• 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 23, 2024 by

What’s on FDA’s new Diagnostics Chief’s mind? Transitions, the LDT rule and cybersecurity

Courtney Lias has been Acting Director of CDRH’s in vitro diagnostics office, known as OHT7, since the departure of Tim Stenzel at the end of 2023. At a conference in DC this week, she confirmed that she is now the permanent Director of OHT7. She takes over the office at a critical time for the office, which is at the forefront of several major regulatory reform efforts.

By Laura DiAngelo, MPH | Aug 20, 2024 10:43 PM CDT

COURTNEY LIAS will now serve as OHT7 Office Director in a permanent capacity

  • Quick background: The Center for Devices and Radiological Health (CDRH) is organized into seven main Offices, of which one – the Office of Product Evaluation and Quality (OPEQ) – is a “super office.” The office, which is responsible for directly reviewing and regulating medical devices and diagnostics, was established under a major restructuring of CDRH in 2018-2019 as part of CDRH’s “Total Product Lifecycle (TPLC)” approach. At a high level, the establishment of OPEQ brought together previously disparate offices involved in pre-market review (the Office of Device Evaluation and its divisions) and post-market work related to compliance and surveillance (the Office of Compliance) and Office of Surveillance and Biometrics). The reorganization also saw the Office that regulated in vitro diagnostics (IVDs), then known as the Office of In Vitro Diagnostics and Radiological Health (OIR), under OPEQ as a division.
  • OPEQ is organized into eight “Offices of Health Technology” (OHTs), which are structured by therapeutic area (e.g., OHT2 is cardiovascular devices). The regulatory oversight of diagnostic products is in OHT7; while this OHT previously included radiological health products, the agency formally established its newest OHT, OHT8 (radiological health and mammography) in 2022, effectively separating out these divisions.
  • OHT7 in transition: The Covid-19 pandemic put extreme pressure on OHT7, with that Office facing high levels of Emergency Use Authorization (EUA) submissions for Covid-19 test products. At the end of 2023, former OHT7 Director TIMOTHY STENZEL retired from the agency. Stenzel had joined the agency as Office Director in 2018 and helmed the OHT7 through the pandemic.
  • Following Stenzel’s departure, COURTNEY LIAS was named Acting Director of OHT7. Lias has been with the agency for over 20 years, and prior to being named Acting Director of OHT7 served as the Director of OHT3 (Office of Gastrorenal, ObGyn, General Hospital and Urology Devices) within OPEQ.
  • At the NextGenDx Summit in DC this week, former OIVD (as it was then called) Director ALBERTO GUTIERREZ announced that Lias will serve as the Office’s permanent Director. Introducing Lias for a plenary at the event, Gutierrez said: “Courtney is in the paperwork for the seminar, she’s put down as the Acting Director. She actually is now officially the Director for the Office of In Vitro Diagnostics, OHT7. So I would like to congratulate Courtney on moving up and becoming the director. I know that she’s got a tough road ahead of her with everything that is going on with [laboratory developed tests] LDTs and everything, but I do also know that Courtney is probably the right person to be leading the office this time.”

Speaking at the NextGenDx summit, Lias provided an overview of the landscape she faces as OHT7 Director

  • Up first: There have been a lot of changes at the top of CDRH. Lias acknowledged the recent retirements of longtime CDRH Director JEFF SHUREN and OPEQ Director BILL MAISEL.
  • Details about the transition of CDRH and OPEQ: Notably, according to Lias, Shuren is still technically at the agency, “currently in a position of Center Director Emeritus up at the agency level to help with the transition to a new Center Director,” she said. Deputy Center Director MICHELLE TARVER is serving as Acting Center Director; “she’s the right person for this acting position, and I’m really thrilled to see what she can do in the next coming months in this role,” said Lias. The agency is currently looking for a permanent replacement for Shuren as the head of CDRH; for now, it’s not entirely clear whether Shuren will stay on until that person is found. Maisel, who has led OPEQ since the super office was established, was replaced on an acting basis by OPEQ Principal Deputy Director OWEN FARIS, but a new permanent Director will be starting in September, Lias confirmed: “They have announced a permanent director who’s scheduled to start in September, Dr. Rusty Segan will be joining as permanent OPEQ Director in about a month,” she said. FDA has not made a formal public announcement, but Lias was referring to ROSS D. SEGAN, a longtime medical device executive who uses the nickname “Rusty” and confirmed the appointment to AgencyIQ.
  • As OHT7 Director, Lias will oversee much of the work in implementing the Laboratory Developed Test (LDT) final rule. This rule, finalized in May 2024, represents a significant change in the way that FDA regulates certain test products known as LDTs. While the agency previously expressed a general enforcement discretion over LDTs (in effect, not requiring their regulatory compliance as medical devices), the agency is phasing out that policy over the next few years. The rule has significant implications for the way that LDTs are regulated in general, but also has additional impact on the use of LDTs in the context of drug development (i.e., tests leveraged during investigations of therapeutic products) and companion diagnostics (CDx).
  • Lias provided a status update on a pilot program administered by OHT7 and the Oncology Center of Excellence (OCE) that sought to formalize a system for LDTs being used as CDx. This project was announced in June of 2023, and the agency has not provided much information about the ongoing work since a December 2023 webinar recapping its design. At a high level, the pilot program was intended to offer sponsors of certain drug products that would need a CDx with the opportunity to submit information about their Clinical Trial Assay (CTA) – the assay they’re using in a pivotal trial – to the FDA [see here for background on that topic]. While the submission of this data would not result in authorization of the CTA as a diagnostic, the FDA would, under the pilot, allow that information to inform a set of minimum analytical performance characteristics on the use of a test with the corresponding drug, which in turn would inform the drug’s labeling. In short, drug sponsors would submit information on the CTAs used in their development programs, the FDA would determine if that information has established clinical validity for use, and the agency would then outline a series of baseline performance specifications for the tests which could be leveraged by additional test developers. The pilot was only open to certain drug sponsors with oncology products regulated by the Center for Drug Evaluation and Research (CDER).
  • Lias confirmed that “this pilot is ongoing” and described some lessons learned. “We’ve been in this pilot for about a year, and we are currently sort of thinking about what’s going well in the pilot, what types of things need to be adjusted,” she said, but stopped short of describing what adjustments those might be. She was able to outline some high-level trends, particularly noting that “we’ve noticed… that it has been difficult for drug manufacturers to get information on the clinical trial assays that they’re using.” She went on, “A lot of the clinical trial assay laboratories are unwilling to provide information on the test performance for the clinical trial assays, and that makes it difficult for us to accept them into a pilot. There’s also been variability across the clinical trail assay sites,” she noted. While the pilot included the opportunity for a retrospective cohort, according to Lias’s presentation it appears that there were additional challenges with accepting programs that had already been done or started into the pilot, given the challenges with getting information out of laboratories. As she explained, “understanding how you don’t have a pre-specified performance criteria, trying to understand the performance from the trial sites is really the only way you can do this, and without the cooperation of laboratories providing that type of validation, it makes it difficult to have them in the pilot.”
  • There are several high-level takeaways from these updates for drug developers. First, Lias put to rest any concerns that the pilot may have been shuttered following the issuance of the LDT final rule. Notably, some lawmakers had recently asked FDA for a status check on the pilot following the final LDT rule’s publication. Second, the operational challenges that Lias has cited with the pilot – in effect, working more closely with clinical laboratories who conduct testing as part of a trial – remain of significant concern to the drug industry under the LDT rule. Per that rule, the investigational device exemption regulations, which outline regulatory expectations for medical devices used in investigations (either of the device itself or a device used in an investigation of another product, like tests or digital health technologies in a drug trial), would come into force during Stage 2 of the final rule in 2026.
  • The reclassification initiative: In February 2024, CDRH announced a new initiative to review the risk-based classifications of “most” IVDs that are regulated as “high risk” (Class III) devices to see if they could be appropriately regulated in a lower risk-based category (Class II, moderate risk). This project, which is focused first on CDx (which are largely Class III products) and IVDs for infectious diseases, was also seen as a key aspect of implementing the LDT rule. Per the LDT final rule, the agency intends to have the reclassification initiative completed by Stages 4-5, which are the pre-market submissions stages of the rule. In effect, the agency should have “most” Class III IVD types re-classified to Class II in advance of when a pre-market submissions would be required by the rule, as the risk-based categorization of the test will inform both when a submission is needed and what application type they will need to prepare. “We have to go through a pretty long process” for reclassification, Lias acknowledged, “but we will be providing updates as we have them, and we’re trying to move through them as quickly as we can,” she said.

Two big things on Lias’s mind for the diagnostics industry: Artificial Intelligence and Machine Learning (AI/ML) and cybersecurity

  • First, on AI/ML, Lias acknowledged that tests and diagnostics are already using AI/ML tools, and the field of AI/ML-based research in diagnostics is advancing quickly. She highlighted FDA’s new authority related to Pre-Determined Change Control Plans (PCCPs), which allows the agency to authorize a medical device along with a plan to make changes to the device after its authorization without needing to re-submit the device. Lias highlighted areas where OHT7 has already made use of these application types, and noted that PCCPs are likely to be of increased relevance to diagnostics going forward, even beyond its use case in AI/ML. “It doesn’t have to be an AI/ML device,” Lias said. “It can be any device… and I will say that IVDs are a little ahead of the curve for PCCPs.”
  • Cybersecurity “is the most important topic because it’s causing a lot of problems for a lot of people right now,” said Lias. She went over the new regulatory changes in the last few years related to cybersecurity for medical devices, specifically the statutory requirement that certain device developers submit cybersecurity information as part of their required regulatory submission to the FDA. This requirement applies to all “cyber devices” [ see AgencyIQ’s explainer here], which Lias described as “a device that includes software that’s validated, installed or authorized by the sponsor as a device or in a device, has the ability to connect to the internet, and contains any such technological characteristics validated, installed or authorized by the sponsor that could be vulnerable to cybersecurity threats.” Per Lias, “it applies to a lot of different devices,” including diagnostic tests. “Cybersecurity mandates apply whether you made the device or not,” she said, “so if you’re a diagnostic test developer and you have a device that you’re using in your system, it needs to be cybersecure even if you didn’t make it,” she confirmed. This includes test or diagnostics that aren’t intended to be connected to the internet, including when their labeling states “do not connect” – per Lias, those “are not going to be adequate mitigations for this type of thing.”
  • What does this mean for diagnostic test developers? “If you take anything away from this talk, I think it is ‘learn more about cybersecurity and what you need to do about it’,” Lias concluded. Specifically for diagnostic test developers, “if you are developing a test and you are making an instrument or some other cyber device, build that cybersecurity in, understand what type of cybersecurity testing and evaluation you should be doing and have it done, and build in the ability to create or to make appropriate cyber security updates,” she urged. She went on, “the second point I want to make is, if you are a test developer and you do not make the instrument that you are using, you should choose one that is cybersecure, and you should choose one that either is separately regulated – for example, it’s an exempt product where they have to meet the cybersecurity requirements on their own, and they’re being inspected, and they have a quality system, and all these requirements applied to them – or you have access to all this information you need to address these cybersecurity questions, because this is going to be a challenge for test developers who are trying to leverage instruments that are not cybersecure and for which they cannot get the information.”
  • According to Lias, this is already an issue. In effect, diagnostic test developers who are using components or devices in their test for which they either do not have the required cybersecurity information (if they did not make the instrument) or for which they did not understand the cybersecurity requirements are already facing challenges when submitting their tests to the FDA. “I think that’s one of the biggest things we’re going to see coming over the horizon as challenges for test developers who are leveraging legacy, outdated or other types of instruments that have not been designed this way,” Lias cautioned.

 

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

  • Next Generation Dx Summit
clarification:This piece has been updated after AgencyIQ confirmed that Ross “Rusty” Segan has been named as the next Director of OPEQ.

Filed Under: Article

August 23, 2024 by

By Laura DiAngelo, MPH

Over a year since its initial draft guidance document on the use of Pre-Determined Change Control Plans (PCCPs) for medical devices that leverage machine learning, allowing sponsors to pre-specific certain post-authorization modifications, the agency has issued a new draft guidance on the use of PCCPs for all medical devices.

Fill out the form to read the full article.

Filed Under: Article

August 23, 2024 by

CDRH unveils new guidance on Pre-Determined Change Control Plans for medical devices

Over a year since its initial draft guidance document on the use of Pre-Determined Change Control Plans (PCCPs) for medical devices that leverage machine learning, allowing sponsors to pre-specific certain post-authorization modifications, the agency has issued a new draft guidance on the use of PCCPs for all medical devices.

By Laura DiAngelo, MPH | Aug 22, 2024 10:13 PM CDT

Intro: Pre-Determined Change Control Plans in medical device regulation

  • Post-market updates to authorized medical devices may, or may not, require a new submission to the FDA. The core process for deciding whether a modification to a device needs to be submitted either through a new marketing application or a supplement (in the case of devices authorized under a Pre-Market Approval (PMA) application) is laid out in a series of three guidance documents. This includes final guidance from 2008 on “ The PMA Supplement Decision-Making Process” and two 2017 guidance documents informing modifications to devices under a 510(k) pre-market notification (one general modifications guidance and another on software updates). This trio of documents are known collectively as the Modifications Guidance documents.
  • Pre-Determined Change Control Plans (PCCPs) represent another way of conducting updates. The general concept of a PCCP is for a device sponsor to submit a series of planned modifications to a medical device, including information on how it would validate, verify, implement and monitor the modifications, to the FDA as part of a marketing application. This would, theoretically, allow FDA to pre-authorize certain modifications to a device. PCCPs were first conceived of as a way to leverage artificial intelligence/machine learning (AI/ML) that would need to be updated over time. In 2019, the agency issued a Discussion Draft on a potential regulatory assessment framework for AI/ML devices that highlighted the specific issues raised by these technologies, including the use of PCCPs.
  • In late 2022, the FDA gained new authority related to PCCPs. As part of the Food and Drug Omnibus Reform Act (FDORA), Congress clarified new statutory definitions related to the development and regulation of PCCPs for medical devices. Specifically, under section 3308 of the legislation, Congress clarified that changes authorized under a PCCP, which would represent the types of significant modifications to an authorized device that would otherwise necessitate a new regulatory submission to FDA, “shall not be required” if they are within the scope of the established PCCP.
  • In March 2023, the FDA issued its initial draft guidance on PCCPs, and specifically PCCPs for machine learning-enabled device software functions (ML-DSF), or the function of a device that leverages ML. That draft guidance introduced the concept of PCCPs formally for the first time. It defined key terms and offered initial thinking on what would – or would not – be a good use case for modifications to a device under a PCCP. As would be expected, this draft guidance focused primarily on the use of PCCPs for AI/ML enabled product, but did offer an initial framework of what a PCCP would look like in practice, including how they should be designed and submitted, how they will be reviewed by the agency, and what the post-market expectations are for a device modified under a PCCP.
  • Expansion of the concept of PCCPs has been a priority for the agency – and industry. As reflected in both the comments on the ML-DSF draft guidance and CDRH’s 2024 guidance agenda, there was a broad expectation that FDA should issue guidance on using PCCPs for more device types than ML-DSF. Per the CDRH guidance agenda, the agency expected to finalize the ML-DSF guidance in 2024, as well as issue draft guidance on PCCPs for medical devices that are not, specifically, AI/ML devices.

New PCCP Draft Guidance: For all device types

  • On August 21, 2024 the FDA released a new guidance document on PCCPs that outlined recommendations for all medical devices, and not just AI/ML-enabled devices.
  • The scope of the new guidance: “This draft guidance recommends that a PCCP describe the planned device modifications, the associated methodology to develop, validate, and implement those modifications, and an assessment of the impact of those modifications,” FDA wrote. The guidance covers PCCP content and process for all device types and for all medical device marketing application types.
  • The guidance does have some specific areas of focus, though. While the guidance applies broadly to all device types, the agency includes a couple of key use-cases for PCCPs in the guidance document, and in particular related to diagnostics. Still, “this draft guidance is not intended to delineate a comprehensive list of modifications FDA would consider appropriate for inclusion in a PCCP for a device,” FDA wrote.
  • How does this new version compare to the guidance on PCCPs for ML-DSF? Many of the same concepts are represented in both guidance documents, and they’re structured similarly. However, as expected, this new draft guidance offers information on PCCPs that applies more broadly and is not specifically focused on AI/ML-related PCCPs. This means that it’s got broader applicability, with less language and content specifically focused on AI/ML issues (e.g., model drift). The overall idea of the PCCP and its consideration align between the two guidance documents – as do the definitions, process, and content expected in a PCCP.
  • In short: Those familiar with the ML-DSF PCCP guidance should be generally familiar with the content of this new draft guidance.
  • At a high level: What is a PCCP? PCCPs are plans, submitted with marketing authorization requests, to make certain changes or modifications to a product under the authorization. PCCPs are comprised of three main components (detailed more below): the Description of Modifications, Modification Protocol and Impact Assessment. In effect, a “PCCP includes those device modifications that generally would otherwise require a new marketing submission. These modifications include those that could significantly affect, or that otherwise affect, the safety or effectiveness of the device, unless those modifications are covered by an authorized PCCP.”
  • The PCCP is authorized as a part of the marketing authorization, and a device sponsor “obtaining FDA authorization of a PCCP as part of a marketing submission… allows a manufacturer to modify its device over time in accordance with the PCCP instead of obtaining separate FDA authorization for each significant change prior to each implementation.” Like making a major modification without a new marketing submission or supplement, making changes outside of the scope of a PCCP could result in the device being adulterated and misbranded.
  • How PCCPs would apply: A PCCP can be authorized through the PMA pathway, 510(k) pre-market notification pathway, or De Novo classification request pathway. For devices cleared under a 510(k) with a PCCP, the version of the device “cleared or approved prior to changes made under the PCCP” would be able to serve as a predicate for a 510(k). Similarly, once such a modification made in compliance with a PCCP is submitted and authorized under a modification application (which is not necessary for the purpose of implementing the change), then that version of the device can be used as a predicate.
  • For combination products, the guidance would apply specifically to the device constituent part of a device-led combination product – a PCCP could not be authorized as part of a drug submission for a drug- or biologic-led combination product. Notably, trade association PhRMA had specifically asked in its comments on the ML-DSF draft PCCP guidance for the FDA to “specifically confirm that sponsors have the flexibility to submit a PCCP as part of the NDA or BLA for drug-device or biologic-device combination products and that a separate device marketing submission is not required in order to submit a PCCP for the device constituent part of such products.” However, this does not appear to be the case.
  • Least burdensome: The agency notes that leveraging a PCCP “may be a least burdensome option” to support device modification. While the system remains voluntary and optional, the use of a PCCP may reduce the overall need for supplements or new submissions.

Building a PCCP and maintaining it over time

  • “PCCPs are specific,” the agency clarifies. PCCPs should “include specific modifications that the manufacturer intends to make over time” but should not list all the modifications that a manufacturer “may possibly make.” Per the agency, sponsors should include “only a few, specific modifications that can be verified and validated” in a PCCP; it goes on to note that the FDA “may not be able to make” an authorization decision if the PCCP includes “numerous modifications, or modifications that range across various aspects of the device.”
  • What application types can sponsors use to establish a PCCP? As described above, PCCPs may be appropriate for devices under a PMA, 510(k) or De Novo classification request. However, it does not need to be an original PMA – PCCPs can also be established through a variety of PMA and supplement submission types (including Modular PMAs, in which a PCCP would be a module, and different supplements). For example, a PCCP comprising a manufacturing change could be submitted as a 135-day supplement, while a minor change that FDA agrees can be achieved in real-time review can be submitted via a Real-Time PMA supplement. For 510(k)s, PCCPs can be established through either the Traditional or Abbreviated 510(k) submission pathways. Only original De Novo requests can be used, however (which makes sense, because there is no other De Novo type).
  • A quick note about following a PCCP – or not. As noted above, implementing changes outside of the scope of a PCCP would be considered the same, in a regulatory sense, to implementing a modification without authorization (i.e., the product as modified would be adulterated or misbranded). However, the agency notes that simply not implementing a change under a PCCP is fine: “FDA would not consider it to be a deviation from the authorized PCCP in situations where a manufacturer chooses not to implement modifications in their authorized PCCP or where a manufacturer chooses to submit a new marketing submission for a device modification in lieu of using their authorized PCCP” (footnote 34).
  • As the PCCP is a technological characteristic, how is this considered in a review of substantial equivalence? For products submitted with a PCCP under a 510(k): “In general, FDA anticipates that the PCCP will primarily be reviewed after FDA finds that the intended use of the subject device and the predicate device are the same, to help determine whether the devices have different technological characteristics that do not raise different questions of safety and effectiveness.”
  • The PCCP in a marketing submission: A focus on labeling. PCCPs should be “a standalone section within the marketing submission” and be “prominently included and discussed in the cover letter” and table of content. It should also be discussed throughout the application – including in the labeling. The new draft guidance offers some additional information about how PCCPs should be addressed in labeling, noting that “information on the device’s authorized PCCP may be necessary for a user to understand changes in the device and to continue to use the device safely and effectively across the intended use populations and intended environments as the device changes pursuant to the authorized PCCP.” Information on PCCPs should also be available, “in sufficient detail,” in public-facing information about the device (e.g., device summaries).
  • The FDA has some specific asks for PCCP-related labeling, including that labeling should include a statement disclosing the PCCP and the PCCP itself include labeling updates that would be implemented as modifications to the device are made under the PCCP. This includes information on the implemented modifications (“including a summary of current device performance, associated inputs/outputs, validation requirements, and related evidence”), how the modifications were implemented, and a description of how users will be informed of the modifications (e.g., updated instructions for use or version history).
  • Using an authorized PCCP to make a modification: In general, “FDA would consider it to be a deviation from the authorized PCCP in circumstances where the PCCP is not followed or cannot be followed”; this includes circumstances in which a modification is not included in their PCCP (in which device modifications guidance should be followed) or a change that is within scope is implemented using a process (methods and specifications) outside of what was described in the Modification Protocol of the authorized PCCP. In these circumstances, the sponsor can either request a modification to the PCCP or they can seek a device modification. If going the second route (i.e., new marketing submission with a modification), then “the manufacturer must also submit the proposed, modified PCCP for the device.”
  • Modifying an authorized PCCP: As with the original authorization of a PCCP, modifications to an authorized PCCP “will generally constitute changes to the device that would otherwise require a new marketing submission” – typically a 510(k), special 510(k) or a PMA supplement. As in the ML-DSF guidance, the agency would want to see a new marketing submission that includes “a summary of the changes to the authorized PCCP, and where practicable, a tracked changes version compared to the authorized PCCP.” Further, (see footnote 70), when the primary change leading to the need for a new marketing submission is to modify a PCCP, “in general, manufacturers may provide references in the marketing submission to prior marketing submissions for content that remains unchanged, as appropriate.” Again, as in the ML-DSF guidance, the agency notes it will “focus its review on the aspects of the device that are most significantly modified.”
  • Version control and maintenance: The FDA seems to be expecting a bit of a learning curve with PCCPs, stating that it expects “interactive” reviews of marketing submissions with a PCCP “as manufacturers gain experience developing and implementing PCCPs.” For now, the agency says it will leverage its existing processes for interactive reviews – or, potentially, through issuing a deficiency letter. This means that PCCPs might be modified while they’re under review, and the agency notes that “a final, revised version of the PCCP should be submitted as a clean copy to FDA” with titles and version numbers. The PCCP (and specific version) are authorized with the device, so “a manufacturer should only have one version of an authorized PCCP for their device” – but new versions can be authorized over time.
  • This section of the guidance also describes some practical information, like how the FDA intends to look at new marketing submissions for products modified under a PCCP (it does not intend to re-review the adequacy of modifications made under a PCCP), and that modifications made under a PCCP authorized in a PMA should be reported under the PMA Annual Report (and include any updated labeling, as applicable).

PCCP Policy: Types of modifications under a PCCP

  • Not all modifications are appropriate for a PCCP. In general, those that are “intended to maintain or improve the safety or effectiveness of the device” and can be “verified and validated” are a good fit for a PCCP. Further, modifications that would alter a device’s intended use, or its indications for use, are likely not appropriate for a PCCP as “FDA believes that most modifications to the indications for use included in a PCCP would be difficult for FDA to assess prospectively to determine whether the device would remain safe and effective.” The agency urges sponsors to submit a Q-submission to discuss potential modifications.
  • Notably, there are some exceptions to the general idea that PCCPs can’t be used to change the labeling or indications for use, clarified in a list of potentially appropriate PCCP use cases later in the document (and again for devices under PMA). These include potential changes to the labeling that focus on “a specific subset of patient population within the originally indicated patient population” (emphasis added), adding information in the labeling/indication for use to specify use of the device with an additional “device, component, or human genetic variant” or updating the device specifically for home use settings.
  • The guidance describes both general recommendations for manufacturers to consider whether their planned modifications are appropriate for a PCCP, and provides some examples. These are intended to work with the guiding principles (outlined above) – but does note that “Ultimately, decisions about the types of modifications to be included in a PCCP are generally fact-specific for each device.”
  • For a 510(k) or De Novo device: In general, modifications under a PCCP for these devices should maintain the device’s intended use. However, “a significant change or modification in design, material, chemical composition, energy source, or manufacturing process may be appropriate for inclusion in a PCCP,” the agency describes, and refers sponsors back to the 510(k) Modifications guidances (for both general changes and software changes). The agency aligns its policy on PCCP-appropriate modifications with those guidance documents, recommending the same general process (see, for example, this flowchart from those guidance documents carried over to this guidance). Specifically, it asks sponsors to conduct a risk-based assessment to determine if a marketing submission would be necessary. For the purposes of a PCCP, a modification that could introduce new risk “are generally not appropriate for inclusion in a PCCP,” but modifications that might modify existing risks “generally may be appropriate for inclusion in a PCCP” when there is adequate risk mitigation between the device’s risk management framework and the manufacturer’s Quality System.
  • The agency offers a list of “certain high-level modifications that generally may be appropriate or are not appropriate for inclusion in a PCCP.” Those that might be appropriate include changes to design (dimensions, performance specifications, wireless communication, components/accessories, user interface), sterilization, transport, expiration dating or packaging (using well-established methods), materials or components (including reagents), software changes related to compatibility or interoperability, software changes to improve device performance, “certain changes to the labeling” to discuss “a specific subset” of the patient population, changes to the labeling or indication for use to specify device use with another device (or component, or human generic variant), or indications for use to allow the device to be used in the home setting. Those that are not appropriate include changes to control mechanisms, design changes that alter the intended use, changing a device from single use to reusable, change or remove contraindications, change from prescription to over-the-counter (OTC) use, changes in indication from general to specific, adding a new patient population, those updates that might need clinical data, changes to address safety issues or changes to a device constituent part that “impacts” a drug or biological part.
  • Quick note: There’s a brief carve out specifically for IVDs. While the list above states that changes requiring clinical data are generally not appropriate for inclusion in a PCCP, “Certain changes that may need new clinical data, such as method comparison data for IVDs, may be appropriate for inclusion in a PCCP” (footnote 90).
  • PCCP modifications appropriate in a PMA Application or Supplement: Similar to the process for 510(k)s, “modifications included in a PCCP must maintain the device within the device’s intended use” although “other modifications that could affect the safety or effectiveness of the device may be appropriate for inclusion in a PCCP.” Per the guidance, these types of changes can include minor changes and manufacturing changes, which “generally may be appropriate for inclusion in a PCCP when the risks of implementing the modification” are mitigated by the risk management framework and the manufacturer’s QS. Again, the agency pulled some of this content from its guidance on modifications for devices subject to PMA (including this flow chart).
  • It also provides a list of examples, differentiating between what is a minor change and what is a manufacturing change. This list generally reflects those that are outlined above, repeated for content on PMAS – minor changes related to materials/components (e.g., new sources for reagents), labeling changes that describe a specific subset of patients within the original indication, or software updates to improve device performance or update interoperability. Manufacturing changes would include those to sterilization, joining or cleaning, changes that can automate existing processes, changes to the “environmental conditions” or “certain changes in methods of manufacture.” Changes that are not appropriate would be those that are significant changes, changes to add new patient populations to the indication/intended use, changes that may need clinical data (except IVDs, see above), to address a safety issue, or to “add, expand, or move the manufacturing or sterilization site of a finished device.”

PCCP Policy: Content

  • As noted above, there are three main components of a PCCP: the Description of Modifications, the Modification Protocol, and the Impact Assessment.
  • Description of Modifications: A PCCP will need to describe “each planned modification” – including information the “specific changes to the device characteristics and performance” from the modifications. This should describe and enumerate the, as previously mentioned, specific individual proposed modifications, the justification for each modification, and “presented in the level of detail that permits understanding of the specific modifications that will be made to the device.”
  • Modification Protocol: This is the component of the document that will describe the methods for “developing, validating, and implementing the modifications” as planned, including full descriptions of the verification and validation activities. “Documentation of modifications verified and validated per the Modified Protocol must be compliant with the Quality System Regulation (QSR), including that the manufacture must document the change in accordance with the manufacturer’s quality system.” The protocol should describe the specifics of modifications, including “appropriate and applicable data, test methods, analysis methods, and specified acceptance criteria used to develop, validate, and implement all proposed modifications” and the process by which they would be implemented (including plans for communication and training on users). It should also include a plan for recordkeeping, risk mitigation strategies, and “be least burdensome” for review. The agency describes the different components of the protocol, including both performance evaluation methods (how to ensure the device will stay within the device’s specifications) and update procedures (the actual plan for implementing a modification). There should also be traceability in the submission, with each part of the Protocol mapped to the Description of Modifications.
  • Impact Assessment: “An Impact Assessment in a PCCP is the documentation of the assessment of the benefits and risks of implementing a PCCP for a device, as well as the mitigations of those risks,” the FDA wrote. It should be based around the manufacturer’s existing quality system. The documentation in a PCCP should include a comparison between the versions of the device (original and under every modification), the benefits and risks of each modification, the verification and validation activities’ ability to reasonably ensure safety and effectiveness, how individual modifications might impact each other and the cumulative impact. In effect, this is the section of the PCCP that should contextualize what each modification will do, how they might interact, and how each modification will impact the overall functionality of the device.

Analysis and what’s next

  • The guidance concludes with a list of illustrative examples related to situations in which a PCCP might be appropriate for certain devices – including for diagnostics. These include adding new sample types or extending stability claims for a test, or changing sterilization methods from an established category A to an established category B. While the list is certainly more comprehensive than was available in the ML-DSF guidance, it’s likely that manufacturers will seek more, and more detailed, information on illustrative examples.
  • When this guidance is final, it will update the considerations on whether to submit a new 510(k) or PMA supplement. When the PCCP policy is fully implemented, it will represent a significant change to the currently available avenues to modify a medical device. The FDA’s assertion is that PCCPs will be the least burdensome option for device modifications, which typically require a new marketing application. However, we’ll likely need to see the full implementation of this policy to understand if, and when, that’s the case – or whether certain types of modifications (e.g., sterilization changes, shelf life or stability claim updates, cybersecurity updates, digital health updates, IVD kit updates) lend themselves better to PCCPs than other types of modifications.
  • FDA leadership has touted the idea of PCCPs for IVDs updates or sterilization changes – both areas facing upheaval after recent policy changes. For IVDs specifically, it’s likely that the diagnostic test industry will seek more nuanced, specific guidance related to IVDs. As the guidance is currently drafted, IVD-specific considerations are sometimes pulled out of the recommendations in the guidance (e.g., some PCCP-appropriate modifications can include clinical data), but additional guidance on the specifics of how the policy can be leveraged for IVD use cases would likely be helpful. For example, guidance specifically on adding sample types, swapping out reagents or requesting a shelf life extension for a test under a PCCP.
  • Process wise: CDRH will need to make Level 2 (clarifying existing process) updates to its existing guidance on “Deciding When to Submit a 510(k)” for a device or for a software change to a device, as well as its guidance on “The PMA Supplement Decision-Making Process” – and “we may also make Level 2 updates” to the PCCP for AI/ML guidance document “and other device-specific guidance documents containing information on PCCPs for consistency.”
  • The FDA is expecting a learning curve here, indicating in the guidance that it expects this to be an “interactive” process while developers and sponsors are still building experience with the PCCP process. Industry has been actively seeking more information about how they can use PCCPs, but the actual implementation of the policy is likely to be a challenge for device developers navigating the new modification process – including both submitting PCCPs and tracking them over time once the device is on the market and in distribution, and under a modification. For example, the guidance notes that “if the labeling states that a modification to the device has been implemented when it has not, the device might be deemed misbranded” (footnote 109). While industry had specifically touted the potential use case for electronic labeling in feedback on the ML-DSF draft guidance (see AdvaMed’s comments), the new draft guidance does not address this issue, despite it being a high priority for industry.
  • Further, the intricacies of reviews with PCCPs going forward are likely to be challenging. FDA says it “does not intend to re-review the adequacy of modifications implemented consistent with an authorized PCCP” but the specifics of a review for a device that is several versions from its originally authorized version, with multiple PCCPs/supplements, will be an organizational challenge down the line.
  • Also: what is a good or a not good fit for a PCCP still needs to be worked out. While the agency provides some principles and a list of examples, it also says that “Ultimately, decisions about the types of modifications to be included in a PCCP are generally fact-specific for each device.” The agency notes that if PCCP review is what’s holding up an application’s authorization, it may authorize the submission if the PCCP is withdrawn, but a better understanding of what types of modifications, and how they’re expected to be appropriate tracked, documented and monitored over time will be critical to the success of this policy.
  • A quick note: Q-sub reliance. The draft guidance is, like the ML-DSF version of the PCCP guidance, highly reliant on the Q-submission process. Multiple times throughout the guidance, the agency urges sponsors to submit a Q-submission to flag their approach in developing their PCCP for regulators. Notably, this is a common refrain from the FDA when implementing a new policy, or when the foundation of a policy relies on “case-by-case” determinations. However, this is not a particularly efficient system; As ThermoFischer Scientific noted in its comments on the ML-DSF draft guidance, “too much flexibility within the review process leads to a lack of transparency and consistency during the decision-making process from the agency,” citing their concerns about “overdependence on Q-submissions” in that guidance.
  • And a final quick note: eSTAR. As many commenters noted in the ML-DSF draft guidance docket, CDRH is moving towards requiring submissions in its electronic submissions template and resource (eSTAR). On August 22, FDA finalized guidance that would require all De Novo applications to be provided in eSTAR format as of October 2025. Many commenters had flagged concerns about navigating the new content type of PCCPs in eSTAR – which will need to be addressed going forward as it becomes mandatory for more submission types.

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

  • Predetermined Change Control Plans for Medical Devices: Draft Guidance for Industry and FDA Staff
  • FDA guidance webinar: September 3, 2024
  • Electronic Submission Template for Medical Device De Novo Requests: Guidance for Industry and Food and Drug Administration Staff
  • AgencyIQ Analysis: FDA’s Pre-Determined Change Control Plan draft guidance: Terminology, content and policy
 

Filed Under: Article

August 16, 2024 by

By Scott Stephens, MPA

The European Commission has issued a call inviting companies to submit tenders for a 1.5-million-euro contract to deliver a pilot project setting up “substitution centers” envisioned to help companies negotiate the convoluted process of substituting hazardous substances with safer alternatives.

Fill out the form to read the full article.

Filed Under: Article

August 16, 2024 by

Are ‘EU substitution centers’ in the cards for quicker substitution of toxic chemicals?

The European Commission has issued a call inviting companies to submit tenders for a 1.5-million-euro contract to deliver a pilot project setting up “substitution centers” envisioned to help companies negotiate the convoluted process of substituting hazardous substances with safer alternatives.

BY Scott Stephens, MPA | Aug 13, 2024 2:45 PM CDT

Regulatory context of the Commission’s initiative

  • An integral part of the EU chemicals regulatory framework is ensuring the substitution of harmful chemicals with safer alternative substances or technologies for human health and the environment when available and technically and economically feasible. At the same time, chemicals substitution is significant for meeting the ambitions under the European Green Deal, including the objectives of the Commission’s Chemicals Strategy for Sustainability (CSS).
  • Industry is required to seek out potential alternative chemicals, as well as create substitution plans where satisfactory alternatives are not yet available, in certain contexts (for example, when companies apply for authorization) under Regulation (EC) 1907/2006 on the registration, evaluation, authorization and restriction of chemicals (REACH). The process of restricting substances under REACH also involves alternatives assessment and substitution planning on the part of the authorities. Similarly, other EU chemicals legislation, such as Directive 2011/65/EU on the restriction of the use of certain hazardous substances in electrical and electronic equipment (RoHS), stipulates related substitution and substitution planning provisions. [ Read this AgencyIQ analysis on substitution and substitution planning in the context of a study the Commission initiated last year that is expected to be completed by late 2024.]
  • The Commission has pointed out that the “pace of addressing” risks of hazardous substances “through substitution is still slow and has addressed only a limited number of substances and uses to date.” The EU executive says this sluggishness can be attributed to the “lengthy procedural requirements” and the related “complexities” of the authorization application process and preparation of REACH restrictions. Such complexities include aggregating “heterogenous uses into a single use.” From start to finish, both procedures, on average, take around three years. By way of example, the Commission referred to applications made by companies for authorization of hexavalent chromium substances and the authorities’ preparation of the universal restriction of PFAS. “Both involve a wide range of uses and a high number of downstream users. The outcome of these challenges risk delaying actions to substitute the substance and missed opportunities for uses where earlier substitution may be possible,” according to the Commission.

Commission launches initiative to forge path for expedited chemicals substitution

  • The EU Commission on July 23 put out a call on Tenders electronic daily (TED) for a contract estimated at 1.55 million euros to deliver a study “on EU Substitution Centre(s) – Providing support to businesses to substitute their use of hazardous chemicals through collaboration, innovation, research and direct assistance.” An undertaking slated to last 2.5 years, the study has as its primary objective to “ demonstrate the feasibility of a European Substitution Centre,” that would serve “to accelerate the substitution of toxic chemicals with safer alternatives.” Planned to be executed as a pilot project, the European Parliament included the study in the 2024 EU budget.
  • The study will comprise four main deliverables, or “lots,” with the first dedicated to the pilot project, determining “how a network of substitution centres for hazardous chemicals could best be set up and identify if additional substitution centres should be created.” The other three lots are focused on supporting projects to promote substitution of specific chemicals, namely per- and polyfluoroalkyl substances (PFAS) in batteries, chromium(VI) substances in hard chrome plating, and substances or substance groups used in paints manufacturing.

Massachusetts experience serves as wellspring of inspiration

  • The pilot project to be delivered in lot 1 is “inspired” by the Toxics Use Reduction Institute (TURI), an initiative that Massachusetts introduced through its 1989 Toxics Use Reduction Act (TURA), according to the tender specifications document. The Commission underscores TURI’s achievements, stating that the institute’s “staff have successfully helped companies, particularly SMEs [small and medium enterprises], to substitute hazardous chemicals with safer alternatives through collaboration and innovation, including research, direct technical and financial assistance, and a focus on finding alternatives that meet a user’s performance and fiscal constraints.” To back this up, the Commission cites several statistics from TURI, including that in 20 years (2000-2020), companies in Massachusetts cut toxic chemical use by 75%, toxic waste by 67%, and toxic releases by 91%. Likewise, from 1990 to 2020, the carcinogen trichloroethylene’s (TCE) use was diminished by 95% and its release by 97%.
  • Under TURA, the Commission highlights, companies are required to report annually their use of substances included in a specified “list of toxic or hazardous substances” when certain quantities are reached. Likewise, they are obligated, every two years, to carry out a “Toxic Use Reduction (TUR) planning.”
  • TURI offers “a wide range of services, including offering grants to businesses, researchers, and communities intending to reduce the use of toxic substances and conducting policy analysis about the potential applications of chemicals in the TURA list,” according to the specifications document.

Other sources of inspiration for enhancing substitution

  • In addition to the experience from across the Atlantic, the project finds inspiration from substitution-related initiatives closer to home. Specifically, the Commission notes activities like the free Marketplace platform, developed and administered by Swedish NGO ChemSec, which seeks “to connect companies in the EU and to allow buyers and sellers of alternatives to hazardous chemicals to interact and forge partnerships.” Other initiatives highlighted include the Swedish Center for Chemical Substitution (RISE), a state-owned undertaking, as well as several other government-run entities that work to advance substitution efforts. In this context, the Commission mentions SUBSPORTplus! of the German Federal Institute for Occupational Safety and Health (BAuA), Ineris Portal of France’s National Institute for Industrial Environment and Risks, and the EU’s Innovation Centre for Industrial Transformation and Emissions (INCITE).
  • Many industry associations have also been involved in efforts to substitute hazardous substances, the Commission notes, as well as “numerous partnerships with academia” and “research partnerships with the purpose of promoting safe and sustainable solutions.”
  • The study also looks to the approach taken under the 2023 Transition Pathway for the chemical industry for guidance on how substitution planning should be developed. Namely, the tender specifications document says, “The purpose of substitution planning is to start discussions at expert level (authorities, current users of the substance, alternative providers and independent experts) on possibilities to substitute and the relative advantages and disadvantages of potential alternatives at an earlier stage.” What results from the discussions should be “an action road map” that has considered “all relevant challenges and opportunities” and lays out “substitution with clear but flexible and adaptable milestones, which can provide guidance for all involved actors on the way forward towards substitution of a substance, or a group of substances.” Comparable to the Transition Pathway, the Commission states, “substitution planning is based on the identification of challenges in a complex transition and a road map or pathway of action lines to achieve the transition.”

Analysis and next steps

  • European regulators are looking at ways to improve hazardous chemicals substitution and substitution planning processes, given EU chemical legislation’s current shortcomings, including EU and member state authorities’ significant provision of time and resources, delays in regulatory decisions, and resulting uncertainties for business. The Commission has already started a project on strengthening the role of substitution planning, the results of which this study is intended to leverage. ( Refer to the AgencyIQ piece cited above under the “Regulatory context” section of this article.)
  • This study is meant to test the waters of a new regulatory direction, setting up a pilot substitution center or network of centers that will aim to “provid[e] scientific, technical and practical advice to companies and authorities to identify safer alternatives and reduce chemical risks through the substitution of hazardous chemicals,” as well as to support “EU companies, in particular SMEs, in the context of innovation and investments into safer alternatives to targeted chemicals in the EU.”
  • The study should “gain experience” with operating “an effective and efficient framework for substitution planning.” According to the Commission, the initiative should advance “collaboration of various actors towards identification of well performing, safer and cost-effective alternative solutions to uses of hazardous chemicals.” Focus should be on those cases where substitution is particularly complex or where regrettable substitution may play a key role. It should also provide support to stakeholders to identify the path, i.e., the action road map, and assist in realizing the concrete steps to take toward implementing alternatives to the hazardous chemicals they use.
  • The Commission is prioritizing the substitution of PFAS in batteries, chromium(VI) in hard chrome plating, and certain, yet-to-be-decided substances used in paints manufacturing. Based on the specifications of the study, the deliverables of the three other lots (i.e., minus the first one, which specifies the pilot project) will entail completing the action road maps for substitution of these three substance-application combinations and implementing them.
  • The Commission is presumably set to introduce significant modifications to how substitution (planning) of hazardous chemicals is handled in the EU. Considering the 1.5-million-euro price tag of this study, in addition to the resources the EU executive has already sunk to realize its other ongoing project addressing the role of substitution planning, these actions appear to signal serious plans for altering the approach to chemicals substitution. It’s still unclear whether such changes will entail new obligations under EU chemicals law or will be of a voluntary nature.
  • Entities interested in submitting tenders to participate in the Commission’s call have until Thursday, September 12, at 4 p.m. CEST to do so.

To contact the author of this piece, email Scott Stephens. ( sstephens@agencyiq.com).
To contact the editor of this item, please email Jason Wermers ( jwermers@agencyiq.com).

Key Documents and Dates

  • Tender specifications on Study on EU Substitution Centre(s) – Providing support to businesses to substitute their use of hazardous chemicals through collaboration, innovation, research and direct assistance (Call for tenders EC-GROW/2024/OP/0038)

Filed Under: Article

August 16, 2024 by

By Amanda Conti

On August 9, Lykos Therapeutics received a Complete Response Letter (CRL) from the FDA, stating the agency will not currently approve its application for midomafetamine (MDMA) to treat post-traumatic stress disorder. Here, AgencyIQ unpacks the announcement and analyzes potential implications throughout the psychedelic enterprise.

A growing body of evidence suggests that psychedelics may provide clinical benefit for certain purposes, especially mental health conditions.

Fill out the form to read the full article.

Filed Under: Article

August 16, 2024 by

The FDA rejected a psychedelic sponsor’s bid for approval. Here are six industry takeaways

On August 9, Lykos Therapeutics received a Complete Response Letter (CRL) from FDA stating the agency will not currently approve its application for midomafetamine (MDMA) to treat post-traumatic stress disorder. Here, AgencyIQ unpacks the announcement and analyzes potential implications throughout the psychedelic enterprise.

By Amanda Conti | Aug 13, 2024 10:00 PM CDT

Regulatory context: Psychedelic regulation and drug development

  • 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”).
  • The road to research on psychedelic products is paved with obstacles. First, nearly all psychedelics are Schedule I drugs under the Controlled Substances Act (CSA). This means that research programs are subject to additional oversight from U.S. Drug Enforcement Administration (DEA) and companies may face difficulties obtaining sufficient supplies for studies. Second, the perceptual disturbances caused by psychedelic substances beget study design challenges like functional unblinding, response expectancy; additionally, the therapeutic presence that traditionally accompanies psychedelic administration comes with “set and setting” nuances that can dramatically affect outcomes. [ Read AgencyIQ analysis here for a detailed discussion of these issues.].
  • FDA released a draft guidance in June 2023 attempting to address many of these unique aspects of clinical research on psychedelic substances. Although the agency maintains that “the substantial evidence standard for establishing effectiveness of psychedelic drugs is the same as it is for all other drugs,” it acknowledges many of the issues with devising an adequate and well-controlled (AWC) clinical study and provides some general input on alternative approaches to research in this area. [ See AgencyIQ’s analysis of the draft guidance here.].
  • An AgencyIQ review of the 223 comments on that guidance shows a disconnect regarding the role of and requirements for psychological support. Many commenters took issue with the guidance’s statement that, “The contribution of the psychotherapy component to any efficacy observed with psychedelic treatment has not been characterized. Psychotherapeutic interventions have the potential to increase expectancy and performance biases. Sponsors should plan to justify the inclusion of a psychotherapy component.” Many stakeholders felt that this statement showed a lack of understanding of the research in this area, in which, at minimum, some form of psychological support is considered standard.
  • Psychotherapy is considered to constitute the practice of medicine, placing regulation of its conduct outside the scope of FDA’s authority. However, the agency can describe a concomitant therapy in labeling for a product it has authority to regulate if it is essential for the therapeutic effect. While the ability to describe and specify aspects of the therapy is limited, the agency can specify the number and licensure of therapists who would participate in the MDMA treatment. The June 2023 guidance recommends that studies involve two monitors (one with graduate-level professional training and clinical experience in psychotherapy, licensed to practice independently) to observe study participants during treatment sessions.

Lykos Therapeutics’ New Drug Application (NDA) for MDMA capsules to treat post-traumatic stress disorder (PTSD)

  • A lengthy regulatory history: The initial investigational new drug application for midomafetamine was filed in 2001. Lykos (formerly MAPS PBC) met with the agency in 2016 for the End of Phase 2 meeting. At that time, FDA flagged concerns with functional unblinding and inherent expectation bias. FDA suggested that the sponsor incorporate an active comparator—a recommendation that was not ultimately taken. Throughout 2017, Lykos sought feedback on the protocol for its first Phase 3 study, MAPP1, via the Special Protocol Assessment (SPA) process. SPA allows sponsors to gain regulatory perspective on whether a proposed trial design is adequate to support a marketing application, though the ultimate approval decision is based on the data submitted for review. Following some back-and-forth regarding statistical analyses and endpoints and abuse potential studies, the agency agreed to a revised SPA in July 2017.
  • FDA accepted the Lykos NDA in February 2024, and the submission received priority review. The application relied on evidence from two multi-site, randomized, double-blind, placebo-controlled trials (MAPP1 and MAPP2) that compared MDMA-assisted therapy (MDMA-AT) to placebo plus therapy. Both studies measured Clinician-Administered PTSD Scale for DSM-5 (CAPS-5) severity score at baseline and two months after the last experimental session, with assessment performed by a “centralized pool of blinded, independent diagnostic assessors.” According to results published by the firm, both studies demonstrated a statistically significant reduction in CAPS-5 total severity score. Lykos also put forth data from the exploratory, non-interventional follow-up study (MPLONG) regarding the durability of the effects.
  • Importantly, midomafetamine was proposed for use in conjunction with psychotherapy. In the clinical studies, this consisted of a four-month course of treatment with three doses of MDMA administered in supervised therapy sessions. This involved preparatory therapy sessions and “integrative psychotherapeutic sessions” between medication sessions. The therapists used the nonproprietary “ MAPS Manual for MDMA-Assisted Psychotherapy in the Treatment of Posttraumatic Stress Disorder,” to guide therapeutic stance, but the practitioners had flexibility in terms of modality.

The advisory committee meeting

  • FDA’s Psychopharmacologic Drugs Advisory Committee (PDAC) met June 4 to weigh in on the Lykos Application. As AgencyIQ discussed in-depth at the time, the committee grappled with data flaws and gaps within the context of positive trials and high unmet need. Specifically, the data showed evidence of functional unblinding, as participants were able to accurately guess their treatment assignment most of the time. In addition, the studies collected limited clinical laboratory data to inform the safety profile and did not systematically document adverse events that could contribute to abuse potential.
  • FDA raised concerns about the psychotherapy component, since its use in both trial arms meant that its specific contribution to efficacy could not be quantified. Panel members flagged potential issues with variability between providers.
  • Separate from statistical concerns, PDAC had questions regarding potential misconduct within the development program. These concerns stemmed from a March 2024 Draft Evidence Report from the Institute for Clinical and Economic Review (ICER) related to the MAPP studies. “Based on public reports, there is no question that, despite the trial requiring dual treatment by one male and one female therapist, boundaries, including sexual boundaries, were severely crossed with at least one patient,” according to the report. In addition, the report alleged that “Concerns have been raised by some that therapists encouraged favorable reports by patients and discouraged negative reports by patients including discouraging reports of substantial harms, potentially biasing the recording of benefits and harms.” Lykos personnel responded by stating that internal investigations led to policy changes and additional training regarding boundary setting for patients and providers. FDA added that the agency had inspections ongoing and reiterated the importance of licensure of study personnel.
  • The agency proposed a Risk Evaluation and Mitigation Strategy (REMS) framework in case of approval. This would be necessary because of the “expected patient impairment from midomafetamine administration.” The proposed elements to ensure safe use (ETASU) plan included four components: (1) dispensing only in certain healthcare settings, (2) dispensing to patients with documentation of safe-use conditions, (3) monitoring each patient using the drug, and (4) enrolling each patient using the drug in a registry. The plan would also include an implementation system and timeline for assessment submission.
  • Ultimately, the panel voted 9-2 and 10-1 against the efficacy and risk-benefit ratio of Lykos’ drug. Lykos responded to the outcome of the meeting in a press release. Further, Lykos expressed confidence in the rigor of its study design, and asserted that clinical laboratory data regarding cardiovascular and hepatotoxicity risks could be addressed in the postmarket environment. Regarding the case of misconduct discussed at the meeting, the firm emphasized that it “reported this violation to Health Canada, the FDA, and the relevant Institutional Review Board and banned the therapist pair associated with this case from all future work.”

In early August, FDA aligned with its advisors and issued a Complete Response Letter (CRL) for the application

  • On August 9, Lykos announced that FDA issued a CRL for its midomafetamine application. The release discussed the agency’s justification for the decision somewhat ambiguously, saying that “the issues expressed in the CRL echo those raised during the FDA Advisory Committee meeting on June 4, 2024.” However, the firm also clarified that “FDA has requested that Lykos conduct an additional Phase 3 trial to further study the safety and efficacy of midomafetamine.”
  • Lykos’ announcement enumerated several of the firm’s concerns related to the PDAC roster and discussion, referring to “concerns around the structure and conduct of the Advisory Committee meeting, including the limited number of subject matter experts on the panel and the nature of the discussion, which at times veered beyond the scientific content in the briefing documents.” The meeting’s roster included five voting members from PDAC, six temporary voting members, and one non-voting industry representative. Areas of expertise included psychiatry, post-traumatic stress disorder, emergency medicine, pharmacy and medication safety, and statistics. Lykos also referenced ongoing efforts within the agency to reform the advisory committee process, although these are general, agency-wide efforts to “tune up our Advisory Committee system in a way that enables our experts to get the best advice possible,” as FDA Commissioner ROBERT CALIFF recently said. [ Read AgencyIQ analysis of a recent listening session on the topic here.].

Where does Lykos go from here?

  • Immediate next steps for Lykos will involve “a meeting with the FDA to ask for reconsideration of the decision and to further discuss the agency’s recommendations for a resubmission.” This meeting will likely be a Type A meeting under the Prescription Drug User Fee Act (PDUFA) [ See AgencyIQ analysis of a September 2023 guidance on these formal meetings.]. Regarding timing, FDA committed in PDUFA VII to respond to Type A meetings within 14 days and schedule them within 30 days of the initial request. After this meeting is held, Lykos says it expects to provide an update.
  • If resubmission is pursued, a new review cycle will be triggered. Resubmissions are categorized into Class 1 and Class 2 resubmissions, with Class 1 submissions requiring a new 2-month review cycle and Class 2 submissions requiring a new 6-month review cycle. The classification of a potential Lykos resubmission could depend on whether a new Phase 3 trial is undertaken.
  • Lykos maintains that a new trial may not be necessary. Lykos CEO Amy Emerson stated in the press release that “While conducting another Phase 3 study would take several years, we still maintain that many of the requests that had been previously discussed with the FDA and raised at the Advisory Committee meeting can be addressed with existing data, post-approval requirements or through reference to the scientific literature.” Still, the design of a new Phase 3 trial would offer insights into how the agency wants to improve the data issues and gaps discussed during the June PDAC meeting. Regarding the psychotherapy component of the protocol, the release stated that “Lykos remains committed to continuing development of this integrated approach.”
  • Lykos currently has other midomafetamine studies registered on ClinicalTrials.gov that could provide some clues regarding future directions. Lykos has contemplated alternative psychotherapy paradigms, as evidenced by the withdrawn Phase 2 clinical study that sought to characterize the feasibility and safety of MDMA-Assisted group therapy. Per the record, the study was withdrawn prior to enrollment of any patients due to “transfer of sponsorship.” Lykos may look again at different approaches to therapy to reduce variability and better characterize its contribution to efficacy. To address issues in the safety realm, the firm has currently registered an active, but not yet recruiting, open-label Phase 1 study that will “collect quantitative data on mood, psychological status, self-compassion, professional quality of life, and professional burnout” in a target of 150 healthy volunteers receiving midomafetamine. This study is planned to start in late 2024 and conclude in late 2025. Lykos also registered an additional open-label Phase 1 pharmacokinetics study of midomafetamine and metabolites in patients with moderate hepatic impairment to understand if a dose adjustment is necessary in that population. Potential hepatic risk was one data gap discussed at the advisory committee meeting. The study is planned to begin in 2026 and conclude in 2028.
  • Per its press release, the firm also plans to “take advantage of agency processes to resolve scientific disagreements.” This implies that Lykos is considering leveraging FDA’s Formal Dispute Resolution (FDR) process [ See AgencyIQ’s in-depth explainer here]. In addition to withdrawing or resubmitting their application, sponsors who receive a CRL can “request review of scientific controversy by an appropriate scientific advisory panel.” If the request is denied, the denial should be communicated by the FDA to the sponsor in writing. A denial may then be escalated to a higher office through a “request for review of the denial” by the FDA’s “Chief Mediator and Ombudsman.” [ For an FDR case study, see AgencyIQ’s analysis of Intarcia’s years-long battle over ITCA-650]. This process could culminate in circling back to yet another PDAC meeting as a final step. According to a 2017 final guidance document on this process, sponsors should not simultaneously pursue several different administrative remedies at the same time. This means that Lykos will likely initiate the FDR process based on the outcome of the abovementioned Type A meeting with the agency.
  • The FDR process is often time- and resource-intensive but does afford companies certain rights, and even potential benefits. As AgencyIQ has previously discussed, the communication obtained through the dispute process can offer additional, favorable information about how to obtain subsequent approval that may not have been previously available or made clear in the CRL process.

Six takeaways: What does the outcome mean for the psychedelic ecosystem?

  • First: Psychotherapy is a major quandary at best, and a landmine at worst. This class-wide issue been an ongoing point of discussion both in the U.S. and in Europe. At a recent workshop addressing regulatory issues with psychedelic medicines, MARION HABERKAMP, an EMA committee member and on staff with the German regulator BfArM, identified the question of whether psychotherapy and psychological support are integral to psychedelic therapy as one of the “main challenges” in the field. The FDA’s MARTA SOKOLOWSKA, who attended the EMA workshop in person, reiterated that the FDA does not regulate the practice of medicine, and hence cannot regulate psychotherapy [ Read AgencyIQ’s full analysis of that workshop here.].
  • Second, the challenges associated with integrating psychedelics with psychotherapy may lead some sponsors to drop the latter. Prior to the Lykos CRL, TIME Magazine reported that the CEO of Compass Pathways, a firm investigating psilocybin for multiple mental health conditions, said, “If and when we get to the point of filing, we will be filing for [only] a drug.” Compass has studied psylocibin for PTSD in an open-label, Phase 2 setting. According to a company press release, the drug was administered with “psychological support” from a licensed medical professional that involves more of a supervisory role in “preparing participants for the treatment session, observing and being present with patients during the session and supporting them after the session.”
  • Next, sponsor-FDA agreement on trial designs via SPA should be taken with a grain of salt. Receiving the agency’s nod through this process indicates “adequacy and acceptability of specific critical elements of overall protocol design (e.g., entry criteria, dose selection, endpoints, and planned analyses) for a study intended to support a future marketing application,” according to a 2018 final guidance on the topic. However, the agency makes clear that “an SPA agreement does not indicate FDA concurrence on every protocol detail.” The SPA did seem to benefit Lykos as they designed the trial. That said, recommendations received during this process should be followed closely.
  • Fourth, sponsors should anticipate a stringent REMS requirement. Through the advisory committee, the field learned FDA’s position that a REMS is unequivocally necessary for midomafetamine due to the perceptual impairment caused by the drug. As a result, most sponsors in this drug class should expect this requirement to come into play. The contours of the midomafetamine REMS laid out by FDA in the proposed ETASU, such as dispensing only in certain healthcare settings, can be incorporated into future study designs.
  • Also, the Lykos outcome could inadvertently chill some academic research. Following the announcement of the CRL, the editors of Psychopharmacology issued a retraction notice for a pooled analysis of Phase 3 trials of MDMA-assisted psychotherapy for PTSD. The notice explained that the retraction follows the discovery of undisclosed “protocol violations amounting to unethical conduct at the MP4 study site by researchers associated with this project” and undisclosed potential competing interest. Separately, in an unusual move made at an interesting moment, the FDA withdrew its plans for a behavioral economics study of real-world use of psychedelics and kratom the same day it issued the Lykos CRL. The exploratory study was intended to study risk and safety considerations for people who use “Kratom and Psychedelics alone and in combination with other substances,” to inform proactive work of the agency’s Controlled Substances Program (CSP) [ Read AgencyIQ analysis of the original announcement here.]. Per the withdrawal notice, “FDA no longer intends to proceed with the proposed study as described because circumstances occurred necessitating changes to the scope of the study.”
  • Last thoughts: Political pressure will likely continue—and heighten. Dozens of legislators had asked the FDA to consider approval for the drug in light of its potential benefits to veterans and others with PTSD. Given this attention, AgencyIQ would not be surprised to see FDA leadership called before Congress to explain the Lykos decision.

Featuring previous research by Alexander Gaffney.

To contact the author of this item, please email Amanda Conti ( aconti@agencyiq.com).
To contact the editor of this item, please email Kari Oakes ( koakes@agencyiq.com).

Key Documents and Dates

  • Lykos Therapeutics Press Release, August 9, 2024

Filed Under: Article

August 16, 2024 by

By Rachel Coe, MSC

Just over a year after it released the draft guidance, the FDA has finalized its guidance on dose optimization for oncology products. In this analysis, AgencyIQ has combed through the newly released final guidance document to tease out what’s new and what’s the same. We will discuss the context and implications of some of the changes in an upcoming second analysis.

For decades, sponsors have turned to maximum tolerated dose (MTD) as the primary method for dose selection in oncology.

Fill out the form to read the full article.

Filed Under: Article

August 16, 2024 by

FDA solidifies its advice on dose optimization for oncology products

Just over a year after it released the draft guidance, the FDA has finalized its guidance on dose optimization for oncology products. In this analysis, AgencyIQ has combed through the newly released final guidance document to tease out what’s new and what’s the same. We will discuss the context and implications of some of the changes in an upcoming second analysis.

By Rachel Coe, MSC | Aug 12, 2024 6:15 PM CDT

Background

  • For decades, sponsors have turned to maximum tolerated dose (MTD) as the primary method for dose selection in oncology. In early-stage development of traditional cytotoxic oncology drugs (i.e., chemotherapies), sponsors have historically pushed the envelope to find the highest tolerable dose when conducting clinical trials for oncology products, since higher doses generally mean increased efficacy for these products. These products have well-characterized pharmacology; sponsors have an understanding of the tradeoff between dose response and toxicity, with each climbing steeply in these products with narrow therapeutic indexes. The MTD represents the point at which the serious toxicities outpace the therapeutic benefits of these products.
  • Even though chemotherapies, both as monotherapy and in combination with other products, have become a mainstay in the treatment of most cancers today, they present many drawbacks. These cytotoxic chemicals have broad distribution throughout the body following administration; by non-selectively targeting rapidly dividing cells, they can cause debilitating and even fatal side effects. Furthermore, these products present risk for serious harm if administered incorrectly.
  • As the field has moved into a new era of oncology products, the MTD approach is less useful. Some treatments with more targeted effects, such as immunotherapies and new molecular targeted agents (MTAs), may present more favorable side effect profiles, while achieving their effects using different—and often more complex—biological pathways. As opposed to the linear dose-response relationships of chemotherapies, the wide therapeutic indexes of many of these newer therapies means that doses can be increased without simultaneously increasing toxicities. However, maximum therapeutic benefit may be reached at relatively low doses for some targeted therapies, before toxicities are experienced. Other issues also arise: Unlike conventional cytotoxic drugs where the relationship between early (e.g., intermediate or surrogate) endpoints and clinical outcome endpoints (e.g., overall survival) are more consistently observed, targeted therapies might improve overall survival without displaying any effect on a commonly measured intermediate endpoint. Further, some newer therapies are associated with immune-mediated adverse events such as cytokine release syndrome that are not always dose-related. Additionally, some immune modulators are meant to be taken chronically, with toxicities that may not appear for months, or even longer.
  • Further, even for conventional cytotoxic therapies, this “highest dose” approach ignores a number of factors, including target interactions and off-target toxicities, longer-term toxicities, and safety considerations outside of dose-limiting toxicities. And regardless of the type of therapy, dose-limiting toxicities may result in dosage interruption, de-escalation, and even discontinuation, in the clinical trial setting as well as with real-world dosing.
  • Returning to long-term considerations, diagnostic and therapeutic advances in oncology have moved many cancer types into the categories of curable or treatable diseases. The areas within oncology that have seen the most success in terms of new and effective treatment options are now entering a new paradigm where the terms “success” and “harm” are being redefined. Previously, burdensome side effects may have been written off as a necessary part of a relatively short course of treatment, with benefits measured in additional weeks or months of survival time.
  • Today, though, patients, clinicians, and regulators expect more. As long-term treatment options become more prevalent, some cancers are being treated like chronic conditions, leading to questions and concerns about long-term effects that can now be measured on a longer time scale as more patients are living longer. The MTD approach evaluates only a few patients at each dose, utilizes a short observation period to observe dose-limiting toxicities (DLTs), and provides limited information on safety beyond DLTs, making it challenging to get a realistic picture of long-term safety and tolerability.

What came before this final guidance

  • Dose optimization for oncology products has been an issue flagged for improvement even prior to the creation of FDA’s Oncology Center of Excellence (OCE) in 2017. However, in recent years, FDA leadership has gradually taken an increasingly stronger stance that the era of simply defaulting to MTD is coming to an end. In parallel, stakeholders have advocated for recognizing the importance of treatment tolerability when determining doses and regimens. The side effects of some newer anticancer treatments can also result in dose interruptions or de-escalations, or even outright discontinuation of an efficacious therapy. These issues are especially apparent in treatments that are intended for chronic use. As a result, products receiving approval due to their demonstrated safety and efficacy in the clinical trial setting may lack real-world effectiveness, with patients failing to see material benefit as they deal with persistent and severe side effects.
  • In 2021, FDA’s efforts on dose optimization came to the forefront with the announcement of Product Optimus. At the time, OCE Director RICHARD PAZDUR remarked that the field of oncology had long suffered from a cultural bias that “more is better” when it comes to dosing oncology products. “We’re not asking anything more than [we are for] any other therapeutic areas,” Pazdur explained, adding that OCE is “going to start making it more of a requirement” that companies conduct dose-finding to facilitate optimization and that it is in companies’ “best interest” to follow this approach.
  • In January 2023, FDA’s work on dose optimization made the leap from research to policy, starting with the release of OCE’s long-awaited guidance on dose optimization for oncology products. In the guidance, FDA outlined why the MTD strategy is particularly inappropriate for targeted therapies, which “demonstrate different dose-response relationships compared to cytotoxic chemotherapy, such that doses below the MTD may have similar efficacy to the MTD but with fewer toxicities,” according to the guidance, which also noted that “Additionally, the MTD may never be reached in certain situations.” The FDA also noted the shifting considerations for evaluating toxicity in modern products meant for long-term or chronic use. Furthermore, the guidance noted that when subsequent indications are being considered for already-approved products, sponsors should recognize that dosages may differ because of differences in the tumor biology, patient population, and concurrent treatments, among other reasons. Accordingly, nonclinical and clinical data will need to be gathered to support a subsequent indication, the guidance document clarifies, adding that “strong rationale for choice of dosage should be provided” before a registration trial for a new dosage is launched. However, the response to FDA’s draft was mixed, ranging between solid support and fervent opposition to the operational approaches proposed by FDA’s guidance. [Read the analysis of the comments submitted here]
  • Since then, this topic has continued to be a priority for the FDA. Just a few months after the draft guidance on dose optimization was published, a handful of OCE staff, including its leaders, published a landmark article in March 2023. Foreshadowing several announcements that followed, the publication casted doubt on several of the most popular surrogate endpoints used for oncology approvals and proposed a few potential explanations for why they sometimes appear to diverge from long-term, outcome-related clinical endpoints. The possibilities cited included magnitude of effect, trial design, inadequate attention to dose optimization, and differential effects on various subpopulations within the overall intent-to-treat population. In particular, the paper stated that the relationship between dosage, toxicity, and efficacy or dose optimization is not always adequately explored by sponsors before initiating randomized trials for drug registration. And as investigational drugs are increasingly delivered orally and for longer periods of time, better dose optimization is critical to maximize adherence. Furthermore, the authors stated that inadequate attention to dose optimization, for both monotherapy and combination regimens, may contribute to randomized trials with potential overall survival (OS) detriments.

What’s new?

  • On Friday, the FDA issued its final guidance on optimizing the dosage of human prescription drugs and biological products in treating oncologic diseases. Per the Federal Register notice, the agency made a handful of changes to the draft based on stakeholder feedback, including an enhanced focus on “FDA’s availability to provide product-specific advice since there is not a one size fits all approach and clarity on the collection and analysis of key pharmacologic data.”
  • Scope: In line with the draft, the final version of the guidance provides general recommendations to sponsors on how and when to perform dose-finding studies to “adequately evaluate a range of dosage(s)” and select appropriate dosages to be further investigated with consideration for “all available clinical data, and a preliminary understanding of dose- and exposure-response (such as for safety, tolerability, and activity).” Likewise, it still omits radiopharmaceuticals, cancer vaccines, cell and gene therapy products, and microbiota-related products from its scope. The final version newly clarifies that oncolytic viruses also fall outside the scope of the document and includes a new disclaimer that the guidance is not intended to address unique considerations of pediatric drug development.
  • In general, the take-home messages from the background and introductory portions of the guidance are the same as they were in the draft. The background section outlines the shortcomings of the long-standing MTD strategy and explains why this approach is particularly inappropriate for targeted therapies, which “demonstrate different dose-response relationships with wider therapeutic indices compared to cytotoxic chemotherapy, such that doses below the MTD may have similar activity to the MTD with fewer toxicities or the MTD may never be reached,” according to the guidance. The FDA makes the additional point that many modern therapies are meant to be taken for long periods of time, even chronically in some cases, shifting considerations for evaluating toxicity. Several minor changes scattered throughout these sections rein in some of the previous draft’s wide-ranging assertions about the process used by sponsors to select a product’s dosage. For example, the draft stated that after identifying the MTD, “Sponsors typically administered the MTD, or a dosage close to the MTD, in subsequent clinical trials without further efforts to optimize the dosage.” The final guidance removes the latter half of this sentence. It also newly adds that, “In some cases, adverse reactions may negatively impact overall survival,” a point that we have heard several times from OCE leadership in recent years.
  • Dosage optimization recommendations: As before, the guidance states that the dosages selected for administration to patients in a clinical trial should be supported by “relevant nonclinical and clinical data,” as well as what’s known about both dose-response and exposure-response for the candidate drug or biologic in terms of both safety and efficacy. The FDA will take a dim view of approaches that select doses “without adequate justification or consideration of all relevant data,” both because of the potential risk to patients and because the trial design might not be able to meet its stated objectives. The guidance emphasizes that identification of an optimized dosage(s) can occur prior to or concurrently with the establishment of the drug’s safety and efficacy; however, this process should occur prior to the submission of a marketing application. Another addition to FDA’s general advice is a recommendation for sponsors to engage early with the FDA “early in clinical development” (as opposed to “at relevant milestone meetings”).
  • Pharmacokinetic (PK) sample and analysis planning: Generally, the recommendations in the final guidance are nearly identical to those contained in the draft, with a few exceptions. Trials involving dose-finding should feature a robust clinical pharmacokinetic (PK) component that includes PK characteristics such as linearity, absorption, and elimination of the product, “following the first dose and at steady-state (or after administration of multiple or repeated doses if steady-state will not be reached) for each dosage evaluated in the trial.” The distinction between first dose and steady state is new to the final version of the guidance. Still, it reiterates that a pre-specified sampling and analysis plan should support population PK and the required analyses of dose-response and exposure-response relationships.
  • Timing of population PK and exposure-response analyses: Another change is the final version now clearly recommends that recommends that these analyses should be initiated early and updated as additional data become available to “identify specific populations (e.g., defined based on weight, age, sex, race and ethnicity, organ impairment, genetic factors) in which the PK demonstrate clinically meaningful differences in exposure.” It further clarifies that the metrics used in these analyses “for evaluating safety (such as laboratory data and adverse events) and activity (such as tumor-assessment based endpoints or other biomarkers) should be appropriate for the stage of development, drug, and disease setting”; also, they should incorporate relevant covariates.
  • To be [administered with] food or not to be? The draft specified that for oral drugs, “the effect of food on PK and safety should be evaluated early in drug development to support the relative administration of the dosage(s) selected for evaluation in a registration trial(s) with food.” The final version refers to “pivotal trial(s)” rather than registration trials, and also tacks on the phrase “when applicable” to the end of the sentence.
  • Model-informed approaches and leveraging previously collected data: The incorporation of this new theme starts with the addition of a recommendation that sponsors should consider whether the FDA’s Model-Informed Drug Development (MIDD) paired meeting program could be utilized. Furthermore, the guidance later (page 5) endorses the use of semi-mechanistic or mechanistic modeling to support the selection of dosage(s) to be evaluated in clinical trials. Regarding the design of trials to compare multiple dosages, it newly adds that “Data from products in similar classes or with the same mechanism of action can also be used, when appropriate, to support the dosages for further evaluation, if relevant” and that “Model-informed or model-based approaches can be helpful to identify and select the dosage(s) to be compared.”
  • Other trial design considerations: The importance of randomization was emphasized in the draft and has been carried through to the final version. Additionally, the final version endorses two new measures to improve the rigor of oncology trials and the analyses of their results. First, it states that “blinding patients and investigators to dosage arm assignment may be considered as there could be bias that higher dosages are associated with greater activity.” Secondly, it notes that “If crossover is permitted, the analysis plan should pre-specify how safety and activity will be assessed to account for crossover.”

What’s next?

  • One of the most subtle yet significant differences between the draft and the final guidance is the language used to define dosage optimization. While the draft stated that the term “optimal dosage” referred to the “dosage that can maximize the benefit/risk profile or provide the desired therapeutic effect while minimizing toxicity,” the final guidance explains the concept differently. Instead of suggesting that a singular, optimal dosage exists for each product—the new guidance discusses dose optimization as the process used by sponsors to arrive at an optimized dosage, with the identical wording to define this term as was used in the draft version. It goes on to acknowledge that the best approach for identifying optimized dosage(s) “depends upon a variety of factors including but not limited to the drug class, proposed indicated patient population, and prior knowledge about the drug that is pertinent to dosing.”
  • This slight adjustment corresponds directly to the concerns of patient advocacy organizations, who suggested that the term “optimal dose” is deceptive. By the reasoning of these groups, since the word “optimal” denotes a singular, best version of something, the phrase “optimal dose” leads patients into falsely believing that sponsors have undertaken a process to identify the singular, best dosage of a product. The result, as articulated by the LUNGevity Foundation, is that it overpromises the results for patients who may “believe that with dose optimization in place, drug toxicities and dose reductions will be things of the past. However, in reality, no dose will be ‘best’ for everyone regardless of how carefully and with how much information it was selected.” In turn, it requested that OCE adopt “more practical language, such as ‘dose selection,’ and be candid about what the new expectations for arriving at the recommended dose will—and will not—mean for patients.”
  • What happens if sponsors do not adequately support the dosage(s) selected for use in clinical trials? Clarifying the consequences of a trial design that FDA deems deficient, the guidance newly adds that this may cause the FDA to place a protocol on clinical hold. Once again, it states that products being developed under expedited pathways will not be given special treatment. The agency makes this clear in no uncertain terms: “Sponsors should note that development of a drug under an FDA expedited program (e.g., breakthrough therapy designation) is not a sufficient justification to avoid identifying an optimized dosage(s) prior to submitting a marketing application.”
  • Stay tuned for Part 2 of Agency IQ’s analysis of the final guidance.

To contact the author of this item, please email Rachel Coe ( rcoe@agencyiq.com).
To contact the editor of this item, please email Kari Oakes ( koakes@agencyiq.com).

Key documents

  • Federal Register Notice: Optimizing the Dosage of Human Prescription Drugs and Biological Products for the Treatment of Oncologic Diseases
  • Final guidance (August 2024)

Filed Under: Article

  • « Go to Previous Page
  • Go to page 1
  • Go to page 2
  • Go to page 3
  • Go to page 4
  • Go to page 5
  • Go to page 6
  • 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