Premarket Submissions: Highlights from FDA issued guidance

The FDA takes steps to promote faster access to new medical technologies that are safe and effective: Streamlining premarket procedures, modernizing the 510(k) program, expediting programs for devices that enhance safety and/or resolve critical unanswered medical needs. The recently issued guides include new pathways, expansion of existing programs, and supporting tools such as checklists and assessment worksheets.

Topics in this Newsletter

510(k) Programs

1. Special 510(k) (UPDATED)

2. Abbreviated 510(k) (EDITED CONTENT)

3. Safety & Performance Based Pathway (NEW)

Additional Premarket Programs

4. Humanitarian Device Exempt (HDE) Program (UPDATED)

5. Safer Technology Program (STeP) (NEW)

More FDA Actions

6. Electronic submissions;

7. Benefit-risk determinations;

8. Acceptance review policy (RTA);

9. Accreditation of testing labs (ASCA)

Adapting the repeatedly criticized 510(k) program to advances in safety and technology; providing clearer requirements and more timely review & response, and lowering workload for FDA and costs for the industry.

Let’s take a short tour of some of the ideas and changes that these documents bring about.

What’s new with the 510(k) program?

The 510(k) program accounts for the vast majority of devices that enter the market each year. It is based on the concept of “substantial equivalence”. The FDA now issues final guidance documents for the Special 510(k) Program, the Abbreviated 510(k) Program, and the Safety and Performance Based Pathway, as well as a Format for Traditional and Abbreviated 510(k)s.

1. The special 510(k) program

This program is intended specifically for changes of own existing device, and only if the methods to evaluate changes are considered well established. FDA reviews special 510(k) applications within 30 days of submission. This final guidance expands the scope of the program.

The significant changes in the program:

• Now, under certain circumstances, changes to the intended use may be made.

• Now, under certain circumstances, changes that alter the fundamental scientific technology may be made.

• If performance data is necessary, it is required that methods are well established and that all data supporting substantial equivalence can be reviewed in a summary or risk analysis format.

NOTE: This pathway therefore may NOT be appropriate,

for example, when a novel sterilization method is used; or methods rely on animal/clinical data; or the changes are complex – with many scientific disciplines involved, change from single-use to re-usable, etc. – so that determining the substantial equivalence depends on the FDA’s interpretation of the underlying data.

2. The abbreviated 510(k) program

An alternative approach to the traditional 510(k): Instead of performing a “head-to-head” comparison with a predicate device in order to demonstrate the substantial equivalence, you declare conformity to voluntary consensus standards / special controls / FDA guidance.

This program is NOT new, but merely received a guidance document separate from the special 510(k) program. It includes a short outline of the content required.

NOTE: This pathway is not necessarily “abbreviated” or shorter.

Also, the review time is 90 days, as for the traditional 510(k). However, under some circumstances, when it is difficult to collect data regarding a predicate, it might be more efficient to submit summary reports describing how to reference standards were used or how the device complies with the special controls. Addressing the FDA early to find out which pathway is appropriate, would be a good idea.

3. Safety and Performance Based Pathway

An optional submission pathway, an extension of the abbreviated program. It is intended for well-understood devices, listed by device type, for which the manufacturer would have to meet objective criteria set by the FDA: criteria that would be consistent with safety and performance characteristics of modern predicates. Along with the guide describing the general pathway, five additional draft guidance documents were issued, describing the required performance criteria for specific devices:

Foley catheters

Recording cutaneous electrodes 03Ø06

Orthopedic spinal plating systems

Non-spinal screws & washers

Magnetic resonance (MR) coils

The basics you should know about this pathway:

• It’s not actually different from the abbreviated 510(k).

• The requirements may be more specific, thus clearer to the

manufacturer (that’s a bonus).

• May be suitable when attaining information regarding an

the actual predicate is impossible/impractical.

fda approval

Additional programs and pathways at focus :

4. The Humanitarian Device Exemption (HDE) Program

HDE program receives updated final guidance, accompanied by the newly revised guidance for Humanitarian Use Device (HUD) Designations. Both follow changes resulting from the “Cures Act”.

The purpose of this program is to encourage the development of devices for the diagnosis and treatment of rare medical conditions.

The main updates:

• Expansion of the HUD designation: The designation is given when the relevant population is up to 8000 individuals per year in the USA, instead of the previous threshold of less than 4000.

• A device may still receive HUD designation in the pediatric population even when the total condition population exceeds 8000 patients if the pediatric population affected does not exceed 8000 patients.

• For diagnostic devices, the threshold applies to not more than 8000 who would be subjected to the diagnosis with the device, including positive and negative results.

• As was before, in order to make use of an approved HUD in patients, oversight of an Institutional Review Board (IRB) in the medical facility is required, and review and approval for the specific individual use are also required. This was previously also required to be performed by the IRB. Now more flexibility is introduced, as an appropriate local committee (ALC), that may be a standing committee with the appropriate expertise, is allowed to review and approve the specific HDE use, and not only the IRB, which may be considered less accessible. IRB facility overview is still required.

• A filing checklist, and tools for the probable benefit-risk assessment are included in the appendices.

5. Safer Technology Program for Medical Devices (STeP)

More action in promoting patient safety:

The draft guidance for this new voluntary program is issued

– complementary to the “Breakthrough Device Program” (BDP), for medical devices that are expected to significantly improve the safety of currently available treatments.

Devices and device-led combination products, that are subject to review under 510(k), De Novo, or Premarket Approval (PMA) submissions, and that are not eligible for the BDP, may enjoy the expedited STeP approach:

• First apply through a Q-submission (pre-sub) to be included in the program;

• Once accepted, go through a prioritized and expedited review of regulatory submissions, with senior FDA management engagement, and assistance with the plan of device and data development.

To apply, the sponsor should explain the innovative approach and how the benefit-risk profile would be improved:

• Does the device reduce the occurrence of a known serious adverse event/device failure mode / user-related hazard or error?

• Does the device improve the safety of another device or intervention?

NOTE: The Breakthrough program which is intended for life-threatening

/ irreversibly debilitating conditions, is mandated by law and would still be prioritized over STeP. This is expressed in the draft with the repeating phrase: “as resources permit”.

Thus, only time will tell if STeP achieves its purpose to open an actual additional expedited pathway.

More FDA actions to streamline, simplify, expedite, clarify…

6. The FDA goes paperless – electronic submissions are on the way

Following the requirements of the FD&C Act, draft guidance regarding electronic submissions was issued. But don’t get rid of your printers just yet – it will take a little while.

The guidance tells us that:

• Application types that would be required to be submitted solely in an electronic format, include 510(k), PMA, De

Novo, Investigational Device Exemption (IDE), HDE, and more.

• Individual, per-type guidance documents will be developed, specifying required formats and implementation timetable.

• IDE compassionate use requests and adverse event reports will be exempt from this requirement.

ATTENTION: In contrast to most guidance documents, which contain nonbinding provisions, this guidance and the following

individual ones – once finalized, will contain both nonbinding AND binding provisions under the statutory authorization of Congress: I.e., sponsors will be required by law to comply with the specified formats & schedules.

7. Clarifying benefit-risk considerations for higher-risk submissions

Two final guides were issued (1, 2), both actually discuss the same subject: determining uncertainty and benefit-risk in PMA, HDE, De Novo, and BDP submissions.

Some key concepts:

• Greater uncertainty may be accepted when there’s a true clinical need: serious/unanswered/small patient population / high anticipated benefit for quick patient access.

• Higher uncertainty  greater need for premarket data; Lower uncertainty data shift to post-market may be allowed.

• Post-market data is always required. It is critical to show that it can be collected in a timely manner, especially as risk/ uncertainty increases.

• A new worksheet is provided as an appendix; Recommended as a basis for the benefit-risk assessment process.

• FDA wants patient perspectives – are the patients really willing to accept the risk and the uncertainty?

8. Saving time for FDA reviewers and for sponsors

– acceptance review policy

Final updated “Refuse to Accept” (RTA) guidance was issued for 510(k) and for De Novo submissions. These outline the preliminary procedure the FDA performs to assure administrative completeness of submissions (including timetable), which takes place before the substantive review that assesses the quality of the information.

Some highlights:

• A checklist of required criteria is provided for each submission type (in the appendices).

• FDA will, within 15 days of submission, electronically notify the submitter if: (a) submission was accepted and under substantive review;

(b) the submission was refused PLUS indication of the missing checklist items, or;

(c) FDA did not complete acceptance review on time, thus transferred

submission to substantive review.

• This notification identifies the lead FDA reviewer assigned for the submission.

• The De Novo guidance includes a recommended content checklist as well, relevant also for the substantive review.

• RECOMMENDED: Follow the checklist, complete it, and include it as part of the application. Do NOT leave sections out (if not applicable – a state so & justify). Letters from subject matter experts are helpful.

9. Accelerating review processes by accrediting testing labs to FDA-recognized standards

Draft guidance for The Accreditation Scheme for Conformity Assessment (ASCA) Pilot Program was issued.

OBJECTIVE: Streamline reviews – once a manufacturer presents results from an accredited body, FDA can accept them without a thorough review. The pilot begins with standards for biological evaluation and for electric safety.

Gsap experts will be happy to assist you in choosing the regulatory track!

suits your product and company, and to support you in the submission process.

This Newsletter Prepared by:

Orly Chillag – Talmor, Ph.D.

Quality & Regulatory Project Coordinator


For more information about our services visit:

ISO 14971-Risk Management with ISO 14971:2019

Risk Management is the systematic application of policies, procedures, and practice in various aspects and processes of a Medical Device in order to achieve a safe and effective product. 

Risk Management allows the manufacturer to understand the controls and design features needed in their Medical Device. 
This process ends only at the end of a Medical Device’s life cycle, so even after the Medical Device has been placed on the market, continuous monitoring and identification of new hazards are required.

For Medical Device companies, this process is often complex. This rises not only due to the device complexity of design, materials, production processes, software used, and device function – but also due to the various stakeholders (Clinical aspects, marketing considerations, manufacturing and available technology, suppliers and sub-contractors), each contributes to the way risk is perceived AND CONSIDERATIONS.

ISO 14971 principles are implemented globally in the Medical Device Industry and conforming to the standard requirements is used to show compliance to regulation all over the world.

Towards the transition to MDR (EU Medical Device Regulation) and IVDR (EU In-Vitro Diagnostic Regulation) and the recognition of ISO 14971:2019 by FDA, and other regulatory bodies, Medical Device companies must assess risk management processes and existing documentation.

Gsap is a leading consultancy firm with accumulated decades of experience in the industry of medical devices and pharmaceutical companies. We work with our customers, corporates to start-up companies in R&D, production, and post-market stages. We deliver the shortest pathway, and consider our clients as partners to success, with tailored service and support.

ISO 14971:2019 meets Regulatory Requirements

Risk management according to ISO 14971:2019 (NEW VERSION) is required according to the new MDR (EU 2017/745) which enters into enforcement in May 2021 and IVDR (EU 2017/746) which enters into enforcement in May 2022.

The transition period from ISO 14971:2012 to ISO 14971:2019 for FDA is December 25, 2022.  After this transition period, only declarations of conformity to ISO 14971:2019 will be accepted by FDA.

ISO 14971 Relations with other standards


ISO 14971 has relationships with various standards: ISO standards such as ISO 9000 (Quality Management Systems), IEC 62366 (Usability), ISO 13485 (Medical Devices – Quality Management Systems), IEC 60601-1 (electrical medical equipment).

Whereas ISO 60601 addresses single fault, ISO 14971:2019 also addresses a combination of fault modes and hazardous situations as a result of a sequence or combination of independent events. Another example of relation with other standards is the Usability Engineering process (IEC 62366). This process is used for the identification of reasonably foreseeable misuse (in addition to use errors and use associated risks). The outputs of the usability engineering process must be fed back into the risk management process and help complete the identification of hazards. This includes System Security (Cyber Security) and breaches of data. IEC 62304 (Medical Device Software – Software Life Cycle Processes) refers to ISO 14971 for the risk management process of software. ISO 14971 adds an identification of hazards that are related to software that needs to be considered in the process, such as confidentiality, the integrity of data, and availability of data.

ISO 10993-1:2018 Biological evaluation of medical devices requires that the evaluation of overall residual risks associated with the medical device acceptability will be part of the risk management file according to ISO 14971.

It is crucial that traceability will be kept and linked between the various related processes.

An example of this relationship is demonstrated in figure 1.

ISO 14971
 Figure 1 – Example of relations of ISO 14971 with other standards

ISO 14971:2019 Vs. ISO 14971:2007

ISO 14971 guidance annexes were removed from the standard and are found in ISO TR 24971:2020. This new version of the ISO TR 24971 document contains all the normative references and is used to guide the proper implementation of the risk management process.

Revised Terms and Definitions:

New terms that are defined in the standard:

• Benefit: The types of benefits to be considered are discussed: the positive impact of clinical outcome, quality of life, diagnosis, public health. The benefit-risk analysis is aligned to meet MDR and IVDR requirements (the MDR mentions benefit over 60 times vs. 2 times in MDD).

• Reasonably foreseeable misuse: There is an understanding that medical devices can be used for a different intention than the device intended use and that use of the medical device by different populations may result in different outcomes – such as use by medical professionals versus use by laypersons.

• State of the art: This does not necessarily imply the latest most advanced technology. Under the ISO 14971:2019 standard principles, when considering the latest most advanced technology, compared with a more established and widely used technology – it is possible that the benefit-risk perspective of the options will be equivalent. Manufacturers must consider state of the art (clause 10.2), and continually monitor and gather information (generally acknowledged state of the art), and understand if the state of the art changes. This concept is considered in the MDR.

ISO 14971:2019 Scope: The scope of the standard has been clarified to avoid misinterpretation and so specifically mentioning software as a medical device (A.2.1), the Risk Management Process can also be applied to data and security (cyber security), and more detail is given to hazards related to these areas and Radiation, Usability and Biocompatibility. 
The standard is not limited to Medical Device Manufacturers, but to products that are not necessarily recognized as Medical Device under Regulation and to Suppliers, Contractors, and Service Providers that are involved in the Medical Device life-cycle (compliance with some or all ISO 14971:2019 requirements).
• Clause 4.1: The diagram representing the risk management process revised to reflect how the role of the risk management plan in the process.
• Clause 4.4: Addition of risk management to include a method for the evaluation of the overall residual risk and requirement to plan criteria of acceptance to this activity (for Medical Device) see clause 8.
• Clause 4.5: traceability
• Clause 5.4: requires the use of multiple risk analysis tools in order to meet the requirement of identifying known and foreseeable hazards (in both normal and fault conditions) and serve as input to the design process (Annex E ISO TR 24971). This clarifies the need for more than single hazard identification tools: Intended Use; Safety-Related Characteristics; Research/Clinical Trials; Preliminary Hazard-Analysis; Fault Tree Analysis; Usability Engineering Analysis/Human Factors Engineering. During design output, the use of single fault analysis is appropriate in risk management on the design, such as FMEA, SW, and biocompatibility analysis and production. Further identification of hazards is done using data from Risk Control Implementation Verification (Design Verification); Risk Control Verification of Effectiveness (Design Validation phase); Complaints and CAPA process (Post Production) which is also part of the suitability evaluation of risk control measures.

• Clause 8 Disclosure of significant residual risks (Annex A.2.8 ISO 14971 and Annex D ISO TR 24971) Discretion for the analysis of risk/benefit has changed to the requirement to perform a benefit/risk analysis.

• Clause 9 Risk Management Review needs to identify who is going to do the review and when to perform it. Note that the risk management review is part of the risk management fie. The review process can be part of product realization (design reviews).

• Clause 10 Production and Post Production Activities: Expanded and is aligned with clause 8 (Measurement analysis and improvement) in ISO 13485:2016 (and GHTF SG3/N18:2010 QMS MD Guidance on corrective action and preventive action and related QMS processes). Emphasis is given to the active process for gaining information (alignment with EU MDR and FDA Requirements) and inclusion of risk management in post-market surveillance.


• Annex C – Since questions for identification of hazards in the previous editions were taken as mandatory even though, the intention of these questions as guidance was taken out from ISO 14971 and moved to annex A in ISO TR 24971:2020 with additional considerations.


• Annex G – techniques – now annex B in ISO TR 24971:2019 (techniques to support risk analysis). Has additional information to clarify misapplication of techniques, and single-tool use such as FMEA in risk management (see above).

New ISO TR 24971:2020

ISO TR 24971:2013 had some information that did not appear in ISO 14971:2007.
The document has been completely revised so it is a very useful guide to risk management and provides guidance with risk analysis, identification of hazards, and evaluation of residual risks – if you follow ISO TR 24971:2020 you can more easily achieve a Medical Device which its’ Risk Management Process conforms with ISO 14971:2019.


Gsap experts will be happy to assist you in updating and preparing your risk management process according to ISO 14971 and related standards.

This Article Prepared by:

Adam Samucha, B.Sc

Medical Device Quality Project Manager


For more information about our services visit:

EU MDR – From Directive to Regulation

The new Medical Device Regulation (MDR) have been approved on May 2017 and the date for its implementation was May 2020, but recently its implementation was postponed due to the Corona pandemic and currently the target date is May 2021.

In this Newsletter, I will briefly highlight the changes in the new regulation related to clinical data collection.Beyond the many changes in aspects such as: new classifications, risk levels, Notified Bodies, certification processes and more, clinical evaluation requirements have been updated as well. The new regulation requires now a constant “live” connection between the risk management and the Clinical Evaluation and requires adjustment of the risk management processes and interphase with the clinical evaluation.                               

The sponsor is essentially required to assess each risk through clinical investigations, clinical evaluations, and of course through post-market clinical follow-up. Overall, referring to risk management, the MDR is in alignment with EN ISO 14971:2019 and EN ISO 13485:2016. While developing or amending the clinical plan, it should be taken into account that any change derived from the new regulation may also have implications on the clinical program. I.e. the question of medical device classification can have such implications and it is therefore advisable to characterize these implications before establishing the clinical program.

The transition from MDD (Medical Device Directive) to MDR has resulted in significant additional information in aspects of clinical data collection which is manifested in 20 new articles (articles 62-82). For Europe these are a fundamental changes and alignment with norms that are accepted worldwide in the medical device arena. The additional articles specify in fact requirements that have been relatively ambiguous so far, among other things, they specify requirements regarding:

the responsibility of the sponsor;

ethical considerations;

subject consent process;

reference to vulnerable populations;

safety information reports;

insurance etc.

In addition to these articles Annex XV, which extend the MDD Annex X, explains what information is required to appear in various core documents such as:

Clinical investigation report,

Investigator`s Brochure,

Clinical Investigation plan and others.

In terms of definitions, the MDR defined “Clinical Evidence”, extend the equivalence requirements to be more stringent and increased oversight of PMS (Post Marketing Surveillance) data. Despite the considerable extensions, more detailed standards and requirements will be found in the ISO 14155 and of course in ICH GCP.

Article 61 provides a concrete requirement for the need to conduct clinical trials with reference to implants or Class III For the other classifications, the requirement for conducting clinical trials is derived from the level of clinical information collected by the sponsor throughout the life cycle of the product. It is the responsibility of the sponsor to characterize the Clinical Evidence required for the product according to its characteristics and intended use. Given that the clinical information is limited, additional data, based on clinical investigation conducted in accordance with the GCP Guidelines, ISO 14155: 2011 and the Declaration of Helsinki would be required to complete gaps.

Article 62 indicates potential purposes for the clinical trial, these operations should be performed under the product design, the “normal” conditions of its intended use, including manufacturing and packaging. The clinical operations may be carried out to characterize the performance, benefits of its use as well as its safety implications. The structure of the trial and its design will be derived from the concrete need. Those operations may be:

relatively small in scope and their purpose will be to continue learning and improving aspects of product performance (Pilot studies) or, alternatively,

more extensive operations designed to present device performance under the conditions and constraints required to its function in the intended environment (Pivotal Study). Pivotal studies will most often be performed when the product is mature and expected to function as required and present its intended performance in accordance with the Claims.

The responsibility for maintaining clinical knowledge about the product on its variety of models and versions is the sole responsibility of the sponsor. When existing clinical knowledge changes or loses its validity as a result of changes in the product, adaptation of the clinical data collection plan should be considered. That process should use existing knowledge to establish updated conclusions and characterize the gap, and clinical investigation may be executed in order to complete the gap. Usually, a solution will be found that combines these two approaches.

In conclusion, for any product intended to be marketed in Europe, a continuous clinical information collection process is required throughout the product life cycle. The aforementioned sources of information include: literature and scientific publications, clinical operations, and information gathering from marketed products. The nature of the information collected and its purpose will be defined and adjusted to the product requirements and to the risk’s assessments. This process importance is further elaborated under the new regulation and plays a critical role in the dynamic assessment of the benefits and risks of using the medical device.

In a nutshell, changes in clinical requirements under the MDR:

Risk-Benefit analysis

Clinical Investigation
under MDR

New class III or Implants requirements

Articles 62-82Annex XV

Clinical Evidence

Equivalence requirements

PMS data
In alignment with EN ISO 14971:2012

In alignment with EN ISO 14155:2011

Added reference for clinical trials

Added – the “What”

Added – the “How”

Defined More stringent

Increased oversight

This Newsletter Prepared by:

Matti Hoggeg, M.Sc,

Clinical Section Manager


For more information about our services visit:

Preps for MDSAP Audit

The International Medical Device Regulators Forum (IMDRF) recognizes that a global approach to auditing and monitoring medical device developers and manufacturers could improve the devices’ safety and the regulators’ oversight on an international scale. Back in 2012, the IMDRF identified a work group to develop specific documents for advancing a Medical Device Single Audit Program (MDSAP).

The MDSAP Program allows an MDSAP recognized Auditing Organization to conduct a single regulatory audit of a medical device manufacturer that satisfies the relevant requirements of the regulatory authorities participating in the program.

Gsap has led MDSAP readiness activities with its Medical Device customers. We have a professional and knowledgeable team to support you in defining and aligning your business regulatory and quality strategy in accordance with the new approach of the MDSAP.

International partners that are participating in the MDSAP:


• Australia’s Therapeutic Goods Administration (TGA)
• Brazil’s Agência Nacional de Vigilância Sanitária (ANVISA)
• Health Canada – Santé Canada – MDSAP is mandatory from January 2019
• Japan’s Ministry of Health, Labour and Welfare (MHLW)
• USA – Food and Drug Administration (FDA)

MDSAP in General

• MDSAP covers regulatory aspects as well as:
o Registration
o Licensing
o Adverse event reporting
• MDSAP participating countries have direct access to the audit reports
• Audit is focused on requirements of ISO 13485:2016 and the regulatory requirements of the applicable jurisdictions (A manufacturer may exclude the requirements of a jurisdiction where the organization does not intend to supply medical devices)
• Audits conducted by approved Auditing Organizations (AO; Updated, official list is available at FDA site)

Preparation for the MDSAP

• Perform gap assessment for all procedures and processes
• Develop and implement risk assessment program
• Reach out to approved Auditing Organizations (AO)
• Fill AO’s questionnaire (to be used for additional assessment)
• Get a quote for services
• List products, registrations and licenses for applicable markets
• List types of products under the scope of the company
• Schedule an audit
• Be prepared for audits – # of days based on # of tasks to be audited

Gsap will be happy to support you in getting ready for the MDSAP requirements!

For more information visit Medical device Industry page

This Newsletter Prepared by:

Marina Lebel, B.Sc, CQE

VP Medical Device


For more information about our services visit:

EU MDR- Timelines and Plan

Who isn’t talking about the EU MDR and the new challenges ahead? At Gsap we want to do as well as talk. Gsap has led MDR readiness activities with its Medical Device customers.  We have a professional and knowledgeable team to support you in defining the business and regulatory strategy in light of the new landscape.

Medical Device Regulation (MDR) requires stringent standards of quality and safety for medical devices which will lead to a higher level of protection for patients and users. It seeks to increase premarket control (especially for high-risk devices), smooth communication between the different EU markets, increase surveillance at the post-market phase, enhance traceability (UDI) and transparency (EUDAMED).

EU MDR for whom?

For beginners in the Medical Device industry, meeting MDR requirements is mandatory for those wishing to sell their products in Europe. Since one of the main changes is the inclusion of devices without a medical purpose (such as esthetic products), there are new players in the Medical Device Landscape.

EU MDR by when?

By May 26th, 2020. No grandfathering, all existing devices must comply with new requirements by this deadline or when the NB certificate runs out (not later than May 26th, 2024).

EU MDR by whom?

Currently there 2 notified bodies approved for MDR audits TUV SUD and BSI.   BSI’s future is questionable with the Brexit, making TUV SUD the only feasible player on the field. 

How can you get ready now for the EU MDR?

Determine the intended purpose of the device for Declaration of conformity including technical documentation by:

  • Medical Device classification (CLASS I \ CLASS IIa \ CLASS IIb \ CLASS III)
  • GSPS (General Safety and Performance Requirements replaces ‘essential requirements’).
  • Traceability by UDI & EUDAMED registration
  • PMS activities
  • Biocompatibility changes

Main changes vs MDD

  • Wider coverage of devices without a medical purpose
  • New obligations for authorised representatives, distributors and importers
  • New European database (EUDAMED)
  • Unique Device Identification (UDI)
  • PRRC – appointment of Person responsible for regulatory compliance
  • Stricter requirements on:
  1. Risk management as a continuous iterative process throughout entire lifecycle of a device
  2. Pre-market controls of high-risk devices
  3. Clinical evaluation and investigation
  4. Use of hazardous substances

Gsap will be happy to support you in getting ready for the MDR storm!

This Newsletter Prepared by:

Marina Lebel, B.Sc, CQE

VP Medical Device


For more information about our services visit:

Medical Cannabis Clinical Trial Considerations

As time passes, keeping track of all the changes in the Global Medical Cannabis market is becoming more and more challenging.
The current newsletter will provide clarity where possible, regarding the design and implementation of Clinical Trials.
Valid Clinical Trials are necessary for the expansion of labeling in the US.
In Europe in general, and in Germany in particular, labeling requirements are less precise and the authorities allow for physicians’ discretion in prescribing Cannabis.

Currently, Israel is known worldwide as a pioneer in Medical Cannabis research and Clinical trials. More than 50 Israeli startups are developing Medical Cannabis products. Most Israeli start-ups are gradually maturing and progressing to clinical trials, mainly for PK, Phase 1, and Phase 2. Since 2011, over 45 clinical trials have been conducted in the field of Cannabis worldwide, with most of these studies being preliminary studies, about 5 phase 3 studies (including GW Pharmaceuticals and Takeda studies), and about 10 phases 2 studies.

The main research question investigated usually is the efficacy of the research product, but most studies also explore safety aspects where the most popular indications are central nervous system disease, Inflammation, and cancer. The quality of the results and the scientific value of these studies are “varied”; therefore, practitioners should develop the ability to monitor the quality of execution and planning of clinical trials. As a result of what we have learned and observed, we believe that proper planning and execution of a Clinical Trial is critical to achieving regulatory approval in a timely manner.
We wish everyone a fruitful and prosperous year, and of course, may it be the year of Cannabis.

Designing medical Cannabis clinical trials

Clinical studies are a significant and necessary portion of the process of developing and approving medical products. Cannabis-based medical products also require clinical trials and the submission of detailed clinical study reports of the clinical trials in order to be included in the Registration file that will be submitted to the authorities. This review will address two key aspects:
• Basic principles in clinical trials design
• Guiding considerations in the design of medical Cannabis clinical trials


Clinical trials should be designed and conducted according to three general principles:


1. Protection of Clinical Study Subject
This principle provides guidance for planning and execution in a way that ensures the patient’s well-being and rights while emphasizing special populations in a way that requires compliance with the ICH E6 Good Clinical Practice requirements and ISO 14155 when the clinical investigation involves medical devices.


2. Scientific Approach in Clinical Study Design Conduct and Analysis
This principle requires a scientific approach in designing the research objectives, executing, collecting the results, and analyzing them, to ensure obtaining reliable data and the required answers for the intended questions.


3. Patient Input into Study Design
Aimed at integrating, receiving feedback, and involvement of patients and patient organizations in the design and implementation of the study. This is done in order to plan and conduct a feasibility study that addresses the real needs of patients.
The characteristics of clinical research in Cannabis, as well as preclinical steps in product development, are derived from the regulatory submission path and therefore great importance should be given to determining the correct regulatory strategy for each product and at the
earliest stage. Although clinical trials are sometimes not mandatory, clinical trials are almost always performed.
These clinical trials will be performed for the purpose of accumulating safety and efficacy information, for marketing needs, financial needs, and or needs to confirm assumptions about the efficacy of the product under different indications, different administration routes, combined with other treatments, etc. This review focuses on the design and conduct of the clinical trials in accordance with the regulatory requirements of a nonherbal pharmaceutical product, these requirements are the most rigorous and demanding and ensure as much as possible the reliability of the scientific insights that can be derived from the research.

Cannabis research requires special attention to these requirements because prior information on cannabinoids / Cannabis use in medical products is limited. This is due to, among other things, a lack of reliable scientific and clinical information in the field of Cannabis research and is the result of conducting studies that are lacking in their design and without the use of common methods to reduce BIAS.
Once the indication has been thoroughly studied and the relevant literature reviewed, the research rationale and assumptions sought by the research can be formulated. In each case, the research and its objectives will be planned with reference to, and in connection with, the broader clinical program and the stages of the project or product.

The research design should focus on the guidelines outlined below at both the planning and execution levels:

1. Study Population
The characteristics of the study target population are derived first from the target product population, the study as a whole will characterize the effect of the investigated product on a particular population and the conclusions, in principle, will be relevant to that population. Later on, the criteria for inclusion and exclusion will also include considerations related to safety, ethics, DDI (Drug-Drug Interaction), background and medical history, etc.

Cannabis studies should also take into account, the extent to which patients have been exposed to Cannabis use in the past, the psychoactive consequences of THC, and the patients’ ability to consume the product as required. In general, the characteristic of the study population can be said to be a delicate balance between reducing the variability between patients in order to increase the chances of demonstrating the therapeutic effect on the one hand, and the implications of these decisions on the ability to recruit patients for research and approve treatment based on research in a population characterized by limited variability on the other hand.


2. Intervention
The dosage, treatment duration, route of administration, and a number of treatments have a major impact on “Product Success / Research”.
Dosage, treatment regimen, and administration have a direct effect on product usability, the absorption of active ingredients, their effectiveness, as well on their safety, and economics. Cannabis studies show a wide variety of forms of administration, ranging from topical ointment form through to nasal and oral vaporizers, sub-lingual, and even rectal administration. Each of the forms of administration has pros and cons. For example, using a vaporizer significantly reduces smoking damage and allows for an “ Entourage Effect” and has close proximity to the traditional Cannabis consumption, but requires patients to be supplied with a vaporizer which significantly increases the operative and regulatory requirements both during and after the study. Oral administration is relatively simple to use and operate but is characterized by a slower and less effective absorption of the active ingredients. Apart from these, the “intervention” consideration also corresponds to the indication itself and the target patient population, which dictates restrictions on how the patient consumes the product being tested.


3. Control Group
The requirement for controlled research is a “gold standard” in clinical studies design and usually, research cannot be carried out with good reliability without a control arm. The need for a control arm/ group results from a phenomenon that has been documented since the 1930s and is called the “Hawthorne effect.” In general, this effect shows that solely just the participation of the subject in the clinical trial results in a change not related to the medication. The purpose of the control group and its value derived from the fact that it allows us to analyze the effects of the treatment only regardless of the effect of other external factors not related to the medication/treatment.
There are different types of controls, for example; a Comparison of the research drug or treatment with another drug or other conventional treatment as well as a placebo. This aspect is particularly challenging in Cannabis trials as it is difficult to create a “placebo” in Cannabis products especially when the tested product is consumed via inhalation and/or contains a THC component that gives a unique feeling and allows for easy differentiation of treatment arms by the patient. Due to this reason, cross-over and double-dummy studies for example are more relevant in the cannabis territory.


4. Response Variables
Endpoints are derived directly from the study objectives and are used to assess and characterize the treatment effects. The endpoints selection has a critical impact on the success of the study and is the result of a complex set of considerations. The endpoints are first and foremost intersected with the indication, regulatory requirements, and statistical considerations, but also take into account broader elements of the clinical development program such as follow-up and future planned studies, patient needs and limitations, competitors and competition, etc. Early studies will usually use measures related to safety and tolerability as a Primary endpoint, while efficacy questions can be also expressed in Secondary endpoints. In the more advanced stages of product development, the priority is changed and the main objective will focus on the question of effectiveness while safety information collection continues throughout the life cycle of the product in order to produce safety knowledge at different levels of product exposure.


5. Reduce or Assess Bias
Bias is an accumulation of factors that can compromise the reliability of clinical trial results.
The bias is derived from the unique research structure and needs to be addressed to reduce it as much as possible. The two most powerful tools for reducing bias are “RANDOMIZATION” and “BLIND”.

Randomization means randomly assigning patients to the treatment arms. This randomization is expressed as an equal probability for each patient to be included in each arm, thus avoiding the conscious and unconscious bias of patient allocation and balancing the baseline characteristics of the treatment groups (The larger the group in the trial, the value of the randomness set increases).

BLIND or MASKING, however, requires that the patient or the research team (SINGLE BLIND \ DOUBLE) do not know to which treatment arm the patient is assigned. Creating a proper blind study creates operative challenges that need to be addressed in order to avoid psychosomatic effects and biases.

Apart from this, every procedure in the study that can influence the results of the study should be considered to ensure that its application is not affected by bias.


6. Statistical Analysis
When the structure of the trial and the selected endpoints are determined, a suitable statistical model should be formulated to analyze the results of the study and a preliminary plan for the analysis of the results should be formulated. It is difficult to overstate the importance of bio-statistician involvement in model selection, sample size, decision making during the study, and of course in analyzing the results and presenting them. As mentioned before, these aspects have great importance in the planning phase and any changes may affect other aspects. Apart from this, during the Study Execution, the Sponsor is expected to ensure that the study conduct is done according to the approved plan and in compliance with the study protocol. In Cannabis trials this challenge is particularly tangible as these are studies with relatively high operative and regulatory complexity. In cases where the design and conduct are properly performed, authentic results are guaranteed which reflect product performance
appropriately.

This Newsletter Prepared by:

This image has an empty alt attribute; its file name is sigalit-1-aspect-ratio-387-442-1-896x1024.jpeg

Sigalit Ariely-Portnoy, Ph.D

CEO


For more information about our services visit:

Medical Cannabis Regulations

Topics in this article:

1.The GMP requirements for Post-Harvest in Europe:

1.1 Medical cannabis manufacturers aiming to sell their products in Europe and specifically in the German market must meet several requirements:


• Compliance with the Herbal Medicinal Products Committee (HPMC) requirements of the European Medicines Agency (EMEA) and Good Agricultural and Collection Practice (GACP) Guidelines for Plant-Derived Starting Materials.

• Compliance with European GMP requirements, EudraLex – Volume 4 – Annex 7, Manufacture of Herbal Medicinal Products. Annex 7 (to EU GMP) presents the requirements for facility, equipment, documentation, and process control for the production of herbal products.
In the Annex, the referencing matrix is presented – for each stage of inflorescence processing, to the relevant quality requirements.

Figure No. 1: Application of Good Practices to the manufacture of herbal medicinal products:

 Medical Cannabis Regulations


1.2 The GMP classification depends on the stage of the inflorescence processing – raw material, intermediate material, or finished product, and the possible impact on the finished product. It is the responsibility of the medical cannabis product manufacturer to ensure that the processing and production processes comply with the relevant GMP requirements.


1.3 Propagation, cultivation, and harvesting of cannabis inflorescence are subject to the GACP guidelines. The GACP guidelines do not directly fall within the GMP guidelines in the traditional sense. However, these guidelines should be the basis for establishing a proper quality assurance system.

1.4 Post Harvesting processes such as Trimming, Drying, Curing, and Hand trimming are considered as a starting material for a medical product and therefore may also be subject to EudraLex part I or part II quality requirements in addition to being subject to the GACP guidelines. Parts I and II of EudraLex Vol. 4 are intended to provide GMP guidelines to the manufacture of medicinal products and active ingredients used as raw materials, respectively. This is to ensure that they meet the quality requirements stated by them.

medical cannabis regulations

2.Submissions of Medical Cannabis Products in Europe

2.1 Generally, there is not a single route for Medical Cannabis approval across Europe. Each country has established its own regulations regarding Cannabis.

2.2 Cannabis is legally authorized in the Netherlands and is decriminalized for use in Germany, Portugal, the United Kingdom, the Czech Republic, Spain, and Estonia.

2.3 In countries such as France, Italy, Poland, Bulgaria, Cyprus, Denmark, Croatia, Finland, Luxembourg, Malta, Romania, Sweden, Austria, Latvia, Slovakia, Slovenia, Lithuania, Belgium, and Hungary Cannabis is still illegal.

2.4 The use of Cannabis-based medications is possible by means of three pathways (in some European countries):
• Cannabis preparations to improve wellbeing – not defined as medicine and without any specific therapeutic indication (wellness).
• A medicinal product – intended for the treatment of a labeled disease, requires clinical trials and in most cases also intended for marketing (e.g. paracetamol – analgesics).
• Medical Cannabis Path for Personal Care – For a specific patient, for certain illnesses accompanied by a doctor. This course is possible in some European countries.


2.4.1 Path No. 1: The cannabis preparations path does not require marketing authorization (MA). Cannabis preparation is originated from the Cannabis plant raw material – for example, inflorescence, oil extraction, and concentrated extraction. Cannabis preparations contain a low amount of THC (the approved level 0.3%, 0.2%) or without THC, thereby eliminating the psychoactive effect. Some countries allow the distribution without medical approval – in such countries where the sale of Cannabis is legal, or in countries where the personal use of Cannabis is decriminalized.

2.4.2 Path No. 2: Medical Cannabis for Personal Care. 
This pathway allows a specific patient to receive special approval for an unapproved drug, usually under a physician’s supervision for the treatment of serious illnesses, illnesses that have not responded to other medications, or as a compassionate treatment (chronic pain, terminal cancer, or degenerative neurological disease). The doctor who provides the prescription is required to monitor and report the results of the treatment as well as any side effects or adverse events. 
Medical Cannabis is supplied by pharmacies and requires EU GMP approval and a valid agreement with a local distributor. (see section 2.6).

2.4.3 Path No. 3 Cannabis-based Medicinal Products Medicinal Product – A substance or combination of substances used to cure or diagnose a disease, or to restore, repair, or alter physiological functions by activating pharmacological, immunological, or metabolic activities. This pathway requires MA for medicinal products. The MA is obtained after submission of a registration file to the health authorities, which includes clinical trials demonstrating effectiveness, efficiency, safety.

2.5 The source of the medicinal product may be synthetic or herbal:

2.5.1 A non-herbal source medicinal product route (e.g. synthetic origin). This path is a regular medicinal product route, manufactured in the pharmaceutical factories (such as paracetamol), and requires all tests to be approved (Clinical and Preclinical Trials).

2.5.4 Herbal-based medicinal product (similar to Sativex®): A medicinal product containing one or more active substances from a plant source or an herbal preparation or a combination of the two. Data should be presented that the herbal material has well-established pharmaceutical use, with over 15 years in the European community, a recognized efficacy, and an acceptable level of safety. In addition, it is necessary to demonstrate the quality and consistency of the drug manufactured in order to ensure that the patient receives the same product every time and is free of contaminants. A long tradition of using herbal medical products enables the reduction of clinical trials to prove the safety of the medicinal product. The need for pre-clinical examinations seems unnecessary, as long as the medical history of the drug and its long-standing traditional use have not caused any harm. However, the competent authorities are entitled to request, where necessary, the data for the safety assessment. In terms of the quality aspects of the medicinal product, the product must be analyzed for physical\chemical, biological, and microbiological tests and to comply with the relevant
quality standards in Europe: Pharmacopeial monograph,  HPMC guidelines, and relevant directives (2004/24 / EC & 2001/83 / EC).

2.6 Obtaining a permit to import Medical Cannabis into Germany

2.6.1 Germany only imports medical Cannabis from farms and facilities operating under the 1961 Convention of Narcotic Drugs.

2.6.2 In order to import Medical Cannabis to Germany, a EUGMP certificate from the EU member state is required.

2.6.3 Manufacturers wishing to distribute Medical Cannabis products in Germany must be in agreement with or be the owner of a domestic importer. The domestic importer will have: 
• Registered business.
• Wholesale distributor drug license, applied at a regional level.
• Federal-level Narcotics handling license.

2.6.4 In order to obtain the necessary licenses, importers should meet the local and federal requirements, such as: employing at least one employee responsible for narcotics, applying extensive security measures, etc.

2.6.5 Regional German inspectors have granted GMP certificates to foreign manufacturers and provided import permits to Germany.

2.6.6 The type of approval depends on the activity planned by the importer:
• Production and import authorization.
• Compliance with EU-GMP requirements.
• Approval of wholesaler-distributors.
• Compliance with Good Distribution Practice (GDP) requirements.

2.6.7 In order to obtain EU-GMP approval, an on-site audit and document set must be submitted in accordance with the requirements of the Authority.

2.6.8 The issues with the approval of Medical Cannabis as an herbal medicine in Europe are due to:
• Medical Cannabis is still included in the narcotic drug regulation in some countries.
• The difficulty in characterizing the full spectrum of the plant’s cannabinoids.
• To demonstrate product stability and purity in terms of microbiological contamination, heavy metals, and pesticides residues.

2.6.8 Table No. 1 lists the requirements for personal care in some European countries. Changes in the legal status of Medical Cannabis use through Europe and the growing demand for approval of medical Cannabis make it difficult to predict the approach to Medical Cannabis in the near future. At any rate, it is advisable to consult with the local health authorities of the country in question in order to assess if it is feasible both financially and logistically.

Table No. 1: Requirements for Personal Care Pathway in some European countries:

 Medical Cannabis Regulations
Insight to the Medical Cannabis Regulations
 Medical Cannabis Regulations

3.Medical Cannabis Product Specifications (Israel, Netherlands, Germany)


3.1 Similarly, to medicinal products, test methods, and specifications have been defined in some countries. The set of methods and specifications for raw material or medicinal is defined as a monograph.

3.2 In several countries where medical Cannabis products were approved, such as in the Netherlands and in Germany,  local monographs for medical Cannabis were defined. We will review the defined requirements in Israel, The Netherlands, and Germany.

3.3 Israel:

3.3.1 The Medical Cannabis unit (IMC) in the Israel Ministry of Health, has issued several guidelines related to the supply chain of Medical Cannabis. SOP 152, “IMC-GMP Quality Requirements for Manufacturing Medical Cannabis Products”, defines the set of tests and specifications required for Medical Cannabis products marketed in Israel. The tests requirements described in SOP 152 are based on the requirements of the European Pharmacopoeia (Ph. Eur.) General Monograph for Herbal Drugs (Ph. Eur., 1433, Herbal Drugs).

3.3.2 According to SOP 152 and the January 2019 notification, there are four main approved medical Cannabis products groups – inflorescences, cigarettes, oils, and cookies.


3.3.3 For inflorescences (flowers or grind flowers): 12 different products are defined, which differ in the concentration of active ingredients, and for THC products also in the source
(sativa / Indica); 

3.3.4 For oils: 9 different products are defined, which differ in the concentration of active ingredients.

3.4 The Netherlands:

3.4.1 The Office of Medical Cannabis (OMC) has defined specifications for 5 different inflorescences varieties that  differ in the concentration of active ingredients (as of January  Bedrocan (Sativa), THC concentration ~ 22%; CBD concentration <1.0%
• Bedrobinol (sativa), THC concentration ~ 13.5%; CBD concentration <1.0%
• Bediol (sativa), THC concentration ~ 6.3%; CBD concentration ~ 8%
• Bedica (indica), THC concentration ~ 14%; CBD concentration <1.0%
• Bedrolite (sativa), THC concentration <1.0%; CBD concentration ~ 9.0%

3.4.2 The Office of Medical Cannabis is also responsible for final batch release to the market.

3.5 Germany:

3.5.1 Germany has two local pharmacopoeia DAB (Deutsches Arzneibuch) and DAC (Deutscher Arzneimittel Codex). DAB has published a monograph for Cannabis inflorescence (flowers) of the female plants of Cannabis Sativa L. The requirement for Assay shall meet 90.0-110.0% of the labeled amount (calculated on a dries basis). Inflorescence storage conditions according to the monograph: tightly closed container, stored below 25°C. The monograph indicates that inflorescence products are divided into three groups: THC >> CBD, THC = CBD, THC << CBD.

3.5.2 In June 2019 a draft monograph of Cannabis oil (extract) monograph was published in DAB and it is planned to become effective during 2020. According to the draft monograph (which is still under review), the Cannabis oil should be prepared by extracting whole or grinded inflorescences of the female plants of Cannabis sativa L. The active ingredient content: THC is in the range of 1-25% and for CBD the range is NMT 10% (m/m). The active substance content shall be within the range of 90.0-110.0% of the labeled amount. The monograph includes applicable manufacturing processes: extraction (such April as ethanol or CO2), dilution of the crude extract with oil (MCT, grapes) in order to reach the target concentration. The material must undergo de-carboxylation where applicable in the process. The storage conditions according to the monograph: tightly closed container, protected from light and refrigerated (2-8 °C) – this probably will be changed in the official monograph. It should be noted that the tests specified in the German Pharmacopoeia (DAB) monographs are in addition to the requirements for Herbal Drugs (Ph. Eur.\ HPMC guidelines).

3.5.3 Table 2 includes a comparison between the specification requirements in the different countries stated above for inflorescences (flowers).

Table 3 includes a comparison for Medical cannabis oils. The tables do not include references to the active ingredient content mentioned above. Also, keep in mind that there may be differences in the test procedures between the different monographs.

Table No. 2: Medical Cannabis Inflorescence

As the world moves to implement Good Manufacturing Practices for the Medical Cannabis industry, the need for standardization is also increased. More and more countries, in Europe and in the rest of the world, are starting to initiate regulation regarding Medical Cannabis. Outside of Europe, we can find Australia, New Zealand, Brazil, and more.

Prepared by:

Sigalit Ariely-Portnoy, Ph.D

CEO


For more information about our services visit:

Skip to content window.TEAMME_CONFIG = { companyNamespace: "bringthemhome", };