• where experts go to learn about FDA
  • Orphan Drugs: The Current Firestorm, a Real Evergreening Issue, and a Possible Solution

    Periodically, legislators and others become concerned about reports citing the high price of some orphan drugs, including drugs that achieve blockbuster status (earning more than $1 billion a year). Several proposals have been introduced in response to such concerns.  In 1990, Congress passed legislation that would have limited market exclusivity in some circumstances, but the President vetoed it.

    ***

    Critics in Congress and in the pharmaceutical industry and patient groups say that while the [Orphan Drug Act] has generally worked, it has proved to be a bonanza for the makers of some very big drugs, allowing them to charge higher prices than there would have been with competition.

    With all of the recent hubbub around orphan drugs and pricing, you might think the two quotes above were ripped from recent stories. In fact, the first quote is taken from a 2010 Institute of Medicine report, titled “Rare Diseases and Orphan Products: Accelerating Research and Development” (see our previous post here).  The second quote is from an April 1990 article in the New York Times, titled “Orphan Drug Law Spurs Debate.”  The fact that you likely could not identify the age of the quotes above means that we’re in the midst of another one of those “periods” referred to in the IOM report, where legislators take a look at the Orphan Drug Act to decide whether or not changes need to me made to the law.

    The latest round of interest started perhaps within the past two years, as legislators began consideration of legislation – the “Orphan Product Extensions Now Act,” or “OPEN Act” – to amend the FDC Act to provide a 6-month extension of exclusivity periods for companies that obtain approval of a previously approved drug for a new, rare condition. (By the by, the OPEN Act was reintroduced in February as H.R. 1223, the “Orphan Product Extensions Now Accelerating Cures and Treatments Act.”)  Then there was an article in the American Journal of Clinical Oncology, titled “The Orphan Drug Act: Restoring the Mission to Rare Diseases,” alleging that companies are “gaming” the orphan drug system established by the Orphan Drug Act “to use the law for mainstream drugs.”  A report from Public Citizen on the OPEN Act, titled “House Orphan Drug Proposal: A Windfall for Pharma, False ‘Cure’ for Patients” (see our previous post here), followed.  Then things calmed down a bit . . . .  until recently.

    In January 2017, Kaiser Health News published a report, titled “The Orphan Drug Machine: Drugmakers Manipulate Orphan Drug Rules To Create Prized Monopolies.”  That report caught the attention of Senator Chuck Grassley (R-IA), who stated  that he would explore possible misuses of the orphan drug program.  Then last week, Sen. Grassley (along with Senators Orrin Hatch (R-UT) and Tom Cotton (R-AR)) sent a letter to the Government Accountability Office (“GAO”) requesting certain information (much of which is already publicly available) and an investigation into “whether the [Orphan Drug Act] is still incentivizing product development for diseases with fewer than 200,000 affected individuals, as intended, and provide any regulatory or legislative changes that may be needed in order to preserve the intent of this vital law.”  The letter to GAO states the Senators’ general concern: so-called “evergreening” of orphan drug exclusivity.

    While few will argue against the importance of the development of [orphan] drugs, several recent press reports suggest that some pharmaceutical manufacturers might be taking advantage of the multiple designation allowance in the orphan drug approval process.

    A review of FDA’s Orphan Drug Designations and Approvals Database shows that there are many, many drugs and biological products with multiple orphan drug designations and/or approvals.  In some cases, there are just a couple of entries on the list for the same drug.  In other cases, such as with Imatinib and Ibrutinib, there are quite a few entries.

    In most cases, a single period of 7-year orphan drug exclusivity extends from a single orphan drug designation granted by FDA’s Office of Orphan Products Development. Each designation covers a different orphan disease or condition.  And once the first period of orphan drug exclusivity expires, FDA may be able to approve an ANDA for a generic version of the drug product with labeling that omits information on a subsequent use protected by orphan drug exclusivity.  This carve-out option has been affirmed by FDA in various Letter Decisions and Citizen Petition response, and by the courts – see, e.g., Sigma-Tau Pharmaceuticals, Inc. v. Schwetz, 288 F.3d 141 (4th Cir. 2002) (here).

    If the reference in the letter to the GAO to “multiple designation allowance” that “some pharmaceutical manufacturers might be taking advantage of” is merely a concern with multiple orphan drug designations that lead to separate grants of orphan drug exclusivity for separate diseases or conditions, then this blogger does not see a particular need for concern. It’s not an evergreening issue at all!  The Orphan Drug Act is working exactly as intended, and generic competition is generally not thwarted because of the ability of an ANDA applicant to carve-out of its labeling (and thus avoid) a period of unexpired orphan drug exclusivity on the brand-name Reference Listed Drug.

    But there may be a real evergreening issue that’s probably been overlooked by most folks. In some cases, a single orphan drug designation can result in multiple periods of orphan drug exclusivity.  (A table of examples is provided at the end of this post.)  FDA explained this concept in the preamble to the Agency’s October 2011 proposed orphan drug regulations:

    The scope of orphan exclusive approval for a designated drug is limited to the approved indication or use, even if the underlying orphan designation is broader. If the sponsor who originally obtained orphan exclusive approval of the drug for only a subset of the orphan disease or condition for which the drug was designated subsequently obtains approval of the drug for one or more additional subsets of that orphan disease or condition, FDA will recognize orphan-drug exclusive approval, as appropriate, for those additional subsets from the date of such additional marketing approval(s).  Before obtaining such additional marketing approval(s), the sponsor in this instance would not need to have obtained additional orphan designation for the additional subset(s) of the orphan disease or condition. [(Emphasis added)]

    In most instances, multiple and staggered periods of orphan drug exclusivity stemming from the same designation do not stymie generic competition. For example, if FDA grants an orphan drug designation for Drug X for Disease Y and the sponsor first obtains approval of the drug for use in adults with Disease Y and then later for the same drug for use in children with Disease Y, FDA would grant two separate periods of orphan drug exclusivity – one for each approval.  An ANDA applicant may obtain approval of the drug for the adult population indication once the initial period of orphan drug exclusivity expires, and then later for the pediatric population indication once that second period of orphan drug exclusivity expires.

    But not all cases are as easy as the one above. You see, indications, like Pokémon, can evolve into something new.  There appear to be a growing number of cases where FDA has granted multiple periods of orphan drug exclusivity based on the same original orphan drug designation, and where the drug’s indication evolves into something new, shedding and subsuming the previous indication statement.  This could occur, for example, as different disease stages or different lines of therapy are approved.  (Some possible examples of this might be in the cases of Ibrutinib, Cinacalcet, Bortezomib, and Bevacizumab.)  As the old labeling is shed, the new labeling may not allow for an ANDA (or biosimilar) applicant to easily (if at all) omit information protected by a new 7-year period of orphan drug exclusivity.

    But is the solution to what may be a real evergreening problem opening up the Orphan Drug Act? This blogger thinks that there could be a better solution.  If the issue preventing a carve-out is the text of the brand-name drug labeling, then one remedy is to have better communication between the Office of New Drugs (“OND”) and the Office of Generic Drugs (“OGD”) during the course of brand-name drug labeling reviews and drafting.  OGD’s experience with labeling reviews and carve-outs should not be overlooked, and can lead to labeling that does not cause carve-out controversies years down the road.  Another possible remedy is for OGD to take a broader view of permissible labeling changes.  That is, considering so-called labeling “carve-ins” that clarify the omission of other labeling information (and effectively return an indication to its previous state).  It’s a topic FDA raised a few years back (see our previous post here), but that the Agency ultimately decided not to address.

    Multiple Orphan Drug Exclusivity Periods Based on a Single Orphan Drug Designation

    Generic Name (Trade Name)Designation (Designation Date)Approved IndicationMarketing Approval Date (Exclusivity End Date)
    adalimumab (Humira)Treatment of juvenile rheumatoid arthritis (3/21/2005)Reducing signs and symptoms of moderately to severely active polyarticular juvenile idiopathic arthritis in patients 4 years of age and older. 02/21/2008 (02/21/2015)
    adalimumab (Humira)Treatment of juvenile rheumatoid arthritis (3/21/2005)Reducing signs and symptoms of moderately to severely active polyarticular juvenile idiopathic arthritis in patients 2 years to 4 years of age. 09/30/2014  (09/30/2021)
    bevacizumab (Avastin)Treatment of fallopian tube carcinoma (11/23/2010)Either in combination with carboplatin and paclitaxel or in combination with carboplatin and gemcitabine, followed by Avastin as a single agent, is indicated for the treatment of patients with platinum-sensitive recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer. 12/06/2016  (12/06/2023)
    bevacizumab (Avastin)Treatment of fallopian tube carcinoma (11/23/2010)In combination with paclitaxel, pegylated liposomal doxorubicin, or topotecan for treatment of patients with platinum-resistant, recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer who have received no more than 2 prior chemotherapy regimens. 11/14/2014  (11/14/2021)
    bevacizumab (Avastin)Treatment of primary peritoneal carcinoma (11/2/2010)Either in combination with carboplatin and paclitaxel or in combination with carboplatin and gemcitabine, followed by Avastin as a single agent, is indicated for the treatment of patients with platinum-sensitive recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer. 12/06/2016  (12/06/2023)
    bevacizumab (Avastin)Treatment of primary peritoneal carcinoma (11/2/2010)In combination with paclitaxel, pegylated liposomal doxorubicin, or topotecan for treatment of patients with platinum-resistant, recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer who received no more than 2 prior chemotherapy regimens 11/14/2014  (11/14/2021)
    bevacizumab (Avastin)Therapeutic treatment of patients with ovarian cancer (2/9/2006)Either in combination with carboplatin and paclitaxel or in combination with carboplatin and gemcitabine, followed by Avastin as a single agent, is indicated for the treatment of patients with platinum-sensitive recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer. 12/06/2016  (12/06/2023)
    bevacizumab (Avastin)Therapeutic treatment of patients with ovarian cancer (2/9/2006)In combination with paclitaxel, pegylated liposomal doxorubicin, or topotecan for treatment of patients with platinum-resistant, recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer who received no more than 2 prior chemotherapy regimens 11/14/2014  (11/14/2021)
    bortezomib (Velcade)Treatment of multiple myeloma (1/15/2003)First-line therapy of multiple myeloma. 06/20/2008  (06/20/2015)
    bortezomib (Velcade)Treatment of multiple myeloma (1/15/2003)Treatment of multiple myeloma patients who have received at least one prior therapy 03/25/2005  (03/25/2012)
    bortezomib (Velcade)Treatment of multiple myeloma (1/15/2003)Treatment of multiple myeloma patients who have received at least two prior therapies and have demonstrated disease progression on the last therapy 05/13/2003  (05/13/2010)
    bortezomib (Velcade)Treatment of mantle cell lymphoma (5/30/2012)Treatment of patients with mantle cell lymphoma who have received at least 1 prior therapy. 12/08/2006  (12/08/2013)
    bortezomib (Velcade)Treatment of mantle cell lymphoma (5/30/2012)Treatment of patients with mantle cell lymphoma who have not received at least 1 prior therapy 10/08/2014  (10/08/2021)
    brentuximab vedotin (Adcetris)Treatment of Hodgkin’s lymphoma (1/30/2007)Treatment of patients with classical Hodgkin lymphoma at high risk of relapse or progression as post-autologous hematopoietic stem cell transplantation (auto-HSCT). 08/17/2015  (08/17/2022)
    brentuximab vedotin (Adcetris)Treatment of Hodgkin’s lymphoma (1/30/2007)The treatment of patients with Hodgkin lymphoma after failure of autologous stem cell transplant (ASCT) or after failure of at least two prior multi-agent chemotherapy regimens in patients who are not ASCT candidates 08/19/2011  (08/19/2018)
    cinacalcet (Sensipar)Treatment of hypercalcemia in patients with primary hyperparathyroidism for whom parathyroidectomy would be indicated on the basis of serum calcium levels, but who are unable to undergo surgery (4/30/2010)Treatment of hypercalcemia in adult patients with primary hyperparathyroidism for whom parathyroidectomy would be indicated on the basis of serum calcium levels, but who are unable to undergo parathyroidectomy. 11/21/2014  (11/21/2021)
    cinacalcet (Sensipar)Treatment of hypercalcemia in patients with primary hyperparathyroidism for whom parathyroidectomy would be indicated on the basis of serum calcium levels, but who are unable to undergo surgery (4/30/2010)Treatment of severe hypercalcemia in patients with primary hyperparathyroidism who are unable to undergo parathyroidectomy 02/25/2011  (02/25/2018)
    cysteamine enteric coated (Procysbi)Treatment of cystinosis (10/24/2006)For management of nephropathic cystinosis in adults and children ages 6 years and older 04/30/2013  (04/30/2020)
    cysteamine enteric coated (Procysbi)Treatment of cystinosis (10/24/2006)To expand the indication to pediatric patients 2-6 years of age with nephropathic cystinosis 08/14/2015  (08/14/2022)
    daratumumab (Darzalex)Treatment of multiple myeloma (5/6/2013)For the treatment of patients with multiple myeloma who have received at least 3 prior lines of therapy including a proteasome inhibitor and an immunomodulatory agent or are double refractory to a proteasome inhibitor and an immunomodulatory agent 11/16/2015  (11/16/2022)
    daratumumab (Darzalex)Treatment of multiple myeloma (5/6/2013)DARZALEX in combination with lenalidomide and dexamethasone, or bortezomib and dexamethasone, for the treatment of patients with multiple myeloma who have received at least one prior therapy. 11/21/2016  (11/21/2023)
    ecallantide (Kalbitor)Treatment of angioedema (2/4/2003)Treatment of acute attacks of hereditary angioedema in patients 16 years of age and older 12/01/2009  (12/01/2016)
    ecallantide (Kalbitor)Treatment of angioedema (2/4/2003)Treatment of acute attacks of hereditary angioedema (HAE) in patients 12 years of age and older 03/28/2014  (03/28/2021)
    everolimus (Afinitor)Treatment of neuroendocrine tumors (2/14/2008)Treatment of adult patients with progressive, well-differentiated, non-functional, neuroendocrine tumors (NET) of gastrointestinal (GI) or lung origin, (excluding pancreatic) with unresectable, locally advanced or metastatic disease. 02/26/2016  (02/26/2023)
    everolimus (Afinitor)Treatment of neuroendocrine tumors (2/14/2008)Treatment of progressive neuroendocrine tumors of pancreatic origin (PNET) in patients with unresectable, locally advanced or metastatic disease 05/05/2011  (05/05/2018)
    factor XIII concentrate (human) (Corifact)Treatment of congenital factor XIII deficiency (1/16/1985)For the routine prophylactic treatment of congenital factor XIII deficiency 02/17/2011  (02/17/2018)
    factor XIII concentrate (human) (Corifact)Treatment of congenital factor XIII deficiency (1/16/1985)Peri-operative management of surgical bleeding in adult and pediatric patients with congenital Factor XIII deficiency. 01/24/2013  (01/24/2020)
    Fomepizole (Antizole)Treatment of methanol or ethylene glycol poisoning (12/22/1988)Use for suspected or confirmed methanol poisoning, either alone or in combination with hemodialysis 12/08/2000  (12/08/2007)
    Fomepizole (Antizole)Treatment of methanol or ethylene glycol poisoning (12/22/1988)As an antidote to ethylene glycol (antifreeze) poisoning, or for use in suspected ethylene glycol ingestion. 12/04/1997  (12/04/2004)
    ibrutinib (Imbruvica)Treatment of nodal marginal zone lymphoma (2/5/2015)Treatment of patients with Marginal Zone Lymphoma (MZL) who require systemic therapy and have received at least one prior anti-CD20-based therapy. 01/18/2017  (01/18/2024)
    ibrutinib (Imbruvica)Treatment of splenic marginal zone lymphoma (2/5/2015)Treatment of patients with Marginal Zone Lymphoma (MZL) who require systemic therapy and have received at least one prior anti-CD20-based therapy. 01/18/2017  (01/18/2024)
    ibrutinib (Imbruvica)Treatment of chronic lymphocytic leukemia (CLL) (4/6/2012)Treatment of patients with chronic lymphocytic leukemia (CLL) who have received at least one prior therapy 02/12/2014  (02/12/2021)
    ibrutinib (Imbruvica)Treatment of chronic lymphocytic leukemia (CLL) (4/6/2012)Treatment of patients with chronic lymphocytic leukemia with 17p deletion who have not received at least one prior therapy 07/28/2014  (07/28/2021)
    ibrutinib (Imbruvica)Treatment of chronic lymphocytic leukemia (CLL) (4/6/2012)Indicated for the treatment of patients with chronic lymphocytic leukemia without 17p deletion who have not received at least one prior therapy (first line therapy). 03/04/2016  (03/04/2023)
    infliximab (Remicade)Treatment of pediatric (0 to 16 years of age) ulcerative colitis (11/12/2003)For reducing signs and symptoms and inducing and maintaining clinical remission in pediatric patients 6 years of age and older with moderately to severely active ulcerative colitis who have had an inadequate response to conventional therapy 09/23/2011  (09/23/2018)
    infliximab (Remicade)Treatment of pediatric (0 to 16 years of age) Crohn’s Disease (11/12/2003)For reducing signs and symptoms and inducing and maintaining clinical remission in pediatric patients with moderately to severely active Crohn’s disease who have had an inadequate response to conventional therapy 05/19/2006  (05/19/2013)
    Iobenguane I 123 (Adreview)For the diagnosis of pheochromocytomas (12/1/2006)To be used in the detection of primary or metastatic pheochromocytomas or neuroblastomas as an adjunct to other diagnostic tests 09/19/2008  (09/19/2015)
    Iobenguane I 123 (Adreview)For the diagnosis of neuroblastomas (12/1/2006)To be used in the detection of primary or metastatic pheochromocytomas or neuroblastomas as an adjunct to other diagnostic tests 09/19/2008 (09/19/2015)
    ipilimumab (Yervoy)Treatment of high risk Stage II, Stage III, and Stage IV melanoma (6/3/2004)For the adjuvant treatment of patients with cutaneous melanoma with pathologic involvement of regional lymph nodes of more than 1 mm, who have undergone complete resection including total lymphadenectomy. 10/28/2015  (10/28/2022)
    ipilimumab (Yervoy)Treatment of high risk Stage II, Stage III, and Stage IV melanoma (6/3/2004)Treatment of unresectable or metastatic melanoma 03/25/2011  (03/25/2018)
    lenalidomide (Revlimid)Treatment of multiple myeloma (9/20/2001)Treatment of multiple myeloma (MM), as maintenance following autologous hematopoietic stem cell transplantation (auto-HSCT) 02/22/2017  (02/22/2024)
    lenalidomide (Revlimid)Treatment of multiple myeloma (9/20/2001)For use in combination with dexamethasone for the treatment of patients with multiple myeloma who have not received at least one prior therapy (first line treatment) 02/17/2015  (02/17/2022)
    lenalidomide (Revlimid)Treatment of multiple myeloma (9/20/2001)For use in combination with dexamethasone for the treatment of multiple myeloma patients who have received at least one prior therapy 06/29/2006  (06/29/2013)
    lumacaftor/ivacaftor (Orkambi)Treatment of cystic fibrosis (6/30/2014)Treatment of cystic fibrosis (CF) in patients age 6-11 year old who are homozygous for the F508del mutation in the CFTR gene 09/28/2016  (09/28/2023)
    lumacaftor/ivacaftor (Orkambi)Treatment of cystic fibrosis (6/30/2014)Treatment of cystic fibrosis in patients age 12 years and older who are homozygous for F508del mutation in the CFTR gene 07/02/2015  (07/02/2022)
    mefloquine HCL (Lariam)For use in the treatment of acute malaria due to Plasmodium falciparum and Plasmodium vivax, and for the prophylaxis of Plasmodium falciparum malaria which is resistant to other available drugs (4/13/1988)Treatment of acute malaria due to Plasmodium falciparum and Plasmodium vivax 05/02/1989 (05/02/1996)
    mefloquine HCL (Lariam)For use in the treatment of acute malaria due to Plasmodium falciparum and Plasmodium vivax, and for the prophylaxis of Plasmodium falciparum malaria which is resistant to other available drugs (4/13/1988)Prophylaxis of Plasmodium falciparum malaria which is resistant to other available drugs 05/03/1989  (05/03/1996)
    Mitoxantrone (Novantrone)Treatment of secondary-progressive multiple sclerosis (8/13/1999)Reducing neurologic disability and/or the frequency of clinical relapses in patients with secondary (chronic) progressive, progressive relapsing, or worsening relapsing-remitting multiple sclerosis (i.e., patients whose neurologic status is significantly adnormal between relapses). 10/13/2000  (10/13/2007)
    Mitoxantrone (Novantrone)Treatment of progressive-relapsing multiple sclerosis (8/13/1999)Reducing neurologic disability and/or the frequency of clinical relapses in patients with secondary (chronic) progressive, progressive relapsing, or worsening relapsing-remitting multiple sclerosis (i.e., patients whose neurologic status is significantly adnormal between relapses). 10/13/2000  (10/13/2007)
    Nitisinone (Orfadin)Treatment of tyrosinemia type 1 (5/16/1995)Treatment of hereditary tyrosinemia type 1 in combination with dietary restriction of tyrosine and phenylalanine. 04/22/2016 (04/22/2023)
    Nitisinone (Orfadin)Treatment of tyrosinemia type 1 (5/16/1995)Adjunctive therapy to dietary restriction of tyrosine and phenylalanine in the treatment of hereditary tyrosinemia type 1 01/18/2002  (01/18/2009)
    Octreotide (Sandostatin Lar)Treatment of severe diarrhea and flushing associated with malignant carcinoid tumors (8/24/1998)Supression of severe diarrhea and flushing associated with malignant carcinoid syndrome. 11/25/1998  (11/25/2005)
    Octreotide (Sandostatin Lar)Treatment of acromegaly (8/24/1998)Reduction of growth hormone and IGF-1 (somatomedin C) in acromegaly. 11/25/1998  (11/25/2005)
    Octreotide (Sandostatin Lar)Treatment of diarrhea associated with vasoactive intestinal peptide tumors (VIPoma) (8/24/1998)Treatment of profuse watery diarrhea associated with VIPoma. 11/25/1998  (11/25/2005)
    ofatumumab (Arzerra)Treatment of chronic lymphocytic leukemia (3/10/2009)Arzerra in combination with fludarabine and cyclophosphamide for the treatment of patients with relapsed chronic lymphocytic leukemia (CLL). 08/30/2016  (08/30/2023)
    ofatumumab (Arzerra)Treatment of chronic lymphocytic leukemia (3/10/2009)Treatment of chronic lymphocytic leukemia (CLL) refractory to alemtuzumab and fludarabine 10/26/2009  (10/26/2016)
    ofatumumab (Arzerra)Treatment of chronic lymphocytic leukemia (3/10/2009)Ofatumumab in combination with chlorambucil, for the treatment of previously untreated patients with chronic lymphocytic leukemia (CLL) for whom fludarabine-based therapy is considered inappropriate. 04/17/2014  (04/17/2021)
    ofatumumab (Arzerra)Treatment of chronic lymphocytic leukemia (3/10/2009)For extended treatment of patients who are in complete or partial response after at least two lines of therapy for recurrent or progressive CLL. 01/19/2016  (01/19/2023)
    oxybate (Xyrem)Treatment of narcolepsy (11/7/1994)Treatment of excessive daytime sleepiness in patients with narcolepsy 11/18/2005  (11/18/2012)
    oxybate (Xyrem)Treatment of narcolepsy (11/7/1994)Treatment of cataplexy associated with narcolepsy 07/17/2002  (07/17/2009)
    pembrolizumab (Keytruda)Treatment of Stage IIB through IV malignant melanoma (11/19/2012)Treatment of patients with unresectable or metastatic melanoma and disease progression following ipilimumab and, if BRAF V600 mutation positive, a BRAF inhibitor. 09/04/2014  (09/04/2021)
    pembrolizumab (Keytruda)Treatment of Stage IIB through IV malignant melanoma (11/19/2012)Treatment of patients with unresectable or metastatic melanoma. 12/18/2015  (12/18/2022)
    polifeprosan 20 with carmustine (Gliadel)Treatment of malignant glioma (12/13/1989)Expanding the indication to include patients with malignant glioma undergoing primary surgical resection. 02/25/2003  (02/25/2010)
    polifeprosan 20 with carmustine (Gliadel)Treatment of malignant glioma (12/13/1989)As an adjunct to surgery to prolong survival in patients with recurrent glioblastoma multiforme for whom surgical resection is indicated 09/23/1996  (09/23/2003)
    ponatinib (Iclusig)Treatment of Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ALL) (11/20/2009)Treatment of adult patients with Philadelphia chromosome positive acute lymphoblastic leukemia (Ph+ALL) that is resistant or intolerant to prior tyrosine kinase inhibitor therapy. 12/14/2012  (12/14/2019)
    ponatinib (Iclusig)Treatment of chronic myeloid leukemia (11/20/2009)Treatment of adult patients with chronic phase, accelerated phase, or blast phase chronic myeloid leukemia (CML) that is resistant or intolerant to prior tyrosine kinase inhibitor therapy. 12/14/2012  (12/14/2019)
    prothrombin complex concentrate (human) (Kcentra)Treatment of patients needing urgent reversal of Vitamin K antagonist therapy for treatment of major bleeding and/or surgical procedures (12/27/2012)Urgent reversal of acquired coagulation factor deficiency induced by vitamin K antagonist therapy (VKA, e.g., warfarin) in adult patients with the need for urgent surgery/invasive procedure. 12/13/2013  (12/13/2020)
    prothrombin complex concentrate (human) (Kcentra)Treatment of patients needing urgent reversal of Vitamin K antagonist therapy for treatment of major bleeding and/or surgical procedures (12/27/2012)Urgent reversal of acquired coagulation factor deficiency induced by Vitamin K antagonist (VKA, e.g., warfarin) therapy in adult patients with acute major bleeding. 04/29/2013  (04/29/2020)
    ramucirumab (Cyramza)Treatment of gastric cancer (2/16/2012)Treatment of advanced gastric cancer or gastro-esophageal junction adenocarcinoma, as a single-agent after prior fluoropyrimidine-or platinum-containing therapy. 04/21/2014  (04/21/2021)
    ramucirumab (Cyramza)Treatment of gastric cancer (2/16/2012)Treatment of advanced gastric or gastro-esophageal junction adenocarcinoma, as a single agent or in combination with paclitaxel, after prior fluoropyrimidine- or platinium-containing chemotherapy. 11/05/2014  (11/05/2021)
    riociguat (Adempas)Treatment of chronic thromboembolic pulmonary hypertension (9/19/2013)Treatment of adults with persistent/recurrent chronic thromboembolic pulmonary hypertension (CTEPH) WHO Group 4, after surgical treatment, or inoperable CTEPH, to improve exercise capacity and WHO functional class 10/08/2013  (10/08/2020)
    riociguat (Adempas)Treatment of pulmonary arterial hypertension (9/19/2013)Treatment of adults with pulmonary arterial hypertension (PAH) WHO Group 1, to improve exercise capacity, WHO functional class and to delay clinical worsening. 10/08/2013  (10/08/2020)
    romidepsin (Istodax)Treatment of non-Hodgkin T-cell lymphomas (9/30/2004)Treatment of peripheral T-cell lymphoma (PTCL) in patients who have received at least one prior therapy 06/16/2011  (06/16/2018)
    romidepsin (Istodax)Treatment of non-Hodgkin T-cell lymphomas (9/30/2004)Treatment of cutaneous T-cell lymphoma (CTCL) in patients who have received at least one prior systemic therapy 11/05/2009  (11/05/2016)
    temozolomide (Temodar)Treatment of recurrent malignant glioma (10/5/1998)Treatment of adult patients with newly diagnosed glioblastoma multiforme concomitatly with radiotherapy and then as maintenance treatment 03/15/2005  (03/15/2012)
    temozolomide (Temodar)Treatment of recurrent malignant glioma (10/5/1998)Treatment of adult patients with refractory anaplastic astrocytoma, i.e., patients at first relapse who have experienced disease progression on a drug regimen containing a nitrosourea and procarbazine 08/11/1999 (08/11/2006)
    trametinib and dabrafenib (Mekinist And Tafinlar)Treatment of Stage IIb through IV melanoma (9/20/2012)TAFINLAR (dabrafenib) in combination with trametinib for treatment of patients with unresectable or metastatic melanoma with BRAF V600E or V600K mutations as detected by an FDA-approved test. This indication is based on the demonstration of durable response rate. Improvement in disease-related symptoms or overall survival has not been demonstrated for TAFINLAR in combination with trametinib. 01/09/2014  (01/09/2021)
    trametinib and dabrafenib (Mekinist And Tafinlar)Treatment of Stage IIb through IV melanoma (9/20/2012)MEKINIST (trametinib) in combination with dabrafenib for treatment of patients with unresectable or metastatic melanoma with BRAF V600E or V600K mutations as detected by an FDA-approved test. This indication is based on the demonstration of durable response rate. Improvement in disease-related symptoms or overall survival has not been demonstrated for MEKINIST in combination with dabrafenib. 01/08/2014  (01/08/2021)

    A New Rulemaking Is Needed for the Intended Use Regulation

    We recently blogged about whether FDA’s recent amendment to the intended use regulation could be considered essentially null and void based upon a failure to comply with the Congressional Review Act.

    Apparently, this suggestion caused quite a stir and even excitement in some quarters, as it seemed like an easy fix for a bad rule. As a result, someone (not us) made inquiry at a high level within the General Accountability Office (GAO) and learned that FDA did comply with the Congressional Review Act.

    Unfortunately, the GAO database has not been updated to include the new rule. It is difficult to tell, because the GAO database seems fairly current with a number of recent rules. But we have been told that the GAO focuses on publishing “major rules” quickly and can fall behind on other rules. (Although the intended use regulation is important to industry, it is not a “major rule.”) So we can expect that the intended use amendment will eventually be included in the GAO’s database.

    With this avenue blocked, we also understand from a source that Congress is unlikely to take up a joint resolution under the CRA to overturn the intended use amendment. Thus, the CRA is not likely to play a role in addressing this midnight regulation from the Obama administration. The new administration will need to decide whether to accept the revised intended use rule or to take administrative steps to revoke it, which could include a new rule‑making.

    In our opinion, if there is to be a new rule-making, it should not be done as a mere purported “clarification” as FDA characterized the most recent amendment (even if going beyond that description). Rather, FDA should begin a rulemaking to fully consider all aspects of the intended use regulation in light of the recent First Amendment case law, due process case law, and other concerns. The regulation dates at least back to 1952. Sixty‑five years later, we are in the age of the Internet. As might be expected, the drug and device industries have evolved significantly in the past half a century or more, as has the dissemination of medical knowledge, and even patient behavior. Reform is badly needed.

    We’ll publish a blog post in the coming weeks with some suggestions about what is wrong with the intended use regulation and how to fix it. We will to try to start a conversation with the aim of bringing forth some fruitful ideas for improvement. It is well past time for FDA to finally modernize its approach to the regulation of labeling and advertising.

    How to Name that Food: The Good Food Institute Petitions FDA for Clarity

    With the expanding market for plant-based foods, often developed as an alternative to the animal-based food products which consumers avoid for various reasons (allergies, health concerns, ethical concerns, environmental concerns, or taste preference), the naming of plant-based foods has become a major issue.

    Although the issue is not limited to non-dairy “milks,” the debate has focused on these foods (e.g. soymilk, almond milk, etc.). In fact, for more than two decades, there has been a debate about including the word “milk” in the name of plant-based non-dairy products. About twenty years ago, in 1997, the soy industry submitted a Citizen Petition asking FDA to issue a regulation recognizing that the name “soymilk” is an appropriate “common or usual name” that had become established through common usage. To date, FDA has not made a determination on this Petition. Meanwhile, the dairy industry has continued in its efforts to persuade FDA to prohibit the use of the word “milk” in the name of plant-based “dairy alternatives,” arguing that these products are not “milk” as that term is defined in FDA’s regulation establishing a standard of identity for milk.

    In 2008 and 2012, FDA issued Warning Letters (here and here) that included comments stating that the agency does “not consider soy milk to be an appropriate common or usual name for a product that does not contain ‘milk’” as defined by the standard of identity for milk.  Yet, just last year, FDA specifically allowed Hampton Creek to keep the name “Just Mayo” for an eggless, plant-based product that does not contain eggs as defined by the standard of identity for mayonnaise.

    Thus far, Courts seem to have sided with the non-dairy industry. False advertising class action lawsuits targeting use of the terms almond milk and soymilk have not been successful. In a December 1, 2015 decision, the District Court dismissed the case alleging that Trader Joe’s has misled consumers and violated FDA standards of identity when it used the term soymilk on food. Gitson v. Trader Joe’s Co., 13-cv-01333, Doc. 139 (N.D. Cal., Dec. 1, 2015) (here); Ang v. WhiteWave Foods Co., 2013 WL 6492353 (N.D. Cal., Dec. 10, 2013) (here).

    However, in December 2016, Reps. Mike Simpson and Peter Welch, and 30 cosigning members of the House urged FDA to more aggressively police the improper use of dairy terms used on labels of plant-based products that do not contain dairy products. The Congressmen asserted that it “is misleading and illegal for the manufacturers of these [foods] to profit from the ‘milk’ name.” In January, 2017, more fuel was added to the fire by the introduction of the DAIRY PRIDE Act (an acronym for the “Defending Against Imitations and Replacements of Yogurt, milk, and cheese to Promote Regular Intake of Dairy Everyday Act”) by Sen. Baldwin. This bill proposed to amend the Federal Food, Drug, and Cosmetic act to include a provision that a product is misbranded “[i]f it uses a market name for a dairy product . . . and the food does not meet the criterion for being a dairy product.” The bill defines dairy product as a food that “contains as a primary ingredient, or is derived from, the lacteal secretion . . . obtained by the complete milking of one or more hooved mammals.”

    In response, the Good Food Institute (GFI) started a campaign, encouraging people to voice their opposition to the DAIRY PRIDE Act by signing a petition to Congress to “Dump” the DAIRY PRIDE Act. The American Soybean Association and the Soyfoods Association of North America (SANA) responded by writing members of the Senate Health, Education, Labor and Pensions Committee to object to the DAIRY PRIDE Act. In addition, SANA sent a letter to FDA arguing in support of the term “soymilk.”  Among other things, SANA points out that “the term soymilk has now been incorporated into government regulations for nutrition assistance programs, federal dietary guidelines, USDA data bases, and communications including but not limited to the Dietary Guidelines for Americans, National School Lunch Program, the Women, Infants, and Children program, and ChooseMyPlate.”

    At the end of January, two new consumer class actions regarding the naming of non-dairy “milks” were filed. Kelley v. WWF Operating Co., 17-cv-117 (E.D. Cal., filed Jan. 24, 2017) (here); Painter v. Blue Diamond Growers, BC 647816 (Los Angeles Super. Ct., filed Jan. 23, 2017) (here). In complaints against Blue Diamond Growers and WhiteWave Foods, Plaintiffs have taken a new approach and assert (among other things) that they have been misled into believing that the plant-based “milk” was nutritionally equivalent to or better than cow milk when the products actually lack many of the essential nutrients and vitamins provided by cow’s milk. Citing FDA regulations, plaintiffs argue that the plant-based product should have been labeled “imitation.”

    On March 2, 2017, GFI filed a Citizen Petition asking that FDA issue regulations that clarify how to name foods. Unlike the Petition by the soy industry in 1997, GFI’s Petition is not limited to “milk” alternatives and does not provide a definition or standard for certain terms. Instead, GFI requests that FDA amend 21 C.F.R. § 102.5, the regulation describing the principles for the common or usual name for nonstandardized foods, to include a provision that addresses the naming of a food by referencing the name of another food. GFI further asks that, in the interim, FDA issue guidance clarifying that foods may be named by referencing names of other foods consistent with the proposed amendment to the regulation. Similar to the 1997 Petition regarding soymilk, the GFI Petition provides a large number of examples of other foods in which FDA has allowed the use of a name that is defined by a standard of identity modified by another term, e.g., bread (defined as made from wheat) vs. rye bread, butter (defined as made from cream/milk) vs. peanut butter and apple butter, and noodles (defined as ribbon shaped products made from wheat flour that must contain egg products) vs. rice noodles. Although the Petition counters the allegations that the plant-based products are imitation products, and therefore must be labeled as such, GFI does not request that FDA amend that regulation.

    We’ll continue to monitor developments in the legislative, administrative, and judicial arenas with respect to this rapidly evolving issue.

    How To Get Rid of The “Totality of the Evidence” Amendment to The Intended Use Regulation

    A few weeks ago, we blogged about FDA’s final rule amending the “intended use” regulation.  The final rule looks very different than the proposed rule, and adopts a new “totality of the evidence” standard that does not resolve the old problems with the regulation but introduces new ones.

    As discussed in the blog post, a trio of pharmaceutical industry groups have filed a request for a stay with FDA. In addition, it appears the rule is vulnerable under the Administrative Procedure Act (APA), because FDA provided an inadequate opportunity to comment on the changes that occurred between the proposed and final rules.

    This blog post discusses a third approach for getting rid of the final rule. Section 801 of the Congressional Review Act (“CRA”) ([5 U.S.C. § 801(a)(1)(A)) provides:

    (a)(1)(A) Before a rule can take effect, the Federal agency promulgating such rule shall submit to each House of the Congress and to the Comptroller General a report containing –

    (i) a copy of the rule;

    (ii) a concise general statement relating to the rule … and

    (iii) the proposed effective date of the rule.

    The submitted reports are maintained by the General Accounting Office (“GAO”) in a searchable database.  Our search of the database did not turn up a report.  Therefore, under the statutory language just quoted, the final rule cannot take effect; the submission of a report is a condition precedent that has not been fulfilled.  If so, the new administration may simply withdraw the rule without going through a notice and comment process as might otherwise be necessary for repeal.

    One argument against enforcing the CRA in this manner (allowing a withdrawal without notice and comment) might be that the failure to submit a report is “harmless error” that should not have such drastic consequences. An obvious counter‑argument, however, would be that this requirement is intended to force agencies to inform Congress of new rules so that they may be potentially disallowed by statute.  If this requirement is not enforceable, that purpose would not be served.  Furthermore, the language is fairly clear that a report must be submitted “[b]efore a rule can take effect.”  What more could Congress have done to express the effect of non-compliance, other than to perhaps add, “and we really mean it”?

    FDA cannot claim ignorance of the requirement, which originated in 1996. A search of the GAO database shows more than 1,200 reports submitted by FDA, many of them recent.  Letting FDA pick and choose without penalty which rules to submit or not would be detrimental to the enforcement of the CRA.  Since FDA chose not to comply with the CRA with respect to this specific rule, it should suffer the consequences spelled out in the plain language of the statute.

    We are not aware of court cases adjudicating this issue one way or the other. So we shall have to wait and see.  The CRA has very much been in the news with Congress invoking other provisions to repeal the Obama administration’s midnight regulations.  With this level of awareness, it is probably only a matter of time before the issue ends up in court.

    A final hot tip: Anyone subject to an enforcement action relying even in part on the new intended use language should raise as a defense that the rule is null and void due to non‑compliance with the CRA.  Perhaps smart government lawyers will find a way to argue to a court that the language quoted above does not mean what it appears to mean.  But, it is they who will have an uphill climb.

    “Eggshell Plaintiffs” Meet Administrative Deference: Ninth Circuit Affirms Dismissal of Proposed Chicken Shed Rule

    Some lawyers old enough to remember carbon paper may also remember, in their archaic law school teaching, the “eggshell plaintiff” rule, accurately described as “a maxim that a tort defendant takes his victim as he finds him.” Also referred to as the “eggshell skull rule,” the principle is named after “an imaginary person who has an extremely thin skull that is as fragile as an eggshell.”  When this imaginary person is hit in the head with a blow that would have only bruised a normal person, he dies. The eggshell plaintiff rule holds “that the person who hit the eggshell-skulled person is responsible for the much greater harm caused by the death, not just the amount of harm that a normal person would have suffered.” Id.

    A different type of eggshell plaintiff (or, more precisely, a “shell egg” plaintiff) brought a case to try to get various federal agencies to create regulatory rules requiring distributors of fresh eggs to label their cartons with information about the chickens’ shed conditions: required disclosure would specify whether the eggs were from “free-range” hens, from “cage-free” hens, or “from caged hens.” (The author of this blogpost uses the term “shed” advisedly, stretching to support the blogpost subtitle.) Lead plaintiff Compassion Over Killing (a nonprofit organization) tried to get FDA, FTC, Agricultural Marketing Service, or Food Safety and Information Service to require this kind of labeling. None of the agencies complied.

    Plaintiffs sued. District Court rejected suit. Ninth Circuit affirmed. Plaintiffs maintained that FDA’s rejection of their proposed rule was arbitrary and capricious, because, they said, “scientific evidence” shows that “egg-laying hens’ living conditions increase the risk of Salmonella-contamination and negatively affect the nutritional value of the eggs.” In commonplace deference to administrative agencies, the Court decided not to “second guess the FDA’s conclusion that these studies were insufficiently reliable.” The Court also held that the “FDA’s explanation for denying Plaintiffs; rulemaking petition barely meets” the “low burden” for administrative agencies rejecting rule-making proposals in these circumstances: the agency “must, at a minimum, clearly indicate that it has considered the potential problem identified in the petition and provide a ‘reasonable explanation as to why it cannot or will not exercise its discretion’ to initiate rulemaking.”

    Omelet you review the decision yourself to see eggsactly the other reasons that the case was dismissed.

    Categories: Enforcement |  Foods

    Rare Disease Week Recap: HP&M’s Frank Sasinowski Briefs the Rare Disease Congressional Caucus

    Every year, hundreds of rare disease patients and caregivers descend on Capitol Hill to participate in Rare Disease Week to learn about federal legislative issues, meet other advocates, and share their unique stories with legislators. As part of this, on March 2, 2017, Hyman, Phelps & McNamara, P.C.’s Frank J. Sasinowski participated in the Rare Disease Congressional Caucus briefing where he spoke to Congressional staff and rare disease advocates about the implementation of the 21st Century Cures Act. This speech covered the following issues that are important for the rare disease community:

    • Patient experience data;
    • Qualification of drug development tools;
    • Priority review vouchers;
    • Regenerative advanced therapies; and
    • Targeted drugs for rare diseases.

    Slides from Frank Sasinowski’s presentation are available here.

    FJS Rare DIsease

    The caucus briefing was organized by the Rare Disease Legislative Advocates in coordination with the Rare Disease Congressional Caucus. The briefing, “Advancing Rare Disease Treatments in the Era of Cures and Health Care Reform,” covered other important topics that were discussed by other speakers:

    • The PDUFA reauthorization process in 2017;
    • The Affordable Care Act repeal and replacement;
    • New models for rare disease drug development; and
    • The role of incentives in the development of orphan therapies.

    More information about the briefing is available here.

    Frank Sasinowski had previously appeared before the House Energy and Commerce’s Subcommittee on Health to present testimony at the first hearing on the 21st Century Cures Initiative (see previous coverage here).

    Categories: Orphan Drugs

    A Decade of the FDA Law Blog!!

    Believe it or not, Hyman, Phelps & McNamara, P.C.’s FDA Law Blog turns 10 years old today (Monday, March 6, 2017). Where have all of the years gone?  It seems like Tuesday, March 6, 2007 was just yesterday.  That’s when we put up our initial post promising to cover “topics of interest to FDA-regulated companies, fellow food and drug and healthcare lawyers and regulatory personnel, as well as people just generally interested in FDA law.”  We think we’ve fulfilled that promise!  (At least that’s what all of the awards we’ve received tell us.)

    Since that day in March 2007, things have never been quite the same. Apparently we’ve become a fixture of the food and drug law bar and required reading for the FDA-regulated industries.  In fact, one of your blogmasters recently mentioned the impending anniversary to a colleague in the food and drug bar.  We were met with: “Only 10 years?  The blog is an institution!”  While we were happy to be labeled an institution, we took (slight) exception to his use of the word “only.”  While the ten years have gone by fast, it’s been grueling work at times (not unlike running a marathon).  We’ve spent thousands of hours researching topics, writing posts (sometimes at unusual hours when we should be sleeping), and creating and updating our popular trackers.  In fact, we’ve written over 3,060 posts!  Assuming each posts averages two pages of text (standard 8.5 x 11 paper), laid out end-to-end we have over 1.06 miles of posts. 

    But all of the work we do has been (and continues to be) worth the effort! We’ve grown a large following – with about 20,000 subscribers – and have had tens of millions of page views and blog website hits.  We’re regularly cited (in the press, in journal articles, and sometimes in judicial decisions) as a reliable authority.  And we appreciate all of the fanmail, kudos, and recommendations we’ve received along the way from interested readers. 

    Most importantly, we’ve had a lot of fun writing for the FDA Law Blog and have learned a lot. Food and drug law may not be the sexiest topic (the Hatch-Waxman genre is though!), and can get quite technical at times.  But we try to spice things up when we can with pop culture references and attention-grabbing headlines.  Where else would you see Godot and FDA called out in the same sentence other than here?  Or learn about “false friends” . . . or get to chuckle over a headline like “Flare-Up Over Generic Herpes Drug Could be Short-Lived” . . . or pick up an earworm for the day (click here and here if you want one).

    In any case, thank you to all of our readers for your attention and support over the past decade. We cannot wait to see what the next decade might bring for FDA regulation, the regulated industries, and the advancements in science and technology that make all of our lives better.

    Categories: Miscellaneous

    Brave New World: The Mutual Recognition of CGMP Inspections

    Last week, the United States and the European Union agreed to recognize each other’s drug cGMP inspections. The agreement reached (see here and here) amends the Pharmaceutical Annex to the 1998 U.S. – E.U. Mutual Recognition Agreement, with a view to avoiding duplicative inspections and saving millions of dollars in repetitive inspections.

    FDA has stated that they believe this “…initiative will result in greater efficiencies for both regulatory systems and provide a more practical means to oversee the large number of drug manufacturing facilities outside of the U.S. and EU.”

    Until now, the EU and FDA sometimes would inspect some of the same facilities within a brief period of time. With this new agreement, such duplication is expected to be the exception, rather than the rule. “By utilizing each other’s inspection reports and related information, the FDA and EU will be able to reallocate resources towards inspection of drug manufacturing facilities with potentially higher public health risks across the globe. This will benefit patients and reduce adverse public health outcomes.”

    Interestingly, many, if not most, products regulated by the Center for Biologics Evaluation and Research at FDA appear to fall outside the ambit of this agreement (as do veterinary immunologicals). “Current good manufacturing practices (CGMPs) inspections of facilities manufacturing vaccines and plasma derived products are not immediately included within the scope of the agreement. The possibility of including vaccines and plasma derived products will be re-evaluated no later than July 15, 2022. Human blood, human plasma, [and] human tissues are not included within the scope of the Amended Sectoral Annex.”

    As some of our readers may recall, we blogged about the negotiations regarding the Mutual Reliance Initiative last summer, here, where we stated that the Food and Drug Administration Safety and Innovation Act of 2012 (FDASIA) allows FDA to enter into arrangements with foreign governments to recognize the inspection of foreign establishments that are registered under the FDCA “…in order to facilitate risk-based inspections…”

    FDASIA section 712 (FDCA section 809):

    (a) INSPECTION.—The Secretary—

    (1) may enter into arrangements and agreements with a foreign government or an agency of a foreign government to recognize the inspection of foreign establishments registered under section 510(i) in order to facilitate risk-based inspections in accordance with the schedule established in section 510(h)(3);

    (2) may enter into arrangements and agreements with a foreign government or an agency of a foreign government under this section only with a foreign government or an agency of a foreign government that the Secretary has determined as having the capability of conduction inspections that meet the applicable requirements of this Act; and

    (3) shall perform such reviews and audits of drug safety programs, systems, and standards of a foreign government or agency for the foreign government as the Secretary deems necessary to determine that the foreign government or agency of the foreign government is capable of conducting inspections that meet the applicable requirements of this Act.

    (b) RESULTS OF INSPECTION.—The results of inspections performed by a foreign government or an agency of a foreign government under this section may be used as—

    (1) evidence of compliance with section 501(a)(2)(B) or section 801(r); and

    (2) for any other purposes as determined appropriate by the Secretary.

    [Emphasis added.]

    The Mutual Recognition Agreement defines an inspectorate that is capable of conducting an inspection of a drug manufacturing facility that meets U.S. requirements, as one that:

    • has the legal and regulatory authority to conduct inspections against a standard for GMP;
    • manages conflicts of interest in an ethical manner;
    • evaluates risks and mitigates them;
    • maintains appropriate oversight of manufacturing facilities within its territory;
    • receives adequate resources and uses them;
    • employs trained and qualified inspectors with the skills and knowledge to identify manufacturing practices that may lead to patient harm; and
    • possesses the tools necessary to take action to protect the public from harm due to poor quality drugs or medicinal products.

    However, according to FDA, the term “capable” does not require that the inspectorate maintain procedures for conducting inspections and overseeing manufacturing facilities that are identical to the FDA’s procedures.

    As we stated last summer, the notion of having a foreign inspectorate perform drug inspections on FDA’s behalf, when the inspections performed by FDA’s own investigators are already so inconsistent, is problematic at best. Indeed, FDA representatives have long acknowledged that the agency doesn’t have an objective method for measuring quality in the drug industry (for instance, at which facilities are cGMPs improving? By how much and in what way?) Nor do they have a reliable method for making cGMP comparisons between facilities manufacturing similar products, or for comparing the results from within a facility over multiple inspections. (This author will acknowledge that the agency has published a draft guidance on Quality Metrics, which has yet to be finalized, and it is also developing a New Inspection Protocol Project, both of which seek to remedy these weaknesses in the agency’s inspection process. However, these projects are in their earliest stages, they remain largely untested, and it is unclear at this point whether they will lead to more consistency in inspectional results.)

    It would seem that rectifying these significant lacunae in FDA’s inspectional responsibilities should be the first order of business for the agency, prior to even considering delegating the responsibility for EU inspections to a foreign inspectorate which is not schooled in FDA’s precise cGMP requirements, and is not required to maintain the same procedures as FDA.

    One final thought, would FDA take an enforcement action against a foreign facility in the absence of having its own FDA investigators gather the evidentiary basis for this enforcement action? This is indeed a brave new world.

    We will continue to keep you posted on all developments in this regard.

    Categories: cGMP Compliance

    Slower than Molasses in January, FDA Moves to Provide Guidance on Product Communications by Pharmaceutical and Device Manufacturers

    In January 2017, FDA issued two Draft Guidance documents concerning communications made by medical device manufacturers about information not expressly contained within a product’s labeling:

    Below is our summary of these two Draft Guidances and an assessment of how it has changed the advice we provide our clients regarding promotional and payor communications.

    Promotional Communication Guidance

    Overview of the Guidance

    FDA stated that medical product manufacturers have expressed an interest in communicating data and information concerning approved or cleared uses of their products that are not contained in such products’ FDA-required labeling—an understatement for sure, but a recognition of the views expressed in various fora over the past several years, including the recent public hearing on manufacturer communications regarding unapproved uses of approved or cleared medical products, held by FDA on November 9 and 10, 2016.

    FDA’s position, as expressed in the Promotional Communication Guidance, is that product promotional communication by a pharmaceutical or medical device manufacturer that is consistent with its FDA-required labeling and truthful and not misleading in any particular would not subject a manufacturer to FDA enforcement action, even if such information were not expressly included in FDA-required labeling.  FDA defined “FDA-required labeling” as the “labeling reviewed and approved by FDA as part of the medical product marketing application review process,” such as the approved U.S. prescribing information for human drugs or biologics or approved labeling for medical devices.  While beauty may be in the eye of the beholder, consistency with FDA-required labeling seems to be only in the eye of the regulator.  However, FDA articulated a number of factors that the agency would use to determine whether a medical product communication is consistent with its FDA-required labeling.  These include:

    • Factor 1: Different Conditions of Use; whether information in a medical product communication is different from the information in the FDA-required labeling regarding:
      • Indication,
      • Patient Population,
      • Limitations and Directions for Handling, Preparing, and/or Using the product,
      • The recommended dosage or use regimen or route of administration.
    • Factor 2: Increases the Potential for Harm; whether the representations or suggestions in a medical product communication negatively alter the benefit-risk profile of the product.
    • Factor 3: Prevents Safe and Effective Use; whether the medical product can still be used safely and effectively in accordance with the directions for use in the FDA-required labeling, given the representations or suggestions in a medical product communication.

    If the analysis of a medical product communication results in the affirmative along any of these factors, then the communication is not consistent with FDA-required labeling. Examples of information included in a medical product communication that could be consistent with FDA-required labeling include:

    • A head-to-head study comparing the safety or efficacy of a manufacturer’s medical product to another medical product approved/cleared for the same, approved indication, when such study does not appear in the FDA-required labeling;
    • Additional context regarding adverse reactions listed in the FDA-required labeling and associated with the approved/cleared uses of the product;
    • Onset of action for the product’s approved/cleared indication and dosing/use regimen;
    • Long-term safety and/or efficacy for those products approved/cleared for chronic use (e.g., longer duration than the product was studied in the clinical trials described in the FDA-required labeling);
    • Effects or use of a product in specific patient subgroups included in its approved/cleared patient population, even when such subgroup analyses were not included in the FDA-required labeling;
    • Patient-reported outcomes when the product is used for its FDA-approved/cleared indication in its approved/cleared patient population;
    • Convenience (e.g., convenient dosing schedule); and
    • Additional context about the mechanism of action described in the FDA-required labeling.

    On the other hand, FDA also provided examples of information included in a medical product communication that the agency would not consider consistent with FDA-required labeling. These include the use of the product for a different, unapproved:

    • Indication;
    • Patient population;
    • Stage, severity, or manifestation of disease;
    • Use alone versus in combination with other product(s);
    • Route of administration;
    • Strength, dosage, or use regimen; or
    • Dosage form.

    In addition to providing examples, FDA also clarified its position on the evidentiary support for medical product communications consistent with FDA-required labeling. FDA stated its view that representations or suggestions made by medical product manufacturers “need to be grounded in fact and science and presented with appropriate context” in order to be truthful and not misleading.  To that end, FDA made some additional recommendations for consideration when presenting information consistent with (but not included in) FDA-required labeling.  FDA stated that material aspects and limitations of study design and methodology should be “clearly and prominently” disclosed for those studies from which information is derived.  Similar to its Medical Reprint Guidance, FDA stated that communication of information consistent with FDA-required labeling “should accurately characterize and contextualize the relevant information about the product, including by disclosing unfavorable or inconsistent findings.” In addition, related information from FDA-required labeling should also be included in the communication.

    Analysis

    The Promotional Communication Guidance contains some helpful clarifications regarding FDA policy on communications consistent with FDA-required labeling (think “near-label” promotion) that industry should find helpful. Pursuant to the Promotional Communication Guidance, manufacturers may engage in certain types of communications, such as that regarding longer-term safety and efficacy and convenience, which are now subject to FDA enforcement discretion if they comply with the requirements imposed by this guidance.

    However, given the recent spate of First Amendment litigation (see, e.g., here and here) concerning off-label promotion, it is unclear why any communications made by medical product manufacturers that are rendered truthful and non-misleading by being grounded in fact and science and appropriately contextualized would be unlawful, and thereby subject to enforcement in the first place. First Amendment issues were not addressed in this guidance, as FDA continues to vacillate on developing and implementing a workable policy (see our post here) in light of the relevant judgments and settlements that have not gone its way.

    Overall, we think the Promotional Communication Guidance is helpful in understanding FDA’s current thinking regarding communication of information that is not included in a medical product’s labeling. We also believe the Promotional Communication Guidance will enable manufacturers to promote their products with additional types of product information (e.g., patient testimonials) compared to what they chose to disseminate previously.

    We do not think the Promotional Communication Guidance goes far enough with respect to truthful and non-misleading medical product communications protected by the First Amendment. However, we recognize that many companies were hesitant to test the boundaries of FDA’s willingness to take enforcement action, even in light of First Amendment litigation that appeared to limit FDA’s ability to curb off-label promotion.

    Payor Communication Guidance

    Overview of the Guidance

    Congress provided a statutory safe harbor for Health Care Economic Information (“HCEI”) communicated to payors, formulary committees, or other similar entities when exercising their responsibilities for selecting approved drugs for coverage or reimbursement. See Food and Drug Administration Modernization Act of 1997, Pub. Law No. 105-115, § 114, 11 Stat. 2312; 21st Century Cures Act, Pub. Law No. 114-255, § 3037, H.R. 34-73.  Pursuant to this safe harbor, claims by pharmaceutical manufacturers meeting the statutory definition of HCEI and presented to payors, formulary committees, or other similar entities were held to the “competent and reliable scientific evidence” standard, as opposed to the more stringent substantial evidence standard that FDA requires for safety and efficacy claims under the Federal Food, Drug, and Cosmetic Act.  The Payor Communication Guidance, issued twenty years after the original HCEI safe harbor was enacted, provides guidance regarding the communication of HCEI to payors about both approved drugs and investigational drugs and medical devices.

    In the Payor Communication Guidance, FDA provided additional clarity regarding the audience for HCEI communications. First, FDA further explained that “payors” refers to any entity “responsible for the financing or reimbursement of costs associated with health care services.”  Second, FDA explained that formulary committees are “multidisciplinary committees [responsible] for the selection of drugs and the management of a drug formulary.”  These entities may range from a technology assessment panel to pharmacy and therapeutics committees that have responsibility for an entire hospital system.  Falling within this definition are committees “constituted to consider HCEI” and make decisions regarding drug selection, formulary management, and/or coverage and reimbursement determinations at the population level through a “deliberative process.”  In accordance with the statute, FDA has expressly excluded health care providers who make decisions for individual patients.

    FDA also addressed the scope of HCEI protected by the safe harbor in the Payor Communication Guidance. One of two major changes to the HCEI statutory safe harbor enacted under the 21st Century Cures Act was the loosening of certain limitations to the analyses protected thereunder.  Broadly speaking, under 21st Century Cures, HCEI must “relate” (as opposed to “directly relate” in the original statutory text) to an on-label indication.  Furthermore, as long as an HCEI analysis does not only relate to an off-label indication, the analysis (and communications associated with the analysis) is protected under the safe harbor.

    In the Payor Communication Guidance, FDA clarified its thinking, through various examples, on what “relates to an approved indication” means. Examples of HCEI that relate to an approved indication include:

    • Duration of treatment; if approved for chronic use and no limitations on its use beyond the duration that the drug was studied in clinical trials, HCEI analyses considering use beyond the duration of such trials;
    • Practice setting; use of a drug in a practice setting different than the setting in which clinical trials were conducted;
    • Burden of illness;
    • Dosing; HCEI analyses that include actual patient use (for approved indications) where the dosing regiment differs from the recommended dosing schedule in the FDA-required labeling;
    • Patient subgroups; HCEI analyses of patient subgroups that were not analyzed in the pivotal clinical trials;
    • Length of hospital stay;
    • Validated surrogate endpoints; HCEI analyses derived from clinical study data using validated surrogate endpoints; and
    • Clinical outcome assessments or other health outcome measures; HCEI analyses derived from studies with patient-reported outcomes or other health economic measures.

    FDA stated that HCEI not considered to be related to an approved indication include analyses of disease modification when the drug is only approved for the treatment of disease symptoms and analyses conducted in patient populations that are not within the patient population covered by the approved indication for the drug.

    One other important 21st Century Cures-related change to the safe harbor included clarification of what is included in HCEI. Prior to 21st Century Cures, FDA created confusion by suggesting that clinical outputs of a health economic analysis were held to the substantial evidence standard even though the economic outputs were held to the lower standard of competent and reliable scientific evidentiary standard.  21st Century Cures expressly included, within the definition of HCEI, the “clinical data, inputs, clinical or other assumptions, methods, results, and other components underlying or comprising the analysis.”  This significant change addressed what had been a fundamental problem with the safe harbor as originally enacted—health economic endpoints cannot be completely disentangled from clinical endpoints in an HCEI analysis and, now, both are protected.  In the Payor Communication Guidance, FDA acknowledged that the competent and reliable scientific evidence standard applies to “all components of HCEI,” including safety and efficacy.

    FDA stated that when medical product manufacturers present HCEI in accordance with the safe harbor provisions, information regarding the study design and methodology, generalizability, limitations, sensitivity analyses, and other information “relevant to providing a balanced and complete presentation” should be clearly and conspicuously presented.

    Concerning presentations by medical product manufacturers regarding investigational products, FDA stated that certain types of information may be presented to payors prior to approval or clearance of a drug or medical device. This information may include:

    • Product information, such as drug class or device design;
    • Indication sought, including the clinical endpoints studied and populations included in clinical investigations;
    • Results of preclinical or clinical studies;
    • Anticipated FDA approval timeframe;
    • Product pricing information;
    • Marketing strategies; and
    • Product-related programs or services provided by the manufacturer, such as patient support programs.

    Such information must be presented in an “unbiased, factual, accurate, and non-misleading” manner. When presenting such information, manufacturers must also provide a “clear statement that the product is under investigation” and information about the product development stage, such as the clinical trial phase.  FDA also suggests that follow-up information should be provided when the previously communicated HCEI becomes outdated.

    It is important to note that FDA stated in the Payor Communication Guidance that it views HCEI as promotional and, therefore, expects that such information will meet FDA’s requirements for the submission of promotional materials, including submission on FDA Form 2253 at the time of first use.

    Analysis

    The most significant changes to a manufacturer’s ability to communicate HCEI to a payor or similar entity came with the passage of 21st Century Cures, which addressed certain fundamental problems with the safe harbor as originally enacted. That is, 21st Century Cures loosened the restrictions regarding whether aspects of an HCEI analysis could relate to off-label uses of the drug and whether the clinical outputs of an HCEI analysis were held to the lower evidentiary standard.  The Payor Communication Guidance acknowledges and operationalizes these changes, along with providing clarity on FDA’s thinking concerning various elements of the safe harbor.

    We found the amendments to the HCEI safe harbor enacted under 21st Century Cures to provide welcome and necessary relief from issues created by the safe harbor as originally enacted. To a large extent, the Payor Communication Guidance furthers the legislative intent of 21st Century Cures and, therefore, not much has changed with respect to our advice to clients preparing these presentations.  The ability to expand the scope of HCEI analyses and include underlying clinical data and outputs in HCEI presentations under the protection of the safe harbor removes some of the risk in HCEI presentations carried out by manufacturers.

    Arguably, the most significant change that the Payor Communication Guidance outlines is the opportunity for manufacturers to provide HCEI for investigational products. We see this as a big win for patients and hold out hope that earlier communications between manufacturers and payors may lead to earlier coverage determinations, which, in turn, may improve patient access.

    As these are Draft Guidances, comments may be submitted to the docket at any time, but FDA requests that they be posted no later than April 19, 2017.

    The Demise of the BPCIA Patent Dance?

    Well, that was quick! Only two weeks after filing, the U.S. District Court of Delaware dismissed Genentech’s Complaint under the Biologics Price Competition and Innovation Act (“BPCIA”) against Amgen.  As we explained here, Genentech sued Amgen for failure to comply with the patent dance provisions of the BPCIA when Amgen provided Genentech with a copy of its aBLA referencing Avastin within 20 days, but refused to provide any information on the manufacturing process as required under 42 U.S.C. § 262(l)(2)(A).

    This action comes after Amgen Inc. v. Sandoz, in which Amgen sued Sandoz for refusing to participate in the exchange of patent information under the BPCIA. There, the Federal Circuit determined that the patent dance was voluntary (see our prior coverage here). Genentech argued vigorously to distinguish this case from the Sandoz case, stating that the Federal Circuit did not foreclose declaratory judgment actions to determine whether an applicant complied with its statutory obligations. In a Letter to the Court, Genentech argued that Amgen is trying to “escalate the stakes” by “forcing Genentech either to produce a list of potentially infringed patents under § 262(l)(3)(A), without the full production of materials or expert assistance that should have informed that list, or sue Amgen for infringement and wait and see whether that lawsuit was proper at some later time.”

    Conversely, Amgen argued in a similar Letter to the Court that the case could not be distinguished from Amgen v. Sandoz, and that any attempt to do so would penalize Amgen’s good faith effort to comply with the BPCIA patent dance.  Amgen argues that Genentech’s only recourse here is a patent infringement lawsuit.

    The District Court did not buy Genentech’s distinction and dismissed Genentech’s action without prejudice after an oral hearing. The Court’s decision implies that the only recourse Genentech has is a patent infringement action.  The court gave Genentech 45 days to amend its complaint.  The Supreme Court isn’t set to hear arguments in Amgen v. Sandoz until April 26, 2017, after the 45 days expires.  We’ll have to wait to see if Genentech goes full throttle with a patent infringement case based on the information Amgen has provided thus far.  But if the Supreme Court decides that the patent dance is mandatory, then the issue of how much information is necessary to fulfill the obligations will surely come up again swiftly.

    The dismissal raises some questions about what it actually means to opt-into the patent dance under the BPCIA. If it’s not mandatory under Amgen v. Sandoz, and there’s no mechanism to challenge compliance for those aBLA applicants who choose to participate, how can reference product sponsors get the information necessary to bring an infringement suit?  Should they just bring an infringement suit and hope to find out more during discovery?  With 35 U.S.C. § 271(e)(2)(C), there’s a presumption of infringement should the aBLA applicant fail to engage in the patent dance, so pleading isn’t an issue, but it’s a costly decision to sue for patent infringement.  Until Amgen v. Sandoz is decided, we’re unlikely to have any answers to a multitude of questions.

    Categories: Biosimilars

    Maybe It Was Worth a Try . . . . Kentucky Defendant, Charged with Illegally Selling “Herbal Supplements,” Loses Motion to Dismiss

    In the absence of blogworthy FDA regulatory pronouncements (draft regulations, draft guidances, and oxymoronically named “tentative final rules”), material for FDA Law Blog posts is harder to come by these days (meaning, since President Trump’s inauguration, not coincidentally). Now comes across our computer screen a district court denial of a motion to dismiss filed by a defendant indicted for illegally selling herbal supplements, which, it is alleged, are illegal misbranded drugs.

    Judge Danny Reeves of the federal district court in Lexington, Kentucky, issued a very brief order, but some of the defendant’s claims prompt sympathetic smiles from attorneys familiar with the Federal Food, Drug, and Cosmetic Act (“FDCA”). The Order, filed February 27, 2017, in United States v. Girod, Criminal Action No. 5:15-87-S-DCR (2017 U.S. Dist. LEXIS 26682), stated that the Defendant in the case basically claimed that the FDCA “lacks a rational basis and, therefore, violates his constitutional rights to equal protection and due process.” That argument failed.

    Next, the defendant contended that “people of ordinary intelligence cannot understand what 21 U.S.C. § 352 means.” The prohibition on distribution of misbranded drugs has confounded many a person of extraordinary intelligence, in light of the interplay of the First Amendment with off-label promotion. But that argument failed, as well.

    Finally, reflecting the frustration that we have heard from many clients, the defendant complained that he was being unfairly singled out. Others, according to the order, “are selling bloodroot and chickweed salve via the internet.” As had other judges in other courts, Judge Reeves rejected that argument, since the defendant had not shown that the “decision to prosecute the defendant was based on any impermissible reason.”

    The decision can be accessed here.

    Categories: Enforcement

    Ten Years On, FDA Still Has Not Eased The Medical Device Reporting Regulatory Burden As Directed by Congress 

    Ten years ago, Congress commanded FDA to ease the burden of Medical Device Reporting (MDR) for most class I and class II devices. Four years ago, we blogged about it here. FDA still has not gotten it done.

    Specifically, Section 227 of the Food and Drug Administration Amendments Act of 2007 FDAAA) amended Section 519 of the Federal Food, Drug, and Cosmetic Act (FDCA), directing that FDA “establish” summary quarterly reporting of malfunction MDRs for most class I and class II devices. It is not clear whether Congress expected FDA to establish the new requirements by amending the MDR regulation or by issuing guidance; the language seems open to either possibility.

    This congressionally ordered change to MDR reporting is sensible. One of the most difficult aspects of MDR reporting is making a probabilistic determination as to whether a malfunction that did not cause a serious injury would be “likely” to do so if it were to recur. We blogged about some aspects of the problem here.

    It is astonishing that FDA has flouted the law for ten years, and almost nothing is even said publicly about it. In such a situation, one is tempted to give up hope that change will ever come.

    But new hope has arrived! On January 30, 2017, President Trump issued an Executive Order (EO) titled, “Reducing Regulation and Controlling Regulatory Costs.” As described in more detail in an earlier blog post, this EO puts the entire Executive Branch on a strict regulatory diet. Every new regulation must be accompanied by removal of two old regulations, and “regulation” includes significant guidance.

    That is where the change to the MDR reporting comes in. Although FDA would technically be issuing a new regulation (or guidance), in doing so it would lift an existing burden rather than impose a new one. Therefore, FDA should easily be able to persuade OMB that issuing this change is deregulatory and gives FDA a credit toward new regulations.

    Perhaps with this brand new incentive dangling in front of it, FDA will at last amend the MDR regulations as required by law. Hope springs eternal!

    Categories: Medical Devices

    ACI’s 29th FDA Boot Camp

    The American Conference Institute’s (“ACI”) popular FDA Boot Camp, now in its 29th iteration, is back in New York at the Millennium Broadway Hotel on March 22-24, 2017. The conference is billed as the premier event to provide folks with a roadmap to navigate the difficult terrain of FDA regulatory law.

    This year’s FDA Boot Camp will provide you not only with the essential background in FDA regulatory law to help you in your practice, but also key sessions that show you how this regulatory knowledge can be applied to situations you encounter in real life.  Back by popular demand, this year’s “Ripped from the Headlines” session that will provide you with updates on the key developments in the FDA regulatory bar, including post-election developments and the impact of the new administration on FDA practice.

    A distinguished cast of presenters will share their knowledge and provide critical insights on a host of topics, including:

    • The organization, jurisdiction, functions, and operations of FDA
    • The essentials of the approval process for drugs and biologics, including: INDs, NDAs, BLAs, OTC Approval, the PMA process and the Expedited Approval Process
    • Clinical trials for drugs and biologics
    • Unique Considerations in the approval of combination products, companion diagnostics, and stem cell therapies
    • The role of the Hatch-Waxman Amendments in the patenting of drugs and biologics
    • Labeling in the drug and biologics approval process
    • Off-Label use and a New World Order
    • cGMPs, adverse events monitoring, risk management and recalls

    Hyman, Phelps & McNamara, P.C.’s Kurt R. Karst, will present with a panel of experts and provide an overview of the Hatch-Waxman Amendments and the Biologic Price Competition and Innovation Act (“BPCIA”).  Mr. Karst will also head one of leading workshops on the program: “Hatch-Waxman and BPCIA in the Trenches: Deconstructing and Constructing an Exclusivity Dispute,” Mr. Karst will deconstruct, in a step-by-step manner, a complex exclusivity dispute, analyzing FDA’s and the disputing parties’ various (and sometimes evolving) positions on exclusivity.  Relevant court decisions will also be analyzed and their practical and future effects discussed.  After the exclusivity case analysis is completed, attendees will have the opportunity to construct their own exclusivity dispute by choosing from various base facts.  Once the case is constructed, Mr. Karst will lead attendees through the exclusivity analysis. 

    FDA Law Blog is a conference media partner. As such, we can offer our readers a special 10% discount. The discount code is: P10-999-FDAB17.  You can access the conference brochure and sign up for the event here. We look forward to seeing you at the conference.

    FDA Meeting to Discuss the Meaning and Use of the Term “Healthy” for Foods

    On February 15, FDA announced a public meeting to give interested persons an opportunity to discuss the use of the term “healthy” in the labeling of human food.

    As described in previous blog posts, since FDA’s issuance of a warning letter to Kind LLC in March of 2015, healthy claims as nutrient content claims have become a topic of discussion. On September 28, 2016, FDA published a request for comments in the Federal Register.  FDA also issued guidance concerning the “Use of the Term ‘Healthy’ in the Labeling of Human Food Products.” As of February 22, FDA had received more than 850 comments. The comment period closes on April 26, 2017.

    On January 27, 2017, the Union of Concerned Scientists submitted a Citizen Petition urging the FDA to establish “disqualifying levels” for added sugars for health and nutrient content claims on food and beverages in sections 21 C.F.R. §§ 101.13 and 101.14. FDA opened a separate docket for the Petition.

    The upcoming public meeting could help inform FDA on rulemaking. Whether any such activity goes forward will depend, in part, on how the Trump administration interprets and applies the recent Executive Order curtailing rulemaking.   The meeting will be held at the Hilton Washington DC/Rockville Hotel, 1750 Rockville Pike, Rockville, MD 20852. There also will be an opportunity for parties who are unable to participate in person to join the meeting via Webcast.

    Drug Debarment Actions: Beware

    FDA recently released a report summarizing its enforcement activities for FY 2016 (October 1, 2015 to September 30, 2016). It shows, among other things, that the number of warning letters from the Center for Drug Evaluation and Research doubled (76 in FY 2015 to 151 in FY 2016), in contrast to the halving of the letters sent by the Center for Devices and Radiological Health (168 in FY 2015 versus 85 in FY 2016). This blog post, however, is focused on a lesser used, but arguably more powerful, enforcement tool: debarment. The FDA report claims that in FY 2016 there was only 1 drug product debarment. Perhaps this low number motivated FDA to kickstart debarment actions because we are only a few months into FY 2017, and there already have been at least three debarment notices posted in the Federal Register.

    What is debarment? Debarment is an enforcement proceeding that bars an individual from working “in any capacity” in the drug industry. Courts have interpreted debarment to be a complete bar, such that “[a]ll direct employment by a drug company, whether in the board room or the cafeteria or somewhere in between” is forbidden. See, e.g., DiCola v. FDA, 77 F.3d 504, 509 (D.C. Cir. 1996). FDA is required by statute to debar an individual who has been convicted of a felony relating to the development or approval of any drug product, or other conduct otherwise relating to the regulation of any drug product under the FDC Act. 21 USC § 335a(a)(2). There are other bases for debarment not discussed in this post.

    Debarment frequently is confused with exclusion, which has a similar concept but is imposed by the HHS Office of Inspector General (OIG) under authority of the Social Security Act. OIG has the authority to exclude individuals from participating in federal health care programs, such as Medicare, Medicaid, and Tricare. No payment can be made for items or services furnished by an excluded individual or entity, which effectively can put that person out of business in the healthcare industry. Exclusion can be triggered for a number of reasons enumerated by statute, some of which require mandatory exclusion and some over which OIG has discretion, and most of which require a showing of fraud. Just last year, however, a court upheld the exclusion of a pharmaceutical executive convicted of a misdemeanor FDC Act violation, even though the elements of the violation did not include evidence of an intent to defraud or mislead. See Bohner v. Burwell, No. 2:15-cv-04088-CDJ, 2016 U.S. Dist. LEXIS 167590 (Dec. 2, 2016).

    But back to debarment and FDA’s use of it. The most recent debarment proceedings are summarized below:

    • 18, 2016: FDA permanently debarred Wesley A. McQuerry related to his role as a clinical trial study coordinator. Mr. McQuerry was responsible for administering the clinical trial and ensuring that trial participants received appropriate remuneration. He was convicted of a felony in February 2015 for knowingly and willfully falsifying data for at least four patients, and he used his own blood, stool, and EKG results, in the stead of fictional patients. His conduct resulted in a loss of over $200,000 to a pharmaceutical company. In October 2015, FDA sent a notice proposing to permanent debar Mr. McQuerry, he waived his right to a hearing by failing to respond, and he was permanently debarred effective March 18, 2016.
    • 14, 2016: FDA permanently debarred Edward Manookian based on his felony conviction for conspiracy to defraud the United States by selling an unapproved drug product. Mr. Manookian was the president of a company that marketed Melanotan II (MII), a peptide used as an injectable tanning product. Despite repeated warnings from FDA to cease sales because MII was unapproved, Mr. Manookian persisted, and thus was convicted in August 2015. He received notice of the debarment action in August 2016, waived his opportunity for a hearing, and was debarred as of November 14, 2016.
    • 15, 2016: FDA permanently debarred Louis Daniel Smith for his conduct related to the sale of a health-related product billed as a Miracle Mineral Solution (MMS), which was composed of sodium chlorite, an industrial chemical used as a pesticide and not intended to be used for human consumption. Mr. Smith obtained chemicals to manufacture MMS and sold MMS as a drug product. He was convicted in 2015 for one count of conspiracy, three felony counts of introducing misbranded drugs into interstate commerce, and one count of smuggling. In August 2016, FDA sent a notice proposing to permanently debar Mr. Smith, and he also waived his right to request a hearing.
    • 15, 2016: On the same day as the notice of Mr. Smith’s debarment, FDA also published notice of the debarment of Paul S. Singh. Dr. Singh was convicted in July 2015 for providing patients with an unapproved birth control method, a copper intrauterine device (IUD), even though Dr. Singh knew of the risk and failed to inform patients of the unapproved nature of the IUD. Dr. Singh received over $83,000 in reimbursement from health care benefit programs by misrepresenting the type of IUD he had inserted. He was convicted on a single count of felony mail fraud. After notice in August 2016 of the debarment proceeding, Dr. Singh failed to request a hearing and thus was debarred effective November 15, 2016.

    Notably all four individuals were debarred after constructive or actual waiver of hearings, and after felony convictions. It is hard to make heads or tails of FDA’s historic reliance on debarment as an enforcement tool.

    FY 20096
    FY 201013
    FY 201116
    FY 201220
    FY 20136
    FY 20141
    FY 201514
    FY 20161
    FY 2017

    (as of 2/20/2016)

    3

    The uptick at the start of FY 2017 serves as a reminder to industry of the collateral consequences of a conviction involving drug regulation. The facts underlying these debarment decisions also may support exclusion by HHS-OIG, as was the case for Mr. Bohner, cited above. Counsel for defendants in criminal cases should be cognizant of these implications in negotiating any resolution with the government.