Chimeric Antigen Receptor (CAR) T Cell Therapy

Chimeric Antigen Receptor (CAR) T Cell Therapy

Introduction

Chimeric Antigen Receptor (CAR) T cell therapy is a revolutionary approach in the field of cancer immunotherapy. This innovative treatment involves genetically engineering a patient’s own T cells to express chimeric antigen receptors, which are artificial receptors designed to target specific cancer-associated antigens. CAR-T cells combine the antigen recognition capability of antibodies with the potent cytotoxic activity of T cells, enabling them to recognise and eliminate cancer cells with remarkable precision. Over the past decade, CAR-T therapy has demonstrated exceptional success in treating certain hematologic malignancies, such as acute lymphoblastic leukaemia and non-Hodgkin lymphoma. The ability to reprogram a patient’s own immune cells to effectively combat cancer marks a paradigm shift in cancer treatment strategies, offering new hope for patients with otherwise refractory or relapsed diseases. As research continues to advance, CAR-T therapy holds promise for broader applications and improvements, paving the way for a new era of personalised and targeted cancer therapies.

CAR Structure

Chimeric antigen receptor (CAR) consists of three parts: an extracellular domain, a transmembrane domain, and an intracellular signalling domain.

  • The extracellular domain contains a single-chain variable fragment (scFv) molecule derived from an antibody to recognise specific cancer cell antigens and a hinge region provides flexibility to the receptor, aiding in optimal antigen binding.
  • The transmembrane domain anchors the CAR within the T cell’s membrane, ensuring stability.
  • The intracellular signalling domain is composed of one or more costimulatory molecules, such as a cluster of differentiation CD28 and 4-1BB, and a stimulatory molecule, CD3 ζ, transmitting activation signals to the T cell’s interior, initiating a robust immune response against targeted cancer cells.

Xin et al., (2022)

CAR Evolution

There are five generations of chimeric antigen receptors (CARs), each building upon the previous to enhance CAR-T cell therapy’s effectiveness.

  • The first generation only contained the CD3ζ derived signalling modules. While functional, these CARs had limited efficacy and persistence.
  • The second generation contained CD3ζ and a costimulatory molecular domain. like CD28 or 4-1BB, alongside the signalling domain. These additions improved CAR-T cell activation, proliferation, and persistence, resulting in more potent anti-cancer responses.
  • The third generation contained CD3ζ and multiple costimulatory molecular domains, including CD28, 4 1BB, OX40 or ICOS, etc, aiming to amplify T cell activity.
  • The fourth generation is called TRUCKs (T cells redirected for antigen-unrestricted cytokine-initiated killing), engineered to secrete immune-stimulating molecules upon antigen recognition. This design aims to recruit and activate other immune cells against the tumour.
  • The fifth generation included simultaneous activation of TCR, costimulatory molecular domain, and cytokine triple signalling to stimulate cell proliferation and enhance its persistence.

Xin et al., (2022)

In-Vitro CAR-T Cell

In this method, T cells are extracted from the patient and engineered outside the body. The extracted T cells are modified to express the CARs, then expanded and cultured to create a large population of CAR-T cells. Once their numbers are sufficient, these cells are infused back into the patient. In-Vitro CAR-T allows for precise CAR design, rigorous quality control, and optimal expansion of modified cells before treatment, but it involves complex cell manipulation processes.

Six FDA Approved CAR-T Cell Products

Product NameTargetsTarget DiseasesTime of Approval
Kymriah® (tisagenlecleucel)CD19B-ALL ; DLBCLAugust 2017
Yescarta® (axicabtagene ciloleucel)CD19DLBCL ; RRFLOctober 2017
Tecartus® (brexucabtagene autoleucal)CD19R/R MCLJuly 2020
Breyanzi® (lisocabtagene maraleucel)CD19DLBCLFebruary 2021
Abecma® (idecabtagene vicleucel)BCMAR/R MMMarch 2021
Carvykti® (ciltacabtagene autoleucel)BCMAR/R MMFebruary 2022

Chen et al., (2022)

B-ALL, B-cell acute lymphoblastic leukaemia; DLBCL, Diffuse large B-cell lymphoma; R/R FL, relapsed or refractory follicular lymphoma; R/R MCL, relapsed or refractory mantle cell lymphoma; R/R MM, relapsed or refractory multiple myeloma.

In-Vivo CAR-T Cell

In this approach, CAR-T cells induced by nanocarriers loaded with CAR genes and gene editing tools are generated within the patient’s body. Once inside the body, these modified T cells start expressing the CAR receptor and target cancer cells. In-Vivo CAR-T offers simplicity and avoids the need for cell collection, engineering, and reinfusion, but controlling the modification process can be challenging.

In-Vivo CAR-T Cell Gene Delivery Systems

Viral Vectors
Viral vectors have good transfection efficiency and are widely used to deliver genes in
various applications, but they suffer from immunogenicity and cellular toxicity.
Example: Lentiviral vector (LV) and Adeno-associated virus vector (AAV)
Non-Viral Delivery Systems
Physical delivery systems have low immunogenicity but cannot target internal organs.
Example: electroporation, needle injection, laser irradiation, and gene guns.
Chemical delivery systems have recently gained worldwide attention. Lipid-based nanoparticles are one of the most attractive non-viral vectors for gene delivery as several formulations of these carriers have been approved to use in the clinic. Some T cell-targeted lipid nanoparticles (LNPs) with plasmid DNA or in vitro-transcribed (IVT) mRNA have been reported.
Example: cationic lipids or polymer-based nanoparticles, gold nanoparticles, silica
nanoparticles, and exosomes.

Wakao et al., (2023)

Challenges & Potential Strategies in CAR-T Cell Therapy

Limitations of CAR-T Cell TherapyPotential Strategies
Antigen escape– Targeting multiple antigens (dual or tandem CARs)
On-target off-tumour effects– Targeting tumour-restricted post-translational modifications
CAR-T cell trafficking and tumoir infiltration– Local administration vs systemic delivery
– Engineering CAR-T cells to enhance penetration through physical barriers (tumour stroma)
Immunosuppressive microenvironment– Combination immunotherapy with CAR-T cells and checkpoint blockade
– Engineering CAR-T cells to provide immunostimulatory signals in the form of cytokines or CARs resistant to immunosuppressive factors.
CAR-T cell-associated toxicities– Altering CAR structure to ameliorate toxicity
– Modifying CAR transduced T cells and neurotoxicity
– CAR “off-switches”

Sterner et al., (2021)


Originally posted by Abnova on https://www.abnova.com/en-global/newsletter/newsletter/content/newsletter2023-09-01

Caltag Medsystems is the distributor of Abnova products in the UK and Ireland. If you have any questions about these products, please contact us.

Chimeric Antigen Receptor (CAR) T Cell Therapy
Tagged on:                 

Contact us