search
close_mark

Speeding Systematic Literature Reviews with AI for CER Compliance

Speeding Systematic Literature Reviews with AI for CER Compliance

19 Sep, 2025

Systematic literature reviews are a central component of Clinical Evaluation Reports. However, searching multiple databases, screening thousands of titles, extracting structured data and preparing audit-ready documentation often absorbs the author’s time and delays progress. When used responsibly, AI can accelerate these repetitive tasks while preserving reproducibility and traceability.

Common Bottlenecks in Traditional SLRs

Teams frequently encounter the same constraints:

  • Running inconsistent and inadequate queries across databases and managing large duplicate sets.
  • Manual title and abstract screening that consumes significant SME/author hours.
  • Extracting study-level fields such as design, population, endpoints and harms into a consistent dataset.
  • Producing PRISMA-style documentation, version control and traceability for audits and Notified Body review.

These efforts are essential for MDR compliance but can also significantly impact timelines and quality.

What AI Contributes, with Safeguards

AI is most valuable for repetitive, pattern-based work. Key capabilities that benefit SLRs include:

  • Automated retrieval and de-duplication across multiple sources.
  • Assisted screening that ranks records by relevance, keeping reviewers in control.
  • Rapid structured extraction into a standardized data dictionary.
  • Automatic generation of PRISMA flows and exportable audit trails.

AI should extend human capability, not replace it. Every AI generated result must link back to the source and remain verifiable by reviewers.

A Regulator-Ready Hybrid Workflow

Below is a practical sequence that balances automation with mandatory human checks:

  • Scope and protocol: Define the clinical questions, populations, comparators and acceptance criteria; pre-register search strings, databases and date ranges.
  • Automated retrieval and clean-up: Run pre-registered searches, remove duplicates and tag records for screening.
  • Assisted screening with adjudication: Use AI scoring to assess records; two reviewers confirm inclusions and resolve disagreements.
  • Full-text extraction and verification: AI extracts data based on pre-defined instructions into templates; reviewers verify critical fields such as endpoints and harms.
  • Quality appraisal and synthesis: Appraise study quality, apply pre-defined criteria for benefit-risk, and conclude study findings accordingly.
  • PRISMA output, traceability and version control: Produce a PRISMA flow diagram, an exportable study table and a traceability matrix linking claims to evidence and risk controls.

This approach reduces the human oversight regulators expect while automating the steps that typically consume the most time.

Mini Case Example

Consider a typical SLR for a Class IIa device that searches five databases and returns 3,500 records. In a manual workflow, title/abstract screening and full-text extraction might require four to six weeks of an SME’s time. With automated retrieval, de-duplication and assisted screening, the initial screening pool can be reduced by more than half within days. Assisted extraction then populates a structured dataset that SMEs can validate in a matter of days rather than weeks. Deliverables such as the PRISMA flow and an exportable study table. The net effect is savings of several weeks in SME time, faster responses to Notified Body queries and a cleaner audit record that documents reviewer verification of AI-extracted items.

Integrating Real-World and Decentralized Data

Regulatory reviewers increasingly expect real-world evidence, registries and decentralized trial outputs to support CER updates. AI can help harmonize heterogeneous inputs by tagging metadata, tracking sources and converting registry exports into structured records for appraisal. The same human-in-the-loop principles apply here, and the analytical validation and reviewer verification are required before RWE is used in benefit-risk conclusions.

Operational Benefits

Adopting this hybrid model delivers measurable improvements:

  • Shorter cycles because searches, de-duplication and extraction are automated.
  • Fewer data-entry inconsistencies due to structured extraction.
  • Better audit readiness through consistent PRISMA outputs and per-item provenance.

These gains free the author and the subject experts to focus on interpretation and clinical judgment rather than repetitive data handling.

Governance and Team Readiness

Introducing AI in your workflow should include clear guidelines. Define standard operating procedures specifying where AI is permitted, which fields require mandatory human verification, and how AI determined scores should be interpreted. Train reviewers to identify common AI errors and record corrections. Periodic audits comparing AI outputs with manual benchmarks help maintain quality and build trust with internal stakeholders and external reviewers.

Below is a quick SLR Checklist when automation is used

  • Pre-specify search strategy and inclusion/exclusion criteria in a protocol.
  • Utilize exportable, audit-ready outputs including PRISMA flows and data dictionaries.
  • Keep reviewers in the loop for screening and extraction verification.
  • Maintain a traceability matrix linking claims, evidence and post-market outputs.
  • Archive logs showing search timestamps, reviewer decisions and data sources.

How CAPTIS® Can Help

Integrated platforms that combine literature search, collaborative review, structured extraction and automated reporting make this hybrid workflow practical on scale. CAPTIS® automates extraction, generates audit-ready reports and preserves references while enabling simultaneous review and simple cross-verification. Features that accelerate compliance include an integrated citation manager that links each extraction to source documents, exportable audit logs for every reviewer decision, role-based review workflows and pre-built templates for traceability matrices. That mix helps teams shorten SLR cycles without sacrificing traceability or regulatory rigor. Read How AI Speeds Up Systematic Literature Reviews by 60% blog here.

Conclusion

A rigorous protocol, clear reviewer responsibilities and tools that produce verifiable outputs make it possible to utilize automation and reduce SLR timelines while improving consistency and auditability. Teams that adopt a balanced AI-plus-reviewer approach will be better positioned to meet Notified Body expectations and keep CERs current with high-quality evidence. By leveraging AI-powered tools like CAPTIS®, medical device professionals can ensure compliance with regulatory requirements while saving time and resources. Stay ahead of the curve by embracing AI in your SLR process. See how our AI-powered solutions can transform your regulatory processes. Contact us at info@celegence.com to learn more.

 

Other Related Articles

View All

Building Audit-Ready CERs — Meet Article 61 & Annex XIV with CAPTIS®

Building Audit-Ready CERs — Meet Article 61 & Annex XIV with CAPTIS®

01 Sep, 2025

Clinical Evaluation Reports are central to EU MDR technical documentation. Article 61 and Annex XIV require that clinical claims be demonstrably supported by up-to-date evidence, linked to risk management, and maintained throughout the device’s lifecycle. This article discusses how an integrated platform such as CAPTIS® helps teams deliver audit-ready CERs with greater speed and consistency.

What Reviewers Now Expect

Notified Bodies and MDCG guidance emphasize transparent methods, reproducible Systematic Literature Review (SLR)/State of the Art (SOTA) outputs, and direct links from claims to source data. Key reviewer expectations include:

  • Clearly defined clinical claims with measurable criteria.
  • A documented SLR/SOTA methodology (search strings, databases, date ranges, PRISMA-style flow).
  • A traceability matrix that links each claim to the evidence, related RMF entries, and PMS/PMCF actions.
  • Version control and an update plan tied to device risk and specific triggers.

If these elements are missing, reviewers commonly issue formal findings — implement a living traceability matrix, strict version control and documented reviewer signoffs to prevent delays.

How to Build a Traceability Matrix

Following the below steps can make the Traceability Matrix operational:

  • Define clinical claims – Phrase each claim which is specific to a patient’s benefit and map it to specific safety and performance endpoints which are measurable.
  • Capture appropriate source information – For every extracted data includes database, DOI, page/location, and so verifiers can find the original quickly.
  • Link to Risk Management – Connect claims to RMF entries showing how residual risks are accepted and mitigated.
  • Tie gaps to PMS/PMCF actions – For any evidence shortfall, document the PMCF objective, planned study or survey details along with expected completion timelines.
  • Date and version of every change – Maintain a clear audit history which identifies the changes along with the rationale. When this matrix is kept current, the CER becomes a single source of truth that accelerates internal review and external assessments.

A Regulator-Ready Workflow

Combine reproducible methods with controlled automation:

  • Protocol and scope: Pre-record clinical questions, inclusion/exclusion criteria, databases and exact search strings.
  • Reproducible SLR: Capture search timestamps and export PRISMA outputs.
  • Structured extraction: Populate a standardized data dictionary that includes fields required for benefit-risk analysis.
  • Customizable verification: Configure a multiple reviewer workflow for confirmation for screening and sign-off on critical extracted fields (endpoints, harms).
  • Traceability and updates: Auto-populate the matrix, identify gaps and record PMCF progress.

This balances efficiency with the level of human review needed for regulatory defensibility.

Practical Benefits and Operational Gains

Using a platform that integrates search, extraction, traceability and review delivers measurable advantages. Automation reduces time spent on repetitive tasks, while structured extraction cuts transcription errors; versioned audit logs and PRISMA outputs improve readiness for NB review. For example, when a Notified Body requests clarification on a performance claim, teams using a living matrix can rapidly cite exact studies, provide extracted data points and show linked PMCF activities already in progress. Rather than reassembling documents, teams can provide a concise package that includes evidence extracts, appropriate references and a documented update timeline — often closing the query with minimal iterations. Another example is Integrating Real-world evidence RWE and Decentralized Data. Real-world data, registries and decentralized trial outputs are increasingly used to strengthen CERs. Harmonizing heterogeneous sources requires careful tracking, analytical validation and reviewer verification before incorporation into benefit-risk conclusions. Platforms that tag metadata and preserve source lineage make these integrations auditable and regulator friendly.

Paired with managed services, this approach shortens project ramp-up and makes costs predictable, enabling more efficient resourcing and a clearer total cost of ownership.

Governance and Team Readiness

As seen above, there are a lot of advantages to integrate automation. However, tools are effective only with the right controls: It is also imperative that the following is considered

  • Establish SOPs that specify which AI-assisted fields need mandatory human verification.
  • Train reviewers to verify source information and resolve uncertain inclusions.
  • Maintain strict version control and exportable logs for each CER revision.
  • Update the traceability matrix with the RMF and PMS outputs on a scheduled basis.

These governance measures can help to reduce errors during an external review and can make lifecycle updates easier.

How CAPTIS® Helps in Practice

CAPTIS® combines integrated literature search, structured extraction and an embedded citation manager, enabling teams to reduce manual effort while preserving auditability. The platform’s version control and role-based review workflows shorten cycles and help avoid oversight. Paired with managed expertise, this delivers faster ramp-up on projects — supporting both operational efficiency and stronger submission outcomes.

Conclusion

Partnering with Celegence provides you access to skilled and experienced regulatory consultants, AI-driven solutions, and access to tailored support to streamline your CER process. From navigating complex regulations to conducting systematic literature reviews, our specialists ensure your CERs are compliant with EU MDR requirements, enabling faster regulatory approvals and confident market entry.

Are you facing challenges with your Clinical Evaluation Reports? Contact Celegence today to learn how we can help you navigate the complexities of CER preparation and ensure regulatory success.

AUTHORED BY

author-image

Associate Manager - Medical Device Services

Dr. Kasturi Rao

Linkdin-image

Kasturi Rao holds a Doctor of Philosophy (Ph.D.) degree in Cancer Research and a Masters degree in Molecular Biology and Human Genetics. She has 12 years of experience in the Life Sciences Industry, including 4 years of experience in Regulatory Writing for Medical Devices. She has authored and reviewed various critical documents such as Clinical Evaluation Plans (CEP), Clinical Evaluation Reports (CER), Post-Market Surveillance (PMS) documentation, Post-Market Clinical Follow-up (PMCF) reports, and Summary of Safety and Clinical Performance (SSCP) documents for medical devices. These documents adhere to EU Medical Device Regulation (MDR) and MEDDEV 2.7/1 Rev 4 guidelines, ensuring compliance and quality. Kasturi Rao has extensive experience in handling end-to-end Post-Market Surveillance (PMS) documentation. This includes conducting literature reviews for Device under Evaluation (DUE) and State of the Art (SoTA), performing searches on clinical trial databases and Adverse Event (AE) databases, authoring SoTA documents, and creating high-level summaries of literature, clinical, and non-clinical test reports. At Celegence, she leads a team of medical writers who specialize in creating various regulatory documents such as CEPs, CERs, PMS Plans, Post-Market Surveillance Reports, Periodic Safety Update Reports, and Post-Market Clinical Follow-up Plans and Reports.

Other Related Articles

View All

Getting MDR Clinical Evaluations Right: What Dental Device Manufacturers Need to Know

Getting MDR Clinical Evaluations Right: What Dental Device Manufacturers Need to Know

26 Aug, 2025

As the EU MDR transition deadlines for legacy devices approach, dental device manufacturers face a heightened burden of clinical evidence. In a recent webinar hosted by Celegence, regulatory experts Priya Ray Chaudhuri and Dr. Manaswitha Boyanagari shared hard-earned lessons from working with dental clients through MDR transitions — and offered clear guidance on how to navigate the clinical evaluation process with confidence and precision.

Here are the key takeaways that regulatory and clinical leaders should prioritize when preparing dental device submissions under MDR.

Why Dental Devices Face Increased Scrutiny

Unlike many other device categories, dental products — particularly implants and restorations — directly interact with human tissue, integrate into bone, and often remain in the body permanently. This raises biological risks — such as inflammation, mechanical failure, or peri-implantitis and Bone loss, making robust clinical evidence not just a regulatory expectation, but a public health necessity.

Certain dental devices have also been reclassified under MDR compared to the earlier MDD framework. Devices previously considered low- to moderate-risk under MDD — for example: Dental implants, Bone replacement materials especially if they have a biological effect or are wholly or mainly absorbed, and certain software tools — are now Class IIb or even Class III. The result? New conformity assessments for all medical devices currently circulating within the European Union Member States, complying with the new regulation and new rules. No grandfathering. Manufacturers must justify safety and performance through new, MDR-aligned clinical evidence and additional requirements like submission of implant cards and summary of safety and clinical performance (SSCP) for implantable and Class III devices.

Clinical Evaluation: A Lifecycle, not a Checkbox

In the webinar, Celegence emphasized the need for a lifecycle approach to clinical evaluations, anchored in four key documents:

  • Clinical Evaluation Plan (CEP): Defines the claims, population, data sources, and appraisal methods. It’s the foundation for all subsequent activities.
  • Clinical Evaluation Report (CER): Synthesizes clinical data, assesses benefit-risk, and demonstrates alignment with the GSPRs.
  • State of the Art (SOTA) Review: Establishes the baseline for comparison and benchmarks claims against current best practices.
  • Post-Market Clinical Follow-up (PMCF): A must-have for most Class IIb and III devices, PMCF provides real-world evidence to support long-term safety and performance.

Building a Robust Evidence Strategy

Most manufacturers won’t have the luxury of new clinical trials, especially for legacy devices. That makes literature appraisal the backbone of many MDR submissions.

So, it’s important that manufacturers:

  • Be precise in defining intended use and claims. Vague terms like “safe” or “effective” won’t pass. Quantify outcomes where possible — e.g., “98% survival at 7 years” — and ensure they match the indications and target population.
  • Design a thorough literature strategy. Use smart filters, search across multiple databases, and include gray literature or implant registries. For dental products, state-of-the-art evidence is particularly critical due to variability in patient response and device longevity.
  • Critically appraise sources. Not all literature is created equal. Use a scoring matrix based on relevance, quality, and alignment with the device in question. The MIDDEF 2.7/1 guidance remains a good starting point.

Common Pitfalls — and How to Avoid Them

Manufacturers frequently run into the same issues when preparing MDR-ready CERs:

  • Claims not linked to evidence. Every claim should tie back to a data point. If the evidence is lacking, that gap must be addressed through PMCF — not ignored.
  • Over-reliance on equivalence. MDR has raised the bar for demonstrating equivalence, especially in terms of material and technical characteristics. A titanium implant is not equivalent to zirconia just because they’re both biocompatible.
  • Outdated SOTA reviews. Even legacy devices need updated benchmarking. SOTA evidence should typically be refreshed every 12–24 months.
  • PMCF treated as an afterthought. PMCF should be risk-driven and strategic, not just a box-checking exercise. Well-designed clinician surveys, registry tracking, or focused case studies can provide meaningful insights — and address notified body feedback proactively.

What Executive Teams Should Take Away

For dental manufacturers, MDR compliance is more than just a regulatory obligation — it’s a strategic imperative. Clinical evaluations must now go beyond generic templates or legacy data. Success requires structured, defensible, and up-to-date documentation.

As Priya Chaudhuri put it during the session: “The CER isn’t just a report. It’s a strategy.”

Leadership teams should ensure they’re resourcing regulatory functions appropriately — with expert partners, robust data collection systems, and cross-functional input from product development and clinical affairs. A well-executed clinical evaluation strategy not only satisfies regulators, it strengthens a company’s competitive standing in a market where performance, aesthetics, and safety all drive success.

Partner With Celegence

MDR clinical evaluations for dental devices don’t need to be overwhelming. With expert support and proven methodologies, you can deliver CERs, SOTAs, and PMCF strategies that align with regulatory expectations and stand up to Notified Body scrutiny.

Work with our regulatory specialists today and simplify your path to MDR compliance with confidence.

AUTHORED BY

author-image

Manager • MD Services

Priya Ray Chaudhuri

Linkdin-image

Priya Ray Chaudhuri is an expert in Clinical affairs with over a decade of experience in medical writing, clinical evaluation, and post-market surveillance for medical devices. As a Manager at Celegence, she oversees global regulatory projects, ensuring alignment with EU MDR, IVDR, MEDDEV 2.7.1 Rev 4, and country-specific requirements.​ Priya brings deep expertise in authoring and reviewing clinical and performance evaluation reports, PMCF plans, PMS reports, and PSURs across a wide spectrum of therapeutic areas. ​ Beyond documentation, Priya supports strategic decision-making by providing regulatory intelligence, training cross-functional teams, and leading high-value proposal development. Her client-centric mindset, backed by deep technical expertise, ensures consistently successful submissions and long-term regulatory success.​

Other Related Articles

View All

MDR Made Easy: How Celegence Helps MedTech Innovators Stay FDA-Compliant

MDR Made Easy: How Celegence Helps MedTech Innovators Stay FDA-Compliant

19 Aug, 2025

In today’s fast-moving MedTech world, regulatory missteps aren’t just setbacks—they’re risks to patient safety, product reputation, and market success. With the FDA’s Medical Device Reporting (MDR) requirements becoming more complex, staying compliant is no longer optional; it’s a strategic imperative.

At Celegence, we help medical device manufacturers turn MDR compliance into a competitive advantage—with expert-driven strategies, streamlined systems, and scalable support tailored to your organization’s needs.

Why FDA MDR Compliance Can’t Be an Afterthought

If you’re marketing a device in the U.S., FDA MDR (21 CFR Part 803) requires you to report adverse events, serious injuries, deaths, and certain malfunctions quickly and accurately. These reports provide critical post-market surveillance data that help regulators monitor device safety and protect patients.

Non-compliance comes with serious consequences, including:

  • FDA warning letters that can disrupt your operations
  • Civil monetary penalties that can escalate rapidly
  • Costly recalls and associated logistics, legal, and reputational fallout
  • Loss of market trust that impacts both patients and healthcare providers

For medical device manufacturers, the real cost isn’t just regulatory—it’s also the risk of losing credibility in a market where safety and reliability drive purchasing decisions.

The key to staying ahead lies in your systems, training, and strategic partnerships that ensure compliance is embedded in your operations from day one.

Celegence: Your Partner in Proactive MDR Compliance

Whether you’re launching a new device or scaling across markets, Celegence provides tailored MDR support that meets you where you are—and takes you where you need to go. We combine regulatory expertise, process optimization, and advanced technology to create MDR systems that are reliable, repeatable, and ready for audits.

Here’s how we help you build a future-proof MDR compliance strategy:

  • Adverse Event Reporting Strategy – Expertly structured workflows to ensure you meet reporting deadlines without errors or omissions.
  • Gap Assessments – Identify risk points in your current process before the FDA does, reducing exposure.
  • QMS & Complaint Handling Integration – MDR seamlessly embedded into your Quality Management System for smoother operations.
  • Audit-Ready Documentation – Organized, consistent, and accessible records that withstand regulator scrutiny.
  • Team Training & Change Management – Equip teams with up-to-date regulatory knowledge and practical tools to execute confidently.
  • Global PMS Alignment – Align FDA MDR processes with EU Vigilance and other international post-market requirements for global compliance consistency.
  • Cross-Functional Communication Enablement – Facilitate seamless collaboration across quality, regulatory, clinical, and technical functions, as well as distributors and affiliates worldwide.

By embedding MDR requirements into every stage of your product lifecycle, we help you protect patients while safeguarding your business objectives.

Common MDR Issues (And How We Help You Avoid Them)

Many manufacturers struggle with MDR implementation because compliance isn’t just about reporting—it’s about building the right systems and culture. Common issues include:

  • Misclassifying reportable events leading to under-reporting or over-reporting
  • Annual entry errors in Form 3500A submissions that create inconsistencies
  • Inconsistent timelines and failure to meet the 5- or 30-day reporting windows
  • Missing documentation that weakens audit readiness
  • Poor SaMD (Software as a Medical Device) traceability for patches and upgrades
  • Disconnected QMS and complaint handling processes that cause reporting delays

Celegence closes these gaps by bringing together people, processes, and technology. Our experts not only interpret regulations but also translate them into practical systems your teams can execute.

MDR & SaMD: What You Don’t Know Can Hurt You

Software-driven devices bring unique reporting challenges. The FDA continues to refine expectations for SaMD, particularly as cybersecurity risks and human factors gain attention. Issues such as software glitches, corrupted data, or confusing user interfaces can directly impact patient safety and fall under MDR requirements.

We help you strengthen your SaMD compliance by:

  • Mapping software risks to MDR reporting requirements to ensure no gaps
  • Building traceability for patches and version control so every change is documented
  • Identifying cybersecurity vulnerabilities that may trigger mandatory reporting
  • Training teams on red flags unique to software such as UI misuse, algorithm errors, or integration failures

By addressing SaMD-specific risks, we enable you to stay compliant while keeping pace with fast-moving software updates.

MDR Compliance That Powers Better Products

A strong MDR program doesn’t just keep regulators satisfied—it strengthens your entire product lifecycle. Proper reporting and data analysis fuel better design, smarter risk management, and continuous product improvement.

With Celegence, you can:

  • Leverage MDR data to detect trends, usability issues, and recurring malfunctions early
  • Strengthen ISO 13485 and ISO 14971 alignment by integrating MDR into risk and quality frameworks
  • Drive continuous improvement across your portfolio, reducing the likelihood of repeat issues
  • Improve customer trust by demonstrating a proactive commitment to safety and compliance

In short, MDR compliance becomes not just an obligation but a driver of innovation and market success.

Building Confidence in FDA MDR Compliance

MDR compliance is more than ticking regulatory boxes—it’s about protecting patients, building trust, and enabling growth. With FDA requirements under constant scrutiny and global markets demanding alignment, medical device companies cannot afford to take chances.

At Celegence, we partner with MedTech innovators to turn compliance into a strategic advantage. By combining expert guidance, proven systems, and scalable technology, we help you achieve confidence in MDR compliance—today and in the future.

Contact us at info@celegence.com to learn how our expert services and solutions can strengthen your reporting processes, improve audit readiness, and ensure ongoing compliance.

AUTHORED BY

author-image

Medical Writer II

Tamana Sahraya

Linkdin-image

Tamanna Sahraya is a skilled medical writer specializing in regulatory documentation for medical devices, with expertise in FDA and EU MDR compliance. She has experience authoring Clinical Evaluation Reports (CER), Post-Market Surveillance Reports (PMSR), and Risk-Benefit Assessments while ensuring adherence to MEDDEV 2.7/1 Rev. 4 and MDR standards. With a strong background in regulatory affairs, literature reviews, and safety analysis, she supports product lifecycle management and clinical data reporting. Her analytical approach and commitment to compliance help streamline regulatory submissions and enhance client collaboration.

Other Related Articles

View All

How the Integrated Search Workflow in CAPTIS® Is Transforming Literature Reviews for Regulatory Teams

How the Integrated Search Workflow in CAPTIS® Is Transforming Literature Reviews for Regulatory Teams

13 Aug, 2025

At Celegence, we know that efficiency, consistency, and traceability are critical when preparing regulatory documents like Clinical Evaluation Reports (CERs). That’s why we’ve adopted and optimized the Integrated Database Search workflow in CAPTIS®, our go-to solution for systematic literature reviews.

This powerful feature allows our medical writers to search PubMed, Europe PMC, and Google Scholar directly within the platform, replacing the need for manually compiling article data in Excel. By eliminating export-import steps and disconnected tracking, it makes literature reviews significantly faster, cleaner, and fully audit-ready.

These integrated searches were among the first features we prioritized when designing our automated literature workflow, because anyone who has reviewed from spreadsheets knows how time-consuming it is to collate article data, especially from sources like Google Scholar. In fact, many manufacturers still outsource this step because of how tedious and error-prone it can be.

What Makes It So Effective?

  • All-in-One Search Access

    Our writers can search across major scientific databases (PubMed, EU PMC, Google Scholar) without leaving the platform. That means fewer clicks, less toggling, and no time wasted.

  • Smart Review Alignment

    Each search can be clearly tagged as part of either the DUE (Device Under Evaluation) or SOTA (State of the Art) workflow. This ensures that literature stays aligned with project goals and regulatory requirements from the start.

  • Built-in Relevance Indicators

    To quickly assess search quality, our teams use positive and negative keywords—a simple but powerful way to check if a search is pulling in the right kind of articles. It helps us know instantly whether a query needs fine-tuning, without wasting review time.

     

  • Audit-Ready Traceability

    Naming searches and maintaining search categories ensures long-term traceability — a huge benefit during regulatory audits or CER updates 2–5 years down the line. This also helps teams align on the overall literature review objectives in the project.

  • Search Editing Without Rework

    Need to tweak your search terms or expand a date range mid-project? No problem. You can edit a saved search, rerun it, and retain all previous screening decisions for overlapping articles. No duplication. No data loss.

  • Auto-Update for Long-Term Projects

    With Auto-Update enabled, the system will automatically refresh your search results weekly or monthly, capturing new publications as they appear — ideal for projects with rolling deadlines or submissions scheduled months ahead.

  • Rapid Reuse Across Projects

    Project cloning combined with search editing lets our team quickly reuse earlier searches — whether it’s extending the date range for a maintenance CER, swapping out key terms to adapt the search for a different device, or reusing the same SOTA strategy across multiple devices. It saves time, ensures consistency, and reduces rework across projects.

 

“We’ve seen measurable time savings and a more focused authoring and review process since adopting this workflow. Our literature teams are not only working faster and more efficiently, but their output is also far less error-prone and more consistently aligned with regulatory expectations. This ultimately strengthens the quality and reliability of our submissions.”

— Dr. Pratibha Mishra, Senior Manager and SME, Medical Device Services

Ready to Work Smarter With Literature Reviews?

This is just one example of how we’ve optimized regulatory work using CAPTIS®. If your teams handle CERs, PERs, or other evidence-based submissions, the right technology can dramatically simplify your workflows.

Get in touch  @ info@celegence.com to see how we can help you scale literature reviews without scaling your headcount.

 

AUTHORED BY

author-image

Technical Account Manager

Shruti Sharma

Linkdin-image

Shruti Sharma is a Technical Account Manager at Celegence with a deep background in regulatory medical writing, specializing in EU MDR-compliant clinical evaluations for medical devices. Drawing on her extensive knowledge of regulatory requirements, she translates industry needs into innovative software solutions that enhance efficiency and compliance in the Life Sciences regulatory space. Shruti is passionate about making technology accessible and impactful, championing tech-enabled process.

Other Related Articles

View All

Staying Compliant: Strategies for EU MDR and Beyond.

Staying Compliant: Strategies for EU MDR and Beyond.

05 Aug, 2025

The regulatory environment for medical devices continues to evolve at a rapid pace. With increasingly stringent requirements under the EU MDR, specific expectations in global markets, frequent updates to regulatory standards, and the complexity of high-risk device portfolios, manufacturers are facing greater challenges than ever in aligning their compliance strategies.

Our recent webinar brought together expert insights on how manufacturers can strategically navigate the evolving regulatory landscape by strengthening their clinical evaluation processes, maintaining audit readiness, leveraging digital tools such as CAPTIS®, and embracing future-focused regulatory foresight.

The Foundation: Understanding the Impact of Regulatory Divergence

Regulatory divergence refers to the differences in medical device requirements and approval processes across countries and regions. These variations impact product development, market access, clinical testing, and launch timelines. Understanding and managing these differences is key to ensuring compliance, accelerating time-to-market, and staying competitive globally.

The Role of Clinical Evaluation in Compliance

A well-defined clinical strategy is indispensable for meeting the diverse regulatory requirements worldwide. Clinical evaluation ensures evidence is purposeful, aligned with the device’s intended use, and effectively supports safety and performance claims. Clinical evaluation plays a crucial role in maintaining ongoing compliance by:

  • Supporting successful Notified Body audits
  • Reinforcing alignment to evolving regulatory standards
  • Mitigating risks of delays or non-conformities.

Leveraging Technology to Drive Efficiency and Accuracy

Technology integration is a strategic advantage for achieving regulatory compliance and audit readiness. Solutions such as Celegence’s CAPTIS® leverage automation, AI, and structured content management to streamline documentation and regulatory workflows.

These digital tools offer:

  • Improved Data Management: Better version control and organization reduce errors and duplication of effort
  • Process Automation: Automating repetitive tasks speeds up document creation and review
  • AI-Driven Evidence Analysis: AI can sift through vast volumes of clinical literature, extracting relevant data accurately and efficiently—a task that is particularly valuable for high-risk devices with complex evidence requirements.

While AI accelerates processes, experienced regulatory professionals remain essential for oversight, quality assurance, and final decision-making within a human-in-the-loop model.

Audit Readiness

Audit readiness is often viewed as a challenging but essential pillar for compliance throughout the medical device lifecycle. A structured, proactive approach can ensure manufacturers are prepared for both scheduled and unannounced regulatory inspections. Key components include:

  • Pre-Submission Consultation: Consulting with notified bodies and regulatory experts early helps identify documentation gaps and align with regulatory requirements, reducing submission delays or rejections
  • Comprehensive Documentation: Maintaining organized, complete, and up-to-date technical files, clinical evaluation reports (CERs), risk management files, and post-market surveillance plans is critical. These documents must be audit-ready—formatted and accessible to facilitate efficient review
  • Team Training and Awareness: Everyone involved, from regulatory affairs to quality assurance, technical and clinical teams, must be well-trained in relevant regulatory requirements and their specific roles during the audits. Familiarity with internal procedures and confident responses to auditors’ inquiries strengthen audit readiness
  • Traceable Evidence: Demonstrating compliance requires clear, traceable documentation aligned with regulatory standards, including clinical data, conformity assessments, and risk-benefit analyses
  • Communication and Logistics: Planning the audit schedule, ensuring key personnel availability, preparing facilities, and establishing audit response protocols all contribute towards smooth inspections
  • Mock Audits: Conducting internal or third-party mock audits acts as a rehearsal, revealing gaps in documentation or processes and enabling corrective actions to be taken ahead of formal audits.

Webinar Highlights – Strategies for EU MDR & Beyond

Future-Proofing Compliance: Regulatory Foresight

The regulatory landscape is dynamic, with frequent updates and evolving expectations—especially in medical devices and digital health sectors. Staying compliant requires a proactive approach which involves not just reacting to changes but anticipating them.

Manufacturers who embed regulatory foresight into their strategies can:

  • Innovate confidently
  • Respond swiftly to regulatory changes
  • Maintain trust with Notified Bodies and end users.

Proactive monitoring of guidance documents and standards, coupled with early integration of regulatory changes, reduces compliance risks and positions manufacturers for long-term success.

Key Takeaways

  • Harmonized and comprehensive EU MDR documentation can be leveraged for other regulatory submissions with minimal modifications, improving operational efficiency and reducing compliance costs
  • Technology platforms, including AI tools, significantly reduce manual effort and increase accuracy in documentation and evidence analysis
  • Human expertise remains essential to interpret AI-generated outputs and make informed regulatory decisions
  • Audit readiness is fundamental for seamless Notified Body audits and maintaining regulatory compliance
  • Clinical evaluation is the compass guiding compliance efforts and ensuring patient safety
  • Regulatory foresight is a necessity in a rapidly evolving environment to sustain market presence and innovation.
Final Thought

Compliance is not a one-time occurrence but an ongoing journey. Robust clinical evaluation combined with strategic use of technology and a future-focused mindset equips manufacturers to navigate this journey with greater confidence.

As echoed in our webinar:
Compliance is a journey, and robust clinical evaluation is your compass.”

By adopting these principles, medical device manufacturers can meet regulatory expectations, enhance operational efficiency, and most importantly, ensure patient safety.

AUTHORED BY

author-image

Manager, Medical Device Services

Dr. Anushree Singh

Linkdin-image

Dr. Anushree Singh has a Doctorate degree in Neuro-oncology and a Master’s degree in Bioscience (Human Genetics). She has 13 years’ experience in the Life Sciences Industry including 5 years’ experience in Regulatory Writing for Medical Devices. She has authored and reviewed EU MDR compliant regulatory documents for medical devices covering a wide range of therapeutic areas. At Celegence, she provides strategic advice to customers on regulatory strategy for clinical evaluation, Post-Market Surveillance (PMS) and Clinical Evidence pathways. She leads a team of qualified medical writers, expert in clinical evaluation of simple-to-complex medical devices, authoring various regulatory documents.

Other Related Articles

View All

Adverse Event Reporting for Medical Devices: Global Regulations, Timelines, and Compliance Insights

Adverse Event Reporting for Medical Devices: Global Regulations, Timelines, and Compliance Insights

09 Jun, 2025

What is Adverse Event Reporting (AER)?

The process of Adverse Event Reporting (AER) involves the systematic recording and submission of information about adverse events that occur during the use of medical products (drugs, devices and vaccines) Such reports contribute significantly to the surveillance of product safety by regulatory agencies, allowing for prompt action when needed to ensure public health and safety.

Importance of Reporting in Medical Devices

Adverse events are any unexpected, harmful or untoward medical occurrence related to the use of the medical product in humans not necessarily caused by the product. Reporting of adverse events in medical devices is a critical part of post-market surveillance aimed at ensuring the continued safety and performance of medical devices after their use. This includes the identification, documentation and reporting of incidents in which the device may have caused or contributed to injury, death or malfunction. Adverse events can be reported by manufacturers, importers, distributors, healthcare providers and patients/consumers.

Reporting Requirements and Key Information

Reporting requirements for adverse events (AEs) vary according to jurisdiction, type of organisation and severity of the event. Serious incidents causing death, serious injury or deterioration of health; requiring medical or surgical intervention or causing permanent impairment should be reported. A recurring device malfunction which has the potential to cause serious harm should be reported. The regulatory authorities usually require information related to reporter, device, date of occurrence, description of the events, outcome and severity. If the reporter is a manufacturer, details related to corrective and preventive actions, recalls, field safety corrective actions, if any are also required. when a medical device adverse event report is submitted. The objective is to present a comprehensive and unambiguous image of the event, device, and patient outcome.

Global Regulatory Frameworks and Reporting Systems

Every nation has set up a different regulatory framework with distinct reporting procedures, specifications, and deadlines for tracking and handling incidents involving medical devices. Manufacturers, medical professionals, and regulatory affairs specialists working in a global market must comprehend these distinctions.

United States – FDA

The Food and Drug Administration (FDA) is in charge of regulating medical devices in the US. The Manufacturer and User Facility Device Experience (MAUDE) database and the MedWatch application are the main reporting platforms. Adverse events must be reported by manufacturers, importers, and device user facilities in accordance with the Medical Device Reporting (MDR) regulation (21 CFR Part 803). The eMDR system must be used to electronically submit reports. The FDA mandates that events requiring immediate corrective action be reported within five workdays, and that serious injuries or deaths be reported within thirty calendar days. The FDA can respond promptly to safety communications and recalls thanks to this strong system. The TPLC and MAUDE database searches are integrated in CAPTIS® platform of Celegence. It enables the medical writers to seamlessly conduct database searches and analysis of the search results to obtain quality outcomes.

European Union – EUDAMED

The Medical Device Regulation (EU) 2017/745, which requires the reporting of serious incidents and field safety corrective actions, governs medical devices in the European Union. EUDAMED (European Database on Medical Devices) is the EU’s central database for medical device vigilance. Incidents must be reported by importers, manufacturers, and authorized representatives; patients and healthcare providers may voluntarily report through the appropriate national authorities. The deadlines are strict: death or a major decline in health must be reported within two days, major public health threats must be reported right away, and other serious incidents must be reported within ten to fifteen days.

United Kingdom – MHRA

The Medicines and Healthcare Products Regulatory Agency (MHRA) was the regulatory pathway that the United Kingdom established. Systems like the Manufacturer’s Online Reporting Environment (MORE) and the Yellow Card Scheme are still used in the UK to report adverse events. Within a certain amount of time—two days for public health threats, ten days for fatalities or serious injuries, and fifteen days for other reportable incidents—manufacturers and their UK Responsible Persons are required to report serious incidents.

Canada – Health Canada

Health Canada is in charge of medical device safety in Canada through the Medical Device Problem Report system and the Canada Vigilance Program. According to the law, distributors, importers, and manufacturers must report unfavorable incidents. Timelines for reporting incidents are set at 30 days for other incidents and 10 days for events that pose a serious risk. Reports may be submitted via specified forms or electronically.

Australia – TGA

The Incident Reporting and Investigation Scheme (IRIS) is run by the Therapeutic Goods Administration (TGA), Australia’s regulatory body. According to Australian laws, healthcare providers may choose to report adverse events voluntarily, but sponsors—including importers and manufacturers—must report them. According to severity, the reporting deadlines are tier-based: two days for major public health threats, ten days for major injuries or fatalities, and thirty days for less important incidents.

Japan – MHLW and PMDA

Medical device vigilance in Japan is a joint responsibility of the Ministry of Health, Labor, and Welfare (MHLW) and the Pharmaceuticals and Medical Devices Agency (PMDA). Marketing authorization holders can report more easily thanks to the J-MEDWATCH system. If an adverse event is considered serious, it must be reported within 15 days; for less serious incidents, reporting must be done on a regular basis. The highly structured regulatory environment in Japan places a strong emphasis on traceability and documentation.

China – NMPA

Adverse event reporting in China is regulated by the National Medical Products Administration (NMPA) via a national electronic monitoring system. Manufacturers, holders of marketing authorizations, and healthcare facilities are required to report. For other kinds of incidents, regular safety updates are necessary, and serious adverse events must be reported within 15 days. In recent years, China’s regulatory framework has undergone significant modernization, bringing it closer to international norms.

India – CDSCO

The Materiovigilance Programme of India (MvPI) is run by the Central Drugs Standard Control Organization (CDSCO), India’s regulatory body, to keep an eye on adverse events linked to medical devices. While hospitals and medical professionals voluntarily participate through Adverse Drug Reaction Monitoring Centers (AMCs), manufacturers and importers are obligated to report incidents. India strives to conform to international reporting standards, despite the fact that timelines are not as clearly stated as in other nations.

Global Challenges in Adverse Event Reporting

A crucial but difficult part of international health systems is the reporting of adverse events for medical devices. A number of factors, including underreporting, inconsistent regulatory frameworks, divided responsibilities, technological constraints, and cultural considerations, contribute to the complexity. A diversified strategy is needed to address these issues, including coordinating global standards, making investments in digital infrastructure, encouraging a transparent and safe culture, and utilizing advanced analytics to improve signal detection.

The Role of Technology in Adverse Event Management

The application of advanced technologies, such as Artificial Intelligence (AI) and Machine Learning, is being investigated for their potential to augment signal identification and pattern recognition within extensive AE (Adverse Event) databases. Furthermore, the fusion of AE documentation with Electronic Health Records (EHRs) can automate data acquisition, alleviate the workload of clinicians, and enhance the precision of the data.

Toward Safer and More Reliable Medical Devices

In conclusion, medical device adverse event reporting is essential to patient safety and preserving the integrity of healthcare systems. It facilitates prompt regulatory actions and ongoing product development by gathering real-world data on device performance and related risks. Its efficacy is hampered by issues like underreporting, disjointed regulatory frameworks, and poor technological integration. In order to get past these obstacles, manufacturers, medical professionals, regulators, and patients must work together. Post-market surveillance will be strengthened and everyone will eventually have access to safer and more dependable medical devices thanks to the development of digital reporting systems, the standardization of international standards, and the promotion of an open culture.

Contact us today at info@celegence.com to learn how we can help your team with global adverse event reporting and strengthen your medical device compliance strategy.

AUTHORED BY

author-image

Associate Manager, Medical Device Services

Deblina Rababi

Linkdin-image

Deblina Rababi is an experienced regulatory professional specializing in medical device compliance under EU MDR. She has expertise in authoring and reviewing Clinical Evaluation Reports (CER), Clinical Evaluation Plans (CEP), Literature Search Reports (LSR), Periodic Safety Update Reports (PSUR), Post-Market Surveillance Reports (PMSR), and Post-Market Clinical Follow-up (PMCF) documentation. As an Associate Manager at Celegence, she ensures the development of high-quality regulatory submissions, supporting medical device manufacturers in achieving compliance. Her detail-oriented approach and regulatory expertise contribute to streamlined documentation and successful audits.

Other Related Articles

View All

Case Study: How a Global MedTech Firm Streamlined Regulatory Writing with CAPTIS® & Celegence

Case Study: How a Global MedTech Firm Streamlined Regulatory Writing with CAPTIS® & Celegence

26 May, 2025

Executive Summary

Regulatory documentation projects often struggle with version control issues, manual processes, and fragmented review cycles. For one global medical device manufacturer, these pain points were slowing down their ability to deliver high-quality, timely submissions. By partnering with our expert medical writing team and using our AI-powered software platform CAPTIS®, they transformed the way they draft, review, and finalize regulatory reports. The result: faster turnaround times, reduced errors, and a more collaborative experience.

The Challenge

Before working with us, the client relied on traditional drafting workflows using Microsoft Word, shared over email. With multiple writers and reviewers involved, the process was fraught with:

  • Frequent document crashes
  • Formatting inconsistencies across Word versions
  • Time-consuming manual updates of citations, abbreviations, and references
  • Confusion from version mismanagement
  • Delayed feedback due to sequential (not parallel) review workflows

Despite having competent internal reviewers, they lacked the tools and processes to streamline document development. Collaboration was siloed, and reviews were often delayed or duplicated.

Our Solution

By combining our experienced medical writing team with our proprietary software CAPTIS®, Celegence helped the client transition to a faster, structured, and AI-augmented documentation process.
Key workflow upgrades included:

  • Centralized Document Management: All literature, documents, source files, and review versions live in one place—with real-time version control.
  • Real-Time Collaboration: Multiple users can draft or review simultaneously, without crashes or version confusion.
  • Automated Formatting & Styling: Templates ensure consistency, removing hours of manual formatting.
  • Integrated Data Dictionary: Updates to a term auto-propagate across all documents, ensuring consistent language and definitions.
  • Abbreviation Table Automation: Abbreviations and their full forms are auto populated, improving clarity and compliance.
  • Citations and Reference Management: Citations are easy to create and the bibliography inserted automatically, with a structured reference pack compiled for each report. Reviewers can open PDFs of references with just a click.

Figure 1: One-click access to reference PDFs directly from the document for seamless verification during review.

 

  • Smart Navigation via Content Links: Built-in navigation and content links make jumping between document sections and referenced documents seamless.

Figure 2: Seamless navigation through clickable content links that connect document sections and referenced source information

  • AI-Assisted Evidence Gathering: Our built-in Copilot offers literature content summarization and Q&A capabilities for faster insights and validation.

Figure 3: AI-Assisted Evidence Gathering

 

  • Review Stage Controls: Users can tag team members, lock sections, and stage reviews, creating transparency and accountability.

Figure 4: View designated reviewers for each review stage

  • Automated Review Initiation Notifications: All reviewers are automatically notified when their designated review stage begins, eliminating manual follow-ups and delays.

Figure 5: Automated review notification alert sent to designated reviewers at the start of their review stage

  • Review Progress Tracking: Reviewers can instantly see which sections they’ve reviewed and what’s still pending, making it easy to pick up where they left off.

Figure 6: Clearly see which sections are reviewed and which are pending. Navigate effortlessly using the section panel

  • Reviewer Approvals Visibility: Client reviewers can quickly view which sections have been approved by other reviewers to avoid duplication and streamline collaboration.

Figure 7: Easily track approved sections to enhance collaboration and prevent duplication

  • Integrated Dashboard: A powerful dashboard allows the client to track the progress of multiple projects in real-time, with clear indicators of where each document stood in the drafting and review cycle, and a quick look at which reviewers had started or completed their tasks.

Figure 8: Track real-time progress across projects with a clear view of drafting stages, review status, and writer/reviewer activity—all in one place

Outcomes and Benefits

Since implementing CAPTIS and collaborating with Celegence’s regulatory medical writing team, the client has experienced measurable improvements:

  1. Faster Turnaround Times: Document drafting and review cycles were significantly shortened through real-time collaboration, automation, and AI support—enabling quicker report finalization and submission.
  2. Full Workflow Transparency & Control: An integrated dashboard and structured review stages gave the client complete visibility into document progress, reviewer status, and project timelines across multiple reports.
  3. Streamlined Collaboration & Reviews: Features like tagging, comments, shared access, and reviewer-specific views reduced communication gaps, avoided duplication, and made it easy for teams to coordinate and review efficiently.
  4. Greater Consistency & Compliance: Centralized document storage, an auto-updating data dictionary, citation tools, and version control ensured accurate, consistent, and audit-ready content across all deliverables.
  5. Smart Automation & AI Assistance: Built-in tools for abbreviation tables, reference management, review stage notifications, and AI-driven literature summarization and Q&A significantly reduced manual work and improved productivity.
  6. Flexibility & Scalability: The system adapted to project changes—like updated terminology or literature search strategies—without disrupting ongoing work, ensuring smooth execution even in dynamic regulatory environments.

Conclusion

Working with Celegence isn’t just about regulatory expertise—it’s about transforming how regulatory documents are created and reviewed. By pairing our deep domain knowledge with a powerful literature review and documentation platform, we helped a major MedTech player gain efficiency, transparency, and confidence in their submissions.

Want to streamline your next regulatory writing project?

Work with us to get not just expert writers – but a smarter, faster way to collaborate.

Other Related Articles

View All

Drug-Led vs. Device-Led Combination Products: Determining the Primary Mode of Action (PMOA)

Drug-Led vs. Device-Led Combination Products: Determining the Primary Mode of Action (PMOA)

20 May, 2025

Once a product is confirmed as a combination product, the next step is identifying its Primary Mode of Action (PMOA). This is defined as the single mode of action that provides the most important therapeutic effect of the product.

If the product’s intended therapeutic benefit is primarily achieved through a chemical or metabolic interaction, then it is typically drug-led. If the effect is driven by physical means, such as mechanical support or energy transmission, it is considered device-led.

The PMOA determines which FDA center will lead the regulatory review. For example:

  • Drug-led products are generally reviewed by the Center for Drug Evaluation and Research (CDER) or the Center for Biologics Evaluation and Research (CBER).
  • Device-led products fall under the purview of the Center for Devices and Radiological Health (CDRH).

Understanding this distinction is crucial because it dictates the regulatory submission pathway, data requirements, and even post-approval compliance expectations.

Comparing Drug-Led and Device-Led Pathways

To illustrate the regulatory differences, the table below outlines key contrasts between the two classifications:

Feature  Drug-Led Combination Product  Device-Led Combination Product 
Primary Mode of Action (PMOA)  Chemical or metabolic action  Physical or mechanical action 
Lead FDA Center  CDER or CBER  CDRH 
Submission Pathway  IND → NDA/BLA  Q-Sub → IDE (if needed) → 510(k), De Novo, or PMA 
Regulatory Focus  Drug efficacy, safety, pharmacology  Device performance, engineering, standards 
Example  Prefilled syringe of a monoclonal antibody  Drug-eluting coronary stent 



In cases where the PMOA is not obvious, the FDA may determine the lead center based on precedent or safety and effectiveness concerns. This is why early interaction with the FDA is strongly encouraged.

Gray Areas and Borderline Scenarios

Innovative therapies don’t always fit neatly into predefined categories. Consider, for example, a therapeutic powder that forms a gel upon hydration and controls the release of a drug physically, without any chemical reaction. While it may appear to be a simple formulation, the polymer mechanism may qualify as a device component. In such borderline cases, even the excipient could be classified as a regulated device.

Another emerging gray area involves digital health integrations. Take the example of an antipsychotic pill embedded with an ingestible sensor to track adherence. Although the drug provides the treatment, the device aspect introduces complexity in classification. FDA treated this as a drug-led combination product with additional review of the device software and hardware elements.

Sponsors dealing with similar situations should not assume a default classification. Instead, they should seek clarity from the FDA early, using the pathways discussed below.

Engaging the FDA: RFD and Pre-Submission Pathways

To avoid ambiguity and regulatory missteps, the FDA offers sponsors two main avenues for determining classification:

  1. Request for Designation (RFD)
    The RFD is a formal process through which the sponsor asks the FDA’s Office of Combination Products (OCP) to determine whether the product is a combination product, and if so, which center will lead the review. The FDA is required to respond within 60 days, providing a binding decision.
  2. Pre-Request for Designation (Pre-RFD)
    This is a less formal, non-binding method to engage FDA on classification questions. Sponsors can initiate Pre-RFD discussions to explore how the agency views their product, receive preliminary input, and prepare more confidently for the full RFD if needed.Engaging FDA through these channels early in development ensures that sponsors follow the correct regulatory path and gather the appropriate data from the start.

Developing a Regulatory Strategy

After classification is determined, sponsors must design a development strategy that aligns with the assigned pathway.

For drug-led products, this typically involves:

  • Filing an Investigational New Drug (IND) application.
  • Conducting clinical studies focused on safety and efficacy.
  • Submitting a New Drug Application (NDA) or Biologics License Application (BLA).
  • Demonstrating that any device component (e.g., injector, delivery system) is safe, functional, and appropriate for the intended user population.

For device-led products, development may involve:

  • Submitting a Pre-Submission (Q-Sub) for FDA feedback.
  • Filing an Investigational Device Exemption (IDE) if clinical studies are needed.
  • Preparing a 510(k), De Novo, or Premarket Approval (PMA) submission.
  • Including supporting drug data to ensure safety of the drug component, even if secondary.

Sponsors must also comply with hybrid quality systems, combining drug GMP (21 CFR Parts 210/211) and device QSR (21 CFR Part 820). The FDA has published 21 CFR Part 4 to provide guidance on integrating these systems for combination product development.

Recent Regulatory Updates (2023–2025)

In the past few years, FDA has introduced several new guidances that significantly impact combination product development:

  • Combination Product User Fees (2024): Clarifies which user fees apply when submitting a single combined application for a combination product.
  • Use-Related Risk Analysis (2024, Draft): Encourages risk assessments tied to user interaction—especially important for auto-injectors and at-home devices.
  • Human Factors Engineering (2023): Offers a Q&A-style guide for incorporating usability studies in combination product submissions.
  • Drug-Device Software (2023, Draft): Discusses regulatory expectations for mobile apps, sensors, and digital interfaces that form part of a combination product.
  • Premarket Pathways for Combination Products (2022): Affirms that most combination products can follow a single submission path, and outlines principles for determining when more than one submission may be needed.

Each of these documents provides valuable insight into FDA expectations, helping sponsors avoid roadblocks and design programs that meet current regulatory standards.

Key Takeaways for Developers in 2025

Combination product development is a multidisciplinary challenge, requiring strategic foresight and active regulatory engagement. Here are four key takeaways for sponsors:

  • Start with clear classification: Use 21 CFR 3.2(e) and, when in doubt, engage the FDA early via Pre-RFD or RFD.
  • Build your development strategy based on PMOA: Whether your product is drug-led or device-led, understand how that affects submission type, testing, and timelines.
  • Address both components: Ensure your development plan includes appropriate testing for both drug and device aspects—neither can be neglected.
  • Stay current with FDA guidance: Regulatory expectations are evolving. Human factors, software integration, and use-risk analysis are now central considerations in many combination product reviews.

With a proactive and informed approach, sponsors can reduce regulatory uncertainty, improve submission quality, and bring combination therapies to patients faster.

Celegence offers comprehensive regulatory consulting services tailored for combination products, providing strategic guidance on classification, regulatory submissions, and compliance with both drug and device regulations. Their expert guidance helps streamline development processes, mitigate risks, and facilitate timely market entry for complex combination therapies.

Other Related Articles

View All

Beyond Europe: The Strategic Value of CERs in FDA Submissions

Beyond Europe: The Strategic Value of CERs in FDA Submissions

14 May, 2025

Clinical Evaluation Reports (CERs) are often associated with EU MDR, but an increasing number of manufacturers are now integrating CERs into their FDA submissions, particularly for devices requiring robust clinical evidence. This approach allows manufacturers to present a clear, comprehensive clinical evidence package, ensuring alignment with FDA expectations and reducing review timelines.

CERs in the Context of FDA Submissions

Unlike the EU MDR, the FDA does not mandate a standalone CER as part of a 510(k) or PMA submission. However, substantiating safety and performance of a device through clinical data is a fundamental expectation of the FDA. Clinical evidence plays a pivotal role in FDA review, especially for:

  • Novel devices without predicates
  • De novo applications
  • Devices with high-risk profile
  • PMA supplements requiring new safety data

What to Watch For: Common Pitfalls

  1. Inadequate Clinical Data
    An unsystematic literature review may result in inclusion of irrelevant or inadequately appraised clinical data, or poorly analyzed data. Additionally, relying on EU-based data without appropriate consideration of its relevance to the U.S. clinical context can further weaken the submission.
  2. Lack of Understanding of Submission Types
    Submitting the wrong type of application may lead to significant delays or rejection. Failing to understand the specific requirements associated with the chosen submission type can lead to missing critical information.
  3. Unsubstantiated Clinical Claims
    If the clinical data does not directly support the intended use or labelling of the device, it may lead to questions from the reviewer or delays. Each safety and performance claim should be clearly substantiated with traceable evidence.
  4. Underutilization of Real-World Evidence (RWE)
    Adverse event trends, registry data, and post-market studies provide real-world evidence that can further enhance the risk-benefit assessments. Not leveraging this data is a missed opportunity to strengthen the submission.
  5. Lack of integration across submission sections
    Lack of alignment between clinical, risk, and labeling sections can make it difficult for reviewers to interpret the submission. Maintaining consistency across all sections is essential.

Considerations for a CER aimed at FDA Review

Although CERs are not mandated by FDA, manufacturers should consider the following when drafting a CER to support regulatory submissions:

  • Discussion of data relevance, especially for U.S. population
  • Clear summary of the clinical evidence, including published literature, registry data, and clinical trials
  • Analysis of risks, adverse events, and benefits in alignment with device labeling
  • Refer to FDA-recognized standards and guidance, like 21 CFR 814 or the Clinical Evidence Premarket Submission guidance

Useful Tools & Resources

Developing CER content for FDA submissions can be time-consuming, particularly when teams are handling multiple submissions or working under tight timelines. The following tools and data sources can help streamline the process:

  • FDA MAUDE Database – for adverse event data and comparator analysis
  • ClinicalTrials.gov – to identify relevant ongoing and completed clinical trials
  • PubMed and Embase – to identify relevant peer-reviewed clinical literature
  • FDA Guidance Documents – to align the CER content with FDA expectations
  • FDA CDRH Learn – to stay updated on submission requirements and review trends

These resources are most effective when used strategically, supported by internal alignment and medical writing expertise.

How Celegence Supports CER Preparation for FDA Submissions

Preparing a CER or clinical justification for FDA review isn’t just about data gathering. It’s about crafting a clear, well-structured narrative supported by robust evidence. This is where Celegence adds value.

We combine regulatory expertise with technology to deliver submission-ready CERs, literature reviews, and clinical summaries—on time and in line with FDA expectations.

What sets us apart?

  • Clinical writers experienced in FDA and EU submissions
  • AI-powered literature reviews and data extraction using CAPTIS®
  • Integration of FDA databases and guidance documents into writing workflows
  • Quick turnaround times for 510(k), PMA, and De novo pathways
  • 99% first-time acceptance rate of clinical documents by regulatory reviewers

Whether you need a literature-based justification, a full CER-style report, or gap analysis of your existing documentation—we offer flexible solutions for every stage of development and submission.

Let’s Work Together

Need help aligning your clinical evidence with FDA expectations? Celegence delivers accurate, regulatory-ready CER support backed by proven expertise and AI tools. Contact our team today to get started.

Other Related Articles

View All