< All Topics
Print

Advanced Therapy Medicinal Products (ATMPs): Gene and Cell Therapy Regulation

The regulatory landscape for Advanced Therapy Medicinal Products (ATMPs) in the European Union represents one of the most complex, scientifically rigorous, and dynamically evolving frameworks in global healthcare governance. For professionals operating at the intersection of biotechnology, regulatory law, clinical research, and advanced manufacturing, understanding the nuances of this framework is not merely an academic exercise; it is a prerequisite for successful development and commercialization. ATMPs—encompassing gene therapy, somatic cell therapy, and tissue-engineered products—challenge traditional regulatory paradigms because they often involve highly personalized interventions, novel mechanisms of action, and manufacturing processes that are intrinsically linked to the patient. This article provides a deep analytical dive into the definition, classification, authorization pathways, evidence expectations, and manufacturing controls governing ATMPs in the EU, with a specific focus on the pivotal role of the Committee for Advanced Therapies (CAT) at the European Medicines Agency (EMA).

Defining the Scope: What Constitutes an ATMP?

To navigate the regulatory requirements, one must first precisely identify the product. The legal definition of ATMPs is anchored in Article 2 of Regulation (EC) No 1394/2007. This regulation established a centralized authorization procedure for ATMPs and created the CAT. The definition is exclusionary in nature; a product falls under the ATMP umbrella if it meets specific criteria regarding biologic function and intended use, and crucially, if it does not fall under the definition of a medicinal product generally or within the scope of other specific regulations (such as devices or blood products).

Regulation 1394/2007 identifies three main categories of ATMPs, plus a fourth “hybrid” category that has become increasingly significant in recent years.

Gene Therapy Products

Gene therapy products are defined as biological medicinal products that have the following characteristics:

  • They contain an active substance which contains or consists of a recombinant nucleic acid.
  • They are administered to human beings with a view to regulating, repairing, replacing, adding, or deleting a genetic sequence.
  • Crucial Distinction: The therapeutic, prophylactic, or diagnostic effect relates directly to the recombinant nucleic acid sequence it contains, or to the genetic sequence of the therapeutic product.

From a regulatory perspective, this includes viral vectors (e.g., AAV, lentivirus), non-viral vectors (e.g., naked DNA, lipoplexes), and genome editing technologies (e.g., CRISPR-Cas9). It is important to note that in vivo and ex vivo approaches are both covered. However, gene silencing products (such as siRNA or antisense oligonucleotides) were historically a point of contention. The European Court of Justice clarified in 2012 (Case C-67/12) that these are generally considered medicinal products within the scope of Directive 2001/83/EC, but they do not fall under the strict definition of gene therapy products in Regulation 1394/2007 unless they modify the genetic sequence permanently. This distinction has significant implications for the regulatory pathway (centralized vs. mutual recognition).

Somatic Cell Therapy Products

Somatic cell therapy products are defined as biological medicinal products that have the following characteristics:

  • They contain or consist of cells that have been manipulated (e.g., expanded, activated, genetically modified).
  • They are administered to human beings with a view to treating, preventing, or diagnosing a disease through the pharmacological, immunological, or metabolic action of those cells.

Examples include CAR-T cells, chondrocytes for cartilage repair, and dendritic cell vaccines. The regulation explicitly excludes cells that are simply isolated from the patient and re-administered without manipulation (homologous use). However, the definition of “manipulation” has been a subject of regulatory debate. The EMA and CAT have issued Guidelines to clarify that minimal manipulation (e.g., centrifugation, freezing) generally falls outside the ATMP scope, provided the cells are used for their homologous function. Conversely, expansion or activation in culture constitutes manipulation.

Tissue-Engineered Products (TEPs)

TEPs contain or consist of cells or tissues that have been manipulated to be used for the same essential functions in the recipient and donor. They are defined by:

  • Containing cells or tissues that have been manipulated to be used for the same essential functions in the recipient and donor.
  • Containing cells or tissues that are not intended to be used for the same essential functions in the recipient and donor (e.g., a scaffold seeded with cells).

These products are often characterized by the use of a scaffold or matrix combined with cells. The regulatory focus here is on the structural and functional integration of the product into the body.

Combined ATMPs (Hybrid Products)

As technology advanced, a gap emerged between pure cell/gene therapies and medical devices. Article 2(1)(c) of the Regulation addresses Combined ATMPs. These are products that:

  1. Contain as an integral part one or more medical devices, or a device constituent part.
  2. The cellular or tissue-engineered part of the product is responsible for the principal action of the product (the “action” must be achieved by the cells/tissues, not the device).

Examples include cells seeded onto a biodegradable scaffold or a gene therapy vector delivered via a specific catheter system. The regulation mandates that the device component must comply with the Medical Device Regulation (MDR) or In Vitro Diagnostic Regulation (IVDR), but the overall product is regulated as an ATMP. This creates a dual compliance burden that requires close coordination between the CAT and Notified Bodies.

The Regulatory Pathway: Centralization and the Role of the CAT

Unlike standard medicinal products, which may utilize various authorization procedures (Centralized, Mutual Recognition, National), ATMPs are subject to the centralized authorization procedure exclusively. This means that a marketing authorization granted by the EMA is valid across the entire European Union (and EEA). This monopoly on the pathway underscores the high level of scientific expertise required for evaluation, which is concentrated at the EMA.

The Committee for Advanced Therapies (CAT)

The CAT is the scientific committee at the EMA responsible for evaluating the quality, safety, and efficacy of ATMPs. It is a multidisciplinary body comprising experts in cell biology, gene therapy, immunology, clinical pharmacology, and regulatory affairs. The CAT does not operate in a vacuum; it works closely with the Committee for Medicinal Products for Human Use (CHMP).

The workflow generally follows this sequence:

  1. Scientific Advice: Developers engage with the CAT/EMA for scientific advice during development. This is highly recommended to align on the evidence generation plan.
  2. Marketing Authorization Application (MAA): Submitted via the EMA’s eSubmission gateway. The EMA validates the application.
  3. Assignment: The EMA assigns the application for evaluation. The CAT is appointed as the Coordinating Committee.
  4. Evaluation: The CAT assesses the Quality, Non-Clinical, and Clinical data. Rapporteurs (appointed by CAT) lead the assessment.
  5. Opinion: The CAT adopts an Opinion (not a decision). This opinion is sent to the CHMP.
  6. Adoption: The CHMP adopts the CAT’s opinion (usually without change). The EMA then forwards the opinion to the European Commission.
  7. Decision: The European Commission issues a legally binding Decision granting or refusing the marketing authorization.

Practical Note: While the CHMP formally adopts the opinion, the CAT is the scientific lead. For ATMPs, the CHMP essentially “rubber-stamps” the CAT’s scientific assessment, acknowledging the specialized nature of these products.

Scientific Advice and Protocol Assistance

Given the novelty of ATMPs, the “one-size-fits-all” approach rarely applies. The EMA offers Protocol Assistance, which is a specific type of scientific advice for orphan medicinal products (many ATMPs are orphan drugs). However, even for non-orphan ATMPs, early interaction is critical. Developers often struggle with the definition of the Quality Target Product Profile (QTPP) and the Control Strategy. Because ATMPs are often “living” products or involve complex patient-specific processes, defining the specifications is difficult. The CAT advises on how to justify release criteria for a product that may have inherent variability.

Evidence Expectations: The “Three Pillars” of ATMP Development

The evidentiary bar for ATMPs is high, but the nature of the evidence differs significantly from small molecule drugs. The assessment rests on Quality, Non-Clinical, and Clinical pillars, all of which are tightly interlinked.

Quality: The “CMC” Challenge

Chemistry, Manufacturing, and Controls (CMC) is often the most challenging aspect of ATMP development. The regulatory principle of GMP (Good Manufacturing Practice) applies, but the interpretation is adapted for ATMPs.

The Starting Material

For cell therapies, the “starting material” is the patient’s own cells (autologous) or donor cells (allogeneic). The quality of the final product depends heavily on the quality of the starting material. The EMA expects a characterization of the donor (for allogeneic) or the collection process (for autologous). For gene therapies, the starting material includes the vector master cell bank.

Vector Shedding and Biosafety

For viral vectors, a major concern is vector shedding (excretion of the virus from the patient) and the potential for replication-competent viruses. The EMA requires rigorous testing for replication-competent viruses (RCV) and detailed risk assessments regarding environmental release (GMO requirements).

Comparability

Because manufacturing processes for ATMPs are often optimized during clinical development, changes are inevitable. Demonstrating comparability (proving that a change in process does not affect quality, safety, or efficacy) is difficult. The CAT requires extensive bridging studies, often involving in vitro assays and sometimes animal data, to support process changes.

Non-Clinical Studies

Traditional toxicology studies (e.g., in rodents) are sometimes not feasible or relevant, particularly for autologous products which are species-specific. The EMA accepts alternative approaches, such as:

  • Use of homologous animal models.
  • In vitro functional assays.
  • Read-across from similar vectors or cell types.

However, biodistribution and integration site analysis (for integrating vectors like lentivirus) are mandatory. The risk of insertional mutagenesis (where the vector integrates into the genome and activates an oncogene) requires long-term follow-up in animal models where possible.

Clinical Evidence: Trial Design and Endpoints

Clinical trials for ATMPs often face challenges regarding patient recruitment due to the rarity of the target diseases. Consequently, trials are often small.

Single-Arm Trials

It is frequently unethical or impractical to conduct randomized controlled trials (RCTs) for life-threatening conditions where no standard of care exists. The EMA accepts single-arm trials with historical controls or external comparators. However, the quality of the data must be exceptionally high, and the selection of endpoints must be robust.

Long-Term Efficacy and Durability

Unlike small molecules that are metabolized quickly, ATMPs can have effects that last for years (e.g., gene therapy) or involve ongoing biological activity (e.g., stem cells). The EMA requires evidence of durability of effect. For gene therapies, this often involves measuring transgene expression levels over time.

Long-Term Follow-Up (LTFU): The Post-Marketing Commitment

Perhaps the most distinct feature of ATMP regulation is the emphasis on Long-Term Follow-Up (LTFU). The risks associated with ATMPs, such as delayed immune reactions or oncogenesis, may not manifest during the relatively short duration of clinical trials.

Regulatory Obligation: A marketing authorization for an ATMP will almost invariably include a specific obligation to conduct post-authorization safety studies (PASS) to monitor long-term safety and efficacy.

Duration and Scope

The standard follow-up period suggested by the EMA is typically 15 years for gene therapy products, particularly those involving integrating vectors. For somatic cell therapies, the duration may be shorter (e.g., 5-10 years) depending on the risk profile.

The LTFU protocol must define:

  • Key Safety Parameters: Oncological surveillance (monitoring for malignancies), immunogenicity, and vector shedding.
  • Efficacy Endpoints: Survival, quality of life, or biomarkers indicating sustained biological activity.
  • Patient Registry: Often, the sponsor is required to establish or participate in a registry to capture data on all patients treated in the EU, including those treated in a compassionate use program.

In practice, managing LTFU is a logistical challenge. Patients may move, withdraw consent, or die from unrelated causes. Sponsors must have robust pharmacovigilance systems and patient engagement strategies to ensure data continuity. The EMA monitors compliance with these obligations through the Pharmacovigilance Risk Assessment Committee (PRAC).

Manufacturing Controls: GMP and the “Bedside” Manufacturing

Manufacturing ATMPs is a logistical feat. It ranges from centralized production of viral vectors to decentralized “bedside” manufacturing of autologous cell therapies.

The Concept of “Marketing Authorization Holder” (MAH) Responsibility

The MAH bears the ultimate responsibility for the quality of the product, even if the manufacturing is outsourced to a Contract Development and Manufacturing Organization (CDMO). For autologous therapies, the chain of identity (Chain of Custody) is critical. A mix-up of cells between patients is a critical GMP failure.

Facility Requirements

Manufacturing must occur in facilities compliant with Annex 2 of EU GMP Guide (specific to ATMPs). This includes:

  • Environmental monitoring (cleanrooms).
  • Segregation of processes (to prevent cross-contamination).
  • Personnel training.

For products manufactured at the point of care (e.g., in a hospital cleanroom), the EMA and national competent authorities (NCAs) have developed specific guidance. The MAH must ensure that the hospital site adheres to GMP standards, often through a Quality Agreement. This blurs the line between the pharmaceutical industry and healthcare providers.

Release Testing

Final product release is a two-stage process for ATMPs:

  1. In-Process Controls: Testing performed during manufacturing.
  2. Final Release: Performed by the Qualified Person (QP) in the EU. The QP must certify that the batch was manufactured in accordance with GMP and the marketing authorization.

For autologous products, “batch” release is often “single-patient batch” release. The QP review must be timely to ensure the product is viable for administration to the patient.

Harmonization vs. National Implementation: The “Hospital Exemption” Debate

While the centralized procedure harmonizes the authorization of ATMPs, the application of these therapies in clinical practice involves national nuances. A major point of divergence is the Hospital Exemption (Article 3(7) of Directive 2001/83/EC).

Definition: Hospital exemption allows for the “preparation, processing, adaptation, or assembly” of an ATMP within a hospital/clinic for an individual patient under the direct responsibility of a medical practitioner, following a medical prescription for a “magistral preparation” or “extemporaneous preparation.”

The Controversy: This exemption was intended for exceptional, one-off cases where a patient has a specific need that cannot be met by a commercially authorized product. However, in some Member States (notably France, Germany, and Italy), hospital exemption has been used extensively to provide access to ATMPs (particularly somatic cell therapies like CAR-T cells) before or instead of formal marketing authorization.

Regulatory Divergence:

  • Strict Interpretation (e.g., UK post-Brexit, EMA guidance): Hospital exemption is strictly for “magistral preparations” (compounding) and should not be used for industrial-scale production or for products that are essentially identical to a commercial product.
  • Flexible Interpretation (e.g., France/Italy): National frameworks allow for “reimbursement” of hospital exemption
Table of Contents
Go to Top