The First Stages of CAR T Cell Therapy Development

The First Stages of CAR T Cell Therapy Development

From Discovery to Preclinical Development: What are the critical stages to look out for?

Introduction

Chimeric Antigen Receptor (CAR) T cell therapy represents a transformative approach in medicine, particularly in oncology. This method offers personalised treatment by harnessing a patient’s immune system to target and eliminate (cancer) cells. These novel therapies undergo a meticulous journey from target discovery to clinical application, starting with three fundamental steps: target identification, CAR design, and preclinical testing. Here, we want to focus on these initial steps, emphasising their crucial role in developing efficacious and safe CAR T cell therapies.

Target Identification

The basis of efficacious CAR T cell therapy is in selecting an appropriate target antigen for respective target cells. The target antigen must meet several criteria to ensure the therapy’s efficacy and safety.

Target Antigens in CAR T Cell Therapy

An antigen is any substance that causes the immune system to raise a specific immune response against it. In CAR T cell therapy, CARs are used to redirect T cells to recognise and eliminate cells expressing a specific target antigen. The target antigen must be highly specific to the target cells to avoid unwanted side effects such as the killing of healthy cells and tissues, which can be a side effect of the treatment. One significant target antigen that has been approved by the EMA (European Medicines Agency) is BCMA (B-cell Maturation Antigen). CAR T cell therapies like Abecma (Idecabtagene Vicleucel) and Carvykti (Ciltacabtagene Autoleucel) have been approved for treating relapsed or refractory multiple myeloma. These therapies modify T cells to target BCMA, which is commonly expressed on multiple myeloma cells.

(https://www.ema.europa.eu/en/news/meeting-highlights-pharmacovigilance-risk-assessment-committee-prac-8-11-january-2024#ema-inpage-item-64904)

Antigen Selection Criteria for CAR T Cell Therapy

The ideal target antigen should be:

  • Highly Expressed on Cancer Cells: Ensuring that the therapy targets most cancer cells.
  • Minimally Present on Healthy Cells: Reducing the risk of off-target effects and associated toxicities.
  • Uniformly Expressed Across Cancer Cells: Providing consistent targeting of all malignant cells. However, marker expression by (solid) tumours may change over time leading to tumour escape from the therapy.

Understanding the characteristics of a potential therapy, including safety, efficacy, and toxicity profile of a CAR T cell product as early as possible is essential for guiding strategic decisions and helps de-risking investment in further clinical development.

Assessing whether a specific antigen is (or is not) expressed on a specific tissue can be done using these methods:

  • In Silico Analysis: Bioinformatics tools analyse gene expression data to predict potential antigens highly specific to cancer cells.
  • In Vitro Studies: Laboratory experiments using cancer cell lines and patient-derived cells help confirming antigen.
  • In Vivo Models: Animal studies, typically in mice, help assessing whether the antigen is expressed on a specific tissue.

Chimeric Antigen Receptor (CAR) Design

The next critical step is generating the CAR construct.

Components of a CAR

A CAR consists of four primary components:

  • Extracellular Domain: Derived from a monoclonal antibody, this domain binds to the target antigen on cancer cells.
  • Hinge Region: Provides flexibility to the CAR, facilitating effective binding to the antigen.
  • Transmembrane Domain: Anchors the CAR to the T cell’s surface.
  • Intracellular Signalling Domain: This part of the CAR includes co-stimulatory domains (CD28 or 4-1BB) and an activation domain (CD3ζ), which trigger T cell activation and proliferation upon antigen binding.

A schematic overview of a CAR can be found in the figure below.

A schematic overview of a Chimeric Antigen Receptor (CAR)

Figure 1. Schematic overview of a CAR. The schematic overview shows key components. The extracellular antigen-binding domain binds to the target antigen on cancer cells. The hinge region, a flexible segment, links this domain to the transmembrane domain, aiding positioning and motility. Within the antigen-binding domain, the extracellular domain identifies the specific antigen. The transmembrane domain anchors the CAR in the T cell membrane, while the intracellular signalling domain transmits activation signals upon antigen binding, triggering the T cell’s response.

Engineering and Optimisation of CAR T Cells

The process of engineering and optimisation of a CAR begins with synthesising the CAR gene. This gene is then incorporated into for example a viral vector, often a lentivirus or retrovirus. Introducing the CAR gene into T cells by using a viral vector is called viral transduction. The engineered CAR is then tested in cell lines to confirm its functionality. The next step is the optimisation of the CAR T cells, which includes but is not restricted to:

  • Affinity Tuning: Adjusting the binding strength of the CAR to the antigen to achieve a balance between efficacy and safety.
  • Signal Modulation: Enhancing the T cell’s response to ensure effective cancer cell destruction without causing excessive activation that could lead to toxicity.
Overview of Chimeric Antigen Receptor (CAR) T cell therapy development

Figure 2. Overview of CAR T cell therapy development. The process starts with harvesting peripheral blood cells from the patient. Then, mononuclear cells are isolated from the blood. If needed, the T cells are isolated from the mononuclear cells. These (isolated) T cells are then genetically modified to express chimeric antigen receptor (CAR) that target specific (cancer) cells. Following activation of the T cells, [Mv1] the insertion of the CAR gene is performed using a viral vector (viral transduction) or non-viral transfection methods. The modified T cells are expanded to clinically relevant numbers. Only after completing quality control testing and certification by a Qualified Person (QP), the CAR T cells can be administered to the patient, providing for a targeted and personalised treatment.

Preclinical Testing

Preclinical testing is used for the evaluation of CAR T cells in controlled environments to ensure their efficacy and safety before proceeding to a first in human clinical trial. The efficacy and safety of CAR T cells are tested using in vitro and in vivo methods.

In Vitro Testing

During in vitro testing, various standard assays are performed to understand more about the characteristics of the engineered CAR T cell, such as its cytotoxicity, proliferation, and cytokine secretion levels.

  • Cytotoxicity Assays: Assessing the CAR T cells’ ability to kill target cells and to leave non-target cells untouched.
  • Proliferation Assays: Measuring the expansion capacity and survival of CAR T cells upon activation by the target antigen.
  • Cytokine Secretion Assays: Evaluating the secretion of cytokines as an indication for T cell activation and potential toxicity​​.

In vivo Testing Following successful in vitro testing, CAR T cells are evaluated in vivo using clinically relevant animal models:

  • Toxicity Studies: These studies assess potential side effects and determine the maximum tolerated dose, which is a challenge as the weight of a mouse is only about 25 grams.
  • Efficacy Studies: Animal models are used to test the CAR T cells’ ability to effectively reduce the tumor burden and improve survival. The safety profile and mechanism of action of the CAR T cell therapy are being assessed.

Regulatory Compliance

Preclinical studies must comply with Good Laboratory Practice (GLP) standards to ensure reliability and reproducibility of the experiments. Comprehensive documentation of findings is crucial for future regulatory submissions, such as an Investigational Medicinal Product Dossier (IMPD) application.

As an academic institution or company aiming to develop a CAR T cell therapy, collaboration with a Contract Development and Manufacturing Organization (CDMO) can be invaluable. CDMOs offer specialised expertise, resources, and capacity, that contributes to streamline the development process, enhance scalability, and ensure regulatory compliance.

Conclusion

The first stage of CAR T cell therapy development, encompassing target identification, CAR design, and preclinical testing, is fundamental in contributing to the therapy’s safety and efficacy. By meticulously planning and executing each step, therapy developers aim to bring safe and effective CAR T therapies from the laboratory to the patient.

€10 Million Awarded to NecstGen, NTrans Technologies, LUMC, and Divvly to Transform Cell and Gene Therapies

€10 Million Awarded to NecstGen, NTrans Technologies, LUMC, and Divvly to Transform Cell and Gene Therapies

• A grant from IPCEI Med4Health will facilitate the development of advanced technologies to improve cell and gene therapy production.

• The consortium will focus on novel technologies and production processes to overcome current bottlenecks in cell and gene therapy manufacturing, enabling cost-effective production and facilitating clinical trials for conditions like Duchenne Muscular Dystrophy and Diamond Blackfan Anemia.

Leiden, the Netherlands, August 28, 2024 – In a major advancement toward bringing cell and gene therapies more rapidly to patients, NecstGen, together with partners NTrans Technologies, Leiden University Medical Center (LUMC), and Divvly, has been awarded a €10 million grant from the European Union’s Important Project of Common European Interest (IPCEI) Med4Cure program. This project aims to transform the treatment landscape for serious medical conditions like Duchenne Muscular Dystrophy and Diamond Blackfan Anemia by pioneering cost-effective production methods, making these treatments more accessible.

Addressing Major Industry Challenges

Cell and gene therapies offer the potential to treat, prevent, and cure devastating diseases by targeting the underlying genetic causes. Yet, their development is often hindered by high costs and complex manufacturing processes, largely due to limited access to advanced production methods. This restricts therapy developers and manufacturers from scaling up production effectively. This project aims to break down these barriers, enhancing the scalability and affordability of therapies and ensuring that they reach patients quicker.

Collaborative Efforts and Impact

NecstGen, a non-profit contract development and manufacturing organization (CDMO), will lead the project, leveraging its comprehensive approach to implementing a complete value chain, from concept to patient. It will focus on innovative cell line and production process technologies to reduce overall production costs. NTrans Technologies will improve treatment effectivenes s through its proprietary gene-editing platform, while Divvly will enhance industry compliance for Good Manufacturing Practices (GMP) with its advanced software solutions. The LUMC, a renowned Dutch medical university, will lead (pre-)clinical trials, offering crucial insights and support, directly impacting therapy development for Duchenne Muscular Dystrophy and Diamond Blackfan Anemia.

“By uniting our strengths and challenging current paradigms in cell and gene therapy manufacturing, we can accelerate the availability of these life-saving therapies,” said Paul Bilars, Chief Executive Officer, NecstGen. “At NecstGen, we are dedicated to transforming these technologies to support therapy developers and ultimately, deliver life-changing treatments to patients, both in Europe and globally.”

About IPCEI
The Important Project of Common European Interest (IPCEI) is an initiative by the European Union designed to support large-scale, cross-border projects that are highly innovative and strategically important for Europe. With 14 highly innovative projects, the IPCEI Med4Cure program aims to enhance drug discovery and pharmaceutical production processes, addressing major health challenges and fostering innovation and collaboration across Europe.

About NecstGen
NecstGen is a non-profit CDMO and centre of excellence for Cell and Gene Therapy, located in a purpose-built GMP facility at the Leiden Bio Science Park, the largest bio-cluster in the Netherlands. NecstGen provides critical contract development, manufacturing and rental services to academic and industrial therapy developers to deliver next-generation therapeutics to patients.

NecstGen offers:

• Full contract manufacturing services for Cell Therapy and Viral Vector development and manufacturing
• Process design, scale-up, optimisation and automation for Cell Therapy and Viral Vector
• Assay development for in-process, release, and potency testing
• Cleanroom rental including services for QA, QC, and QP.

For more information about our project, please visit our website or contact us directly. If you are a therapy developer or industry professional interested in partnering with us or learning about our innovative solutions, please reach out to:
https://necstgen.com/contact-us/

For media inquiries, please contact:
bd@necstgen.com

This project has received funding from Rijksdienst voor Ondernemend Nederland (Netherlands Enterprise Agency).

Collaboration UMC Utrecht for Bone Regeneration Therapy

Collaboration UMC Utrecht for Bone Regeneration Therapy

Leiden, Netherlands — NecstGen, the Netherlands Center for the Clinical Advancement of Stem Cell and Gene Therapies, is proud to announce a collaboration with Dr. Debby Gawlitta of the Department of Oral and Maxillofacial Surgery & Special Dental Care at UMC Utrecht. This collaboration aims to develop a pioneering therapy for bone defects, leveraging the power of mesenchymal stromal cell (MSC)-derived spheroids.

Dr. Gawlitta’s research has demonstrated promising results in preclinical studies, showcasing successful endochondral bone regeneration in rat and goat models. These findings show the potential of devitalised bone marrow-derived MSC-spheroids in treating bone defects, a significant advancement in regenerative medicine.

Through this collaboration, NecstGen will support Dr. Gawlitta’s project by offering development support of this innovative therapy.

“We are excited about the potential of this collaboration with Dr. Gawlitta,” said Paul Bilars, CEO of NecstGen. “Our mission is to accelerate the delivery of innovative medicinal products to patients. This project exemplifies our commitment to support the next generation of therapies that can significantly impact patients’ lives.”

The project focuses on the GMP manufacturing of human MSC-derived spheroids, targeting maxillofacial and spinal fusion defects. It encompasses an extensive process, including expanding MSCs, spheroid formation, and rigorous quality control assays to ensure the required safety and efficacy standards.

This collaboration marks a significant step forward in bone regeneration therapy and reinforces the importance of partnerships between academia and industry in advancing healthcare innovations.

Dr. Gawlitta has received a RegMed XB/Dutch Cardiovascular Alliance Thematic Tech Transfer Voucher sponsoring this collaborative activity to further develop her innovative findings into treatments. “This subsidy of RegMed XB/DCA for propelling advanced therapies to clinical implementation is coming at the right time to explore and take our next steps in the translational axis. The expertise and experience of NecstGen in this field is instrumental in designing our envisaged bone regenerative treatments.”

For more information about NecstGen, please visit necstgen.com.

About NecstGen

NecstGen is a non-profit CDMO and centre of excellence for Cell and Gene Therapy, located in a purpose-built GMP facility on the largest bio-cluster in the Netherlands, Leiden Bio Science Park. NecstGen provides critical contract development, manufacturing and rental services to academic and industrial therapy developers to deliver a new generation of therapies to patients.

About RegMed XB/Dutch CardioVascular Alliance TTT Voucher

The Thematic Tech Transfer (TTT) vouchers from RegMed XB/Dutch CardioVascular Alliance support the valorization trajectory of promising academic research within the regenerative and/or cardiovascular medicine fields. For more information, please visit www.tttransfer.nl and contact our impact officers.

NecstGen Services

We offer process development solutions, contract preclinical/ GMP manufacturing and cleanroom rental for Cell and Gene Therapies to clinicians, academic, small, medium, and large commercial organisations worldwide.

Viral Vector Manufacturing & Development

Cleanroom Rental

Cell Therapy Manufacturing & Development

About Us

NecstGen is a non-profit CDMO and Centre of Excellence for Cell and Gene Therapy located in a purpose-built GMP facility in Leiden, The Netherlands. Dedicated to the field of Cell and Gene Therapies, we provide expertise and
capacity to focus on patient solutions.

NecstGen

Team

Partners

History

Facility