “AcneApp” and “Acne Pwner,” claimed that they could treat acne with colored lights emitted from the mobile devices.  According to the FTC, the marketers alleged that the blue and red lights emitted by the apps kill acne-causing bacteria.  The apps advised consumers to hold the display screen next to the area of skin to be treated while the app was activated.

    The FTC alleged that the acne treatment claims made for both apps were unsubstantiated.  The marketers of the apps have agreed to stop making claims about the products in order to settle charges with the FTC, which would bar them from making certain health-related claims without supporting scientific evidence.

    The market for mobile medical apps appears to have grown exponentially recently, and one can only wonder whether FTC’s interest in this area will also intensify.  If you worry about FDA, enforcement actions may not be at the top of its agenda quite yet; the FDA regulatory landscape for these products, although somewhat defined, is still being carved out.  We previously blogged that FDA has proposed to exert regulatory authority over select mobile medical apps that meet the “device” definition in the Federal Food, Drug, and Cosmetic Act, and that are either used as an accessory to a regulated medical device, or transform a mobile platform into a regulated medical device.  The FDA is now seeking public input on that proposal, and it is holding a public workshop on mobile medical apps on September 12-13, 2011.

    Is a Bioengineered Food “100% Natural”?

    By Ricardo Carvajal

    That question is presented in class actions recently filed against ConAgra Foods, Inc. in California and New York.  The complaint in the California case alleges that ConAgra’s labeling and advertising of Wesson Oils as “100% natural” violates California law because the oils are “derived from plants grown from GMO seeds,” and such plants are not “natural.”  In support of their position, plaintiffs cite Monsanto Company’s definition of “genetically modified organism (GMO)” (“Plants or animals that have had their genetic makeup altered to exhibit traits that are not naturally theirs…”), as well as the World Health Organization’s definition of that term (“organisms in which the genetic material (DNA) has been altered in a way that does not occur naturally…”).  The complaint in the New York case includes similar allegations.  We note that FDA disfavors use of the term “genetically modified organism,” preferring instead the term “bioengineered foods” to describe foods derived from plant varieties that are developed using rDNA technology.

    Surprisingly, FDA does not appear to have publicly addressed the question of whether a bioengineered food can properly be labeled as “natural” under the agency’s policy on the use of that term (FDA interprets “natural” to mean that nothing artificial or synthetic (including colors regardless of source) is included in, or has been added to, the product that would not normally be expected to be there).  For its part, ConAgra maintains that “[p]laintiff’s claims do not depend on the FDA’s definition of ‘natural.’”  ConAgra contends in part that, under FDA’s regulatory framework for bioengineered foods, “[n]o special labeling requirements apply to foods made from bioengineered plants because those foods are not meaningfully different from other foods.  If a label is appropriate for food made from plants developed by older methods of genetic selection, then that same label – with each of the representations it makes, whether about the name of the product, the ingredients it contains or whether it is natural, artificial or an imitation – is appropriate for the food made from bioengineered plants too.”  

    The broader issue of GMO labeling could gain greater visibility as the result of an upcoming 2-week march on Washington, DC planned by a number of consumer groups, businesses, and trade associations who assert that bioengineered foods should be labeled as such.  The marchers’ desired outcome would require new federal legislation – a doubtful prospect heading into 2012.

    HHS Issues Final Conflict of Interest Rules for PHS-Funded Research That Increases Burden to Institutions and Investigators

    By Nisha P. Shah

    The U.S. Department of Health and Human Services recently revised conflict of interest rules for Public Health Service ("PHS") related grants and research (i.e., the National Institutes of Health (“NIH”)) in response to congressional and public concerns over potential conflicts between investigators and companies, such as drug and medical device manufacturers.  The final rules implement changes to regulations issued in 1995 on the Responsibility of Applicants for Promoting Objectivity in Research for which Public Health Service Funding is South and Responsible Prospective Contractors (42 C.F.R. Part 50 and 45 C.F.R. Part 94).  Below are some of the highlights of the final rules that will result in increased monitoring, disclosure, and reporting requirements for institutions and investigators.

    Definitions (42 C.F.R. § 50.603, 45 C.F.R. § 94.3)

    The most significant changes to the definitions are the changes to the definition of significant financial interest ("SFI") and the inclusion of a definition of financial conflict of interest ("FCOI").

    • For SFI, the minimum threshold decreases from $10,000 to $5,000 for payments and/or equity interests, including non-publicly traded entities, related to their institutional responsibilities.  SFI also now includes certain intellectual property rights and interests and most reimbursed or sponsored travel.  Under the final rules, SFI excludes income from investment vehicles and income from seminars, lectures, or teaching, and service on advisory or review panels for government agencies, institutions of higher education, academic teaching hospitals, medical centers, or research institutes affiliated with an institution of higher education.
    • An FCOI is an SFI that could directly and significantly affect the design, conduct, or reporting of PHS-funded research.  In this context, HHS explained that “significantly” means that the financial interest would have a material effect on the research.

    Responsibilities of Institutions Regarding Investigator Financial Conflicts of Interest (42 C.F.R. § 50.604, 45 C.F.R. § 94.4)

    • While the 1995 regulations required an institution to maintain a conflict of interest policy, the new rules add that an institution must make such policy available via a publicly accessible website.  If an institution does not have a presence on a website, the institution must make the policy available in writing within 5 business days of any request.
    • The 1995 rules did not have a training requirement; the new rules require each institution to train each investigator on the institution’s conflicts of interest policy and the new regulations prior to engaging in research related to any PHS-funded grant or contract, at least every 4 years, and under specific circumstances, such as when the investigator is new to an institution. 
    • Each investigator who is planning to participate in the PHS-funded research must disclose to the institution the investigator’s SFI (and those of the investigator’s spouse and dependent children) no later than the date of submission of the institution’s proposal to the NIH, and thereafter, the investigator must update disclosure within 30 days any newly discovered or acquired SFI and at least annually of any unreported SFI. 

    Management and Reporting of Financial Conflicts of Interest (42 C.F.R. § 50.605, 45 C.F.R. § 94.5)

    • While the 1995 rule did not require public disclosure of SFI, the new rules now require an institution to either post on a publicly accessible website or make available in a written report within 5 days of a request information concerning any SFI disclosed to an institution that meets the following 3 criteria: (1) the SFI was disclosed and is still held by senior/key personnel; (2) the institution determines that the SFI is related to the PHS-funded research; and (3) the institution determines that the SFI is a FCOI.  Information that must be made publicly available include (but is not limited to): the investigator’s name, the investigator’s title and role regarding the research project, the nature of the SFI, and the approximate dollar value of the SFI (ranges are permissible).  The institution must update this information at least annually and within 60 days of the institution’s receipt or identification of information concerning any additional SFI.
    • While institutions implemented a “management plan” under the 1995 rules, the final rules now require that an institution must review all investigator disclosures of SFIs, determine whether a SFI is a FCOI, and, if so, develop and implement a management plan that specifies the actions that will be taken to manage such FCOI.  Examples of actions under a management plan include: public disclosure of FCOI when presenting or publishing research, disclosure of FCOI to participants in a research project involving human subjects, modification of the research plan, and change of personnel or personnel responsibilities.
    • The number of requirement elements in the FCOI report submitted to the NIH has increased and now includes (but is not limited to): the name of the investigator with FCOI, the nature of the financial interest, the value of the financial interest, a description of how the financial interest relates to the PHS-funded research, and a description of the key elements of the institution’s management plan.
    • If an investigator does not disclose a SFI in a timely manner, the institution must implement a management plan within 60 days and within 120 days must complete and document a retrospective review as to whether any portion of the PHS-funded research was biased.  If bias is found, the institution must notify and submit a mitigation report to the NIH.

    The effective date of the final rule is September 26, 2011, and the compliance date is no later than August 24, 2012 and immediately upon making its FCOI policy publicly accessible.

    Citing Imminent Hazard to Public Safety, DEA Publishes Notice of Intent to Temporarily Places Synthetic Cathinones Into Schedule I of the CSA

    By Karla L. Palmer & John A. Gilbert

    On September 8, 2011, the Drug Enforcement Administration (“DEA”) published a Notice of Intent to temporarily place into Schedule I of the Federal Controlled Substances Act (“CSA”) three synthetic cathinones.  75 Fed. Reg. 55616 (Sept. 8, 2011).  The action is based on a finding by the DEA Administrator that the substances mephedrone (methylcathinone), methylone (3,4-methylenedioxy-N-methylcathinone), and MDPV (3,4-methylenedioxypyrovalerone) are an imminent hazard to the public safety.

    As reported here in December 2010 and March 2011, DEA has authority (as delegated by the U.S. Attorney General) to temporarily place a substance into Schedule I of the CSA for a one-year period (subject to a six-month extension) without having to comply with the usual scheduling requirements under 21 U.S.C. § 811(b) if the agency makes a finding that such action necessary to avoid imminent hazard to the public health.  DEA last invoked its temporary scheduling authority in December 2010 when it published a notice of intent to temporarily schedule five synthetic cannabinoids in Schedule I, and finalized that temporary scheduling in March 2011.  Prior to its emergency action concerning synthetic cannabinoids, DEA last invoked its temporary scheduling authority in 2004.

    As described in yesterday’s Notice of Intent, in order to temporarily place a substance in Schedule I of the CSA to avoid an imminent hazard to the public safety, the DEA Administrator is required to consider three of the eight factors set forth in 21 U.S.C. § 811(c).  In this case, the Administrator considered the following three factors: (4) the substances’ history and current pattern of abuse; (5) the scope, duration and significance of abuse; and, (6) what, if any, risk each poses to the public health.  DEA explained that  consideration of these factors also “includes actual abuse, diversion from legitimate channels, and clandestine importation, manufacture, or distribution.” (Citing 21 U.S.C. § 811(h)(3)).

    With respect to the first factor that the Administrator considered – the abuse history and current pattern of abuse – the pharmacological effects of synthetic cathinones are similar to those of highly abused products such as methamphetamine, cathinone, methcathinone and MDMA.  They cause, among other effects, agitation, tachycardia, dilated pupils, hyperthermia, sweating, and hypertension.  Thus, the abuse of synthetic cathinones is likely similar to those highly abused substances.  The DEA also noted that the three substances identified by DEA in its Notice of Intent represent more than 98% of the reported synthetic cathinones recently seized by law enforcement.

    As to the second factor that it considered – the scope, duration and significance of abuse – the DEA cited to the recent emergence of these substances on the U.S. illicit drug market, yet noted that the products have been popular in Europe since 2007.  The three products (marketed as bath salts, plant food, or research chemicals; and labeled “not for human consumption”) are typically sold in smoke shops, head shops, adult book stores,  gas stations, and are routinely purchased via the internet.  The DEA noted that retailers promote that routine drug tests will not detect their presence in the body.  Surveys and other research also indicates that the substances are being widely abused, marketed to teens and young adults, and sold for their psychoactive properties – as alternatives to stimulants like cocaine and MDMA.  The DEA Notice of Intent describes that the methods of administration of the illicit substances include snorting, swallowing, and “bombing” (i.e., wrapping powder in a paper wrapper and swallowing), and are also reported to be used as  “binge” products.

    Citing their rampant increase in popularity since these synthetic products appeared on the U.S. drug market scene back in 2009, DEA states that poison control centers have already received 4,137 calls relating to synthetic cathinones in the first seven months of 2011; which is up from the 303 calls received for such substances in all of 2010.  In addition, the U.S. Customs and Border Patrol has encountered at least 96 shipments of synthetic cathinones at one border; most of which originated from India or China and are being shipped throughout the United States.  Due to the potential for abuse, as of July 2011, at least 33 states have emergency scheduled or taken action to control synthetic cathinones.  All members of the European Union have placed controls on the possession or sale of them as well.

    As to the required third factor in DEA’s consideration – the risk to the public health — the DEA stated that these substances have been the subject of serious abuse, and are associated with numerous emergency room admissions.  Given the frequent reports of adverse and significant health incidents (including three deaths) associated with the use of these substances, their high potential for abuse, and because there is no acceptable medical use in treatment in the United States for the substances, the DEA found it necessary to provide the requisite 30-day notice that the substances are subject to  expedited temporary scheduling as set forth in 21U.S.C. § 811(h).  The DEA Administrator will issue a final order temporarily scheduling the three substances into Schedule I upon the expiration of the 30-day notice period, or after October 11, 2011.  The temporary scheduling is effective for up to 18 months pending the DEA’s completion of the scheduling process.  After the expiration of the 30-day notice period, MDPV, mephedrone and methylone will be subject to the “regulatory controls and administrative, civil  and criminal sanctions applicable to the manufacture, distribution, possession, importing and exporting of a Schedule I controlled substance under the CSA.”

    Last-Ditch Effort to Jettison “The Dog Ate My Homework Act” from the America Invents Act Fails

    By Kurt R. Karst –      

    On September 8th, the U.S. Senate passed, by an 89-9 vote, H.R. 1249, the Leahy-Smith America Invents Act.  Final passage of the bill, which will make significant changes to the U.S. patent system, was preceded by a contentious vote on, you guessed it, Section 37 (often referred to as “The Dog Ate My Homework Act” or the “Medco fix”), which would legislatively resolve The Medicines Company’s (“MDCO’s”) decade-long battle to obtain a Patent Term Extension (“PTE”) for U.S. Patent No. 5,196,404 covering ANGIOMAX (bivalirudin). 

    As we previously reported (here, here, and here), as the prospects of patent reform grew over the past few months (after the House passage of H.R. 1249), so too did lobbying efforts to ensure inclusion of Section 37 in the Senate-passed bill.  Those efforts took the form of portraying Section 37 as a law of general applicability affecting multiple companies and products (3 that we know of, not including ANGIOMAX) rather than as single company (including a law firm and its malpractice insurer) legislation. 

    That portrayal rankled some Senators.  Earlier this week, Senators Jeff Sessions (R-AL), Joe Manchin (D-WV), Tom Coburn (R-OK), and Mike Lee (R-UT) proposed Senate Amendment 600, the text of which states: “On page 149, line 20, strike all through page 150, line 16.”  Those line references are to entire Section 37 of the House-passed H.R. 1249, which states:

    SEC. 37. CALCULATION OF 60-DAY PERIOD FOR APPLICATION OF PATENT TERM EXTENSION.

    (a) IN GENERAL.—Section 156(d)(1) of title 35, United States Code, is amended by adding at the end the following flush sentence:

    ‘‘For purposes of determining the date on which a product receives permission under the second sentence of this paragraph, if such permission is transmitted after 4:30 P.M., Eastern Time, on a business day, or is transmitted on a day that is not a business day, the product shall be deemed to receive such permission on the next business day. For purposes of the preceding sentence, the term ‘business day’ means any Monday, Tuesday, Wednesday, Thursday, or Friday, excluding any legal holiday under section 6103 of title 5.’’.

    (b) APPLICABILITY.—The amendment made by subsection (a) shall apply to any application for extension of a patent term under section 156 of title 35, United States  Code, that is pending on, that is filed after, or as to which a decision regarding the application is subject to judicial review on, the date of the enactment of this Act.

    Senators Sessions and Coburn urged the Senate in a September 7th “Dear Colleague Letter” to support passage of their amendment, stating:

    While many of us had supported the Senate version of H.R. 1249, the  “Leahy-Smith America Invents Act,” the House version contains a controversial special interest provision (Section 37) that was added via amendment during a confusing and highly-unusual floor debate. Initially, the amendment failed by a vote of 209 to 208.  When some Members objected that they had not been able to vote, the original vote was vacated.  The second time around, the amendment was adopted by 19 votes.  We believe this apparent confusion is attributable to the fact that Section 37, or the “Medco fix,” looks like a benign, technical change to an obscure section of the U.S. Code. But it is actually an illustration of Washington at its worst – a bailout for a well-connected, big law firm and its malpractice insurer and a brand pharmaceutical manufacturer, which have hired an army of lobbyists. The provision is a special interest matter which is currently in litigation and cannot be justified.

    The letter goes on to provide several reasons why the Senate should vote to strike Section 37: “First, this case is in active litigation. . . .  Second, this provision addresses a problem that does not exist. . . .  Third, if this language becomes law, it would permit Medco to delay the launch of a generic version of Angiomax by an additional five years (2015). . . .  Finally, Congress has a private relief process for dealing with cases such as this.” 

    Despite these pleas, the Senate rejected, by a 51-47 vote, Senate Amendment 600, paving the way for a final vote on, and passage of, H.R. 1249.  (The September 8th Senate floor debate on Senate Amendment 600 is available here.)  If President Obama signs the Leahy-Smith America Invents Act into law, the ongoing battle in the U.S. Court of Appeals for the Federal Circuit will presumably be mooted, and the last chapter in this story may very well have been written.

    As we say goodbye (or is it “hello”?) to “The Dog Ate My Homework Act” (although there may still be more interesting things to come on this drug), we leave you with a list of all the FDA Law Blog postings on this topic (a blogography of sorts).  Queue up Green Day’s Time Of Your Life:

    FDA Grants 60-Day Extension to Comment on NDI Guidance

    On September 9, 2011, FDA will publish in the Federal Register a notice granting a 60-day extension, until December 2, 2011, to file comments on the draft guidance on New Dietary Ingredient (“NDI”) notifications that the Agency issued on July 5, 2011 (see our previous post on the draft guidance here).  In late July, Hyman, Phelps & McNamara, P.C. filed a request for a one-year comment period to allow affected businesses adequate time to respond to the lengthy and controversial draft guidance (see our previous post here).

    NORD Petition Requests FDA Policy Statement on Orphan Drug Review Flexibility

    By Kurt R. Karst –      

    The National Organization for Rare Disorders (“NORD”) announced the submission of a Citizen Petition to FDA  requesting “that a documented policy be established regarding the review of potential treatments for people with rare diseases.”  The petition comes on the heels of a Report to Congress FDA issued earlier this year and required by Section 740 of the Agriculture, Rural Development, Food and Drug Administration, and Related Agencies Appropriations Act of 2010 (Public Law No. 111-80) (see our previous post here), in which the FDA reports on the findings and recommendations of two Agency groups to improve the current regulatory/scientific armamentarium to facilitate the development of products for rare and neglected diseases. 

    In addition to the Report to Congress, Section 740 of the Appropriations Act also requires FDA to “issue, not later than 180 days after submission of the report to Congress . . . . guidance based on such recommendations for articles for use in the prevention, diagnosis, and treatment of rare diseases and for such uses in neglected diseases of the developing world,” and to “develop, not later than 180 days after submission of the report to Congress . . . internal review standards based on such recommendations for articles for use in the prevention, diagnosis, and treatment of rare diseases and for such uses in neglected diseases of the developing world.”  “Because the report was issued to Congress June 27, 2011, this guidance document is due to be issued no later than December 27, 2011,” says the NORD petition. 

    NORD requests that FDA’s guidance explicitly include:

    • Acknowledgement that the conduct of clinical trials for most orphan drugs is qualitatively and quantitatively different from the conduct of trials for drugs that treat common conditions.
    • Acknowledgement that FDA review of marketing applications for most orphan drugs is accordingly qualitatively and quantitatively different from FDA review of applications for articles that treat common conditions.
    • In recognition of the above, and notwithstanding the unchanged requirements that articles for rare diseases must demonstrate both efficacy and safety, we request a statement that it will now be FDA official policy to afford special flexibility to the regulatory review of submissions for all orphan drugs.

    “This petition does not request any itemization of past actions – rather, through the language of forthcoming guidance, we request the establishment of a policy to direct future actions,” says NORD in the Citizen Petition.

    In addition, NORD requests that FDA “incorporate mandatory training in this new policy and other matters related to orphan drug development for all full-time FDA review professionals,” and that this training become a requirement of FDA’s core curriculum.  As we recently reported, the Proposed PDUFA V Reauthorization Performance Goals and Procedures for Fiscal Years 2013 through 2017 includes an initiative, styled as “Advancing Development of Drugs for Rare Diseases,” that would require FDA to, among other things, “develop and implement staff training related to development, review, and approval of drugs for rare diseases.  The training will be provided to all CDER and CBER review staff, and will be part of the reviewer training core curriculum.”

    In addition to the Report to Congress, NORD states in the petition that the organization’s requests are consistent with the recommendations of a recent finding of the Institute of Medicine, which released a report in October 2010, titled “Rare Diseases and Orphan Products: Accelerating Research and Development,” and with the testimony delivered by Hyman, Phelps & McNamara, P.C. Director and Chair of the Board of NORD Frank J. Sasinowski at a June 2010 FDA Part 15 hearing. 

    New Twist In “Natural” Lawsuit

    By Riëtte van Laack

    On August 24, 2011, another class action suit regarding “natural” food labeling claims was filed in the Southern District of California.  The defendants include Kashi Co. (“Kashi”) and its general manager, as well as Kashi’s parent Kellogg Co. and its President (the complaint also threatens to sweep in numerous other defendants, potentially including certain suppliers).  Plaintiffs, a national class of consumers who purchased Kashi products, claim defendants falsely and misleadingly labeled virtually all Kashi products as “all natural” or containing nothing artificial even though the products allegedly do not conform to applicable federal regulations and policies on “natural.”  The federal regulations and policies referenced in the complaint include the National Organic Program’s ("NOP’s") regulatory definition of “synthetic,” an inaccurate statement of FDA’s policy on “natural,” USDA’s policy on “natural,” and FDA’s regulatory definition of artificial flavor.  Plaintiffs further allege that the products in question do not conform to Kashi’s own definition of “natural,” which states:

    Natural food is made without artificial ingredients like colors, flavors or preservatives and is minimally processed.

    A natural ingredient is one that comes from or is made from a renewable resource found in nature.

    Minimal processing involves only kitchen chemistry, processes that can be done in a family kitchen and does not negatively impact the purity of the natural ingredients.

    Based on the referenced regulations and policies, as well as Kashi’s definition of “natural,” plaintiffs assert that a reasonable consumer understands the term “natural” to mean than none of the ingredients are synthetic or artificial, and none of the ingredients have undergone excessive processing.

    In what appears to be a new twist, plaintiffs draw on manufacturing and other information from various sources to argue that Kashi’s products contain a number of “unnatural substances,” including but not limited to bromelain, plant sterols, potassium bicarbonate, xantham gum, yeast extract, inulin, cholecalciferol, ascorbic acid, calcium caseinate, lactic acid,  high oleic safflower oil, and a number of soy products.  The manufacturing information is drawn from NOP regulations (primarily 7 C.F.R. § 202.605(b)), a variety of food additive approval and GRAS listing regulations, the National Library of Medicine’s Hazardous Substances Data Bank, and the Environmental Protection Agency’s pesticide and hazardous substance regulations, among other sources.

    Plaintiffs allege that they suffered not just economic injury, but also ingestion of “a substance that is generally harmful to their health, their children’s health, or their unborn fetus’s health,” and being “forced to unwittingly support an industry that contributes to environmental, ecological, or health damage.”  Plaintiffs allege numerous causes of action, including unfair business practices, false advertising, breach of express and implied warranty, negligent misrepresentation, and even assault and battery.  They request damages, statutory penalties, restitution, punitive damages, attorneys’ fees and expenses, and costs, and an injunction.

    FDA Issues Proposed PDUFA V Performance Goals & Procedures

    By Kurt R. Karst –      

    Last week, FDA issued its highly anticipated Proposed PDUFA V Reauthorization Performance Goals and Procedures for Fiscal Years 2013 through 2017, which were hammered out between FDA and industry after long negotiations.  The proposal includes several new provisions compared to the PDUFA IV Reauthorization Performance Goals.  We’ll focus on a couple of them here. 

    Although many of the goals for priority and standard NDA and BLA review (including Class 1 and Class 2 resubmissions, efficacy supplements and manufacturing supplements) remain the same as under PDUFA IV, the proposed PDUFA V agreement establishes a new review model that will apply to all New Molecular Entity (“NME”) NDAs and original BLAs received from October 1, 2012, through September 30, 2017, including applications that are resubmitted following a refuse-to-file action.  The new review model, referred to as “the Program” in the PDUFA V proposal, is intended to “promote greater transparency and improve communication between the FDA review team and the applicant” and to “improve the efficiency and effectiveness of the first cycle review process and decrease the number of review cycles necessary for approval, ensuring that patients have timely access to safe, effective, and high quality new drugs and biologics.”

    The parameters of the Program include, among other things, a pre-submission meeting (which the agreement says is “strongly encouraged”), a mid-cycle communication “to provide the applicant with an update on the status of the review of their application,” and a late-cycle meeting at which the FDA review team and the applicant will discuss the status of the review of the application.  At the pre-submission meeting, “FDA and the applicant will agree on the content of a complete application for the proposed indication(s), including preliminary discussions on the need for risk evaluation and mitigation strategies (REMS) or other risk management actions.”  In addition, “FDA and the applicant may also reach agreement on submission of a limited number of application components not later than 30 calendar days after the submission of the original application,” such as the submission of updated stability data.  The proposed agreement cautions that “[i]f the applicant does not have a pre-NDA/BLA meeting with FDA, and no agreement exists between FDA and the applicant on the contents of a complete application or delayed submission of certain components of the application, the applicant’s submission is expected to be complete at the time of original submission.”

    As part of the Program, priority and standard NME NDAs and original BLA will be subject to different review goals as compared to other applications.  As shown in the table below, the proposed agreement says that FDA will review and act on 90% of standard NME NDA and original BLA submissions within 10 months of the 60 day filing date, and 90% of priority NME NDA and original BLA submissions within 6 months of the 60 day filing date.  Use of the filing date instead of the receipt date essentially means that the reviews are slated to take place within 12 months (priority) and 8 months (standard) from submission.

    PDUFAVTable 

    Another change of note is the treatment of “major amendments” to pending applications.  Under PDUFA IV (and FDA’s regulations at 21 C.F.R. § 314.60), “[a] major amendment to an original application, efficacy supplement, or resubmission of any of these applications, submitted within three months of a goal date, may extend the goal date by three months.  A major amendment to a manufacturing supplement submitted within two months of the goal date extends the goal date by two months” (emphasis added).  Under the proposed PDUFA V agreement, however, “[a] major amendment to an original application, efficacy supplement, or resubmission of any of these applications, submitted at any time during the review cycle, may extend the goal date by three months” (emphasis added), and “[a] major amendment to a manufacturing supplement submitted at any time during the review cycle may extend the goal date by two months” (emphasis added).  The proposed PDUFA V agreement says that a major amendment may include, among other things, “submission of a REMS with [elements to assure safe use (ETASU)] not included in the original application; or significant amendment to a previously submitted REMS with ETASU;” however, “changes to REMS that do not include ETASU and minor changes to REMS with ETASU will not be considered major amendments.”

    The PDUFA V proposal includes several items intended to enhance regulatory science and expedite drug development, including: (1) initiatives to enhance FDA-sponsor communications (e.g., the agreement says that “FDA will develop a dedicated drug development communication and training staff within the Office of New Drugs in CDER and augment the manufacturers assistance staff in CBER”); (2) approaches and methods for the conduct of meta-analyses; (3) advancing the use of biomarkers and pharmacogenomics; and (4) advancing the development of patient-reported outcomes and other endpoint assessment tools. 

    One initiative, styled as “Advancing Development of Drugs for Rare Diseases,” appears to show the growing importance of orphan drugs.  According to the proposal:

    • By the end of FY 2013, FDA will complete a staffing and implementation plan for the CDER Rare Disease Program within the Office of New Drugs and a CBER Rare Disease liaison within the Office of Center Director.
    • FDA will increase by five the staff of the CDER Rare Disease Program and establish and fill the CBER Rare Disease liaison position.
    • On an ongoing basis, the staff in the Rare Disease Programs of the two Centers will develop and disseminate guidance and policy related to advancing and facilitating the development of drugs and biologics for rare diseases, including improving understanding among FDA reviewers of approaches to studying such drugs; considering non-traditional clinical development programs, study design, endpoints, and statistical analysis; recognizing particular challenges with post-market studies; and encouraging flexibility and scientific judgment, as appropriate, on the part of reviewers when evaluating investigational studies and marketing applications for drugs for rare diseases.
    • By mid-FY 2014, FDA, through the Rare Disease Program, will conduct a public meeting to discuss complex issues in clinical trials for studying drugs for rare diseases, including such questions as endpoint selection, use of surrogate endpoints/Accelerated Approval, and clinical significance of primary endpoints; reasonable safety exposures; assessment of dose selection; and development of patient-reported outcome instruments.
    • By the end of FY 2015, FDA will develop and implement staff training related to development, review, and approval of drugs for rare diseases. The training will be provided to all CDER and CBER review staff, and will be part of the reviewer training core curriculum.
    • By the end of FY 2016, FDA, through the Rare Disease Program, will develop an evaluation tool to evaluate the success of the activities of the Rare Disease Program, including the reviewer training.

    Peter L. Saltonstall, president and CEO of the National Organization for Rare Disorders, applauded the PDUFA V proposal, saying that “[t]he document reflects a clear recognition that drugs for rare diseases warrant special consideration and special staff training.”

    Additional Reading: