• Skip to primary navigation
  • Skip to main content
  • Skip to footer
AgencyIQ by POLITICO
  • Solutions
  • About Us
  • Events
  • Blog
  • Newsletters
    • FDA Today: Life Sciences
    • FDA Today: Food
    • EMA Today
    • The Periodic
    • Overview
Login
Try a Demo

Article

August 9, 2024 by

By Scott Stephens, MPA

The European Commission plans to implement exemptions introduced under the Stockholm Convention’s 2023 ban of UV-328, an ultraviolet-light absorbing persistent organic pollutant (POP). Specifically, the EU executive’s proposal seeks to harmonize the POPs Regulation, carving out exclusions for mechanical separators in blood collection tubes, triacetyl cellulose film in polarizers, and photographic paper, among other exempted articles containing UV-328.

Fill out the form to read the full article.

Filed Under: Article

August 9, 2024 by

Commission proposes exemptions to UV-328 ban under POPs Regulation

The European Commission plans to implement exemptions introduced under the Stockholm Convention’s 2023 ban of UV-328, an ultraviolet-light absorbing persistent organic pollutant (POP). Specifically, the EU executive’s proposal seeks to harmonize the POPs Regulation, carving out exclusions for mechanical separators in blood collection tubes, triacetyl cellulose film in polarizers, and photographic paper, among other exempted articles containing UV-328.

BY Scott Stephens, MPA | Aug 6, 2024 8:17 PM CDT

The Stockholm Convention and its implementation in the EU

  • Adopted in 2001, the Stockholm Convention is an international treaty aimed at limiting the production, release, and damaging effects of persistent organic pollutants (POPs). These organic compounds are resistant to environmental degradation and can present hazards to humans and wildlife far from their point of origin. This is largely due to their capacity to travel great distances and across international borders, transported by wind, water and transitory species.
  • The convention currently lists 34 entries, categorized among three levels of varyingly strict annexes. Annex A requires the production and use of 30 entries to be eliminated, with some specific exemptions. Annex B restricts the production and use of the two listed entries – DDT and PFOS, its salts and PFOSF – allowing certain exemptions that should otherwise be eliminated. Annex C requires stakeholders to reduce “unintentional releases” of the seven listed entries with the objective of “continuing minimization and, where feasible, ultimate elimination.” Some Annex C entries are repeated from Annex A.
  • Most recently, in May 2023, the conference of the parties adopted amendments adding the substances dechlorane plus, methoxychlor, and UV-328 to Annex A of the convention. The substances were introduced in accordance with their respective decisions, including Decision SC-11/11 (note: document downloads to local drive) for UV-328, which includes several specific exemptions.
  • Regulation (EU) 2019/1021 (POPs Regulation) serves to codify the Stockholm Convention’s commitments into EU law. Pursuant to Article 1, the regulation “prohibit[s], phas[es] out as soon as possible, or restrict[s] the manufacturing, placing on the market and use of” the POPs “subject to the Stockholm Convention on Persistent Organic Pollutants,” and to the Aarhus Protocol.

EU executive is seeking feedback on proposed amendment to POPs Regulation banning UV-328

  • On July 30, 2024, the Commission published a draft delegated regulation on its “Have your say” portal, allowing four weeks for stakeholders and interested parties to contribute feedback on the proposal to align EU law with the convention by including UV-328 in Part A of Annex I of the POPs Regulation; this part reflects the substances listed in the convention. Article 15(1) of the POPs Regulation empowers the Commission to amend annexes “to adapt them to changes to the list of substances set out in the Annexes to the Convention or the Protocol.”

Identity, use profile, regulation in the EU

  • UV-328 (CAS RN 25973-55-1) belongs to a class of substances called phenolic benzotriazoles (BZTs). The substance absorbs the full spectrum of ultraviolet (UV) light. UV-328 is used in a wide range of applications as a UV absorber and stabilizer.
  • UV-328 is regulated as a substance of very high concern (SVHC) on Annex XIV, the authorization list, of Regulation (EC) 1907/2006 on the registration, evaluation, authorization and restriction of chemicals (REACH). However, according to Recital 5 of the draft regulation, no application was ever submitted to obtain authorization for the continued use of UV-328 beyond its November 2023 sunset date, indicating that the substance is no longer manufactured in the EU.
  • Nevertheless, it appears the substance appears is still imported into the EU in great quantities, given that active registration dossiers exist at annual tonnage levels between 100 and 1,000 metric tons. According to the registrations, the substance is used in many different consumer, professional use and industrial applications, including polymers, coatings, air care products, adhesives and sealants, lubricants and greases, polishes and waxes, and washing and cleaning products.

Proposed exemptions

  • The EU executive is proposing to implement several exemptions as stipulated in the convention to account for certain products containing UV-328 imported into the EU “until the substance is fully substituted.”
  • Five-year exemptions for placing on the market and use are carved out for articles containing the substance in (1) land-based motor vehicles, including cars, motorcycles, agricultural and construction vehicles, and industrial trucks; (2) mechanical separators in blood collection tubes; (3) triacetyl cellulose film in polarizers; and (4) photographic paper.
  • Also exempted are spare parts for (1) land-based motor vehicles; (2) stationary industrial machines for use in agriculture, forestry and construction; and (3) liquid crystal displays in instruments for analysis, measurements, control, monitoring, testing, production and inspection, other than for medical applications. These spare parts may continue to be used until the end of their service life or until 2044, whichever comes first.
  • A separate exemption applies to spare parts used in liquid crystal displays for medical and in vitro diagnostic medical devices. They may be used until the end of their service life.
  • All exempted articles may continue to be used that were already in use before or on the day their respective exemptions expired.
  • Finally, the proposed regulation establishes an exemption for unintentional trace contaminants of UV-328 present in substances, mixtures or articles, where concentrations of the substance are equal to or below 1 mg/kg (0.0001% by weight). This exemption is in accordance with Article 4(1)b of the POPs Regulation.

Analysis and next steps

  • Companies importing products that contain UV absorbers or stabilizers into the EU will want to determine whether their products contain UV-328. If so, they will need to make sure they are covered by the proposed exemptions. Based on the quantities and wide scope of applications documented in the active registrations for UV-328, it seems plausible that some products containing the substance could fall outside these exemptions, meaning their import will be prohibited when this delegated regulation begins to apply.
  • The regulation will begin to apply starting on February 26, 2025, according to the Commission’s proposal. Accordingly, the five-year exemptions expire on February 26, 2030.
  • The Commission has opened a four-week consultation to gather feedback on the proposed regulation, with comments being accepted until August 27.
  • Adoption of the delegated regulation was originally expected in the third quarter of 2023, according to the “Have your say” initiative. Considering the proposed application date, its adoption will likely happen toward the end of this year, or early next, with publication in the Official Journal of the EU soon after.

Featuring previous analysis by Rayan Bhargava.

To contact the author of this piece, email Scott Stephens ( sstephens@agencyiq.com).
To contact the editor of this piece, email Kari Oakes ( koakes@agencyiq.com).

Key Documents and Dates

  • “Have your say” published initiative on persistent organic pollutant UV-328, July 30, 2024
  • Regulation (EU) 2019/1021 of the European Parliament and of the Council of 20 June 2019 on persistent organic pollutants
  • Stockholm Convention on persistent organic pollutants
 

Filed Under: Article

August 9, 2024 by

By Sebastian Godoy, MPH

Earlier this year, the EMA hosted a multi-stakeholder workshop to discuss regulatory issues surrounding drug development of psychedelic products. Despite intense interest in their use, no psychedelic products are approved in the European Regulatory Medicines Network, with drug developers facing legal and regulatory obstacles, including a dearth of guidance. EMA has now published a report on the workshop.

Fill out the form to read the full article.

Filed Under: Article

August 9, 2024 by

EMA’s psychedelic workshop leaves stakeholders with as many questions as answers

Earlier this year, the EMA hosted a multi-stakeholder workshop to discuss regulatory issues surrounding drug development of psychedelic products. Despite intense interest in their use, no psychedelic products are approved in the European Regulatory Medicines Network, with drug developers facing legal and regulatory obstacles, including a dearth of guidance. EMA has now published a report on the workshop; read on for AgencyIQ’s analysis.

By Sebastian Godoy, MPH | Aug 6, 2024 5:32 PM CDT

Background: Psychedelic drug development and regulatory context

  • A growing body of evidence suggests that psychedelics may provide clinical benefit for certain purposes, especially mental health conditions. Psychedelic drugs primarily influence the way the brain processes the chemical serotonin, which can lead to vivid visions and alterations to a person’s sense of self, according to the National Institutes of Health. Classic psychedelics include serotonergic agonists such as psilocybin (from psychedelic mushrooms) and lysergic acid diethylamide (LSD). Other substances that also typically fall into this category are entactogens or empathogens, such as 3,4-methylenedioxy-methamphetamine (MDMA, or “ecstasy”). However, the road to research for these products is paved with obstacles.
  • For example, the actual experience before, during, and after a single dose of a psychedelic substance may significantly alter the participant’s long-term outcomes. Research indicates that the effects from these substances for the treatment of mental health disorders may be both rapid in onset and long-lasting. This suggests that patients may only require a single dose, or few repeat doses, to achieve therapeutic benefit. However, most experts agree that the experience after drug administration, often referred to as “set and setting,” can dramatically affect outcomes. Most research to date has provided doses in a highly controlled, positive environment, often with a licensed mental health practitioner present to help guide the participant through the experience. Many studies have also employed a follow-up session for the participant to discuss their experience with the practitioner. This contrasts dramatically with the experience of using these substances in a home or social setting.

Status check on European psychedelic landscape

  • Currently, there is no E.U. psychedelic-specific guidance. As AgencyIQ previously reported, the closest psychedelic-specific regulatory resource is the draft guideline on clinical investigation of medicinal products in the treatment of depression – Revision 3. This draft guideline, which was open for public comments from September 15, 2023, through March 31, 2024, aims to modernize its set of criteria for developing medicines to treat depression since the guideline has not been updated since 2013. Curiously, psychedelics considerations are addressed in only two sub-sections of the methodology section.
  • The section on psychedelics clarifies that the EMA’s definition includes “classical hallucinogens,” such as psilocybin, LSD, and mescaline, as well as entactogens like MDMA and dissociative anesthetics such as ketamine and esketamine. The guideline recommends beginning investigation in “a more severely affected population,” such as patients with treatment resistant depression.
  • The EMA lays out several challenges in studying psychedelics, including the risk of functional unblinding and difficulty choosing a placebo or comparator, finding an appropriate dose, showing maintenance of effect, and establishing a safe trial environment. Importantly, the draft guideline also addresses the issue of psychological support that ordinarily accompanies administration of psychedelics. Recognizing that extensive pre- and post-administration support, as well as attendance during an hours-long dosing experience, is a customary part of psychedelic administration, the EMA recommends that applicants address these and other issues unique to the drug classes by seeking scientific advice before beginning trials. [ See AgencyIQ’s analysis on EMA’s draft guidance on clinical trials to treat depression].
  • Zooming out, the legal classification of psychedelics in the European Medicines Regulatory Network (EMRN) is primarily left to Member States. The legal classification of psychedelics is not within the EMA’s remit. Instead scheduling is dependent on the “ specific substance and the national legislation in each EU Member state.” although most are generally classified as controlled substances and their production and access are often highly restricted. All Member States have signed onto the United Nations Convention on Psychotropic Substances of 1971, which classifies psychedelic drugs as Schedule 1 substances, meaning that there is little to no therapeutic benefit and there is a high potential for abuse. Although the E.U. has adopted some measures to spur Member States to harmonize their approach towards regulating the possession and research use of narcotics, the specifics nonetheless vary from jurisdiction to jurisdiction.
  • This legal paradigm often hinders the spread of psychedelic clinical trials but there is still substantial interest in growing their presence. For example, there are pathways for continued research under existing E.U. funding frameworks, such as Horizon Europe and the Innovative Health Initiative. Through these programs, the EMRN has seen a rise in the number of psychedelic clinical trials that address various central nervous system conditions; from February 2023 to April 2024, the number of ongoing psilocybin trials grew from 11 to 19, MDMA trials grew from four to seven, and LSD trials grew from one to three.
  • EMA has not offered any more updates guiding psychedelics developers since it released its third revision of the major depressive disorder guideline for public comments. Many of the challenges identified in that document featured prominently in a recent EMA-hosted workshop on regulation of psychedelics, where attendees shared both regulatory challenges and innovative approaches to studying therapies in this area.

An overview of the EMA’s psychedelic workshop

  • The EMA recently gathered an array of psychedelic stakeholders to discuss developing psychedelic medicinal products. The April 2024 multi-day workshop showcased presentations and panels from various regulators, trade groups, academics and patient advocates. Notably, in addition to five academic presentations, the workshop featured presentations from the U.S. FDA’s Deputy Center Director for Substance Use and Behavioral Health MARTA SOKOLOWSKA, the Australian Therapeutic Goods Administration’s Chief Medical Officer for its Health Product’s Regulatory Group ROBYN LANGHAM, and the German regulator BfArM’s Head of Neurology, Psychiatry, and Ophthalmology MARION HABERKAMP. (Haberkamp was the chief author of the EMA’s draft guideline for depression treatments.) Additionally, representatives from trade groups and professional societies – like the European Federation of Pharmaceutical Industries and Associations (EFPIA) and the European Psychiatric Association (EPA) – participated in several panels discussions alongside patient advocacy groups like the Psychedelic Participant Advocacy Network (PsyPAN).
  • During the workshop, presenters and participants primarily discussed how psychedelic clinical research can fit in the EMA’s existing regulatory guidelines. In addition to highlighting ongoing research, some of the most important regulatory topics covered included study design approaches, accurate evaluation of data, and the role of drug regulation versus clinical regulation, among other topics. To capture the insights from the multiple presentations and discussion, EMA recently shared a report summarizing the major points from the event. The main takeaway from the report is that the EMA recognizes that psychedelics stakeholders still face mighty R&D and legal obstacles but that “ measures can be implemented to mitigate their impact.”
  • At a high level, the report highlights psychedelic study design challenges, EMA’s measures to mitigate the challenges, and stakeholder feedback. The opportunities highlighted in the report center around the promise of addressing a prevalent, unmet need and further integrating innovative technology, like using artificial intelligence to “ provide simultaneous translation and text creation while collecting analytics on the data.” However, the report primarily focuses on the factors that are bottlenecking the expansion of psychedelic clinical trials. Among the list of challenges the report outlines, AgencyIQ pulled out three key themes – bias, psychotherapeutic interventions, participant selection – that capture the complexity of the psychedelic landscape.

The problems of functional unblinding and expectancy bias

  • Functional unblinding poses a significant study design challenge for sponsors because it blurs the line between comparator and treatment groups. The report notes that properly designed and conducted clinical trials are the gold standard for sponsors to demonstrate a drug’s efficacy and safety and psychedelic drugs are no exception. However, the unique effects of psychedelic products can present a barrier to adequate blinding. Additionally, many patients enrolled in these studies may have preconceived expectations related to the upcoming experience, referred to as response expectancy, leading patients to correctly guess their study arm assignment, either validating expected effects of the study drug or provoking frustration that the perceived therapeutic effects aren’t experienced. Further, psychedelic clinical trials may be more likely to enroll patients who have had previous experience with psychedelics. All of these factors can make it difficult for investigators to achieve the aims of a randomized, double-blind placebo-controlled trial and complicate sponsors’ interpretation of efficacy and safety data.
  • The report offers several strategies to mitigate bias in the study design. The report proposes three strategies to mitigate the effect of functional unblinding, including: (1) remote, blinded independent raters (which was used in Janssen-Cilag International NV’s development program for its antidepressant, Spravato [esketamine]); (2) post-participation blinding questionnaires given to both groups to assess the impact of functional unblinding; and (3) administering different dosage levels to “introduce some uncertainty in the treatment allocation.” Additionally, regarding possible solutions, the report also briefly highlighted ongoing clinical trials evaluating the effectiveness of ketamine and midazolam as a comparator to psilocybin for treatment-resistant depression and in cancer patients. Lastly, the report noted that effective blinding is most important when subjective scales are used for assessment; it also may be less critical when the treatment effect is since blinding is “ substantial.”
  • Stakeholders were critical of EMA’s proposed mitigating measures. Although stakeholders, such as the European College of Neuropsychopharmacology (ECNP) and the European pharmaceutical trade association EFPIA recognized that the Agency’s outlined blinding measures could be helpful, they countered that the mitigating measures do not overcome a central issue – “ functional unblinding may bias efficacy interpretation.” Further, other representatives from academia and industry questioned whether regulatory agencies place too much emphasis on blinding given that obstacles related to effective blinding are not unique to psychedelics. Additionally, stakeholders from academia, the Psychedelic Access and Research European Alliance (PAREA), and the European Brain Council (EBC) observed that the existing frameworks are designed primarily designed for conventional medicines and proposed that the “ gold standard of randomized controlled trials may not fit psychedelics.”

Psychotherapeutic interventions

  • Psychedelic products are often accompanied with some variation of “psychological support or psychotherapy.” The EMA differentiates between these terms, holding that psychological support is focused on ensuring the safety of a patient, such as by “ chaperoning them during active treatment,” while psychotherapy involves an additional “ psychotherapeutic in intervention,” moving beyond safety concerns to integrating the psychotherapy as an integral part of treatment. Despite their differences, these roles are understood to contribute to the “set and setting” of a psychedelic clinical trial (these terms were coined early in the course of psychedelic research to refer to patient mindset and the physical and social environment, respectively). In many trials, both psychological support and psychotherapy have similar baseline frameworks with three stages: (1) offering preparatory sessions prior to administration; (2) administrating the sessions; and (3) following up with a patient to assess their experience and subsequent insights (i.e., integration sessions).
  • From the EMA’s perspective, psychological support aimed solely at ensuring patient safety in support of marketing authorization applications (MAAs) for psychedelics can be “ described and standardized” in a clinical trial’s protocol and setting description.
  • Conversely, pairing psychotherapy alongside psychedelic administration adds more burden to a sponsor’s study design; the EMA report observed that sponsors would “likely” need to use a factorial study design to assess the relative contributions of psychotherapy and the psychedelic to “ relevant efficacy endpoints.”
  • Another wrinkle in adoption of psychotherapy is that regulating the practice of medicine falls outside of the EMA’s remit. The role of a medicinal products regulatory authority like the EMA is to ensure that health care professionals and patients can access safe and effective medical products; however, the role of determining how clinical support (psychological support versus psychotherapy) is integrated into existing clinical practice guidelines or which patients would benefit from a specific type of support does not fall within the EMA’s scope. Accordingly, the agency cannot define an optimal psychotherapy approach, and the EMA noted that the burden of scoping out such an approach should fall onto the professional bodies or learned societies that regulate and guide clinician practice. Additionally, the EMA spotlighted the U.S.’s FDA approach to the issue of medical products vs clinical regulation. According to the FDA, once a psychedelic treatment is available to the public, it does not “ regulate the medical practice that would be applying such treatments.” However, the FDA did note that it has worked with state medical boards on their approach towards regulating psychotherapy in clinical settings where psychedelics are used.
  • Stakeholders were divided on the utility of psychological support versus psychotherapy. Overall, stakeholders agreed that adopting a standardized, empirical framework to integrate phycological support or psychotherapy is a challenge for all sponsors. Additionally, representatives from PAREA noted that a patient’s level of support during a psychedelic administration is highly variable and may depend not only on the type of psychedelic, but also on its intended therapeutic indication. However, representatives from EFPIA advocated against the use of psychotherapy in psychedelic clinical trials, since other than MDMA-assisted psychotherapy for PTSD, most drug development programs do not involve this clinical practice and the psychotherapy component may introduce bias in the assessment of the drug’s efficacy. Conversely, representatives from the PsyPAN said that in their experience, most trial participants want (or need) additional therapeutic services to “help them achieve lasting change and prolong response and remission.”
  • A possible solution to address the varying levels of patient support, proposed by PAREA and endorsed by PsyPAN, would be to establish a pan-European multidisciplinary advisory board. This group could establish a comprehensive E.U. mental health support framework that recommends models of care, crafts ethical guidelines and sets safety standards.

Participant selection

  • Clearly defining comprehensive inclusion and exclusion criteria to select trial participant is difficult given that there are many unknowns. The EMA report explained that to establish a positive benefit-risk balance, sponsors should consider several key factors, such as disease severity, responses to prior treatments(especially for those patients facing treatment resistance), demographics (like age or sex), concomitant medicines and comorbidities.
  • However, sponsors may face significant challenges selecting in patient selection: Psychedelic products are often evaluated for severe psychiatric diseases, such as treatment-resistant depression (TRD) and PTSD. Since polypharmacy is common among this category of patients, the report also emphasizes the importance of inclusion and exclusion criteria for patients taking concomitant medicines. Although sponsors are responsible for assessing whether a potential drug-drug interaction could impact the results of their evaluation or cause participants harm, not much is known about how and to what extent psychedelics may interact with other psychiatric medicines. For example, some literature suggests that selective serotonin reuptake inhibitors (SSRIs) may diminish the therapeutic effects of psychedelics, although it does not appear that other medicines have a negative impact on psilocybin.
  • The EMA recommends that sponsors focus on selecting s participant population with high, unmet medical needs, to increase the probability of treatment effect. One example of such a group would be patients with TRD. Although studying a broader population is possible, the EMA advises against sponsors taking this approach because generating confirmatory safety and efficacy data may be more challenging. Data could later be applied to a “broader patient population with a less severe disease” and help gain more insight into an optimal treatment paradigm. However, the EMA emphasizes that it is still the responsibility of learned societies to offer treatment guidelines for patient cohorts.
  • Discussions from stakeholders emphasized that a lack of understanding of disease burden makes selecting an “ideal” patient cohort a challenging feat, even in a smaller patient group. Although the EMA noted that enrolling patients with a specific, high unmet need will likely yield the best clinical trial data, an industry representative countered that early intervention for a potentially broader patient group is key because “psychiatric diseases such as depression have a higher risk of further episodes.” A clinician also explained that depressive episodes can be neurotoxic and possibly lead to a reduction in volume of the brain’s hippocampus.

A snapshot of global psychedelics regulation

  • At the workshop, several global regulators also weighed in on their national efforts to advance the marketing of psychedelic products. The report highlighted presentations by the Australian Therapeutics Goods Administration (TGA), Health Canada and the U.S. FDA sharing timelines of their regulatory work.
  • In Australia, certain psychedelics have been rescheduled to allow approved psychiatrists to administer the drug. Prior to 2023, all psychedelics were labeled under Australia’s scheduling policy framework as a schedule 9 “prohibited substance,” meaning that drug was deemed to have “ no therapeutic value” and that the “benefits of use are substantially outweighed by the risks.” However, following a two-year effort where regulators and other government officials weighed the opinions of expert groups, analyzed existing evidence, and sought public consultation, Australia moved to reschedule MDMA and psylocibin to a schedule eight “controlled drug,” still conferring stringent safeguards. Therefore, as of July 1, 2023, patients can be prescribed MDMA and/or psylocibin, in combination with psychotherapy, to manage TRD and PTSD in certain cases. Importantly, this rescheduling did not register MDMA or psylocibin as medicines. Consequently, the TGA developed a mechanism, named the Authorized Prescriber pathway, to allow certain psychiatrists to apply to be able to administer one (or both) of the schedule 8 psychedelic drugs. As of April 16, 2024, there are nine authorized prescribers who can prescribe MDMA for PTSD, or psylocibin for TRD, or both.
  • Health Canada’s case study demonstrates how legislative reversals can impact access to psychedelics. From 2013-2021, the Canadian government classified psychedelics as a restricted drug in Part J of the Food and Drug Regulations, meaning that Canda did not recognize any approved medical uses. Health Canada does have a specific pathway – the Special Access Program (SAP) – where restricted or unapproved drugs can be given to patients who have life threatening conditions where “ conventional therapies have failed, are unsuitable or are unavailable”; however, due to regulatory amendments to the Food and Drugs Regulations in 2013, psychedelics were the only the class of drug that could not be requested through the SAP.
  • Fast forwarding to 2020, Health Canada moved to amend the Food and Drugs Regulation to once again restore psychedelic drugs’ inclusion in SAP; after a lengthy public consultation period and expert evaluation of more recent psychedelic research, Health Canada ultimately did move MDMA and psilocybin back into SAP. As of January 2022, health care professionals can apply to provide specific patients psychedelic-assisted psychotherapy. Since adopting the amendments to the Food and Drug Regulation, 200 Canadian patients have received psilocybin for either major depressive disorder (MDD) or end-of-life psychological distress, and 40 patients have received MDMA for PTSD, all under SAP. Additionally, Health Canda is working to establish a scientific mental health expert committee to “ examine the Canadian clinical context for treating different mental health conditions.”
  • The FDA, and other federal agencies, have been addressing how psychedelics are studied and regulated. In the U.S., nearly all psychedelics are classified as Schedule I drugs under the Controlled Substance Act (CSA), meaning that research programs evaluating these substances are subject to additional oversight from U.S. Drug Enforcement Administration. Also, when a substance is scheduled under the CSA, companies may find it difficult to obtain sufficient supplies to run clinical trials and gather data and evidence to support their marketing efforts. However, in December 2023, the National Defense Authorization Act for Fiscal Year 2024 included a specific mandate that the Department of Defense investigate the therapeutic impact of certain psychedelics on patients with PTSD or traumatic brain injuries.
  • Additionally, as AgencyIQ previously reported, the FDA in June 2023 released a draft guidance on development of psychedelic medicines for public comment. Safety and risk mitigation are prominent components of the draft guidance, with discussion on topics ranging from assessing abuse potential, to conducting regular echocardiograms, to requiring study facilitators to have specific training, experience, and credentials. AgencyIQ also reviewed the 223 comments that FDA received; a key takeaway is that there is a large disconnect between the FDA and stakeholders regarding best practices. However, new clinical trial research is still ongoing, especially given that since 2017, some psychedelics under development to treat mental health conditions have been granted breakthrough therapy designation by the FDA.

What’s next

  • Despite the fact that the EMA consistently reminded workshop participants that regulating the practice of medicine (such as defining the optimal treatment paradigm regarding psychotherapy) is outside of its remit, stakeholders still sought direction. For example, the European Psychiatric Association (EPA) called for more guidance from the EMA on how to account for factors such as integration sessions in the design of psychedelic clinical trials.
  • The current lack of direction on such specifics is compounded by the lack of a specific guideline for development of psychedelic therapies. The most current guidance is the slim section addressing psychedelics within the third revision of EMA’s draft anti-depressant guideline. Additionally, EMA has also not addressed the question of developing psychedelic drugs for any indication other than depression, potentially leaving developers who wish to investigate treatment of PTSD, anxiety, or other psychiatric developers in the dark about key questions such as endpoint selection, study population, trial duration, long-term follow-up, and more.
  • Regarding this lack of specific guidelines, the EMA shared that further guidance may be introduced once a “ sufficient evidence base has been established to identify the optimal approach to support drug development and characterize the benefit-risk.” Until this changes, sponsors will likely need to request scientific advice early and consult frequently with regulators throughout their studies.
  • The workshop’s inclusion of other leading regulators was noteworthy. Sokolowska, a senior FDA official, attended in-person, continuing a conversation among global regulators about how to address the regulatory challenges of psychedelics as medicinal products. In the fall of 2023, the EMA’s FLORENCE BUTLEN teamed with a representative from U.S. industry to anchor a psychedelics-focused plenary session at the annual Regulatory Affairs Professionals Society (RAPS) Convergence meeting. The issues raised in that session mirrored those in the spring workshop and the recent EMA report, showing that even if global regulators haven’t yet figured out how to resolve some conundrums in psychedelic drug development, they are at least in agreement about where the problems lie.
  • In contrast to the situation in Europe, the U.S. has published psychedelic-specific guidance and has several ongoing psychedelic clinical trials. Additionally, the EMA’s report did highlight how the FDA has been communicating with state boards on their regulation of psychotherapy performed concomitantly with psychedelic use. The report considers a slightly different approach that would collaborate with regulators and professional societies to set these parameters and foster pan-European collaboration.
  • Additionally, the Australian TGA case study highlights a potentially applicable pathway to better integrate psychotherapy into psychedelic interventions. The TGA also shares the EMA’s and FDA’s view on regulatory remit. However, their case study highlights how a high degree of collaboration between medicines and clinical regulators can help clinicians participate in the development of psychedelic therapies.
  • One area that was mentioned throughout many presentations but not prominently featured in the report is the administrative and financial burdens of conducting these clinical trials. Given that psychedelics are not universally legalized, sponsors can have trouble accessing national funds or even the drugs themselves. For example, during PAREA’s presentation on the impact of the E.U.’s legal status on psychedelic R&D, they highlighted a quote from a psychedelic researcher, DAVID NUTT, that underscores the administrative challenges of carrying out a trial: “Our first psilocybin depression trial took 32 months to get permissions to buy and import psilocybin for all largely due to regulations.”
  • Additionally, the high cost of these trials was a point that was raised by an industry representative during the European College of Neuropsychopharmacology’s (ENCP) discussion on conducting trials in combination with psychotherapeutic interventions; especially for larger trials, these expenses could represent disincentives for firms to continue investigation of psychedelics. The report did not offer any concrete solutions to these administrative and financial burdens. However, during Haberkamp’s presentation on the European regulatory perspective, she highlighted that in January 2024, the E.U. funded a planned large psychedelic study for patients with incurable diseases, information also reported by our colleagues at POLITICO. This study, set to begin in January 2025, received 6.5 million euros from the E.U.’s Horizon Europe program; it may serve both as an important data generation source and as a regulatory proving ground as Europe finds its way in this complicated field.

Featuring previous research by Chelsey McIntyre and Kari Oakes.

To contact the author of this item, please email Sebastian Godoy ( sgodoy@agencyiq.com)
To contact the editor of this item, please email Kari Oakes ( koakes@agencyiq.com).

Key Documents and Dates

  • EMA multi-stakeholder workshop on psychedelics – Towards an EU regulatory framework, April 16 and 17, 2024
  • EMA multistakeholder workshop on psychedelics – Workshop report, July 16, 2024

Filed Under: Article

August 9, 2024 by

What we expect EU chemical regulators to do in August 2024

Welcome to AgencyIQ’s monthly roundup of EU chemical sector activities. This recurring feature compiles information from across EU agencies and institutions to deliver an overview of chemicals-related regulatory actions likely to happen in the month ahead, including planned legislation, consultations, webinars, meetings, and more.

By Scott Stephens, MPA | May 29, 2024 4:05 PM CDT | Updated Jul 31, 2024 2:24 PM CDT

Highlights of upcoming chemical regulatory activities

  • Several consultations on regulatory procedures under the REACH and CLP Regulations are coming to an end in August, despite the summer lull (REACH is the Registration, Evaluation, Authorization, and Restriction of Chemicals Regulation; CLP is the Classification, Labeling and Packaging Regulation).
  • Under REACH, these address testing proposals for five entries, including trimethylolpropane diallyl ether and 5-(hydroxymethyl)-2-furaldehyde. Likewise, a call for evidence soliciting information on uses and alternatives for certain hexavalent chromium substances, for which a restriction proposal is being prepared, is ending on August 15.
  • Under the CLP, four substances, including flonicamid and 1-ethoxy-2-(2-methoxyethoxy)ethane, have been put forward for harmonized classification and labeling (CLH).
  • Other consultations ending in August include two addressing potential candidates for substitution and exemption conditions under the Biocidal Products Regulation (BPR), including for the substance DBNPA. Two others concern 1,3-propanesultone and EDB, for which the authorities are in the process of establishing occupational exposure limit (OEL) values.
  • During a slow month for regulatory events, Keller & Heckman is holding a free webinar on August 28 dedicated to the newly published Ecodesign Regulation and how it relates to EU chemical legislation.
  • “Have your say” feedback periods are winding down in August, including one updating the lists of hazardous substances regulated under Regulation (EU) 649/2012, which implements the Rotterdam Convention on the prior informed consent (PIC) procedure.

ECHA consultations under the REACH Regulation closing in August

REACH: Testing proposals

  • Testing proposals are part of registration requirements under REACH. They are subject to 45-day consultations during which third parties are invited to submit relevant scientifically valid data addressing the substance(s) and hazard endpoint(s).

Name

CAS RN

Hazard endpoint proposed for vertebrate testing

Deadline

Eleven testing proposals for five chemical entries, including for 5-(hydroxymethyl)-2-furaldehyde and spiro[piperidine-1,1′-pyrrolidinium] bifluoride.

(See ECHA’s Current Testing Proposals webpage for details on these entries and to provide comments)

Various

Various

August 26

 

REACH: Call for comments and evidence

  • These actions are intended to offer stakeholders opportunity to contribute to the preparatory phase of a REACH Annex XVII restriction before the competent authorities (i.e., member states or ECHA) have completed and published the restriction proposal.
  • They are meant for parties not initially identified or consulted by ECHA and, therefore, would not otherwise be able to contribute to shaping the proposed restriction. The calls supplement ECHA’s public consultations on restrictions conducted pursuant to REACH’s Title VIII restriction procedure. They do not replace them.

Name

CAS RN

Subject of the call

Deadline

Certain chromium(VI) substances

–

Uses and alternatives for certain hexavalent chromium substances to support the preparation of a restriction proposal

August 15

 

REACH: Restrictions under consideration

  • These comment periods under REACH’s Title VIII restriction procedure allow interested parties to weigh in on the opinions authored by ECHA’s risk assessment (RAC) and socio-economic analysis (SEAC) committees on restriction proposals.

Name

CAS RN

Restriction Information

Deadline

–

–

–

–

 

REACH: Identification of substances of very high concern (SVHCs)

  • Interested parties are invited to provide feedback on the chemicals put forward for identification as SVHCs. Once identified as SVHCs, these chemicals are added to the Candidate List. Being placed on this list represents the first step toward eventual inclusion in REACH’s Annex XIV, the authorization list, and triggers certain legal obligations for affected companies.

Name

CAS RN

Reason for proposing

Deadline

–

–

–

–

 

REACH: Recommendations related to the Authorization List

  • These consultations provide interested parties a way to contribute to ECHA’s regular assessment of the substances on the Candidate List that are prioritized and, subsequently, recommended for inclusion or amendment (for existing entries) in the authorization list (Annex XIV).

Name

CAS RN

Deadline

–

–

–

 

REACH: Applications for authorization (AfAs)

  • Each application for authorization under REACH involves an eight-week public consultation inviting interested third parties to provide information on alternative substances or technologies for the uses of the Annex XIV substances included in the AfA.

Name

CAS RN

Number of AfAs

Deadline

–

–

–

–

 

ECHA consultations under the CLP Regulation closing in August

CLP: Harmonized classification and labeling (CLH)

  • Consultations on classification and labeling harmonization (CLH) are open for 60 days. They seek to elicit feedback from interested parties on the hazard classes proposed by the dossier submitter, as well as on any others that the dossier submitter did not find warranted classification.

Name

CAS RN

Hazard classes open for commenting

Deadline

1-ethoxy-2-(2-methoxyethoxy)ethane

1002-67-1

Reproductive toxicity

August 9

flonicamid (ISO); N-(cyanomethyl)-4-(trifluoromethyl)pyridine-3-carboxamide

158062-67-0

Explosive

Acute toxicity, oral

August 9

O-isopropyl ethylthiocarbamate

141-98-0

Reproductive toxicity
Hazardous to the aquatic environment

August 9

Silica, amorphous, fumed, cryst.-free; Pyrogenic, synthetic amorphous silica, nano [1]

Silica gel, pptd., cryst.-free; Precipitated silica, silica gel, colloidal silica, amorphous, nano [2]

112945-52-5

[1]

 

112926-00-8

[2]

 

Specific target organ toxicity — repeated exposure

August 9

 

ECHA consultations under BPR closing in August

BPR: Potential candidates for substitution (CfS) and derogation conditions

  • These consultations support the evaluating competent authority in assessing the availability of substitutes or alternatives to active substances found to be potential candidates for substitution in accordance with Article 10 of the BPR.
  • The consultations also seek additional information from third parties to determine if relevant substances subject to Article 5(1) exclusion criteria may qualify for exemptions under Article 5(2) of BPR.

Name

CAS RN

Product type

Deadline

2,2-dibromo-2-cyanoacetamide; [DBNPA]

10222-01-2

11 (Preservatives for liquid-cooling and processing systems)

August 26

Reaction products of ammonium bromide and sodium hypochlorite, generated in-situ;

re-named in analytical methods, physico-chemical properties working group (APCP WG), IV-2023. Previously: Bromide activated chloramine (BAC) generated from ammonium bromide and sodium hypochlorite

—

11 (Preservatives for liquid-cooling and processing systems); and

 

12 (Slimicides)

August 26

ECHA consultations under occupational, safety and health (OSH) rules closing in early August

CAD/CMRD: Occupational exposure limit value (OELV) proposals

  • ECHA contributes to establishing OELs in support of the Chemical Agents Directive (CAD) and the Carcinogens, Mutagens or Reprotoxic substances Directive (CMRD). Consultations are held to allow interested parties to contribute to this process.

Name

CAS RN

Deadline

1,3-propanesultone

1120-71-4

August 12

Ethylene dibromide (EDB) or 1,2-Dibromoethane

106-93-4

August 12

 

ECHA consultations on persistent organic pollutants (POPs) proposals

  • These eight-week consultations provide opportunity for interested parties to contribute feedback on draft reports for substances anticipated to be, or already proposed for, listing as POPs under the Stockholm Convention.

Name

CAS RN

Deadline

–

–

–

 

Meetings, trainings, webinars, calls for tenders and other events in August

Event title (+ link)

Hosting entity

Date/Deadline

Type

REACH 30/30 Webinar Topic: New Ecodesign Regulation From a Chemical Perspective

Keller & Heckman

August 28

Free webinar

Contract research organization days – bridging in vivo laboratories and regulatory scientists

 

ECHA

19-20 November

(NOTE: Registration deadline is August 31)

In person (Helsinki, Finland)

 

Chemical legislation that the Commission is preparing

  • The European Commission is responsible for proposing legislation under EU law. Below is a list of the yet-to-be-published legal acts that the Commission has flagged on its “Have your say” initiatives portal for future adoption.
  • Under ordinary legislative procedure (OLP), once adopted, a Commission proposal is forwarded to the EU’s co-legislators, the Council of the EU, representing the bloc’s 27 member states, and the European Parliament (EP).
  • Each co-legislator takes up the proposed legislation for discussion and debate, possibly amends it, and adopts a position on the (amended) proposal. The legislation can only be finalized and, subsequently, published in the Official Journal if the co-legislators are able to come to an agreement on the final text and adopt the common version agreed to by both institutions.
  • Delegated and implementing legal acts, conversely, are not taken up by the co-legislators after they are proposed by the Commission. Unlike under OLP, such legislation is subject to a faster procedure. For delegated acts, the EP and Council usually have two months to object to the proposals but are not allowed to amend them. For implementing acts, the Commission is required to consult with a committee, on which every EU member state is represented, before adopting them.
  • An important caveat: The adoption timeframes that the EU executive indicates on the “Have your say” portal are not always strictly adhered to and may be subject to delay.

Planned legislation

Description

Type of act

Link

European Chemicals Agency (ECHA) – updated fees and charges

Adoption planned for Q1 2024

 

Initiative amends Commission Regulation 340/2008 on the fees and charges payable to ECHA. It will:

– Adjust fees in line with inflation
– Introduce other measures to increase sustainability of ECHA financing by reducing administrative burden on ECHA linked to the ex post verification of the size of companies (used to determine their eligibility for SME fees and charge rebates).

Proposal for Implementing Regulation

Initiative entry

European Chemicals Agency – proposal for a basic regulation

Adoption was planned for Q2 2023

This initiative will propose a basic regulation for ECHA. It aims to: strengthen ECHA governance and adapt it to its future role, as well as to streamline the working methods of ECHA bodies and make their financing more sustainable.

Proposal for Regulation

Initiative entry

Restriction updating Annex XVII REACH regarding CMRs (2023)

Adoption was planned for Q4 2023

The initiative aims to protect consumers by adding newly classified CMR (carcinogenic, mutagenic, toxic to reproduction) substances to the lists of CMR substances restricted for consumer use under the Regulation on the registration, evaluation, authorization and restriction of chemicals (REACH).

Proposal for Regulation

Initiative entry

Chemicals regulation – update of EU rules for test methods

Adoption was planned for Q4 2023

EU legislation on chemicals provides for checks on chemicals that may be hazardous for humans, animals, or the environment. The current rules include a list of approved methods for testing chemicals.

This initiative will add new/updated methods to that list. These methods could: reduce the number of animals needed to test for chemical hazards; and refine certain tests to obtain more reliable/precise results on chemical hazards.

Proposal for Regulation

Initiative entry

Ecodesign – European Commission to examine need for new rules on environmental impact of photovoltaics

Adoption planned for Q1 2024

Given the role that photovoltaic products are expected to have in decarbonizing the EU’s energy system, it is essential that newly installed products in the EU are environmentally friendly.

Therefore, the Commission is assessing the need for regulation to manage the environmental impacts of photovoltaic products.

Proposal for Regulation

Initiative entry

Chemical pollutants – limits and exemptions for perfluorooctanoic acid (PFOA)

 

Adoption planned for Q3 2024

Perfluorooctanoic acid (PFOA), its salts and PFOA-related substances are generally banned in the EU and globally, but there are some time-limited exceptions.

This measure will postpone the date of the ban for fire-fighting foams and propose new concentration limits as an unintentional trace contaminant.

Proposed delegated regulation

Initiative entry

Persistent organic pollutants – dechlorane plus

Adoption was planned for Q4 2023

Regulation (EU) 2019/1021 on persistent organic pollutants implements the EU’s international commitments under the Stockholm Convention on persistent organic pollutants (chemical substances). Following a decision to add dechlorane plus to the Convention, this initiative amends Annex I to the Regulation to include dechlorane plus as a substance subject to certain restrictions.

Proposal for Delegated Regulation

Initiative entry

Draft act

Persistent organic pollutants – PFOS limits and exemptions

Adoption was planned for Q1 2023

This act reduces the maximum PFOS concentration allowed as unintentional trace contaminant in substances, mixtures and articles and removes the last specific exemption allowed in the EU since it is no longer needed.

Proposal for Delegated Regulation

Initiative entry

Persistent organic pollutants – UV-328

Adoption was planned for Q4 2023

Following a decision to add UV-328 to the Convention, this initiative amends Annex I to the Regulation to include UV-328 as a substance subject to certain restrictions.

Proposal for Delegated regulation

Initiative entry

Persistent organic pollutants – methoxychlor

 

NOTE: The Commission adopted this regulation on July 22; it is now under EU Parliament and Council scrutiny, after which it will be published in the Official Journal.

Following a decision to add methoxychlor to the Convention, this initiative amends Annex I to the Regulation to include methoxychlor as a substance subject to certain restrictions.

Proposal for Delegated Regulation

Initiative entry

Persistent organic pollutants – polybrominated diphenyl ethers (PBDEs)

Adoption was planned for Q1 2022

This act concerns PBDEs (included in Annex I of Regulation (EU) 2019/1021). It amends the limit value for the presence of PBDEs in mixtures or articles.

Proposal for Regulation

Initiative entry

Related documents

Chemicals legislation – revision of REACH Regulation to help achieve a toxic-free environment

 

This initiative will likely be postponed until the next Commission after June 2024.

The European Green Deal sets out the ambition to reach zero pollution for a toxic-free environment.

As part of this ambition, the chemicals strategy for sustainability announces actions to revise the rules governing the registration, evaluation, authorization and restriction of chemicals in the EU

Proposal for Regulation

Initiative entry

EU chemicals strategy for sustainability – Cosmetic Products Regulation (revision)

Adoption was planned for Q4 2022

 

This initiative will likely be postponed until the next Commission after June 2024.

The EU chemicals strategy for sustainability aims to better protect human health and the environment as well as boost innovation for safe and sustainable chemicals.

This initiative focuses on cosmetic products, in particular on various actions and potential measures to improve the efficiency and effectiveness of the current rules on cosmetic products.

Proposal for Regulation

Initiative entry

Preventing terrorism – new rules on the marketing and use of high-risk chemicals

Adoption was planned for Q3 2023

This initiative establishes rules on the marketing and use of high-risk chemicals.

It aims to increase security in the EU by reducing the risk of dangerous chemicals being acquired by terrorists or other criminals to carry out attacks.

Proposal for Regulation

Initiative entry

Revision of EU rules on food contact materials

Adoption was planned for Q2 2023

 

This initiative will likely be postponed until the next Commission after June 2024.

EU food safety policy includes rules on food contact materials (e.g., food packaging, kitchen and tableware and food processing equipment).

This initiative aims to modernize the rules, to: Ensure food safety and a high level of public health protection; reduce the presence and use of hazardous chemicals; take account of the latest science and technology; support innovation and sustainability by promoting safe reusable and recyclable solutions; and help reduce the sector’s environmental impact.

Proposal for Regulation

Initiative entry

Hazardous substances in electrical and electronic equipment – list of restricted substances (update)

Adoption was planned for Q4 2023

Following a technical assessment, this initiative proposes adding tetrabromobisphenol A (TBBP-A) and medium-chain chlorinated paraffins (MCCPs) to the list of restricted substances.

Proposal for a Delegated Directive

Initiative entry

Protecting workers from exposure to carcinogens, mutagens or reprotoxic substances – codification of Directive 2004/37/EC (CMRD)

Expected Commission adoption not provided

This initiative codifies the 2004 directive, which has been frequently amended over the years, to render this legislation clearer and more transparent.

Proposal for Directive

Initiative entry

Protecting workers against cancer-causing substances at work (CMRD Revision)

 

Adoption planned for Q1 2024

Sixth revision of Directive 2004/37/EC on the protection of workers from the risks related to exposure to carcinogens, mutagens and reprotoxic substances at work.

Latest scientific and technical developments considered for the following chemicals, groups of chemicals or process-generated substances:

– welding fumes
– polycyclic aromatic hydrocarbons
– isoprene
– 1,4-dioxane
– cobalt and inorganic cobalt compounds

Proposal for Directive

Initiative entry

Microplastics pollution – measures to reduce its impact on the environment

Expected Commission adoption not provided

This initiative aims to tackle microplastics unintentionally released into the environment. It will focus on labeling, standardization, certification, and regulatory measures for the main sources of these plastics. It seeks to reduce environmental pollution and potential health risks, while respecting the principles of the single market and encouraging competitiveness and innovation.

Proposal for Regulation

Initiative entry

Hazardous substances – exemption for use of lead to create superconducting magnet circuits in specific devices

Adoption was planned for Q3 2021

This initiative allows lead to be used in metallic bonds to create superconducting magnetic circuits in superconducting quantum interference device (SQUID) detectors.

Proposal for Delegated Directive

Initiative entry

EU fertilizing products – biodegradability criteria for polymers and other technical amendments

 

 

NOTE: The Commission adopted this regulation on July 15; it is now under EU Parliament and Council scrutiny, after which it will be published in the Official Journal.

This initiative aims at introducing biodegradability criteria for polymers (coating agents, water retention agents, mulch films and other polymer-based technical additives) in EU fertilizing products. It also introduces other technical amendments.

Proposal for Delegated Regulation

Initiative entry

 

European Commission ‘Have your say’ consultations closing in August

  • A public feedback period is launched to elicit feedback once the Commission proposes a legal act (e.g., regulation, directive, decision). Below are open consultations related to proposed chemicals legislation that are ending this month.

Issue area or planned legislation

Description

Deadline

Link

Export and import of hazardous chemicals – review of the list of chemicals subject to international trade rules

The Commission intends to update the list of hazardous chemicals subject to certain rules when internationally traded.

The aim is to support non-EU countries in preventing unwanted imports, and to ensure that information on hazards, risks and safe handling is exchanged when these chemicals are exported. Once a chemical has been put on the list, it can only be exported if certain conditions are met.

August 16

Initiative entry

Revision of the EU explosives precursors legislation

This initiative aims to revise the legislation on the marketing and use of explosives precursors. Current law aims to prevent terrorists and criminals from acquiring and using explosives precursors to produce homemade explosives to stage attacks. The revision will address the shortcomings identified by the current evaluation. It will also expand its scope to cover high-risk chemicals which can be misused to stage attacks.

August 20

Initiative entry

To contact the author of this analysis, please email Scott Stephens ( sstephens@agencyiq.com).
To contact the editor of this analysis, please email Kari Oakes ( koakes@agencyiq.com).

Filed Under: Article

August 9, 2024 by

By Amanda Conti

In its inaugural meeting, FDA’s Genetic Metabolic Diseases Advisory Committee heard Zevra’s second try for arimoclomol to treat ultra-rare lysosomal storage disorder Niemann-Pick Type C. The committee voted 11-5 in favor of the drug’s effectiveness, leaving FDA to test the limits of its regulatory flexibility toward post hoc analysis and lackluster confirmatory evidence.

Fill out the form to read the full article.

Filed Under: Article

August 9, 2024 by

FDA’s new metabolic diseases committee gives hesitant nod to ultra-rare disease drug

In its inaugural meeting, FDA’s Genetic Metabolic Diseases Advisory Committee heard Zevra’s second try for arimoclomol to treat ultra-rare lysosomal storage disorder Niemann-Pick Type C. The committee voted 11-5 in favor of the drug’s effectiveness, leaving FDA to test the limits of its regulatory flexibility toward post hoc analysis and lackluster confirmatory evidence.

By Amanda Conti | Aug 6, 2024 7:22 PM CDT

Background: Niemann-Pick Type C (NPC)

  • Niemann-Pick disease Type C (NPC) is an autosomal recessive lysosomal storage disorder that typically results in progressive neurologic symptoms and organ dysfunction. NPC is caused by variants in the genes encoding the NPC1 or NPC2 proteins, which are involved in cholesterol metabolism. Without functional versions of these proteins, lipids accumulate in tissues, according to the National Institute of Neurological Disorders and Stroke (NINDS).
  • NPC is an ultra-rare disease, with prevalence estimates ranging from 0.35 to 2.2 per 100,000 births across countries. Although it is traditionally considered a childhood-onset disease, some patients display NPC later in life.
  • Symptoms and outcomes vary considerably. NINDS explains that the associated neurological complications can include extensive brain damage that can cause movement difficulty and progressive loss of vision and hearing, among other symptoms. Generally, diagnosis earlier in life as well as having certain genotypes forecasts more severe disease.
  • The heterogeneity of the disease, and its slow progression, have challenged therapeutic development. The FDA has participated in several discussions on how to overcome these obstacles. At a January 2022 workshop on NPC endpoint development, the FDA’s Director of the Center for Drug Evaluation and Research (CDER), PATRIZIA CAVAZZONI, explained that endpoints used in trials for ultra-rare conditions such as NPC may not reflect disease presentation or clinically meaningful improvements for all patients. In addition, “there are only a limited number of patients with NPC who are available to participate in clinical trials and this may restrict the study design,” Cavazzoni explained.
  • The NIH-developed NPC Clinical Severity Scale (NPCCSS) used in most trials has some issues, according to participants in an August 2022 webinar that followed the January workshop. The scale measures clinician-reported outcomes in 17 domains, though the five most important domains are used in the abbreviated 5-domain NPC Clinical Severity Scale (5DNPCCSS). These domains are Ambulation, Swallow, Cognition, Speech, and Fine Motor Skills. At that webinar, JACKIE KARP, lead physician at the FDA’s Division of Rare Diseases and Medical Genetics (DRDMG), outlined validity concerns with the scale’s cognition domain for short trials because the “rate of decline does not align with trial durations.” [ Read full AgencyIQ analysis here.]
  • As a result of these challenges, there are currently no approved therapies for NPC in the U.S. However, miglustat (brand name: Zavesca), which was approved by the EMA to treat NPC in 2002, has been reported to be used off-label in the U.S. to help preserve swallowing function in NPC.

Regulatory context: Arimoclomol aims to be the first NPC therapy to receive FDA’s nod

  • Arimoclomol is an oral small molecule that is intended to amplify the body’s natural heat shock response (HSR); the drug crosses the blood-brain barrier. This is thought to prevent protein misfolding and facilitate lysosomal function, which in turn prevents cell death associated with NPC.
  • In June 2021, Orphazyme received a Complete Response Letter (CRL) in response to its first New Drug Application (NDA) filing for arimoclomol. Side note: The product was acquired by KemPharm, Inc., in 2022, which changed its name to Zevra Therapeutics in 2023.
  • That application relied on data from a single prospective randomized, double-blind, placebo-controlled, phase 2/3 study (NCT02612129). Patients were randomized 2:1 to receive arimoclomol or placebo as an add-on therapy to their current treatment. According to the results published in the Journal of Inherited Metabolic Disease, the 12-month study enrolled 50 patients ranging from age 2 to 18. “Given the ultra-rare condition, and expected rate of progression, sample size and trial duration were primarily informed by feasibility and not by a formal sample size calculation,” the paper states. Forty-two patients completed the study.
  • The study reported modest but positive results in its primary endpoint, which was change in the 5DNPCCSS score from baseline to 12 months. The study found a treatment difference of −1.40 points (95% confidence interval [CI], −2.76 to −0.03; P = 0.046) favoring the arimoclomol group. In terms of safety, the researchers judged that, “The majority of TEAEs [treatment-emergent adverse events] were assessed by the investigators to be related to NPC rather than the investigational product.”
  • According to a firm presentation, the CRL highlighted three information gaps: (1) evidence needed to support use of the NPCCSS as the primary instrument in measuring NPC disease progression, (2) additional analysis related to how missing data are handled for statistical analysis, and 3) additional support and data related to confirmatory evidence of efficacy. According to the presentation, “FDA did not request additional efficacy data in the CRL.”
  • The presentation also summarized the discussion at a Type A End-of-Review Meeting that took place in October 2021. The firm writes that at the meeting, the agency agreed to three actions: (1) reanalysis of the 5DNPCCSS with the removal of the cognition domain, (2) rescoring and a reassessment of the swallowing domain including further validation, and (3) further discussions regarding NPCCSS.
  • Zevra resubmitted the application in December 2023. While the application initially had a target PDUFA date of June 21, 2024, an updated Zevra press release explained that the subsequent review and associated information requests necessitated an extension to September 21, 2024. In the same press release, Zevra stated the agency’s intention to convene an advisory committee meeting regarding the application.

On August 2, FDA’s Genetic Metabolic Diseases Advisory Committee (GeMDAC) heard the case for arimoclomol’s second chance

  • An upfront note: This was the first meeting of the newly minted GeMDAC. The committee was announced in December 2023, with a charge to provide “independent, knowledgeable advice and recommendations on technical, scientific and policy issues around medical products for genetic metabolic diseases.” [ Read AgencyIQ’s analysis of the announcement here.]
  • The committee has slowly staffed up, with the roster indicating that six of its 10 spots have been filled. The August 2 meeting featured a significant number of temporary members. Temporary member ROBERT ALEXANDER, a psychiatry professor from the University of Arizona, served as the Acting Chairperson of GeMDAC. Alexander is no stranger to the agency’s advisory committee system, having previously served as the chair of the Peripheral and Central Nervous System Drugs Advisory Committee (PCNS) in an acting capacity.

The post hoc reanalysis

  • As suggested by FDA, Zevra completed a post hoc reanalysis of the clinical trial results, with the primary endpoint changed from the 5DNPCCSS to a rescored 4-domain Niemann-Pick disease type C Clinical Severity Scale (R4DNPCCSS). This meant that the cognition domain was omitted from the score. NAOMI KNOBLE, Associate Director of FDA’s Division of Clinical Outcome Assessment, explained that published natural history study data showed that, “…it would take more than one year to observe changes in the NPCCSS Cognition domain score, making it unsuitable to show stabilization and/or changes as part of the primary efficacy endpoint. Ratings were also dependent on the patient’s environment, such as whether they were receiving educational services, which is not a clear indicator of cognitive functioning.” In addition, the agency raised concerns that the original swallow domain did not accurately reflect linear progression of the disease. For example, the original rubric did not differentiate between patients who require feeding tubes sometimes and all the time. The swallowing domain was therefore rescored based on consultation with experts.
  • During Zevra’s presentation on the clinical background of NPC, Mayo Clinic child neurologist MARC PATTERSON further contextualized the endpoint. He pointed out that there are no established surrogate endpoints or measurable biomarkers for NPC. “It’s important to underscore that the NPC clinical severity scale is the only validated and appropriate tool to specifically measure Niemann-Pick disease type C severity and progression across multiple domains,” he said. He also provided perspective on its real-world use, saying, “We must bear in mind that we are dealing with neurologically impaired children across a broad range of ages. If you do not actually spend time sitting in an examination room with a child, particularly a neurologically impaired child, it may seem very reasonable to use a large number of scales to try to assess different aspects of the disease, but it’s important to recognize that is not a realistic goal for most children with NPC.”
  • In addition, the reanalysis addressed missing data called out in the CRL. The initial analyses excluded data from one patient who died and other patients who took an “early escape” route due to rapid disease progression. “It is notable that the excluded data indicated disease worsening,” stated the agency. Various imputation methods were used to account for these data in the post hoc analysis.
  • Taken together, these changes resulted in modest treatment differences favoring the arimoclomol arm, with effect sizes ranging from -1.5 to -1.2 points depending on the specific methods used. Importantly, FDA’s presentation states that “These analyses provide smaller treatment difference estimates and wider confidence intervals compared to those from the prespecified analysis.”
  • Several committee members expressed reservations regarding post hoc analyses in general. KENNETH FISCHBECK, Distinguished Investigator Emeritus at NINDS commented, “My understanding from statisticians is that post hoc analysis [is] not statistically valid,” but also acknowledged that this was done at the request of the FDA. Zevra emphasized that the prespecified analysis reached statistical significance and favored arimoclomol, and that the new analysis did not change the direction of the results.

The confirmatory evidence

  • Quick background: When seeking approval from the FDA, companies must demonstrate “substantial evidence,” as defined under the Federal Food, Drug, and Cosmetics (FD&C) Act, that their product is safe and effective. Traditionally, FDA has interpreted the need for “well-controlled investigations” to mean at least two clinical trials for applications for new drugs or supplemental indications. Over time (with the help of litigation and the passage of the 1997 FDA Modernization Act (FDAMA)), the definition has been updated so that one “adequate and well-controlled investigation” can be sufficient to demonstrate safety and effectiveness if it is supported by “confirmatory evidence.”
  • FDA released draft guidance in September 2023 describing data sources that could serve as confirmatory evidence and the situations in which they would be appropriate. Importantly, the agency stated that the volume of confirmatory evidence needed is inversely proportional to the strength of the single adequate and well-controlled study. In addition, the single trial and confirmatory evidence are intended to be “considered together” to assess effectiveness. [ Read AgencyIQ’s complete analysis of the guidance here.].
  • Over time, the FDA’s acceptance of single pivotal trials has become more common, especially given the increased submission of applications for products intended to treat, cure or prevent rare diseases or life-threatening conditions for which there is an unmet clinical need. In a recent analysis, AgencyIQ found that the majority of new molecular entities approved each year since 2020 have relied on a single pivotal trial.
  • At the meeting, GeMDAC considered Zevra’s bolstered confirmatory evidence package, which consisted of results from the Open-Label Extension (OLE) phase of the original trial, plus additional nonclinical studies.
  • Forty-one of the 50 total participants in the initial double-blind trial chose to continue to the OLE phase. Twenty-nine of those patients ultimately completed 48 months of treatment in the extension. Withdrawal reasons included caregiver preference, adverse events, death, and physician decision. The data alone “appears to show relative slowing/stability of disease progression with arimoclomol,” though a subset of patients declined rapidly on treatment. No single common characteristic was identified in the rapidly advancing subset, though factors such as early symptom onset and double-null mutations were explored as potential drivers.
  • Given the lack of a control group in the OLE phase of the study, the data were compared to NIH natural history study data as an external control. Participants from the two groups were matched based on variables including baseline age, sex, and miglustat use. FDA reported that these comparisons “numerically favored arimoclomol” without reaching statistical significance.
  • Multiple committee members, including Chair Alexander, were concerned that the OLE and natural history study represented an “apples to oranges” assessment given the updates to the endpoint made in Zevra’s study. Zevra clarified that it was able to convert the NIH data to the four-domain scale to enable comparison of scores. However, the rescoring of the swallowing domain could not be performed using the NIH data. As a result, Zevra confirmed that the same four-domain methodology was used on both groups.
  • The CRL described the original nonclinical data as “weak and contradictory,” and Zevra conducted additional work in this area for the resubmission. The new nonclinical studies consisted of in vitro mechanistic data and in vivo studies in two mouse models.
  • Arimoclomol is thought to work by amplifying the body’s natural HSR, and Zevra hypothesizes that this is done via upregulation of genes within the Coordinated Lysosomal Expression and Regulation (CLEAR) network. Specifically, arimoclomol is hypothesized to activate specific transcription factors that bind to CLEAR gene promoters during cellular stress response. The new cell culture studies in the resubmission demonstrated that arimoclomol treatment leads to small increases in nuclear localization of these transcription factors, as well as increased expression of CLEAR genes. However, these results were observed when the drug was given at 30 to 60 times the human clinical dose. Additional pharmacodynamic biomarker studies were limited due to their non-specificity to NPC and arimoclomol.
  • The new animal studies involved mouse models of both gene interruption and point mutation, which are akin to infantile and delayed onset forms of NPC, respectively. At a high level, FDA described the results as “small, variable, and lacked a dose-relationship and/or failed to repeat when re-tested.” The studies assessed survival, motor function, and biochemical endpoints. Of note, the arimoclomol doses were administered through drinking water. FDA’s Acting Lead Pharmacologist SHAWNA WEIS explained that “…the applicant did not measure water consumption or [pharmacokinetics] to evaluate either the dose delivered or the exposures achieved, which make these data very difficult to interpret.” The GeMDAC members picked up on additional potential issues related to dosing arimoclomol in drinking water, as NPC itself is associated with swallowing difficulties that could further influence the dose.

The two discussion questions and the vote

Discussion Question: Discuss your assessment of the efficacy results of trial CT-ORZY-NPC-002 (NPC002). In your discussion, please comment on: (a) The 5-domain Niemann-Pick disease type C Clinical Severity Scale (5DNPCCSS) and the rescored 4-domain Niemann-Pick disease type C Clinical Severity Scale (R4DNPCCSS), and (b) Your assessment of whether the trial results demonstrate a treatment effect of arimoclomol on the treatment of Niemann-Pick disease type C (NPC).

  • In general, the committee felt that the severity scales represented the best available option for an endpoint to assess efficacy. That said, committee members envisioned improvements to the scale in terms of increasing its utility, either by incorporating additional aspects of the full 17-domain scale or by increasing the granularity of the ratings within each domain. CHERYL COON, Vice President of the Clinical Outcome Assessment Program at the Critical Path Institute, summarized, “…We should not let perfect get in the way of good enough. And I think in this case, this is good enough. We can certainly get information out of this instrument as it’s administered.”
  • The committee agreed that the trial results showed a small, incremental treatment effect, but members had varying opinions on the meaning and reliability of this effect. For some, the effect represented stabilization, which can be meaningful for progressive diseases. Consumer Representative SARAH CHAMBERLIN expanded on this, saying that “…that one-point difference to them is a bonus on top of what they’re really looking for—a treatment to keep their family members from getting worse than what they are.” On the other hand, concerns stemmed from the sample size of the study, which was necessarily small in the ultra-rare disease context. Notably, changes in a small number of patients contributed to large changes in the overall treatment effect.

Discussion Question: Discuss your assessment of other data (specifically the additional clinical and nonclinical data) with respect to support for the effectiveness of arimoclomol.

  • “It’s all approximately the same,” said WENDY CHUNG, chief of pediatrics at Boston Children’s Hospital, who felt reassured by the similarity in the additional data regarding a small effect size and lack of new safety concerns. “For me, it was at least consistent internally,” she said. This sentiment was echoed by others. SUSAN ELLENBERG, professor emerita of biostatistics, medical ethics and health policy at the University of Pennsylvania, explained that while the new data did not help much, they also did not detract from the clinical trial results.
  • Thomas Jefferson University Professor WALTER KRAFT pointed out that the lack of safety signals could have positive or negative ramifications. He spoke to the drug’s hypothesized mechanism of action, noting that a small molecule working through transcription factors would likely have off-target effects. “So, the fact that we don’t have side effects actually can be a little bit worrisome,” he said.
  • Other committee members poked holes in the rigor and value of the additional clinical and nonclinical data. JEAN BAPTISTE LE PICHON from the University of Missouri-Kansas City pointed out that the NIH registry used as an external control included patients on a variety of treatment regimens, saying, “I don’t think it’s a comparison that should have happened.”
  • Several members were frustrated by the lack of translatability of the mouse studies to humans. University of Michigan neuropathologist ANDREW LIEBERMAN was concerned by the need to use a much higher dose than that in humans to see effects in mice. Fischbeck felt that the data were limited because the treatment in mice was started prior to disease manifestation, which does not typically occur in humans. Chung found some utliity in these data, referencing family perspectives discussed during the public hearing regarding multiple siblings with NPC. “It was interesting through that lens to see the data on the mice in terms of what might be going on in the future,” she said.

VOTING QUESTION: Do the results of trial NPC-002 in concert with the other data (clinical and nonclinical in particular) support a conclusion that arimoclomol is effective in the treatment of patients with NPC? Provide a rationale for your vote. If you voted no, provide recommendations for additional data that may support a conclusion that arimoclomol is effective.

  • The committee voted 11-5 in favor of arimoclomol’s effectiveness for NPC, with no abstentions. Prior to the vote, FDA’s Acting Director of the Division of Rare Diseases and Medical Genetics, CATHERINE PILGRIM-GRAYSON, clarified that the question is asking about arimoclomol “as it happened in the clinical trial,” meaning as an add-on treatment to the background or standard-of-care therapy
  • The committee members who voted yes saw a small but beneficial treatment effect in the totality of the data. Le Pichon summarized, “It was not an enormous effect, but it was an effect that is clinically significant.” Duke Professor of Pediatrics PRIYA KISHNANI highlighted the importance of stabilization in the context of a progressive disease, and Ellenberg was particularly impressed by the improvement observed in a small group of patients, which she believes makes the treatment worthwhile to pursue.
  • Other considerations contributing to “yes” votes included arimoclomol’s safety profile and the critical unmet need. Several members were assuaged by the safety profile, especially over the duration of the extension phase of the study. Retired Child Neurologist JOHN MINK explained the reasoning for his “very reluctant yes,” saying, “…the bulk of the data favored a slightly positive effect. I think the unmet need is very clear. I’m not sure that this meets that need, but again, on balance, I voted yes.”
  • On the other hand, the “no” votes could not see past “problematic” aspects of the data. While most agreed that the clinical data suggested a modest effect, the confirmatory evidence was not compelling enough to support an overall conclusion. These members found noteworthy deficiencies in the data from the mouse models. Kraft assumed the regulator perspective, stating his view that the resubmission package “did not meet the evidentiary standards for approval.” He raised concerns about downstream effects, saying, “I do worry about approval of drugs for which there is not unequivocal evidence of efficacy that is not without harm, also in terms of diversion of resources and activity within the space.” Regarding next steps, the “no” votes pushed for more rigorous nonclinical studies.

Analysis

  • FDA has more than six weeks to draw its final conclusion, with a Zevra press release indicating an action date of September 21, 2024, for the application. The agency is not required to align with the opinion of its advisors, and the mixed result of the vote makes FDA’s next steps less clear. FDA, the sponsor and the advisory committee agreed that NPC represents an area of high unmet need, but the first application’s CRL indicates that this alone is not enough to get arimoclomol across the finish line. While the additional confirmatory evidence was lackluster, it is notable that most advisors found the strength of the entire evidence base to be somewhat greater than the sum of its parts.
  • If arimoclomol is ultimately approved, Zevra will receive a valuable voucher. The product has received Rare Pediatric Disease designation, meaning it is eligible for a priority review voucher (PRV) upon approval. PRVs, which can be redeemed for priority review of another product application, are a highly valuable commodity. Because the vouchers offer the potential for a company to bring their drug to market up to four months faster, they offer the potential to accelerate commercial sales, make use of more of a product’s useful patent life, and cement a lead for a product before a competing product can come to market. Individual vouchers regularly sell in excess of $100 million. [ See AgencyIQ’s Priority Review Voucher Tracker here.] The vouchers may rise further in value, as the program is set to expire at the end of September unless it is reauthorized by Congress.
  • Back to the basic (science). A major takeaway from the meeting was that the agency and advisors struggled with issues stemming from an unclear mechanism of action and poorly designed nonclinical studies. While this reflects the brisk pace of development in the rare disease space, it may continue to throw a wrench in arimoclomol’s approvability. This sequence of events also underscores the importance of translational and regulatory science research to identify appropriate animal models and biomarkers.
  • This meeting adds an interesting use case to FDA’s ongoing discussions related to the use of external controls. In late 2023, FDA finalized guidance offering extensive new recommendations on considerations for the development of drugs and biologics for rare diseases. That document expressed the agency’s willingness to accept external controls in cases where there are unmet medical needs, a well-documented and highly predictable disease course, and a large drug effect [ Read full AgencyIQ analysis here.]. The ultra-rare NPC certainly checks the unmet need box, though its course is notoriously heterogeneous. It remains to be seen how arimoclomol’s modest drug effect will factor into this calculus.

To contact the author of this item, please email Amanda Conti ( aconti@agencyiq.com).
To contact the editor of this item, please email Jason Wermers ( jwermers@agencyiq.com).

Key Documents and Dates

  • August 2, 2024: Meeting of the Genetic Metabolic Diseases Advisory Committee (meeting materials and recording linked here)
  • FDA Docket No.: FDA-2024-N-2930

Filed Under: Article

August 9, 2024 by

By Laura DiAngelo, MPH

Last month, the FDA published a new draft guidance document on diversity action plans. While the new version of the guidance is greatly expanded as compared to an earlier version from 2022, it has left plenty of important questions unanswered. In this piece, AgencyIQ answers some of the most common questions we’ve received about the guidance – and raise a few of our own.

Fill out the form to read the full article.

Filed Under: Article

August 9, 2024 by

FDA’s Diversity Action Plan: Questions, answers, and what we (don’t) know so far

Last month, the FDA published a new draft guidance document on diversity action plans. While the new version of the guidance is greatly expanded as compared to an earlier version from 2022, it has left plenty of important questions unanswered. In this piece, AgencyIQ answers some of the most common questions we’ve received about the guidance – and raise a few of our own.

By Laura DiAngelo, MPH | Aug 2, 2024 6:30 PM CDT

A refresher on FDA’s Diversity Action Plan (DAP) guidance:

  • In April 2022, the FDA issued a draft guidance document on diversity in clinical research programs. At a high level, the guidance laid out a policy whereby sponsors of certain products would voluntarily submit a Race and Ethnicity Diversity Plan as part of their development program that would outline and justify their approach to recruiting and retaining a “representative” research population. [ Read AgencyIQ’s extensive analysis of the draft guidance here.]
  • In December 2022, as part of the Consolidated Appropriations Act, 2023, Congress provided the FDA with new authority to require what the law referred to as Diversity Action Plans (DAPs) for certain clinical studies. Section 3601 of the law amended the Federal Food, Drug and Cosmetic (FD&C) Act to expressly grant the FDA the statutory authority to require DAPs for 1) Drugs: for “a new drug that is in a phase 3 study” or “as appropriate, another pivotal study of a new drug (other than bioavailability or bioequivalence studies)”; 2) Medical devices: As part of an Investigational Device Exemption (IDE) submission or “for any clinical study.”
  • DAPs are a plan, developed by the product sponsor, that would need to include specific enrollment goals, the rationale by which these goals were developed, and “an explanation of how the sponsor intends to meet such goals.” The statutory provision also allows for certain research programs to be exempted from the DAP requirement through a waiver system, which could be initiated by the FDA or “at the request of a sponsor.”
  • The law also directed the FDA to “update or issue guidance” on DAPs. Per Section 3602 of the Consolidated Appropriations Act, this guidance should include information about the specific format and content of the plans, such as the rationale for enrollment goals (e.g., estimated prevalence of the condition of interest, any relevant pharmacokinetic or pharmacogenomic data, demographic factors) and how they intend to meet those goals, as well as operational and process factors such as how they should be submitted and how the “action plan” may be updated over time.
  • The agency issued a new draft guidance on DAPs in June 2024, several months after the Congressional deadline had passed. While the new draft guidance document and FDA’s original 2022 draft guidance document share a federal docket – meaning that the comments submitted on the original 2022 draft guidance are in the same online folder that comments on the new draft will be submitted to – the new draft is essentially a complete rewrite of the 2022 version, and not a simple revision.
  • A quick recap of the draft guidance: The new June 2024 draft guidance describes the various elements of a DAP (enrollment targets, rationale to justify those targets), the basics of their formatting, and how to submit a DAP to the FDA – including both the information they expect and when/how those documents should be sent in. The guidance also includes a section describing the process for sponsors to seek a waiver from the DAP requirements. Notably, FDA continues to maintain that DAP waivers should be “rare” – previewing its expectations that it will receive just five waiver requests for drugs/biological products and five waiver requests for medical devices each year. [Read AgencyIQ’s full analysis of the July 2024 DAP draft guidance here.]

Answering key questions about the Diversity Action Plan guidance document

  • Although the 2024 draft guidance on DAPs includes significantly more detail than the 2022 version, there are still significant outstanding questions that it leaves unanswered.
  • Following a recent AgencyIQ webinar, we collected dozens of questions from our subscribers and webinar attendees to put together this list of commonly-asked questions, as well as our attempts to answer them using available information. We’ve also collected additional questions that we have about the guidance.

What programs or products need a DAP?

  • According to the guidance, the following drug studies need a DAP: Clinical investigations of a new drug “that is a phase 3 study” or “another pivotal” clinical study except for bioavailability or bioequivalence (BA/BE) studies will need a DAP. For these studies, the DAP should be submitted to the relevant Investigational New Drug (IND) “as soon as practicable but no later than the date on which the sponsor submits the protocol to FDA for the phase 3 study” or “another” pivotal study.
  • What does that mean: “Phase 3”? The actual legal definition of a Phase 3 study can be found at 21 CFR 312.21, and the DAP authority references this specific definition: “Phase 3 studies are expanded controlled and uncontrolled trials. They are performed after preliminary evidence suggesting effectiveness of the drug has been obtained, and are intended to gather the additional information about effectiveness and safety that is needed to evaluate the overall benefit-risk relationship of the drug and to provide an adequate basis for physician labeling. Phase 3 studies usually include from several hundred to several thousand subjects.” The agency may also require a DAP for an “other pivotal clinical study” of any kind (besides a BA/BE). However, it doesn’t go into depth about what studies, exactly, they mean here – beyond its function as either a Phase 3 or “another pivotal study.”
  • What does that mean: a “new drug”? As defined in statute, the DAP requirement for drug products applies to clinical studies for a “new drug.” The statute does not offer a specific definition for new drug, although the FDA has its own extensive definitions of the term (21 CFR 310) that have been set by previous legislation. Per those regulations, the “newness of a drug may arise by any reason (among other reasons) of: (1) the newness for drug use of any substance which composes such drug, in whole or in part, whether it be an active substance or a menstruum, excipient, carrier, coating, or other component. (2) The newness for a drug use of a combination of two or more substances, none of which is a new drug. (3) The newness for drug use of the proportion of a substance in a combination, even though such combination containing such substance in other proportion is not a new drug. (4) The newness of use of such drug in diagnosing, curing, mitigating, treating, or preventing a disease, or to affect a structure or function of the body, even though such drug is not a new drug when used in another disease or to affect another structure or function of the body. (5) The newness of a dosage, or method or duration of administration or application, or other condition of use prescribed, recommended, or suggested in the labeling of such drug, even though such drug when used in other dosage, or other method or duration of administration or application, or different condition, is not a new drug.”
  • In guidance, FDA has noted that it has “recognized, and courts have accepted, that drug can be interpreted, among other possible meanings, to mean ither drug product or drug substance” and that because the definition is broad, the statute “delegates to FDA the task of determining how to apply the definition in particular statutory provisions.” However, it’s possible that its interpretations may be on shakier ground following the landmark Supreme Court decision in Loper Bright earlier this year, which determined that courts are not obligated to defer to FDA judgments. In other words, it’s possible that companies may disagree with FDA regarding the “newness” of their drug products – for example, whether certain studies intended to inform new indications, formulations, or other adjustments to an approved drug product may cross the line into being a “new” drug and therefore need a DAP.
  • Per the guidance, the following device studies need a DAP: Investigations for products under an Investigational Device Exemption (IDE), as well as studies that are not under an IDE but are intended to be the primary basis of evaluation by the FDA; no IDE-exempt study is required to submit a DAP. For products under an IDE, DAP information must be submitted with the IDE. For products not under an IDE but that are not IDE exempt and are intended to serve as the primary basis of FDA’s review, the DAP must be submitted with a marketing application for the product.
  • What about combination products? Combination products are defined by the FDA as: “A product comprised of two or more regulated components (i.e., drug/device, biologic/device, drug/biologic, or drug/device/biologic” that are either combined/mixed into a single entity or are co-packed or cross labeled. In general, combination products generally do not need separate regulatory authorizations for each constituent part, but are authorized under a marketing application for the part that provides the primary mode of action (PMOA) of the combination product; the part that provides the PMOA is considered to “lead” the combination product. The DAP guidance does not include any information about combination products – this will likely need to be addressed by the FDA in future clarifications. From regulatory precedent, it seems likely that the way that a DAP would be submitted for a combination product would follow the rules of the “lead” part; in effect, a DAP would be submitted under the IND for a drug-led combination product, or the IDE or in the marketing application of a device-led combination product. However, it’s not entirely clear at this point how that would affect enrollment target setting or rationale.
  • What about companion diagnostics (CDx)? CDx are diagnostic products that are “essential for the safe and effective use of a corresponding therapeutic product” and are authorized to reference that/those therapeutics. Unlike combination products, CDx are authorized separately from the associated drug product, and need their own marketing application. However, depending on study design, the investigations into a CDx might not need their own investigational submission. While investigations of CDx typically do require an IDE application to the FDA (and therefore require a DAP per statutory requirements), a sponsor “may seek to submit an IND alone, or both an IND and an IDE.” In the case that a CDx is included in the IND (rather than getting its own IDE), it’s not clear if the DAP requirement for the CDx as a standalone product would be waived out, or whether the sponsor would need to submit a DAP for the CDx to the IND. Notably, there are significant developments on CDx policy expected in the next few years, and it’s not clear if clarification needed in this are will come in a DAP guidance or in a CDx-specific guidance (or if it’s something the agency has thought through just yet).

Global trials, enrollment targets, and how to account for a DAP population

  • Quick definition: What are the enrollment goals? The DAP is comprised of three basic content elements: Enrollment goals, the rationale for the enrollment goals, and the measures to meet enrollment goals. FDA’s 2024 draft DAP guidance includes an Appendix summarizing these three elements. The section of the guidance on enrollment goals begins on page 11 of the document. The enrollment goals are the baseline framework underlying a DAP; in effect, they’re intended to break down the sponsor’s “goals” in enrolling a participant population by demographic subgroups.
  • How to set enrollment goals: the U.S. population should be the baseline. “Generally, enrollment goals should be informed by the estimated prevalence or incidence of the disease or condition in the U.S. intended use population for which the medical product is being studied” (lines 232-234). More specifically: “The estimated prevalence or incidence of the disease or condition by demographic characteristics in the U.S. population for which the medical product is being investigated should generally inform enrollment goals” (lines 267-269). In effect: The enrollment goals should be focused solely on the intended use population in the U.S.
  • What does this mean for global, or multinational, trials? The agency acknowledges “the importance of global medical product development and supports the use of well-designed and conducted multi-regional clinical studies, when appropriate.” However, the DAP requirements still apply for multinational trials.
  • The guidance offers some information on what DAPs for multinational trials will look like: These recommendations can be found at lines 295 to 318 of the guidance. While the setting of enrollment goals should be based on the U.S. intended use population, the DAP’s enrollment goals “must describe participant enrollment goals for the entire study and should not be limited to U.S.-enrolled participants” – in effect, the enrollment goals themselves should be set using U.S.-specific data to inform the goals themselves, and then should incorporate the participation enrollment for the entire study, including multi-national geographies. Or, as the FDA explains at line 243, “the sponsor’s enrollment goals specified in the Diversity Action Plan for each study should consider how individual clinical studies may fit into an overall clinical development program for the medical product (i.e., for a particular indication or intended use), and how such individual studies should help generate data representing the clinically relevant population’s demographic characteristics consistent with the incidence or prevalence in the disease population for the program.”
  • How should sponsors of global trials collect data on patient demographics? As the guidance acknowledges, sponsors are expected to collect DAP data using U.S.-specific categorizations of race and ethnicity (as defined by OMB Directive 15). However, this is likely to be challenging – something noted in the guidance. “FDA recognizes that the lack of uniformity across the globe in the use of population descriptors across the globe in the use of population descriptors such as race and ethnicity may pose challenges when setting enrollment goals for international sites” the guidance states (lines 305-307). In other words: The challenge is that sponsors need to describe the enrollment of non-U.S. participants using demographic descriptors that may not be available (or used, or legally permitted to be collected) in the data in that region.
  • What data can be – or shouldn’t be – used to set enrollment targets? As mentioned above, demographic information must be in the U.S. formatting for racial and ethnic categories (a subject on which more clarity would be welcome). Further, the agency urges sponsors to use “appropriate available sources” – such as certain registries, “publicly available” epidemiological surveys or published literatures – but notes that “non-publicly available sources” can also be used. In these cases, sponsors should “provide the rationale for the approach, a synopsis of the analysis used, and citations for the source(s) for these data.” So far, it’s not yet clear what level of granularity the agency will be looking for in information about non-public sources.

Enforcement and post-market implications:

  • A key point of the statutory authority: FDA has the authority to require the submission of DAPs, but it’s less descriptive about what happens after that. The statutory authority to FDA that was granted under FDORA to require DAPs is limited to their submission – not their completion or FDA’s enforcement of specific provisions.
  • How might FDA enforce DAPs? One option: Refuse to accept, refuse to file. The first consideration is that, upon the implementation of this policy, DAPs will be a required component of certain submissions and therefore, will be necessary for the FDA to accept or file the submission. In effect, missing a DAP would be missing a required element of the submission, and therefore the application would theoretically not pass (appropriate, by submission) technical review and not be accepted or filed by the FDA.
  • What happens if you don’t meet your goals? That is a significant question, and not addressed in the guidance. As AgencyIQ has previously discussed, the 2022 version of the draft guidance (which was not backed up by statutory authority) included the following footnote, which was the subject of much discussion in comments on the document: “In the event that recruitment goals are not met despite best efforts, sponsors should discuss with FDA a plan to collect this data in the post-marketing setting” (footnote 24). The new 2024 version of the document, notably, does not include any information on the subject, and does not address the question of what happens if studies under DAPs are unable to reach their goals in the pre-market setting.
  • There is one place where “postmarketing clinical studies” are mentioned: Medical device Post-Approval Studies (PAS) and section 522 studies. In footnote 31 of the new draft guidance, the agency states that “although medical device postmarketing clinical studies are outside the scope of this guidance, FDA considers diversity and representative patient enrollment to be important in postmarketing clinical studies of devices.” As AgencyIQ noted in analysis of the 2022 version of the diversity plan guidance, the framework for the diversity plans in these guidance documents aligns operationally with agency guidance on these two study types that were drafted just a year before the original diversity plan guidance (PAS guidance, 522 guidance), and were finalized in October 2022. These include similar elements like the establishment of pre-study enrollment goals, the requirement that sponsors/manufacturers provide rationale on how they intend to meet those goals, and time-based milestone check-ins.
  • However, those two guidance documents go further than the DAP guidance does (even the 2024 version), recommending specific enrollment milestones (e.g., first subject enrolled within 6 months of the date that the study protocol is approved, 20% enrolled by 12 months). The new DAP draft guidance does not include any such specific expectations, but could preview work in future years should the agency raise similar concerns with DAPs as it did with PAS/522 studies not being sufficiently enrolled. Further, it’s not entirely clear what impact, if any, a DAP will have on enrollment goals under a PAS/522.
  • For devices, will this be an eSTAR module? In the U.S., most medical device submissions are now either required to be submitted, or can be voluntarily submitted, via the electronic submission template and resource (eSTAR). If and when DAPs become required for certain medical device submissions, the content would either need to be built into the template or the FDA would need to provide more information on where it goes. For example, the guidance states that DAPs for drugs under an IND would need to be submitted in eCTD module 2.5 (Clinical Overview).

Timeline, analysis, and what’s next

  • When should we expect the guidance to be finalized? For now, the guidance comment period runs through September 2024; it’s not yet clear if the FDA will extend the comment period. If it doesn’t, there’s a quick turnaround on getting the guidance finalized. Under FDORA (and notwithstanding that the agency blew past the 12-month timeline for a draft guidance), the final guidance would theoretically be due in June 2025.
  • When would the guidance go into effect? Under the statute, the DAP requirement would go into effect for studies “for which enrollment commences after 180 days from the publication of the final guidance” – assuming the final guidance is issued on time, this would be December 2025.
  • Are there any exemptions? The agency says “It does not expect” a DAP for certain projects in three specific scenarios: 1) when a drug product’s protocol is submitted “within 180 days” after a final guidance is published and in which enrollment will begin in the 180 days (i.e., during the implementation period); (2) device studies for which an Investigational Device Exemption (IDE) is received within 180 days of the publication of the final guidance; and (3) device studies for which an IDE is not required but which are approved by an institutional review board (IRB) or independent ethics committee (IEC) within 180 days of the final guidance publication.
  • There are significant remaining operational questions, such as who at FDA will be reviewing DAPs (and whether reviews will differ by review division), what FDA enforcement will involve, the mechanics of these submissions, and implications for combination products and CDx. Overall, it’s likely that this new draft guidance will garner significant feedback from industry, particularly on how to interpret some of the more ambiguous sections.
  • Can sponsors submit a diversity plan in advance of final guidance? Yes, although these would presumably count as voluntary diversity plans (per the 2022 draft guidance), rather than DAPs (which are statutorily required under a provision of the law that has not yet been implemented). This might give sponsors a little bit more time to get a feel for how the FDA will be interpreting key provisions in advance of when submission would be required.

To contact the author of this item, please email Laura DiAngelo ( ldiangelo@agencyiq.com)
To contact the editor of this item, please email Alexander Gaffney ( agaffney@agencyiq.com)

Key Documents and Dates

  • FDA just published its Diversity Action Plan guidance. Here’s what you need to know.
  • Diversity Action Plans to Improve Enrollment of Participants from Underrepresented Populations in Clinical Studies

Filed Under: Article

  • « Go to Previous Page
  • Go to page 1
  • Interim pages omitted …
  • Go to page 3
  • Go to page 4
  • Go to page 5
  • Go to page 6
  • Go to page 7
  • Interim pages omitted …
  • Go to page 21
  • Go to Next Page »
AgencyIQ by POLITICO
  • About POLITICO
  • POLITICO Pro
  • E&E News by POLITICO
  • Privacy Policy
  • Terms of Service
  • Do Not Sell or Share My Personal Information
LinkedIn
© POLITICO, LLC