Neeta RatanghayraSeptember 18, 2024
The major players in the CAR-T cell therapy market include Novartis, Gilead, and Bristol Myers Squibb. Notably, Bristol Myers Squibb is a leading patent filer in this field, reflecting its significant role in advancing CAR-T cell therapies.
While CAR-T cell therapy has achieved significant success in treating hematological cancers, its application to solid tumors—such as those in the stomach, pancreas, and lungs—is still in the experimental phase. The main challenges for using CAR-T therapy in solid tumors include the absence of suitable CAR-T antigens and the tumors' immune-suppressive and complex microenvironments, which hinder effective CAR-T cell trafficking.
Current research is focused on enhancing CAR-T cell infiltration into solid tumors by administering the therapy directly at the tumor site rather than systemically. As of now, over 1,100 trials involving CAR-T therapies are underway, with approximately 150 of these targeting solid tumors. Additionally, emerging therapies like CAR-NK and CAR-M cells are being developed as potential alternatives or complementary approaches for treating solid tumors.
Despite the significant advances CAR-T cell therapies have brought to cancer treatment, they are not without their challenges. One major issue is the heightened risk of severe immune reactions. These include cytokine release syndrome (CRS), which can cause fever, difficulty breathing, low blood pressure, nausea, and vomiting. Additionally, patients may experience CAR-T cell-related encephalopathy syndrome and immune effector cell-associated neurotoxicity syndrome. These toxicities complicate the safety profile of CAR-T therapies, making their management challenging.
Another critical challenge lies in antigen selection and tumor resistance. CAR-T therapies rely on the presence of specific antigens on tumor cells. However, tumors may lack these antigens or exhibit variability in antigen expression, which can undermine the effectiveness of the CAR-T cells. Furthermore, tumor cells can develop resistance mechanisms, such as downregulating antigen expression or increasing immune inhibitory factors in response to the CAR-T cell attack. This adaptability reduces the long-term effectiveness of the therapy and poses an ongoing hurdle.
Efforts to address these challenges have led to significant advancements, including the exploration of multiple antigen targets, improvements in CAR-T cell design, and adjustments in drug dosages. Despite these innovations, the issues of treatment tolerance and safety have not been entirely resolved. Continued research is essential to refine CAR-T cell therapies further and enhance their efficacy and safety.
Future Directions
Ongoing research aims to enhance the effectiveness and safety of CAR-T cell therapy through several innovative approaches. Key areas of focus include:
· Adjustable Switching Systems: Introducing mechanisms that allow for the regulation of CAR-T cell activity could help mitigate adverse reactions. These systems would enable clinicians to start or stop CAR-T cell activity as needed, potentially reducing the incidence of severe side effects.
· Multiplexed and Bispecific CARs: Exploring the use of multiple CAR structures to target different antigens or employing bispecific CARs to recognize two antigens simultaneously may help overcome immune escape mechanisms and enhance the therapy's efficacy against diverse cancer types.
· Gene Editing Technologies: Utilizing advanced gene editing tools to precisely modify CAR-T cells could improve their activation, viability, and antitumor effects. These modifications aim to address and overcome mechanisms that tumors use to evade immune detection.
· CAR-NK Cells: In addition to CAR-T cells, CAR-NK (Natural Killer) cells are being investigated as a promising alternative. Enhanced NK cells, which bypass the inhibitory signals from killer immunoglobulin-like receptors, could potentially provide effective antitumor responses.
· Tumor Microenvironment: Research is focused on improving CAR-T cell survival and efficacy within the challenging tumor microenvironment. Strategies include targeting specific molecular markers or using antibodies on CAR-T cells to better navigate and overcome immunosuppressive elements in tumors.
Antibody-drug conjugates (ADCs) represent a cutting-edge approach in cancer treatment, designed to induce targeted cell death in cancer cells. Each ADC consists of three crucial components: an antibody, a payload, and a linker. The antibody is engineered to bind specifically to an epitope on the target substance, the payload is responsible for delivering a cytotoxic effect, and the linker connects the antibody to the payload. This targeted approach allows ADCs to selectively attack cancer cells while minimizing damage to healthy cells.
Since the clinical introduction of the first ADC, Gemtuzumab ozogamicin, in 2000, this class of drugs has rapidly advanced and proven to be highly effective. ADCs leverage their ability to deliver potent chemotherapeutic agents directly to cancer cells, demonstrating significant therapeutic activity in refractory diseases and enhancing progression-free survival (PFS) and overall survival (OS) in earlier stages of treatment.
As of January 2024, eleven ADCs have been approved by the FDA for use in treating various cancers, including acute myeloid leukemia, lymphoblastic leukemia, multiple lymphoma types, and cancers of the breast, stomach, lung, urothelium, cervix, and ovary. These ADCs have demonstrated enhanced anti-tumor efficacy compared to standard treatments across a broad range of indications. In addition to targeting antigen-expressing tumor cells, some ADCs have been engineered to extend cytotoxic effects to low-antigen-expressing or antigen-negative cells within heterogeneous tumors. However, ADCs are not without side effects. Common issues include myelosuppression due to the cytotoxic payload and unique effects related to specific tissue antigens, such as cardiac toxicity with HER2-targeting ADCs and hemorrhagic complications with Tisotumab vedotin, which targets tissue factor.
Developing tumor-specific antigens and highly specific, high-affinity monoclonal antibodies for antibody-drug conjugates (ADCs) presents significant challenges. While various tumor-specific antigens have been identified, including glycoproteins, extracellular matrix components, and cell surface proteins, several issues remain unresolved. High affinity for an antigen does not always translate to effective tumor penetration. Additionally, the distribution of cell surface antigen expression can influence the therapeutic window, and high antigen levels in a tumor do not guarantee that an ADC will be effective.
The complex pharmacokinetic and pharmacodynamic profiles of ADCs further complicate their design. The clearance of each component of an ADC is influenced by various factors. Antibodies, for instance, are primarily cleared through the mononuclear phagocyte system and recycling via the neonatal Fc receptor (FcRn), which prolongs their half-life by exporting them to extracellular compartments for recycling. Conversely, the cytotoxic payload is metabolized in the liver and then excreted through the kidneys or in feces, making it susceptible to variations in liver and kidney function, as well as potential drug-drug interactions.
Drug resistance is another significant challenge in ADC development. Resistance mechanisms include reduced efficacy of the payload, downregulation of antigen expression, and alterations in intracellular trafficking pathways. For example, ATP-binding cassette (ABC) transporters such as multidrug resistance 1 (MDR1), multidrug resistance-associated protein 1 (MRP1), and breast cancer resistance protein (BCRP) can actively pump out chemotherapeutic agents and common cytotoxic payloads like MMAE, DM1, and ozogamicin. Chronic exposure to T-DM1 can lead to downregulation of HER2 expression and upregulation of MDR1 and MRP1 drug efflux pumps. In contrast, T-DXd remains effective in HER2-positive cancers that express high levels of MRP2 and BCRP following resistance to T-DM1, due to its payload being a poor substrate for ABC transporters.
The ADC market is projected to grow substantially, from $6.8 billion in 2024 to $15.4 billion by 2029. This growth is driven by increased research and development collaborations between global pharmaceutical companies and biotechnology firms.
Building on the preclinical and clinical successes of existing ADCs and ongoing research, future advancements will focus on two main areas: enhancing ADC design and delivery and exploring novel payloads with immunotherapy or radiation properties.
One key direction is improving the tolerability and efficacy of ADCs to broaden their therapeutic window. Strategies under investigation include developing peptide-drug conjugates (PDCs), where peptides replace monoclonal antibodies, potentially allowing for better tumor penetration. Additionally, immune-stimulating antibody conjugates (ISACs) are being explored, which can trigger an influx of pro-inflammatory cytokines to activate dendritic cells and stimulate a robust anti-tumor T-cell response. Another innovative approach involves incorporating radioactive isotopes into ADCs to boost their cytotoxic activity.
References
1. Iyer KA, Ivanov J, Tenchov R, et al. Emerging Targets and Therapeutics in Immuno-Oncology: Insights from Landscape Analysis. J Med Chem. 2024;67(11):8519-8544.
2. Alturki NA. Review of the Immune Checkpoint Inhibitors in the Context of Cancer Treatment. J Clin Med. 2023;12(13):4301.
3. Sun Q, Hong Z, Zhang C, Wang L, Han Z, Ma D. Immune checkpoint therapy for solid tumours: clinical dilemmas and future trends. Signal Transduct Target Ther. 2023;8(1):320.
4. Johnson PC, Gainor JF, Sullivan RJ, Longo DL, Chabner B. Immune Checkpoint Inhibitors - The Need for Innovation. N Engl J Med. 2023;388(16):1529-1532.
5. Wang JY, Wang L. CAR-T cell therapy: Where are we now, and where are we heading?. Blood Sci. 2023;5(4):237-248.
6. Wang JY, Wang L. CAR-T cell therapy: Where are we now, and where are we heading?. Blood Sci. 2023;5(4):237-248.
7. Sun D, Shi X, Li S, Wang X, Yang X, Wan M. CAR‑T cell therapy: A breakthrough in traditional cancer treatment strategies (Review). Mol Med Rep. 2024;29(3):47.
8. Song CH, Jeong M, In H, Kim JH, Lin CW, Han KH. Trends in the Development of Antibody-Drug Conjugates for Cancer Therapy. Antibodies (Basel). 2023;12(4):72.
9. Gogia P, Ashraf H, Bhasin S, Xu Y. Antibody-Drug Conjugates: A Review of Approved Drugs and Their Clinical Level of Evidence. Cancers (Basel). 2023;15(15):3886.
Neeta Ratanghayra is a freelance medical writer, who creates quality medical content for Pharma and healthcare industries. A Master’s degree in Pharmacy and a strong passion for writing made her venture into the world of medical writing. She believes that effective content forms the media through which innovations and developments in pharma/healthcare can be communicated to the world.
Contact Us
Tel: (+86) 400 610 1188
WhatsApp/Telegram/Wechat: +86 13621645194
Follow Us: