• 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

July 26, 2024 by

EU executive consults on proposed update to PIC chemical lists

Regulation (EU) 649/2012 (PIC Regulation) is the main act on the export and import of hazardous chemicals between the European Union and third countries. It also implements EU commitments to the Rotterdam Convention on the prior informed consent (PIC) procedure for certain hazardous chemicals and pesticides in international trade.

BY SCOTT STEPHENS, MPA | JUL 23, 2024 10:04 PM CDT

Regulatory background

  • Regulation (EU) 649/2012 (PIC Regulation) is the main act on the export and import of hazardous chemicals between the European Union and third countries. It also implements EU commitments to the Rotterdam Convention on the prior informed consent (PIC) procedure for certain hazardous chemicals and pesticides in international trade. It seeks to protect human health and the environment, while reaffirming the sovereignty of third nations in deciding when hazardous chemicals are allowed to enter their borders.
  • Annex I of PIC Regulation consolidates all hazardous chemicals which are subject to special export and import procedures. Part 1 of the annex lists substances that carry the obligation to submit an export notification. This is accomplished using ECHA’s ePIC platform, allowing non-EU countries advance notice of hazardous substances potentially arriving in their countries.
  • Parts 2 and 3 of annex I list substances which, in addition to needing export notification, require prior informed consent. The importing country must grant consent through ePIC for the shipment to progress.
  • The chemicals listed in part 3 exactly match those included in annex III of the Rotterdam Convention. Any other chemicals that the EU determines should be subject to the same heightened export procedure, though not yet recognized by the Rotterdam Convention, are added to part 2. Where the Rotterdam Convention subsequently adds chemicals already listed in part 2, these chemicals are deleted from part 2 and added in part 3 in the next PIC update to avoid redundancy.
  • Annex V of PIC Regulation lists substances added to annexes A and B of the Stockholm Convention and are subject to a complete export ban. Part 1 of annex V contains the convention-listed persistent organic pollutants (POPs), while part 2 comprises the chemicals other than POPs listed in the convention’s annexes.
  • The Commission is mandated to review PIC Regulation’s annex I list of chemicals at least once a year, harmonizing it with new regulatory developments under the Rotterdam Convention and EU chemicals legislation regulating related hazardous substances. Specifically, PIC Regulation implements import and export regulatory measures based on the effective restrictions and bans placed on pesticide active substances under Regulation (EC) 1107/2009 on plant protection products (PPPR); biocidal active substances pursuant to Regulation (EU) 528/2012 on biocidal products (BPR); industrial chemicals under Regulation (EC) 1907/2006 on the registration, evaluation, authorization and restriction of chemicals (REACH); and persistent organic pollutants in accordance with Regulation (EU) 2019/1021 on POPs (POPs).

Commission proposes aligning PIC lists with amendments to PPPR, BPR, REACH and POPs

  • The Commission on July 19 published a draft delegated regulation on its “Have your say” portal, launching a four-week pubic consultation to solicit feedback on proposed updates to annexes I and V listing hazardous substances subject to PIC’s import and export provisions.

Proposed updates to PIC’s parts 1, 2, and 3 of annex I and annex V

  • In part 1 of annex I, the proposal is adding 41 new entries and making minor changes to three existing ones. See the entries (and accompanying CAS identifiers) below that are being added. The lion’s share of these substances have been added because they were either not renewed or not approved under PPPR – effectively banning them from use as pesticide active substances – and have been assigned harmonized classification and labeling under Regulation (EC) 1272/2008 on classification, labeling and packaging of substances and mixtures (CLP), demonstrating “sufficient evidence that” these substances “[raise] concerns for human health or the environment.”

Chemical

CAS RN (or EC No.)

1,2-benzenedicarboxylic acid, dihexyl ester, branched and linear

68515-50-4

1,2- benzenedicarboxylic acid, di-C6-10-alkyl esters or mixed decyl and hexyl and octyl diesters

68648-93-1

68515-51-5

Abamectin

71751-41-2

Acrolein

107-02-8

Asulam-sodium

2302-17-2

Bendiocarb

22781-23-3

Benfluralin

1861-40-1

Benthiavalicarb

413615-35-7 177406-68-7

Calcium phosphide

1305-99-3

Chloridazon

1698-60-8

Chlorsulfuron

64902-72-3

Clofentezine

74115-24-5

d-Allethrin

231937-89-6

Difenacoum

56073-07-5

Dihexyl phthalate

84-75-3

Dimethomorph

110488-70-5

Dimoxystrobin

149961-52-4

Fenpropimorph

67564-91-4

Flusilazole

85509-19-9

Fuberidazole

3878-19-1

Ioxynil

1689-83-4

3861-47-0

Ipconazole

125225-28-7 115850-69-6 115937-89-8

Mepanipyrim

110235-47-7

Methylene dithiocyanate

6317-18-6

Metiram

9006-42-2

Molinate

2212-67-1

Oxadiazon

19666-30-9

Oxamyl

23135-22-0

Penflufen

494793-67-8

Profenofos

41198-08-7

Quinoclamine

2797-51-5

Silver copper zeolite

130328-19-7

Silver sodium hydrogen zirconium phosphate

265647-11-8

Silver zeolite

130328-18-6

S-metolachlor

87392-12-9

Sodium perborate, perboric acid, sodium salt

15120-21-5

10332-33-9

10486-00-7

13517-20-9

90568-23-3

11138-47-9

125022-34-6

Sodium peroxometaborate

7632-04-4

Spirotetramat

203313-25-1

Tralkoxydim

87820-88-0

Triadimenol

55219-65-3

Triflusulfuron-methyl

126535-15-7

  • The existing entries for cyanamide (CAS RN 420-04-2), warfarin (CAS RN 81-81-2), and terbufos (CAS RN 13071-79-9) are updated to reflect the fact that the first two are being added to part 2 of annex I (i.e., they are now subject to the provisions qualifying them for PIC notification) and the final substance is being moved from part 2 to part 3 of the annex, because terbufos was included in May 2023 in annex III of the Rotterdam Convention).
  • The substances proposed for listing in part 1 of annex I, except for abamectin (CAS RN 71751-41-2) and difenacoum (CAS RN 56073-07-5), are also being included in part 2 of the same annex. Cyanamide and warfarin, as mentioned above, are also being incorporated in part 2.
  • Terbufos (CAS RN 13071-79-9) is being added to part 3 of annex I, as explained above.
  • A minor change is being made to the existing entry for polychlorinated biphenyls (PCB) in part 3 of annex I to expand the entry to sufficiently cover all members of this substance group, including homologs, congeners and mixtures, as reflected in the corresponding annex III entry to the Rotterdam Convention.
  • Part 1 of annex V is being harmonized with the amended POPs Regulation, encompassing an entry for perfluorohexane sulfonic acid (PFHxS), its salts and PFHxS-related compounds (355-46-4 CAS RN and others), as well as a minor update to the existing entry for endosulfan, to which additional CAS numbers have been added.

Analysis and next steps

  • The deadline to provide feedback on the Commission’s proposed regulation is August 16 under the “Have your say” consultation. Because the proposal is a delegated act, aimed at updating “non-essential” elements of substantiative legislation, the EU Parliament and the Council are only minimally involved in the regulation’s development, empowered only to object to the regulation during a scrutiny period that will follow the consultation.
  • Once adopted and having successfully completed scrutiny, the regulation will be published in the Official Journal. Affected stakeholders will then have until January 1, 2025, when the regulation begins to apply, to prepare for compliance with the requirements triggered by the listing of the new substances in annexes I and V of PIC Regulation. For the 41 substances listed above, this will entail new reporting obligations, including export notifications, via the ePIC dossier submission tool.

Featuring previous analysis by Rayan Bhargava

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

Key Documents and Dates

  • Export and import of hazardous chemicals – review of the list of chemicals subject to international trade rules; Have your say initiative; July 19, 2024
  • Regulation (EU) No 649/2012 of the European Parliament and of the Council of 4 July 2012 concerning the export and import of hazardous chemicals
  • Rotterdam Convention on the Prior Informed Consent Procedure for Certain Hazardous Chemicals and Pesticides in International Trade

Filed Under: Article

July 26, 2024 by

BY COREY JASEPH, MS, RAC

The newly available May 2024 meetings of the Medical Device Coordination Group provide insight into the European Commission’s efforts to fully implement the European medical device and diagnostic regulations. AgencyIQ took a look at the doings of the group and its subcommittees that are most pertinent for device manufacturers.

The Medical Device Coordination Group (MDCG) is an expert group addressing “key issues from the medical devices sector” (including IVDs).

Fill out the form to read the full article.

Filed Under: Article

July 26, 2024 by

Top IVDR Notified Body concern: Lack of E.U. reference laboratories for high-risk diagnostics

The newly available May 2024 meetings of the Medical Device Coordination Group provide insight into the European Commission’s efforts to fully implement the European medical device and diagnostic regulations. AgencyIQ took a look at the doings of the group and its subcommittees that are most pertinent for device manufacturers.

BY COREY JASEPH, MS, RAC | JUL 25, 2024 11:26 PM CDT

Quick regulatory background on the MDCG

  • The Medical Device Coordination Group (MDCG) is an expert group addressing “key issues from the medical devices sector” (including IVDs). Article 103 of the Medical Device Regulation (“MDR”, Regulation (EU) 2017/745) established the MDCG to “deal with key issues from the medical devices sector.” Article 105 of the MDR outlines the tasks of the MDCG, including “to contribute to the development of guidance aimed at ensuring effective and harmonized implementation of this Regulation.” The MDCG also addresses issues related to in vitro diagnostic devices and the In Vitro Diagnostic Regulation (IVDR; Regulation (EU) 2017/746).
  • Thirteen subgroups address different aspects of the regulations by providing “advice and draft guidance on their expertise field;” each subgroup has one expert representative (with designated alternates) from each Member State. Subgroups address Notified Body oversight, standards, clinical investigation/evaluation, post-market surveillance and vigilance, market surveillance, borderline and classification, new technologies, unique device identification (UDI), IVDs, nomenclature, Annex XVI products, EUDAMED, and international matters. These groups “meet regularly with the EU Commission as Chair.”
  • There is a yearly calendar of meetings for the group that sets out when the MDCG plans to meet, when it will meet with stakeholders, and the subgroup meeting schedule. Stakeholders are largely industry groups (e.g., APPLiA, AESGP, BioMed Alliance, MedTech Europe), but also include associations representing Notified Bodies, authorized representatives, healthcare professionals (e.g., ECOO, HOPE, ESC) and patients (e.g., EPF). Additional information about the meetings can be found here; this includes meeting attachments, general agendas, minutes and presentations. These resources provide a look into what each group is working on before the policy, regulation or guidance document being developed is ultimately published.

AgencyIQ walked through the newly available May meeting notes to highlight key developments for the device and diagnostic industries

  • The MDCG met privately and with stakeholders in May 2024 to discuss implementation of the MDR and IVDR; these minutes have just been made public. This analysis focuses on a handful of the projects with the biggest impact for device and diagnostic manufacturers, including the E.U. Reference Laboratories, a recent poll of Notified Bodies on their views of MDR/IVDR implementation challenges, the Medical Device Single Audit Program (MDSAP) and overlap with other E.U. regulations, specifically the Substances of Human Origin (SoHo) regulation and the AI Act.
  • Some news on E.U. Reference Laboratories: E.U. Reference Laboratories, or EURLs, are a key component of both assessing high-risk diagnostic devices and ensuring they maintain performance, by testing them against common specifications or other validated tests. [ Read more about what EURLs are and what they do here.] The Commission designated the first five EURLs in December 2023, but these covered just four of the eight categories of high-risk diagnostic tests – hepatitis or retrovirus, herpesvirus, bacterial infection, and respiratory virus infection. The other four categories do not yet have designated EURLs (arboviruses, hemorrhagic fever and other biosafety level 4 viruses, parasites and blood grouping).
  • In February 2024, the Commission launched a new expression of interest to find more EURLs, asking for labs that could cover the four remaining diagnostic categories. If enough interest was generated, the Commission would launch a formal second call. Labs had until April 30, 2024 to contact the Commission. As of May 6, 15 Member States responded that they found 14 laboratories from eight countries that were interested in applying to be EURLs. Based on that information, the Commission proposed launching a second call for parasites and blood grouping testing labs; however, it didn’t find enough expertise or coverage for arboviruses and biosafety level 4 virus testing. As of this week, the Commission hadn’t yet launched the formal second call.
  • NBCG-Med poll of Notified Bodies: NBCG-Med is a Commission expert group composed of Notified Bodies with observer status for the MDCG. In April 2024, the group held a plenary meeting at which it took the opportunity to poll its members on their greatest MDR and IVDR concerns. NBCG-Med presented the results of the poll at the MDCG/stakeholders meeting in May.
  • MDR poll concerns: The top concern from Notified Bodies was the amount of effort it was taking Notified Bodies to implement Regulation (EU) 2023/607 (the mid-2023 extention and transitional provisions for IVDR/MDR implementation). Notified Bodies complained about receiving MDR applications from manufacturers with no real intention to transfer to MDR, as well as the burden of handling change requests for legacy devices and surveillance of devices with expired certificates. The next biggest concern was manufacturers’ overall lack of preparedness – including those not submitting technical documentation, not responding to Notified Body requests, threats of legal procedures, and not submitting clinical data. Next on the list of concerns was harmonization among Notified Bodies on questions of substantial change, depth of manufacturer and product assessment, clinical requirements and interpreting MDCG documents. The last concern was MDR implementation, wherein some parts of the regulation were difficult to implement and there had been an unrealistic expectation that all Notified Bodies would be designated and would have reviewed and certified all devices within four years.
  • IVDR poll responses: By far the top concern for IVDR-designated Notified Bodies was the EURLs; in particular, the first batch of designations took a long time, and even then the first group designated covered only half of the high-risk diagnostics. The first group isn’t yet operational, but is expected to start testing activities by October 1, 2024. The second major concern was the amount of effort Notified Bodies are having to put toward implementing Regulation (EU) 2024/1860, which further lengthened the transition period for IVDs and allowed for a gradual rollout of the EUDAMED database, rather than focusing on IVDR implementation. The next concern was the IVD designation codes outlined in Regulation (EU) 2017/2185, with Notified Bodies complaining that five-factor coding was too granular, IVT codes (which are intended to describe technologies) were not all relevant to IVD manufacturing processes, and there was a lack of clarity in how to apply codes. The fourth area of concern was the Notified Body designation process, the lack of pathways for innovation and the lack of manufacturer preparedness.
  • Update on Europe and the Medical Device Single Audit Program (MDSAP): MDSAP participation means that a single regulatory audit can cover multiple jurisdictions; Europe is not currently a participant in the program, but is an observer has been an observer but not a participant. [ Read more about the MDSAP program and who participates here.] The Commission has been evaluating participating fully in the program for some time, and the NBCG-Med took the opportunity at the May 2024 MDCG meeting to tout its benefits. The group suggested that adding the E.U. to the MDSAP certificate might increase acceptance of MDR/IVDR and lower trade barriers, as the Global Harmonization Task Force did in previous years. NBCG-Med also pointed to its predictable and uniform audit process and reduction of inspectors. For Notified Bodies also certified for MDSAP, it would allow for uniform audit checklists, planning, and reports, thereby reducing administrative burden. The group did acknowledge more groundwork would be needed, to include guidance on performing MDR/IVDR audits under MDSAP and an assessment as to whether an extra MDR/IVDR QMS assessment would be needed on top of MDSAP audit.
  • The interaction of the Substances of Human Origin (SoHo) Regulation on MDR and IVDR: The SoHo sector relies on diagnostics to have safe products for use in humans. The use of human tissues and tissue derivatives in devices now need to take into account the SoHo regulation. Blood bags and the chemical classification of bisphenol A, di(2-ethylhexyl)phthalate (DEHP) can impact SoHo and their collection and use. Critical devices for the SoHo space are those diagnostics used for testing the blood supply and devices that separate blood components for use in stem cell transplants, CAR-T cell therapy and other applications. At the May 2024 meeting, MDCG members and stakeholders received updates on the “state of play of the new EU legal framework” under the SoHo regulations. These regulations enter into force during 2024 but have a three-year implementation period. As reflected in the meeting minutes, “stakeholders highlighted the importance of involving all the necessary stakeholders for the proper implementation of the new legislation.”
  • And finally: The AI Act. The long-awaited AI Act was formally published in the Official Journal earlier this month. As of the May 2024 MDCG meeting minutes, the Commission had “provided an update on the AI Act and the interplay with MDR and IVDR.” While the Commission has committed to prioritizing guidance on this topic – specifically a Q&A document – there are other things that needed to happen first: “The Commission explained that the prioritisation of these activities depended on the appointment of the AI board, which would adopt their Terms of Reference during their first meeting in June 2024.” While industry is still awaiting a formal Q&A guidance, AgencyIQ has a two-part analysis of the implications of the AI Act for device and diagnostics companies available here: Part One, Part Two.

Summary and analysis

  • Once again, a dive into MDCG meetings uncovers information that isn’t easily available elsewhere. Summing up the details:
  • The next call for EURLs won’t cover all eight categories of high-risk diagnostics. At best, it will cover two of those four, parasites and blood grouping. But in the EURL meeting presentation, the Commission noted that there will likely be a drop-off in the number of labs that meet all the EURL requirements, similar to what happened with the first call. This leaves arboviruses and biosafety level 4 viruses with no designated laboratories, though the Commission is quick to note that EURLs aren’t required for conformity assessment where laboratories aren’t yet available.
  • Not all manufacturers that submitted MDR notifications really meant it. Notified Bodies are concerned about manufacturer readiness for the MDR and IVDR, but they also struggle with implementing the regulations themselves. A new issue that hasn’t been discussed much is Notified Bodies receiving MDR applications from manufacturers that don’t plan to actually complete the MDR transition. This may be related to the complaint that manufacturers aren’t submitting documentation or responding to Notified Body inquiries. There wasn’t much other information besides the poll responses at the NBCG-Med plenary meeting, but if this is a real concern, we should start to see it raised more publicly soon.
  • Europe keeps saying it wants to join MDSAP, but when? The MDCG has had many discussions on the viability of adopting the MDSAP audit model and participating in the certification program. The NBCG-Med pointed out benefits to doing so at the May 2024 MDCG meeting, including the possibility of increasing the overall acceptance of MDR and IVDR. But we have yet to see discussions on how to operationalize MDSAP within the MDR/IVDR or any firm plans for Europe to become a full member of MDSAP.
  • The policy landscape continues to increase in complexity. With regulations like SoHo and the AI Act impacting the device and diagnostics industries, and intersecting with the ongoing MDR/IVDR challenges, industry is facing a complicated landscape ahead.

To contact the author of this item, please email Corey Jaseph ( cjaseph@agencyiq.com).
To contact the editor of this item, please Laura DiAngelo ( ldiangelo@agencyiq.com).

Key Documents and Dates

  • Medical Device Coordination Group (MDCG) meeting, May 13, 2024
  • Meeting between the Medical Device Coordination Group (MDCG) and Stakeholders, May 13, 2024

Filed Under: Article

July 26, 2024 by

BY AMANDA CONTI

FDA has fulfilled its commitment under the Over-the-Counter Monograph Drug User Fee (OMUFA) program to issue final guidance on how sponsors can electronically submit monographs and other documents. The final document reflects the agency’s efforts to develop reference guides for its upload portals.

Fill out the form to read the full article.

Filed Under: Article

July 26, 2024 by

FDA finalizes guidance on electronic submissions for OTC products

FDA has fulfilled its commitment under the Over-the-Counter Monograph Drug User Fee (OMUFA) program to issue final guidance on how sponsors can electronically submit monographs and other documents. The final document reflects the agency’s efforts to develop reference guides for its upload portals.

BY AMANDA CONTI | JUL 24, 2024 9:57 PM CDT

Background: Modernizing regulation of over-the-counter (OTC) products

  • Nonprescription products, also called over-the-counter (OTC) products, must demonstrate the ability to be used safely and effectively without the supervision of a qualified healthcare professional. These products are regulated differently from traditional prescription drugs.
  • The FDA’s regulatory paradigm for these products has undergone modernization in recent years following the passage of the Over-the-Counter Monograph Safety, Innovation, and Reform Act within the Coronavirus Aid, Relief, and Economic Security Act (CARES Act). The law made key changes to how nonprescription products are regulated and also established the Over-the-Counter Monograph Drug User Fee (OMUFA) program to support the agency’s activities in this space. [ Read AgencyIQ’s explainer on the CARES Act here.]
  • Prior to CARES Act reforms, FDA relied on a monograph process through which firms could bring OTC drugs to market without FDA approval so long as they adhered to pre-set terms under the monograph. The monographs functioned as rule books for specific classes of OTC drugs, describing active ingredients, doses, indications, labeling and tests required for covered OTC products to be generally recognized as safe and effective (GRASE).
  • The new system replaced the monograph rulemaking process with an administrative process, allowing the FDA to approve a drug without first having to issue a proposed and final regulation for public comment. This enabled more efficient procedures for both agency- and sponsor-initiated operations. Additionally, if a change to a monograph is requested by the sponsor, the sponsor will be required to submit a new submission type called an OTC Monograph Order Request (OMOR).
  • The law also required the electronic submission of OTC monographs and other OTC related submissions—a requirement intended to further increase efficiency. This includes the submission of OMORs, public comments to administrative orders, formal meeting requests and packages, dispute resolution requests, administrative hearing requests, record request responses, as well as updates to drug listing information.
  • According to the OMUFA commitment letter, the FDA committed to the publication of a draft guidance on electronic monograph submissions by October 1, 2022, which must then be finalized by April 1, 2024.

FDA published draft guidance on electronic OTC submissions in late September 2022

  • The draft guidance, Providing Over-the-Counter Monograph Submissions in Electronic Format, explained that OTC monographs should be submitted through the CDER NextGen Portal or through the OTC Monographs@FDA portal. Further, “For OTC monograph submissions not explicitly described in this guidance, interested parties should check the OTC Monographs@FDA portal to see whether FDA has provided instructions on how they should be submitted electronically. If FDA has not provided instructions on how an OTC monograph submission not explicitly described in this guidance should be submitted, contact FDA at druginfo@fda.hhs.gov.”
  • What should be submitted to which portal? Per the draft guidance, OMORs, meeting requests and packages, formal dispute resolution requests, administrative hearing requests and responses to record requests should use the CDER NextGen portal. On the other hand, data, information and comments to a proposed or final order should use the OTC Monographs@FDA portal. Sponsors should continue to submit updates to drug listing information using FDA’s Electronic Drug Registration and Listing System (eDRLS).
  • CDER NextGen portal requires an applicant to create an account and does not allow editing once submitted. As the draft guidance noted, the receipt date for an electronic OTC monograph is based on passing validation checks on file properties such as file size and a virus scan. If successfully submitted, the applicant will receive confirmation both in the portal and as email confirmation with the receipt date. The guidance warned: “The receipt date for an OTC monograph submission is the date on which the request is deemed to have arrived at FDA. The receipt date should not be confused with the date of FDA’s subsequent decision to file a request.” Questions related to the portal can be submitted to the agency via email.
  • Sponsors should be aware the monograph order process is generally public. If any information is considered confidential, sponsors should follow instructions under the CDER NextGen portal or OTC Monographs@FDA portal on how to submit confidential information.
  • The draft guidance received a single comment from the Consumer Healthcare Products Association (CHPA) with mostly technical requests. CHPA asked for clarification for the file types and maximum file sizes accepted by each portal, along with more communication regarding workarounds (e.g., compressed files) and reasons for failed uploads. “Sponsors should know in advance if their file sizes will need additional CDER assistance prior to submission,” CHPA wrote, continuing, “A submission rejection because of file size limitation may lead to confusion and may ultimately cause the sponsor to miss their submission timeline date.” The organization also requested additional information in the guidance regarding portal accounts, particularly asking for further instruction if an individual does not have a secure email or certain other identifying information.

Now, FDA has finalized this document with minor changes

  • The draft guidance was brief and generally limited in content, and the final version is no different. Published July 25, 2024, the final guidance retains the vast majority of content from its predecessor, with a handful of wording changes and additional explanations. For example, rather than listing the information that should be submitted to different portals in paragraph form, the guidance now includes bulleted lists to increase readability.
  • The biggest change: the guidance contains many references to the CDER NextGen Portal Reference Guides. In places where the draft guidance encouraged sponsors to reference FAQ sites or email the agency directly, the guidance now provides links to reference guides that elaborate on information such as file naming conventions, acceptable file formats, and file size limitations. These guides represent a significant investment on FDA’s part in developing these resources. These address many of the questions raised by CHPA while giving FDA more leeway to update the guides as technology changes.
  • That said, the final guidance adds expanded instructions on “next steps” for sponsors once a CDER NextGen Portal account is created. Per the guidance, the portal will lead submitters through the process to establish a monograph file (MGF), which involves submission of a Pre-Assignment Request. Once this is reviewed, an MGF number is assigned, and the OTC monograph submission may proceed.
  • What’s next? With this guidance publication, FDA has fulfilled its commitment under OMUFA. Still, the agency is over three months late on the timeline set in the commitment letter. A couple of additional guidances agreed to under the user fee program have yet to be finalized, though they are likely to be forthcoming in the coming months. First, the agency agreed to finalize guidance on formal meetings between FDA and sponsors or requestors for OMUFA ingredients and drug products, which was released in draft form in February 2022 [ See AgencyIQ analysis here.]. Final guidance was expected in July 2023, according to the commitment letter performance goals. and the most recent update Second, a draft guidance on best practices for appeals of decisions regarding a monograph was published in June 2023 [ See AgencyIQ analysis here], targeted for finalization in February 2024 in the commitment letter.

Featuring previous research by previous AgencyIQ researcher Kedest Tadesse.

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

Key Documents and Dates

  • Final Guidance: Providing Over-the-Counter Monograph Submissions in Electronic Format
  • FDA Docket No.: FDA-2022-D-2059

Filed Under: Article

July 26, 2024 by

BY LAURA DIANGELO, MPH, RACHEL COE, MSC

Meeting a biosimilar user fee commitment, the FDA is expanding on its recommendations for biosimilar and interchangeable product applicants asking the FDA for post-approval manufacturing changes. The new draft guidance gives considerations for applicants seeking new dosage form or strengths, and those manufacturing the product in a facility with other versions of the biological product. When finalized, this will supersede recommendations from a 2021 Q&A guidance.

Fill out the form to read the full article.

Filed Under: Article

July 26, 2024 by

FDA’s new guidance on postapproval manufacturing changes for biosimilars focuses on current practice, new dosage forms

Meeting a biosimilar user fee commitment, the FDA is expanding on its recommendations for biosimilar and interchangeable product applicants asking the FDA for post-approval manufacturing changes. The new draft guidance gives considerations for applicants seeking new dosage form or strengths, and those manufacturing the product in a facility with other versions of the biological product. When finalized, this will supersede recommendations from a 2021 Q&A guidance.

BY LAURA DIANGELO, MPH, RACHEL COE, MSC | JUL 23, 2024 9:54 PM CDT

Biosimilarity and interchangeability: A quick recap

  • The Biologics Price Competition and Innovation (BPCI) Act of 2009 intended to increase the number of biologic products on the market by creating two types of approvals for biosimilar products. Both types of approvals (biosimilars and interchangeables) utilize the same 351(k) pathway but have slightly different requirements. Biosimilars are biological products that are highly similar (but not identical) to a previously approved biologic, referred to as the reference product. Because of the complexities involved in the production of biological products from living organisms, minor differences between a reference product and its biosimilars are anticipated. Instead, approval hinges on the ability of sponsors to demonstrate there are no “clinically meaningful differences” between the proposed biosimilar and the reference product.
  • Interchangeable products were established by the BPCI as a subset of biosimilars that are demonstrated to produce the same clinical result as the reference product in any given patient. In practice, the biggest difference between interchangeable products and biosimilars is that pharmacists can substitute interchangeable biosimilars without consulting the prescriber (though different state laws may apply). For sponsors, interchangeable products have historically required additional clinical studies (i.e., switching studies). These studies are intended to ensure that if products are used more than once by patients, “the risk in terms of safety or diminished efficacy of alternating or switching between use of the [proposed interchangeable products] and the reference product is not greater than the risk of using the reference product without such alternation or switch.” In switching studies, a two-arm trial design is commonly employed. During the lead-in period, all patients are assigned to the reference product followed by randomization to either the “switching arm” or “non-switching arm” of the study.
  • Recently, the FDA has openly supported doing away with the interchangeable distinction. In its Fiscal Year 2025 budget proposal, the FDA requested that Congress “eliminate the statutory distinction between the approval standard for biosimilar and interchangeable biosimilar products” and “deem all approved biosimilars to be interchangeable with their respective reference products.” As the agency explained in the request, “The statutory distinction between biosimilars and interchangeable biosimilars has led to confusion and misunderstanding, including among patients and healthcare providers, about the safety and effectiveness of biosimilars and about whether interchangeable biosimilars are safer or more effective than other biosimilars.” [Read AgencyIQ’s analysis of this issue here]
  • In the absence of Congressional action on this topic, the FDA has been working to tackle this issue itself. In a September 2023 guidance, the agency proposed eliminating the interchangeability statement from product labeling entirely, stating that all products, whether biosimilar or interchangeable, should be referred to as “biosimilar” on the product label. As AgencyIQ discussed in December 2023, the FDA moved to implement the aforementioned guidance – which was still in draft form and in its comment period – almost immediately. In June 2024, the FDA published a new draft guidance signaling its elimination of the costly and time-consuming switch study requirement for interchangeability, making it significantly easier for sponsors with biosimilar products to gain interchangeability status and reap the reimbursement benefits.

Postapproval manufacturing changes for biological products

  • During clinical development or following a product’s approval, sponsors and/or manufacturers may identify the need for a change in the product, production process, quality controls, equipment, facilities, responsible personnel, or labeling. Broadly, these topics all fall under the umbrella of chemistry, manufacturing, and control (or CMC) changes. In these instances, the first step sponsors should take (per the FDA’s many guidances on manufacturing changes) is to perform a risk assessment to determine whether the modification has potential to impact the product’s quality. These assessments should be performed prior to any type of manufacturing change regardless of the stage of product development (i.e., including well before and after product approval). For additional information on this process, sponsors should refer to International Council for Harmonisation (ICH) guidelines Q8, Q9 and Q10 on the development of quality risk management processes, and the steps to conducting the risk assessment.
  • For approved products, one of the next steps (following completion of the risk assessment) is to determine how the FDA should be notified of the proposed change and what information should be submitted in support of the change. For major changes—those with substantial potential to have an adverse effect on the product—a Prior Approval Supplement (PAS) should be submitted. In these instances, described in 21 CFR 314.70(b), the FDA must review and approve the change before the altered product can be distributed. Changes that have a moderate potential to have an adverse effect on the identity, strength, purity, or potency of a drug constitute moderate changes. These require notification through either a Changes Being Effected in 30 Days (CBE-30) or Changes Being Effected (CBE) supplement. The CBE and CBE-30 supplements operate more like a notification of a change to the agency, with the CBE-30 providing a 30-day window between the notification and the changes being implemented. The FDA can stop a manufacturer from continuing to distribute the product if it disapproves. Lastly, minor changes with a very small potential to impact product quality can be described in the sponsor’s next Annual Report to the FDA.
  • Risk classification is highly dependent upon the product type and the manufacturing process. Generally, any changes that the FDA would categorize as warranting the completion of a bioavailability (BA) / bioequivalence (BE) study as well as those made to the “synthesis or manufacture of the drug substance,” or to the sterilization methods used during manufacturing would be considered major. Likewise, multiple smaller (or insignificant) changes could raise the overall level of risk to that of a major change. For instance, manufacturing a product at a different site which uses identical equipment to perform an identical process could be a minor change. However, a change in manufacturing site which then entails the use of different equipment could be considered a moderate change, and lastly, if the different equipment entails changes in the overall manufacturing process, it might rise to the level of a major change.
  • If a change could impact product quality, a comparability study will likely be required. The ICH Q5E Guideline defines “comparable” as the conclusion that products have “highly similar quality attributes before and after manufacturing process changes and that no adverse impact on the safety or efficacy, including immunogenicity, of the drug product occurred.” To determine the effect of a manufacturing change, samples from both batches (i.e., pre- and post-manufacturing changes) should be analyzed using product quality attributes and process parameters along with predefined acceptance criteria to assess comparability.
  • Any CMC changes (e.g., in equipment) for products that are more complex or aren’t as well-characterized, such as antibody drug conjugates or recombinant DNA products, will be considered higher risk than they would if the change was made to the production of standard, small molecule products. There is also a higher burden when it comes to switching the source or process used to manufacture active pharmaceutical ingredients or drugs that are required to be sterilized or labeled as sterile, including products sterilized via filtration or other means, those aseptically processed, and terminally sterilized drug products)
  • One thing that sponsors can do to preemptively decrease the burden for reporting future manufacturing changes is to develop and submit a comparability protocol (CP) during clinical development or at the time of application submission. A CP is a well-defined, detailed, written plan for assessing the effect of specific CMC changes in the identity, strength, quality, purity, and potency of a specific drug product. These protocols are used to describe the change and present all the tests and studies to be performed along with the acceptance criteria to ensure the continued safety and efficacy of the product. In some cases, sponsors with approved products may also have the option to leverage a pre-specified CP in order to reduce the reporting requirements for manufacturing changes. The idea here is that sponsors with pre-specified CPs have already worked with the FDA to develop an agreed-upon plan for how product comparability should be demonstrated in case different types of CMC changes are made in the future. As a result of securing FDA’s blessing on the plan ahead of time, sponsors with these plans in place may be able to notify the agency of some manufacturing changes using a less burdensome approach (e.g., in the annual report instead of via a post-approval supplement).
  • FDA’s guidance on biosimilar product development includes one Q&A on postapproval manufacturing changes. Last finalized in 2021, the guidance “ Questions and Answers on Biosimilar Development,” includes one question and answer on postapproval manufacturing changes – question Q.I.20. The content in that guidance is limited, but generally points sponsors to ICH guidelines on the subject, saying that they should “follow the principles outlined in the ICH guidance for industry Q5E Comparability of Biotechnological/Biological Products Subject to Changes in their Manufacturing Process.” The Q&A then summarizes some of the key aspects of that document, including comparability assessment contents and the use of reference materials. That final guidance, however, does note that “FDA intends to provide specific recommendations for postapproval manufacturing changes to interchangeable biological products in future guidance.”
  • The user fee commitments called for guidance on this subject. As part of the current Biosimilar User Fee program (BsUFA III), the FDA committed to the publication of “a draft guidance on the nature and type of information, for different reporting categories, a sponsor should provide to support post-approval manufacturing changes to approved biosimilar and interchangeable biosimilar biological products” by September 30, 2024 – the end of federal fiscal year 2024.

The FDA has now issued a new draft guidance on the subject of postapproval changes for biosimilar and interchangeable biosimilars

  • First up, why this guidance now? According to the agency, the guidance – which is in Q&A style – “provides answers to commonly asked questions from applicants and other interested parties… regarding postapproval manufacturing changes.” In effect, the agency appears to have been fielding questions about the topic, and is looking to give answers in one place. The guidance “is intended to inform prospective and current applicants of the nature and type of information that applicants should provide in support of manufacturing changes to licensed biosimilars and licensed interchangeable biosimilars in different reporting categories.”
  • The new draft guidance is fairly short, clocking in at 13 pages total, about seven of which are the Q&A – with four individual questions. The policy recommendations are broken into two parts: recommendations for reporting categories and recommendations for product quality data. The bulk of the guidance is in the latter category; only Q1 addresses “Recommendations for Reporting Categories,” while the rest of the Q&A (i.e., Q2-Q4) is focused on recommendations for product quality data. The guidance goes beyond the scope of the information offered in Q.I.20, including information on introducing a biosimilar/interchangeable biosimilar to a manufacturing site or area where other products are being manufactured (Q3) and what to do when seeking supplement approval for a new dosage form or strength (Q4).
  • Q1: What information does FDA recommend, by reporting category, to support manufacturing changes for these products? Again, the three reporting categories for post-approval changes are a PAS, CBE-30/CBE-0, or Annual Report. The new draft guidance goes over the basics of when each report type is appropriate, and asks applicants to “clearly identify the reporting category under which the change is being reported.” For a PAS/CBE-30/CBE-0 (i.e., a supplement submission for a manufacturing change), then “the applicant should specify the supplement as a CMC supplement,” and cover letters should include a comprehensive list of the changes. Manufacturing changes that impact labeling should have those labeling changes in the CMC supplement.
  • The rest of the guidance, Qs 2-4, is on recommendations for product quality data. As in the FDA’s 2021 guidance on biosimilar development more broadly (see above, Q.I.20), the agency continues to recommend applicants follow the principles in ICH’s Q5E Comparability of Biotechnological/Biological Products Subject to Changes in their Manufacturing Process guidance for industry. The new draft guidance goes a little bit deeper on the subject, referring to Q5E’s call for applicants to conduct a comparability exercise (see more on the subject from that guidance here) and references ICH’s definition of quality attributes (data and information necessary to establish comparability that is commensurate with the type of manufacturing change). At a high level, the new draft guidance summarizes many of the key points from Q5E on the goals of comparability exercises (including side-by-side analytical comparisons of “a sufficient number of lots” and stability data), the need for comparative stability studies and how they are “especially important when the proposed manufacturing changes can alter protein structure or purity and impurity profiles.” The new draft guidance also points applicants to 2019 draft guidance on the Development of Therapeutic Protein Biosimilars (comparative analytical assessments) and the ICH Q6B guidance on test procedures and acceptance criteria.
  • The draft guidance explains that “Data and information submitted in support of manufacturing changes should demonstrate that quality attributes remain comparable among prechange and postchange products and should demonstrate that consistency in the quality, safety, and efficacy is predictable.” This may mean the product will need to be evaluated at “multiple stages of manufacture,” as it should be “evaluated at the process step most appropriate to detect a change in the quality attributes.” Comparisons of analytical data “from the postchange material to the historical analytical data (i.e., prechange material) may be sufficient to support a manufacturing change if the quality attributes of the prechange and the postchange material are comparable,” it notes, and goes on to describe the comparative analytical assessment (CAA, as described in the 2019 draft guidance cited above) expectations. If applicants are using a subset of “all available historical data” they should provide a justification, and if there has been a change in analytical assay, then “adequate assay bridging data on assay performance should be provided.”
  • Back to the questions: Q2, reference materials. This guidance uses the following definition for in-house reference material: “appropriately characterized material prepared in-house by the manufacturer from a representative lot(s) for the purpose of biological assay and physiochemical testing of subsequent lots” (see ICH Q6B). As recommended in the 2021 guidance, the new draft guidance again recommends the use of “a well-qualified, in-house reference material in the comparability exercise” (note: The 2021 guidance uses “standard” instead of “material”). The new draft guidance, however, goes on to further describe the importance of this in-house reference material, which is “an important calibration point for the evaluation(s) conducted in a comparability exercise.” If the applicant asserts, and the FDA agrees with, an “informed pr diction” that there is no adverse impact expected postchange, FDA may not require additional information beyond the in-house reference material comparability exercise – “However, demonstration of comparability to support manufacturing change(s) should provide adequate assurance that a postchange product remains biosimilar to or interchangeable with a reference product.” If there are differences in quality attributes between prechange and postchange material, “comparison to the data from the reference product submitted to support licensure should be considered to help assess the potential impact and acceptability of these differences.”
  • Q3: How should proposals to introduce a biosimilar/interchangeable “into a multiproduct manufacturing area or a multiproduct contract manufacturing facility be reported?” Notably, this question is not addressed at all in the 2021 guidance on biosimilar product development. However, the agency does open its answer to this question by pointing to other guidance documents that can provide more information: Changes to an Approved Application for Specified Biotechnology and Specified Synthetic Biological Products (July 1997), CMC Postapproval Manufacturing Changes for Specified Biological Products To Be Documented in Annual Reports (December 2021), and Chemistry, Manufacturing, and Controls Changes to an Approved Application: Certain Biological Products (June 2021).
  • Q3 continued: The agency goes into the types of risks that might be associated with introducing a licensed biosimilar or interchangeable biosimilar into a multiproduct manufacturing area or multiproduct contract manufacturing facility, including “cross-contamination or product mix-ups” in sites or areas where the reference product or another biosimilar/interchangeable is manufactured. While the agency generally recommends that identity testing can detect and control risks, “a respective single identity test for each product might not always be able to distinguish between the different products” when they are all the same reference biological product and its biosimilars/interchangeables. When this is the case, “in addition to identity tests for each product, current good manufacturing practice requirements, including applicable manufacturing and procedural controls (e.g., separate manufacturing areas, control of personnel, process, and material flow, and control of materials, as applicable) to prevent cross-contamination or mix-ups, must be followed.”
  • Q4: What is the “nature and type of CMC information” recommended to support supplement approval for a new dosage form or strength? When submitting a supplement for a new dosage form or strength for a biosimilar or interchangeable biosimilar product, “applicants must include information demonstrating, among other things, that the dosage form and the strength of the proposed biosimilar or the proposed interchangeable biosimilar product “are the same as those of the reference product.” This is considered a major change – and thus, typically requires a PAS.
  • Supplements of this type “generally should include the following CMC-related information: (1) adequate comparability data between a licensed biosimilar or interchangeable biosimilar (i.e., prechange product) and the proposed biosimilar or interchangeable biosimilar with the new dosage form or strength (i.e., postchange product), (2) comparative analytical assessment (CAA) data, and (3) manufacturing data (e.g., process validation) to support the proposed postchange product,” according to the new draft guidance. However, the “extent” of the data (comparability, CAA) will depend on the risk assessment of differences – and, in “some cases,” additional data like pharmacokinetic studies might be needed. While the agency notes that previously-submitted CAA studies might provide a model for the design of these studies for a new dosage form or strength, “applicants should assess whether additional CAA studies with the new dosage form or the new strength are appropriate.”
  • The draft guidance goes on to give two examples: In one, an applicant’s supplement proposes a new strength with the same administration, dosage form, excipients, and for the same patient population and indication as a biosimilar or interchangeable biosimilar already licensed. In this case, the previously submitted CAA and a “risk-based comparability exercise… may be reasonable.” In the second, the supplement seeking a new dosage form/new strength is also intended for a different patient population/indication than an already-licensed biosimilar or interchangeable biosimilar. In this case, the applicant can use the previously-submitted CAA but “should also include a risk-based targeted CAA between the proposed product with the new dosage form and strength and its reference product and include a risk-based comparability exercise.” In general, the agency recommends that sponsors come to the agency to discuss their approaches here.

Analysis and What’s Next

  • The issuance of the guidance does fulfill the BsUFA III commitment, referenced above, to issue a draft guidance on this subject by the end of federal fiscal year 2024. Per the BsUFA commitment letter, the agency should “work towards the goal of publishing final guidance within 18 months” after the comment period, which would anticipate a final guidance around March 2026 – unless, of course, the agency chooses to issue a revised draft guidance, depending on feedback on this round. Comments are due September 23, 2024.
  • If and when this new draft guidance is finalized, Q.I.20 from the final guidance cited above will be removed: “FDA intends to withdraw Q&A I.20 from that guidance when this guidance becomes final.” In effect, this new draft guidance will carve out the information offered in Q.I.20 (above) into its own guidance.
  • The new draft guidance includes information on both biosimilar products and interchangeable biosimilar products, noting in a statement on the new draft guidance that “The agency has since [the 2021 guidance] determined the principles that apply to postapproval manufacturing changes to licensed biosimilar products are also relevant to licensed interchangeable biosimilar products and today’s guidance provides recommendations for both.” This is an expansion of scope, compared to the 2021 Q&A, which stated at the time that the documents would be separate.
  • The guidance also offers additional information beyond what was available in that 2021 guidance, but many of the key themes are carried through; this includes the FDA’s continuing recommendation that applicants refer back to the ICH Q5E document on comparability of biotechnological/biological products subject to changes in their manufacturing process, and reliance on ICH definitions of key terms like quality attributes and the conduct of comparability exercises. Still, the new draft guidance does offer additional information that was not included in that 2021 Q&A guidance, such as expectations for considering risk in multiproduct manufacturing areas/multiproduct contract manufacturing facilities. Whether the agency’s guidance goes deep enough into those topics to satisfy industry’s questions, however, remains to be seen.

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

Key Documents and Dates

  • Postapproval Manufacturing Changes to Biosimilar and Interchangeable Biosimilar Products Questions and Answers
  • Docket FDA-2024-D-2581
  • Comments due September 23, 2024
 

Filed Under: Article

July 22, 2024 by

BY SCOTT STEPHENS, MPA

At the REACH and CLP regulators’ meeting in July, the European Commission revealed specifics about its formal schedule to eliminate animal testing used for chemical safety assessment. The ban was promised last summer in response to a European Citizens’ Initiative urging faster action to rid the EU of all animal testing; now, the EU executive has provided a clearer picture of the timeline and path toward fulfilling this ambitious objective.

Fill out the form to read the full article.

Filed Under: Article

July 22, 2024 by

Commission unveils details on plan for phasing out animal testing for chemicals safety

At the REACH and CLP regulators’ meeting in July, the European Commission revealed specifics about its formal schedule to eliminate animal testing used for chemical safety assessment. The ban was promised last summer in response to a European Citizens’ Initiative urging faster action to rid the EU of all animal testing; now, the EU executive has provided a clearer picture of the timeline and path toward fulfilling this ambitious objective.

BY SCOTT STEPHENS, MPA | JUL 18, 2024 2:48 PM CDT

Animal testing rules and non-animal test method development in the EU

  • Avoiding animal testing is one of the main objectives of the EU’s cornerstone Regulation (EC) 1907/2006 on the registration, evaluation, authorization and restriction of chemicals (REACH), which includes registration testing requirements for registrants assessing the intrinsic properties of their substances for safety. While at times animal testing may be unavoidable to ensure compliance with these requirements, the cross-sectoral legislation emphasizes that such tests should be conducted “ only as a last resort,” when all other possible non-animal testing avenues have been exhausted. Accordingly, REACH requires registrants to avoid the use of vertebrate animals for generation of toxicity data whenever possible, considering, instead, alternatives like read-across methods, (quantitative) structure-activity relationship ((Q)SAR) models or in vitro methods. Where REACH stipulates certain methods to be used for animal testing, it mandates that these methods continually be subject to review and further development “with a view to reducing testing on vertebrate animals and the number of animals involved.” The rules also require testing proposals to be submittedwhen animal tests are needed, and oblige stakeholder sharing of information on already registered chemicals and, whenever possible, reducing unnecessary duplication of testing.
  • A total ban on animal testing for cosmetics has been in place in the EU since 2013 under Regulation (EC) No 1223/2009 on cosmetic products (Cosmetics Regulation). Article 18 prohibits the marketing of cosmetic products that involved animal testing and extends to products using ingredients or combinations of ingredients that were subject to animal testing. Animal testing conducted within the EU on cosmetic products, ingredients and combinations of ingredients is also prohibited.
  • The EU has provided significant funding for research in non-animal testing and alternative measures. According to the Commission’s July 2023 communication, over the past two decades, the EU executive has spent over 1 billion euros across more than 300 research projects investigating alternatives to animal testing. The EU Horizon and Horizon 2020 Initiatives provided generous funding to work on reducing animal use in chemicals assessment (e.g., the PARC partnership with a budget of 400 million euros).

European Citizens’ Initiative (ECI) “Save cruelty-free cosmetics”

  • The Commission last year promised to set out a blueprint for systematically addressing animal testing after a European Citizens’ Initiative (ECI) advocating for the EU executive to “commit to a Europe without animal testing” obtained more than one million signatures within one year, a threshold triggering the Commission’s mandatory consideration of the initiative and a response, pursuant to Regulation (EU) 2019/788.
  • The initiative called on the Commission to undertake three main actions, introducing provisions that underpin and expand the article 18 ban on animal testing under the Cosmetics Regulation; calling for concrete steps for the development of human-relevant alternatives to animal testing, including actual timelines, funding opportunities, and cross-sectoral support, for the EU chemicals management regulatory framework; and urging the Commission “to prioritise a transition to non-animal approaches as an integral part of all EU research, innovation, and education initiatives,” while, at the same time, appreciating that the EU law protecting animals used for scientific purposes (i.e., Directive 2010/63/EU) “does not in and of itself represent a roadmap towards full replacement.” [See AgencyIQ’s article here for more details on ECIs in general and this initiative in particular.]

The Commission’s response to the initiative

  • On July 25, 2023, the EU executive published a communication and accompanying press release, stating its intention to “accelerate phasing out of animal testing in response to a European Citizens’ Initiative.” The Commission said it aimed to accomplish this with both legislative and non-legislative changes, envisioning actions undertaken as part of key pieces of chemical regulation such as REACH, the Biocidal Product Regulation (528/2012/EU), and the Plant Protection Products Regulation (1107/2009/EC).
  • The Commission further stated that it would “immediately” begin work on a roadmap to further reduce animal testing and transition to an animal-free regulatory framework. The roadmap, it stated in the press release, would provide milestones and specific actions to further reduce animal testing, on the path to an eventual complete phase-out of animal testing. The “core of the roadmap will be to analyse and to describe necessary steps to replace animal testing in pieces of legislation that currently require animal testing for chemical safety assessment,” according to the Commission communication. The EU executive had originally expected to finalize the roadmap “in the first quarter of the term of the next Commission,” or the first quarter of 2025.

The Commission followed up with an update on the roadmap’s development

  • The EU executive presented on its roadmap at the July 1-2 meeting of REACH and CLP regulatory experts (CARACAL), offering concrete details about its plans for developing the roadmap. Below, AgencyIQ summarizes the accompanying slides, highlighting the most important aspects of this presentation.

Key commitments

  • The roadmap will likely be delivered in the form of a Commission communication. According to the presentation slides, it will represent “a navigation plan” or “schedule” that “is listing actions and milestones necessary to reach the goal of phasing out animal testing.” The roadmap will be “applicable to all relevant pieces of EU chemical legislation that might lead to animal testing for chemical safety assessments.”
  • The core elements of the “navigation plan” will encompass (1) an “analysis” describing “the steps to replace (reduce/refine) animal testing”; (2) an outline of “the path to expand and accelerate the development, validation and implementation of non-animal methods”; and (3) a description of “the path for uptake across legislations.” In this context, the Commission emphasized that, “for many endpoints,” unknowns endure about which non-animal testing methods, or combinations of them, can replace animal methods, indicating a consensus that “we need to think of designing a new regulatory system based on non-animal testing.”
  • Other elements of the roadmap will include a determination about the “need” or “feasibility of an advisory scientific committee on non-animal methods”; an outreach component, bolstering links to multilateral organizations, with the aim of “strengthening the acceptance of non-animal testing” in the Globally Harmonized System of Classification and Labeling of Chemicals (GHS); considering avenues for expediting “the acceptance and validation of new non-animal methods, while taking into account the importance of mutual acceptance of data across different jurisdictions.”

Development of the roadmap

  • Which parts of the Commission will develop the roadmap is laid out visually in the presentation on slide 10. The EU executive foresees assigning main responsibility for its drafting to an “Interservice Group” with representation from the directorates general (DGs) for the environment (ENV), internal market (GROW), research and innovation (RTD), health (SANTE), and the Commission Secretariat-General (SG), as well as from ECHA, the European Food Safety Authority (EFSA), the European Medicines Agency (EMA), and the Joint Research Centre (JRC). Individual tasks will be carried out by three separate working groups (WGs) for human health, environmental safety assessment, and change management. Several stakeholders outside the Commission, including EPAA, ASPIS, PARC, member state competent authorities, and NGOs will also be key sources of feedback, input, and ideas leading to the draft roadmap. Planned public consultations will also contribute to its development.

Status of roadmap development today

  • So far, the Interservice Group has met six times, discussing terminology, structure, and overarching organization of the roadmap development. Likewise, the members have decided that 15 areas or pieces of legislation will comprise the regulatory scope included in roadmap development. The presentation, however, does not specify which areas. Finally, one of the group’s meetings was dedicated to the topic of how to accelerate non-animal method validation.
  • The initial activities of the WGs were also summarized. Among other activities, the WGs on human health and environmental safety assessment initiated analyses on currently used animal methods and available non-animal methods; discussed “short-term solutions and methods requiring development”; and examined the level of protection provided under the EU’s current testing scheme versus that desired in the new framework.

Acceleration of validation

  • The presentation underscores the steps that the Commission has taken thus far to ensure that the pace of validating non-animal testing methods is hastened. Actions taken toward this end include a December 2023 workshop session (Day 2, Session 4), for which, the slides note, a workshop report “will be published soon.” The Commission’s Interservice Group dedicated meeting, which took place in May of this year, discussed issues related to validation, including funding, mutual acceptance of data, and “learnings from the EMA qualification process to enhance the acceptance of new methods.”

Analysis and next steps

  • The roadmap is expected to be finalized sometime in late 2025, or early 2026, according to the presentation. An “implementation phase that implements the planned actions” will follow its release.
  • Several stakeholder events are in the pipeline in support of the roadmap’s development, including a call for evidence, which the presentation indicates will be announced shortly; a targeted consultation in the fourth quarter of 2024, with focus on validation acceleration; and a follow-up workshop organized by the Commission, planned for October 25.
  • The presentation makes clear that the Commission’s exercise of creating a “navigation plan” toward the ultimate elimination of animal testing in favor of yet-to-be-developed non-animal methods is no simple task. Expected to take at least a year and a half to complete from today, just the roadmap is already behind schedule by almost a year, compared to the Commission’s original expectations laid out in the 2023 communication. This fact alone hints at how long the journey could potentially be to actually realize the Commission’s commitments made to phase out animal testing.

Featuring previous analysis by Kirsten Messmer and Rayan Bhargava

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

Key Documents and Dates

  • Commission Roadmap towards phasing out animal testing for chemical safety assessment; CARACAL-52; July 1, 2024
  • Factsheet summarizing Commission’s response to the European citizens’ initiative ‘Save cruelty-free cosmetics – Commit to a Europe without animal testing’; July 25, 2023
  • Q&As: Commission’s response to Citizens’ Initiative “Save Cruelty-Free Cosmetics – Commit to a Europe without Animal Testing”
  • European Citizens’ Initiative – “ Save Cruelty-Free Cosmetics – Commit to a Europe without Animal testing”; first registered June 30, 2021, initiative answered July 25, 2023

Filed Under: Article

  • « Go to Previous Page
  • Go to page 1
  • Interim pages omitted …
  • Go to page 5
  • Go to page 6
  • Go to page 7
  • Go to page 8
  • Go to page 9
  • 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