IVDR CE marking NGS: MDx Case Study with Fulgent

At a glance

  • Outcome: CE mark granted by TÜV SÜD for an end-to-end Class C germline NGS solution (wet lab + bioinformatics).
  • Scope: Furthermore, panel covering 4,600+ clinically relevant genes with a validated PLM (Pipeline Manager) software component; later expanded to >7,000 genes using a new probe set.
  • What we did: Specifically, we built an ISO 13485 QMS from the ground up, prepared full Annex II + III technical documentation, validated bioinformatics under IEC 62304/82304, split documentation into two Basic UDI-DIs (wet lab vs. software), and guided Stage I/II audits.
  • Why it matters: Ultimately, this demonstrates a repeatable pathway to IVDR certification for large NGS services and software—something that had no clear precedent.

Read the announcements: For details, read the Fulgent press release and Citeline case study.

The challenge: certifying a service-based, large-scale NGS system under IVDR

To begin with, FulgentExome is a service-based NGS solution that integrates wet-lab workflows with the Fulgent PLM bioinformatics pipeline. As a result, pursuing IVDR certification meant converting a mature CLIA/CAP testing service into a CE-marked IVD system with robust evidence across scientific validity, analytical performance, and clinical performance—for thousands of genes. In particular, key hurdles included: defining intended use at scale; validating third-party components; proving scientific validity across 4,600+ genes; above all fully validating the bioinformatics pipeline under medical device software standards.

MDx approach: a playbook for complex NGS + software

1) Build the right QMS, fast

First, we performed an IVDR GAP assessment. Next, we designed and implemented an ISO 13485-compliant QMS with risk management, supplier control, document control, internal audits, and management review—migrating from a CLIA/CAP model to IVDR-ready operations.

2) Engineer a defensible intended use

Meanwhile, the intended-use statement evolved iteratively—from an initial ~300-gene scope to whole-exome, finally landing on 4,600+ genes aligned to available clinical and analytical evidence. In the end, the final language was future-proofed to support rapid updates as evidence expands.

3) Split wet lab and software into two regulated products

Afterward, following round 1 review feedback, we separated the documentation into two Basic UDI-DIs—FulgentExome (wet lab) and Fulgent PLM (software)—to align with IVDR expectations for traceability and lifecycle control. Consequently, this restructuring sharpened conformity assessment and accelerated subsequent approvals.

4) Validate the informatics stack like a medical device

In parallel, we validated PLM under IEC 62304/82304, including architecture, version control, cybersecurity, verification/validation, and integration with external databases. Therefore, the result was a fully evidence-backed bioinformatics pipeline capable of reproducible, high-confidence variant calling and reporting.

5) Make “evidence at scale” practical

  • First, Scientific validity: Three-tier strategy combining validation of exome sequencing as an approach, reliance on curated public databases, and deep exemplars for a large subset of genes.
  • Second, Clinical performance: Leveraged routine testing experience (thousands of positives) to focus clinical evidence on high-prevalence genes, and instituted a robust PMPF strategy to continuously strengthen low-prevalence areas.

6) Orchestrate TÜV SÜD audits to success

  • Initially, Stage I confirmed documentation readiness, scope, Basic UDI-DIs and integration of IVDR processes into daily practice.
  • Subsequently, Stage II verified on-the-floor implementation of SOPs, training, competence, CAPA and change control—closing findings on short cycles to hit NB timelines.

Results that move the market

  • CE mark granted for FulgentExome & Fulgent PLM—among the first end-to-end Class C germline NGS solutions under IVDR.
  • Certified scope covers 4,600+ genes, positioning Fulgent as a reference lab for comprehensive hereditary disease testing serving European patients.
  • Post-certification, the platform scaled to >7,000 genes using a new probe set—demonstrating the inherent scalability built into the certified system (process, documentation, and change control).
  • Strengthened competitive standing in the EU diagnostics market; public communications highlight the magnitude of this achievement for large NGS panels.

Read more in the Fulgent press release and Citeline’s in-depth article.

What this means for labs and IVD developers planning large NGS submissions

If you operate an LDT today: you’ll need to translate CLIA/15189 practices into an ISO 13485 framework, document design controls, and produce a full PER (PEP/PER), APR, SVR, PMS/PMPF, SSP, and labeling/IFU aligned to GSPR. Expect to validate any bioinformatics pipeline as SaMD with IEC 62304/82304 and cybersecurity controls.

If your panel is “large”: you likely won’t have uniform clinical data across every gene. A structured tiered evidence model + PMPF can satisfy Notified Bodies while keeping your roadmap scalable.

If you combine wet lab + software: plan for separate Basic UDI-DIs and documentation sets. Treat the pipeline as a product with its own requirements, verification, and risk controls.

Why MDx

  • End-to-end IVDR expertise: From regulatory strategy & classification to Annex II/III technical files, PER/SVR/APR, training, and mock NB reviews.
  • Clinical performance studies: We design, run, and report ISO 20916 studies (protocols, eTMF, monitoring, biostats, PER alignment), and we can act as delegated sponsor for multi-country submissions—100% submission success rate to date.
  • Operational scale: ISO 9001 clinical QMS, EU/US partner network, multilingual CRAs, and a repeatable process honed on 60+ performance study submissions for top IVD and pharma clients.

Project timeline

Our joint project with Fulgent spanned July 2023–July 2025, with overlapping tracks for QMS creation, technical documentation, NB review, and Stage I/II audits—a coordinated plan that allowed rapid closure of findings and post-certification scaling.

Client perspective

The program demanded evening/weekend execution across regulatory, documentation, and project management to meet Notified Body timelines—effort that, in the client’s words, made all the difference in achieving what initially “seemed almost impossible.

Planning IVDR for your NGS panel? Here’s a quick readiness checklist

  • Intended use aligned to evidence (and future updates)
  • ISO 13485 QMS with software lifecycle integration
  • PER (PEP/PER), SVR, APR mapped to gene-level strategy
  • PLM/DR pipeline validated per IEC 62304/82304 (+cybersecurity)
  • Separate documentation/UDI for wet lab vs. software (if applicable)
  • PMS/PMPF plan to mature low-prevalence evidence post-market
  • Mock NB review + Stage I/II audit readiness

(Our team can lead or co-author each artifact above.)

Talk to us

Whether you’re certifying a focused oncology panel or pushing the limits with exome-scale content, MDx brings the cross-functional regulatory, clinical, quality, and software depth to make it possible—on a timeline that keeps your business competitive.

Written by:
Carlos Galamba

Carlos Galamba

CEO

Senior regulatory leader and former BSI IVDR reviewer with deep experience in CE marking high-risk IVDs, companion diagnostics, and IVDR implementation.
Industry Insights & Regulatory Updates

The future of in vitro diagnostics

The future of in vitro diagnostics is being written at the intersection of tighter regulation, workforce pressure, and data‑driven innovation. Under the EU In Vitro Diagnostic Regulation (IVDR), evidence expectations and lifecycle obligations have risen sharply, changing how products are designed, validated, and maintained in the market. For manufacturers, success now depends on pairing scientific advances with stronger clinical evidence strategies, interoperable data flows, and operational resilience across supply, quality, and post‑market systems.

Demand is rising while systems are stretched

Backlogs from the pandemic have converged with long‑standing workforce shortages, particularly in diagnostic specialties, delaying access and lengthening diagnostic pathways. The OECD’s Health at a Glance: Europe 2024 highlights these shortages as a structural risk to access, quality, and system resilience—evidence that the pressure to do more with fewer people is not easing soon. For diagnostics leaders, that reality elevates the value case for automation, near‑patient testing, and real‑world evidence that proves earlier, faster decisions.

Market momentum is real—if evidence and access align

IVDs remain a cornerstone of Europe’s medtech economy and the largest segment globally by sector share, signaling robust demand for better, earlier diagnostics. But translating that momentum into market access requires credible performance evaluation, clear intended‑purpose claims, and a plan for post‑market performance follow‑up that stands up to Notified Body scrutiny. (MedTech Europe DataHub – Market). 

IVDR is raising the bar—and capacity is still normalizing

IVDR’s higher evidence threshold is now a constant, but Notified Body (NB) capacity and throughput continue to shape time‑to‑market. The European Commission’s latest Notified Bodies Survey shows progress on designations and certifications under MDR/IVDR while acknowledging persistent bottlenecks—practical context for planning dossier quality, NB engagement, and transition timelines. Manufacturers that front‑load clinical evidence planning and close gaps against GSPR, PER, and PMPF requirements are better positioned to move through review without costly rework. 

What the next decade looks like for IVD innovators

Expect faster iteration cycles powered by cloud connectivity and AI‑assisted analytics, paired with stronger governance of data provenance, cybersecurity, and change control. Procurement and HTA bodies will demand interoperable outputs that feed clinical systems and population analytics. In practice, this means designing for the 4P future—predictive, preventive, personalized, and participatory—while proving clinical performance and patient‑management claims under IVDR. Aligning technical files, labeling, and performance evaluation with clinical utility (not just analytical superiority) will increasingly differentiate winners.

What leaders should do now

Treat clinical evidence as a product pillar from day one—map intended purpose, target population, and clinical benefit to a coherent performance evaluation plan that integrates literature, device‑generated data, and targeted clinical performance studies. Build for integration and reuse of data across care settings to ease adoption and payer evaluation. Engage NBs early with complete, audit‑ready files. And make post‑market performance follow‑up a source of competitive insight, not a compliance afterthought.

How MDx CRO accelerates IVD market readiness

MDx CRO helps IVD manufacturers compress time‑to‑evidence and navigate IVDR with confidence—from regulatory strategy and technical documentation to clinical performance studies and post‑market performance follow‑up. We translate regulatory expectations into practical study designs and submission‑ready deliverables, then stay with you through NB interactions and lifecycle monitoring. Explore our IVD regulatory services and clinical research support, or contact us to scope a market‑access plan tailored to your portfolio.

Written by:
Carlos Galamba

Carlos Galamba

CEO

Senior regulatory leader and former BSI IVDR reviewer with deep experience in CE marking high-risk IVDs, companion diagnostics, and IVDR implementation.
Industry Insights & Regulatory Updates

How many MedTech companies are there in Europe?

Europe’s MedTech Landscape in 2025: 38,000+ Companies Driving Innovation

Europe is home to one of the world’s most dynamic and diverse medical technology ecosystems, with more than 38,000 MedTech companies operating across the continent. The vast majority—over 90%—are small and medium-sized enterprises (SMEs), playing a critical role in driving innovation, improving healthcare outcomes, and fueling economic growth .

What Defines Europe’s MedTech Industry?

According to MedTech Europe, the sector includes manufacturers of medical devices, in vitro diagnostics (IVDs), and digital health solutions, spanning everything from surgical tools to AI-powered diagnostic platforms. In total, the industry employs over 930,000 people directly, making it one of the largest employers in Europe’s life sciences space .

Why It Matters for Clinical and Regulatory Success

With the growing complexity of EU MDR and IVDR regulations, these companies—especially SMEs—face increasing pressure to:

  • Prove clinical evidence and safety of their technologies
  • Navigate Notified Body reviews and CE Marking
  • Manage post-market surveillance (PMS) and performance evaluations

That’s where MDx CRO steps in.

Supporting Europe’s MedTech Growth

At MDx CRO, we specialize in helping MedTech innovators—from start-ups to established manufacturers—successfully plan, execute, and submit their clinical and regulatory strategies across the EU and global markets. With proven expertise in:

We partner with both large diagnostic leaders and agile SMEs to deliver compliant, high-quality, and market-ready solutions.

A Pan-European Presence

With offices in Barcelona, Madrid, Lisbon, and London, and a network of CRAs and regulatory experts across Europe, MDx provides localized insight with global reach—helping MedTech companies meet requirements faster and smarter.

The Bottom Line

The European MedTech sector is growing—but so are its regulatory challenges. Whether you’re launching a new diagnostic product or preparing for a Notified Body audit, MDx CRO is here to support your success every step of the way.

Let’s talk about your next clinical or regulatory challenge.

Industry Insights & Regulatory Updates

IVDR Transition for Precision Medicine: How MDx CRO Enabled a Seamless Portfolio Upgrade

Introduction

IVDR transition for precision medicine programs can stall when portfolios span liquid biopsy, RNA-based sequencing, and comprehensive tumor profiling. A leading US-based precision medicine company asked MDx CRO to migrate its oncology diagnostics from self-declared IVDD and FDA pathways to full IVDR certification—without disrupting European market access. This blog shares how we planned the transition, selected the right Notified Body, rebuilt regulatory files, and safeguarded ongoing clinical and CDx development in Europe.

The Challenge

  • Convert complex files from FDA/IVDD to IVDR. Multiple assay types (liquid biopsy, RNA-seq, tumor profiling) required re-evidence and restructuring under IVDR Annexes.
  • Select the optimal Notified Body. The client needed a partner capable of reviewing a diverse portfolio efficiently and cost-effectively.
  • Regulatory documentation lift. We had to redevelop key documents: analytical & clinical performance (including CPS reports), risk and design files, and labeling—while maintaining business continuity.

MDx CRO’s Approach

1) Strategic IVDR roadmap and portfolio triage

We assessed intended purpose, risk class, and evidence gaps for each product, then prioritized quick-win files to protect revenue while scheduling deeper re-verification work for complex assays. This created a clear IVDR transition for precision medicine timeline across the portfolio.

2) Notified Body strategy

Leveraging our knowledge of NB capacity and focus areas, we strategically selected a Notified Body that balanced approval probability, cost, and credibility. Early technical consultations reduced surprises and kept reviews on track.

3) Robust regulatory files

We generated comprehensive IVDR documentation:

  • Analytical and clinical performance reports, including scientific validity and performance evaluation reports
  • Risk management aligned to Annex I GSPRs
  • Design and development files with clear traceability
  • Usability and labeling aligned with intended purpose and user context

4) Operational partnership and sponsor duties

The client expanded our role into delegated sponsor responsibilities for clinical studies. We served as EU Legal Representative, oversaw clinical operations, and implemented streamlined processes for biomarker and CDx study submissions across Europe.

Results

  • Successful IVDR transition for priority diagnostics with uninterrupted market access in Europe.
  • Competitive advantage: stronger operational readiness helped the client attract pharma partners for clinical trial biomarker testing and CDx development.
  • Ongoing partnership: MDx CRO manages clinical studies, maintains sponsor duties, and continues the portfolio-wide IVDR journey.

Client Testimonial

“Working with MDx’s Precision Medicine Team has been a pleasure. As a U.S.-based company operating in Europe, I consider them our EU extension. Their expertise and responsiveness keep us ahead in a dynamic market, and the consistency of their delivery has shaped our current and future plans.”

Why This Matters

IVDR raises expectations for evidence, documentation, and lifecycle controls—especially for precision medicine diagnostics. Success requires portfolio triage, NB strategy, and regulatory files that stand up to scrutiny while your teams continue running trials and supporting pharma partnerships. MDx CRO brings integrated regulatory, clinical, and diagnostic know-how to keep your transition moving.

Call to Action

Planning an IVDR transition for precision medicine diagnostics? Let’s protect your market access, cut RFI cycles, and ready your files for Notified Body review.

Industry Insights & Regulatory Updates

IVDR Lab Readiness: Step-by-Step Transition Checklist

The IVDR Shift and What It Means for Clinical Laboratories

The in Vitro Diagnostic Regulation (IVDR) (EU) 2017/746 came into force on 26 May 2022, representing a paradigm shift for diagnostic testing in Europe. Its purpose is clear: ensure safety, traceability, and performance of all in vitro diagnostic devices (IVDs). Unlike its predecessor, the IVDD (98/79/EC), the IVDR applies far-reaching obligations not only to manufacturers but also to clinical laboratories that develop and use their own in-house IVDs (IH-IVDs).

A cornerstone of this new landscape is Article 5(5), which sets conditions under which health institutions may continue manufacturing and using in-house devices without CE marking. While this exemption acknowledges the clinical need for tailored diagnostics, it also imposes new responsibilities.

This blog provides a step-by-step readiness checklist for laboratories to guide you through the transition.

What exactly is an in-house IVD under the IVDR?

An in-house IVD (sometimes called a laboratory-developed test or LDT) is any in vitro diagnostic device manufactured and used only within a health institution, not supplied to another legal entity, and not manufactured on an industrial scale

Examples include:

  • PCR assays where the lab develops its own probes.
  • Custom-developed software tools for diagnostic interpretation.

Excluded are:

  • General laboratory supplies.
  • RUO (research use only) products – unless repurposed for diagnostic use. If an RUO product is used for diagnostic purposes (i.e., results are communicated to the patient for medical decision-making), it ceases to be RUO and must comply with IVDR Article 5(5), thereby becoming subject to the same obligations as an in-house IVD/LDT.
  • Commercially available CE-marked IVDs (which must be purchased and used as intended) – unless it is modified, combined or used outside it’s intended purpose.

You must determine whether you are using an in-house IVD. If you are modifying, combining, or using CE-marked diagnostic tests outside their intended purpose, or if you are repurposing RUO products for diagnostic use, you must ensure compliance with Article 5(5).

Who is entitled to the Article 5(5) exemption?

Only health institutions may use in-house IVDs. According to the IVDR, a health institution is an organization whose primary purpose is patient care or public health. This includes:

  • Hospitals
  • Clinical laboratories
  • Public health institutes

Importantly, the recognition of health institutions may depend on national legislation. For instance, some countries require formal registration or accreditation to benefit from Article 5(5).

Always check your national laws to confirm whether your laboratory qualifies as a “health institution” and whether additional national restrictions or obligations apply.

Should your lab buy CE-marked tests or continue with in-house ones?

Under IVDR, labs face a strategic decision:

  • Purchase CE-marked IVDs: These carry regulatory assurance but may not always exist for niche diagnostic needs, and market withdrawals could limit supply.
  • Develop and use in-house IVDs: Allowed under Article 5(5) if your lab demonstrates compliance with conditions (e.g., GSPR, QMS, technical documentation).

From 31 December 2030, labs must justify why an equivalent CE-marked device is not suitable if they want to continue using their in-house test (article 5(5)(g))

Begin analyzing your portfolio now. Which tests could be replaced by CE-IVDs, and which must remain in-house due to clinical need?

What technical documentation requirements already apply?

Since 26 May 2022, all in-house devices must comply with Annex I of the IVDR (GSPR). This includes:

  • Risk management system covering patient, user, and use error risks.
  • Performance evaluation based on scientific validity, analytical performance, and clinical performance.
  • Traceability and identification (lot numbers, production dates).
  • Appropriate instructions for use and safety information

Treat your in-house tests with the same rigor as CE-marked devices. Maintain documentation to always prove compliance with the GSPRs.

What does IVDR require for quality management when operating under article 5.5?

Since 26 May 2024, labs must manufacture and use in-house devices under an appropriate Quality Management System (QMS). For in-house IVDs, this generally means compliance with EN ISO 15189 or equivalent national provisions

However, note:

  • ISO 15189 covers quality in medical laboratories but not necessarily manufacturing processes.
  • Therefore, supplement with elements of ISO 13485 for design and production control.
  • In addition, laboratories must address the QMS requirements described in Article 10(8) IVDR, which outline the minimal aspects of a system covering risk management, manufacturing documentation, monitoring, corrective actions, and communication with authorities.

Expand your QMS to cover risk management, manufacturing documentation, monitoring, and corrective actions, and the additional QMS obligations set out in Article 10 IVDR. Note that ISO 15189 alone is not sufficient; relevant elements of design and manufacturing from ISO 13485 must also be considered, as the IVDR introduces further QMS requirements that must be fulfilled.

Do labs need to publish information about their in-house devices?

Article 5(5)(f) IVDR requires health institutions to draw up and make publicly available a declaration for each in-house device. This obligation has applied since 26 May 2024, following the end of the initial transition period.

What must the declaration contain? At minimum:

  • Name and address of the health institution manufacturing the device.
  • Details necessary to identify the device (e.g., designation, type, internal code).
  • A declaration of compliance with Annex I (GSPR), or where full compliance is not possible, a reasoned justification explaining the deviations.
  • Confirmation that the device is manufactured under an appropriate QMS.

This declaration must be kept up to date and made easily accessible, typically via the laboratory or hospital’s website This transparency ensures accountability and facilitates oversight.

Prepare standardized declarations for each in-house device. A practical tool exists: the IVDR Taskforce Guidance on LDTs (2020) provides a template (Appendix B) for the declaration that can be directly adapted by laboratories.

What role do regulators play?

Competent authorities may request documentation or even audit your lab to verify compliance. Labs must be prepared to show:

  • Design, manufacturing, and performance documentation of their in-house devices.
  • Clinical justification for developing or using the test instead of a CE-marked alternative.
  • Ongoing performance review and vigilance records, including corrective actions and monitoring of clinical use.
  • Evidence of an appropriate Quality Management System (QMS), as required since 26 May 2024.

The degree of oversight varies across Member States. For example, Belgium and Ireland already operate registration portals where laboratories must register their in-house tests. In other countries, legislation is still under development (Spain) or practices remain vague.

Anticipate audits. Keep a compliance file for each in-house IVD.

What happens in 2030?

From 31 December 2030, labs must justify why the specific needs of their target patient group cannot be met by a CE-marked device – Article 5(5)(g).

This justification may be based on:

  • Technical aspects (e.g., higher sensitivity).
  • Biological aspects (e.g., pediatric vs adult reference ranges).
  • Clinical needs (e.g., unmet diagnostic gaps).

Start now by mapping your portfolio and identifying tests likely to face challenges in proving non-equivalence.

Why are many labs struggling?

Challenges highlighted in recent analyses include:

  • Lack of dedicated regulatory staff.
  • Limited time and budget for documentation.
  • Unfamiliarity with regulatory terminology.

Seek structured support, whether through consultants, digital tools, or peer networks, to avoid non-compliance.

Step 1: Perform a GAP Assessment

  • Map your current situation: List all in-house IVDs and how they are used in your lab.
  • Check national status: Verify if your institution qualifies as a “health institution” under national law, and review whether national legislation imposes additional obligations such as mandatory QMS accreditation (e.g., ISO 15189), registration of in-house IVDs with competent authorities, or other reporting requirements that go beyond the IVDR.
  • Compare requirements vs. practice: Review the IVDR Article 5(5) obligations and identify where your lab already complies (e.g., risk management, traceability) and where gaps exist (e.g., QMS documentation, technical documentation).
  • Prioritize risks: Highlight critical areas (such as missing QMS procedures or incomplete Annex I documentation) that could block compliance in an inspection.

Step 2 – Take Action to Close the Gaps

  • Strategic choice: Decide whether to replace tests with CE-IVDs or maintain in-house versions. Document the rationale.
  • Annex I (GSPR): Ensure all in-house IVDs comply with General Safety and Performance Requirements (effective since 26 May 2022).
  • Quality Management System: Implement or update your QMS to align with ISO 15189, supplemented with elements from ISO 13485 and Article 10(8) IVDR.
  • Compliance documentation & oversight readiness: Compile and maintain a compliance file for each in-house IVD, including full technical documentation (design, manufacturing, risk management, and performance evaluation). Ensure these files are audit-read and can be provided upon request by competent authorities.
  • Vigilance & corrective actions: Set up procedures for monitoring performance, handling incidents, and implementing corrective/preventive measures.
  • Public declaration: Draft and publish a declaration for each in-house device (mandatory since 26 May 2024). Use available templates from guidance.
  • 2030 justification: Start documenting why no equivalent CE-IVD meets the needs of your patient population to support continued in-house use after 31 December 2030.

Closing Thoughts

The IVDR sets high expectations for laboratory-developed in-house IVDs, transforming informal diagnostic practices into rigorously controlled processes. While compliance requires effort, resources, and cultural change, it also strengthens quality, safety, and patient trust. For laboratories, the transition is not optional, it is an opportunity to embed regulatory excellence into daily operations and secure the future of innovative diagnostics. Are you ready for the IVDR transition? Start today with a gap analysis, QMS reinforcement, and documentation plan. The earlier you act, the smoother your path to compliance will be.

At MDx CRO, we specialize in helping clinical laboratories navigate the IVDR, from gap assessments to QMS implementation and technical documentation. We support laboratories in demonstrating compliance with Article 5(5) for in-house IVDs by assisting with:

  • Gap assessments: Mapping all in-house IVDs, comparing current practice with IVDR Article 5(5) requirements, and identifying compliance gaps.
  • QMS alignment: Extending ISO 15189-based systems with manufacturing and design elements from ISO 13485, plus additional QMS obligations under IVDR.
  • Technical documentation: Preparing complete compliance files per device.
  • Public declarations: Drafting and publishing Article 5(5)(f) declarations using recognized templates, ensuring accessibility and consistency.
  • Regulatory readiness: Preparing for competent authority oversight, including audits and requests for documentation.
  • Strategic portfolio decisions: Advising whether to replace tests with CE-IVDs or justify continued in-house use, including preparing 2030 equivalence justifications.
  • Vigilance systems: Setting up monitoring, incident reporting, and corrective/preventive actions in line with IVDR obligations.

Our team knows the pitfalls and the solutions. Let us support you in achieving full compliance. Contact us today to discuss how we can help.

Written by:
Hugo Leis, PhD

Hugo Leis, PhD

Training & Quality Manager

Quality & Training Manager and Senior IVDR consultant with expertise in CE marking, Clinical Laboratories, SaMD, Precision Medicine, Quality Assurance, and academic lecturing.
Industry Insights & Regulatory Updates

Navigating IVDR for NGS Assays: Challenges and Solutions with MDx CRO

Next-Generation Sequencing (NGS) has revolutionized molecular diagnostics by enabling simultaneous analysis of hundreds or thousands of genes across diverse clinical applications. These include germline testing for hereditary disorders, somatic mutation profiling in oncology, infectious disease characterization, and transcriptomic gene expression analysis.

A particularly impactful advancement is liquid-biopsy NGS, which allows non-invasive detection of tumor-derived nucleic acids—such as circulating tumor DNA (ctDNA) or RNA—from blood or other bodily fluids. This method now supports cancer screening, minimal residual disease monitoring, and therapy stratification.

NGS also powers Comprehensive Genomic Profiling (CGP). These assays assess a wide spectrum of biomarkers—single nucleotide variants (SNVs), insertions and deletions (indels), copy number alterations (CNAs), copy number losses (CNLs), gene fusions, and splicing events—across large panels in a single run. Many workflows also integrate microsatellite instability (MSI) and tumor mutational burden (TMB).

Assays can range from targeted panels to whole exome sequencing (WES) or whole genome sequencing (WGS). Each format carries unique validation needs and bioinformatics requirements. The mix of technologies, analytes, sample types (e.g., blood, plasma, FFPE, cfDNA, RNA), and clinical contexts increases regulatory complexity.

Under the EU In Vitro Diagnostic Regulation (IVDR; EU 2017/746), you must define each intended use clearly and support it with comprehensive evidence of scientific validity, analytical performance, and clinical performance. That requirement calls for a holistic, coordinated validation and documentation strategy.

For CE-marking manufacturers and clinical laboratories operating under Article 5(5), IVDR demands structured validation, clear documentation, and lifecycle management. For NGS-based assays, compliance becomes even more demanding due to scientific, technical, and operational intricacies.

Key Challenges in IVDR Compliance for NGS

1) Complex Gene Panels & Variant Diversity

NGS panels often include multiple genes and variant types, each with distinct performance characteristics. You must demonstrate analytical performance—sensitivity, specificity, LoD, and robustness—per variant class. This tailoring increases the scale and complexity of testing.

2) Defining a Clear Intended Use

A precise, testable intended purpose statement anchors the program. Define analytes, clinical context, sample types, output format, and role in patient care. Any ambiguity risks misclassification or validation gaps.

3) Scientific Validity Across Many Analytes and Conditions

Establishing scientific validity grows challenging when one test targets dozens or hundreds of genes. Under IVDR, link each analyte to a clinically relevant condition. That linkage often requires extensive literature review, database referencing, and written justification for inclusion.

4) Clinical Performance Evidence

With broad genomic scope, comprehensive clinical studies may be infeasible. A pragmatic approach combines routine diagnostic data, published literature, and a clear link to Post-Market Performance Follow-up (PMPF) plans to support claims over time.

5) Complex Bioinformatics Pipelines

Bioinformatics sits at the core of NGS diagnostics. Validate every step—from base calling to variant annotation. Implement version control, clear revalidation triggers, and change management to maintain consistent performance after software updates.

6) Use of Third-Party Reagents and Instruments

NGS workflows often incorporate off-the-shelf reagents and platforms not originally CE-marked as part of the IVD system. Document compatibility, performance, and traceability of third-party components to meet IVDR expectations.

7) Labelling Without a Physical Device

Many NGS assays function as software-driven services or LDTs without a packaged device. You still must meet Annex I labelling and Instructions for Use (IFU) requirements—even without physical labels or packaging.

How MDx CRO Supports Your IVDR Journey

MDx CRO brings specialized expertise to guide NGS programs through IVDR across the full lifecycle:

  • Gap Assessments: Identify regulatory shortfalls and prioritize remediation.
  • Performance Evaluation Plan (PEP): Craft PEPs that balance analytical rigor with operational feasibility.
  • Analytical Study Oversight: Design statistically robust studies tailored to complex panels.
  • Bioinformatics Validation: Map and validate each software component under IEC 62304 and ISO 13485.
  • QMS Integration: Build audit-ready documentation, risk management, and traceability.
  • PMS & PMPF Strategies: Establish real-world evidence systems that sustain compliance and support clinical claims.

Conclusion

Achieving IVDR compliance for NGS assays poses a multi-dimensional challenge that blends regulatory discipline with scientific depth. From defining intended use to managing software changes and clinical claims, every step benefits from clarity, structure, and foresight.

MDx CRO partners with diagnostics developers and clinical laboratories to turn regulatory complexity into actionable validation strategies—accelerating time to market while protecting long-term compliance and patient safety.

Written by:
Marketa Svobodova, PhD

Marketa Svobodova, PhD

Regulatory Director, Precision Medicine

Expert in Precision Medicine, NGS & CDx, combining technical and regulatory expertise to guide IVDs through CE certification.
Industry Insights & Regulatory Updates