What Immune cell therapy are being developed?

17 March 2025
Overview of Immune Cell Therapy

Immune cell therapy is an innovative and rapidly evolving branch of immunotherapy that harnesses the innate and adaptive functions of immune cells to mediate therapeutic effects against cancer, autoimmune diseases, infections, and other pathological conditions. The fundamental premise of these therapies is to either augment the natural ability of the immune system or to re-engineer cells ex vivo so that, upon re-administration, they can recognize and eliminate diseased or malignant cells. This overview section sets the stage by defining immune cell therapy and describing its underlying mechanisms, before outlining the various types currently under investigation.

Definition and Mechanism

At its core, immune cell therapy involves the isolation, modification (when needed), expansion, and reinfusion of immune cells into patients. These cells are often genetically reprogrammed or selected to enhance their specificity, cytotoxic potential, and ability to persist when facing the immunosuppressive tumor microenvironment. The general mechanism relies on three major aspects:

1. Recognition: Immune cells—whether they be T cells with native or engineered receptors, natural killer (NK) cells, or other effector cells—identify and interact with target antigens expressed on tumor cells or diseased tissues. In many cases, such recognition is mediated by receptors such as the T cell receptor (TCR) or by genetically introduced chimeric antigen receptors (CARs).

2. Activation: Once the target antigen is recognized, intracellular signaling cascades are initiated, leading to the activation of cytolytic processes. Cytokines, perforins, and granzymes are secreted to induce cell death in the targeted cancer cells or to modulate the local immune environment. Some therapies also incorporate costimulatory signals to reinforce sustained activation.

3. Expansion and Persistence: For effective in vivo activity, these immune cells must proliferate after reinfusion and persist over time. Strategies such as genetic modification of progenitor cells (e.g., using induced pluripotent stem cells in NK cell therapy) or integration of survival signals into CAR designs have advanced the longevity and reproducibility of these therapies.

Overall, immune cell therapies transform immune cells into “living drugs” that can adapt, propagate, and respond dynamically during treatment, thus providing enduring therapeutic benefits.

Types of Immune Cell Therapies

Several immune cell therapies are in development, each engineered to exploit distinct immunobiological principles. The main categories include:

- CAR-T Cell Therapy: T cells are genetically modified to express chimeric antigen receptors that recognize tumor-associated antigens independent of major histocompatibility complex (MHC) presentation.

- TCR-T Cell Therapy: T cells are re-engineered with specific T cell receptors (TCRs) that target intracellular antigens presented by MHC molecules, allowing these cells to recognize a wider array of tumor antigens.

- NK Cell Therapy: Natural killer cells, which are part of the innate immune system, can be used either in their native form or genetically modified (e.g., CAR-NK cells) to target malignant cells with high cytotoxic potential, often with fewer adverse events compared to T cell–based therapies.

Additional emerging modalities include cytokine-induced killer (CIK) cells, dendritic cell vaccines, and hybrid approaches combining cell therapies with gene editing or drug delivery systems. Each type exploits distinct recognition or functional pathways to overcome immune evasion and tumor heterogeneity. In this way, immune cell therapies represent a highly modular and adaptable platform for treating a wide range of diseases.

Current Developments in Immune Cell Therapy

Using advanced bioengineering and genetic modification techniques, researchers have made remarkable strides in the design, optimization, and therapeutic application of immune cell therapies. In this section, we review the current state of three core types of immune cell therapies under development: CAR-T, TCR-T, and NK cell therapies, while examining the strategies behind their continued evolution.

CAR-T Cell Therapy

CAR-T cell therapy has revolutionized the treatment of hematological malignancies and is being actively developed to address challenges in solid tumors as well. The workhorse of this field, CAR-T therapy involves modifying autologous T cells with a chimeric antigen receptor (CAR) that is typically composed of an extracellular single-chain variable fragment (scFv) to specifically bind target antigens and intracellular costimulatory domains such as CD28, 4-1BB, or ICOS that drive T cell activation and persistence.

Researchers have refined the design over several generations:
- First-generation CARs incorporated only the CD3ζ signaling domain, thereby offering limited in vivo expansion due to a lack of costimulatory signals.
- Second- and third-generation CARs have integrated additional costimulatory domains to improve proliferative capacity and durability, enhancing antitumor responses.
- Advanced designs now incorporate “smart” CARs that can adjust the intensity of signaling to reduce cytokine release syndrome (CRS) and off-target toxicity while also co-expressing molecules that improve cell persistence.

Current developments are not limited to hematologic cancers; many research groups are investigating strategies to overcome the limitations seen with solid tumors. Approaches include dual-targeting CARs, which recognize more than one antigen to prevent tumor escape due to antigen loss, and combination strategies that integrate checkpoint inhibitor mechanisms to bolster efficacy. Furthermore, methods focusing on manufacturing improvements using allogeneic cells, or “off-the-shelf” CAR-T cells, are emerging as a viable solution to reduce costs and variability associated with autologous cell collection. Recent patents discuss “enhancing and maintaining CAR-T cell efficacy” by incorporating features that reduce cytotoxic effects and prolong persistence in vivo. These innovations also address critical issues such as immune rejection via gene editing techniques and process optimization for large-scale manufacturing.

TCR-T Cell Therapy

Whereas CAR-T cells are designed to recognize surface antigens independently of MHC, TCR-T cell therapy is based on genetically modifying T cells with specific T cell receptors capable of recognizing intracellular antigens presented by the MHC. This allows TCR-T cells to target a broader array of proteins, including neoantigens and cancer-testis antigens such as NY-ESO-1, MART-1, and WT-1.

Recent advances in TCR-T therapy have focused on:
- Augmenting TCR affinity and specificity: Engineering high-affinity TCRs to enhance recognition of low-abundance antigens, particularly in solid tumors, while ensuring that off-target effects are minimized.
- Personalized neoantigen targeting: Emerging clinical trials are investigating the use of individualized TCR-T cells that are directed against patient-specific neoantigens derived from somatic mutations—a concept that holds promise for a more tailored and effective immune response.
- Technical improvements and safety switches: To avoid mispairing with endogenous TCR chains, researchers are exploring modifications such as single-chain TCRs and safety mechanisms (e.g., suicide genes) that can terminate the activity of TCR-T cells if adverse reactions occur.
- Overcoming MHC restrictions: Although TCR-T therapies inherently depend on the patient’s MHC genotype, efforts are underway to develop TCR-like chimeric receptors that may bypass such restrictions, thereby broadening the patient population eligible for these treatments.

TCR-T cell therapies have achieved encouraging clinical results in both hematological and solid tumors, though challenges remain concerning persistence, on-target off-tumor toxicity, and scalability.

NK Cell Therapy

Natural killer (NK) cell therapy is gaining increasing attention due to the innate cytotoxic potential of NK cells and their ability to mediate antitumor responses without prior sensitization. Unlike T cells, NK cell mechanisms do not require antigen presentation by MHC, meaning they are less affected by tumor downregulation of these molecules—a common mechanism of immune evasion.

Current developments in NK cell therapy include:
- CAR-NK cells: Similar to CAR-T cell strategies, NK cells are being modified with chimeric antigen receptors to enhance their specificity and cytotoxic functions. CAR-NK cells have shown promising preclinical activity and are being evaluated in multiple clinical trials. They tend to have a favorable safety profile, as NK cells are less likely to cause cytokine release syndrome and graft-versus-host disease compared to CAR-T cells.
- Allogeneic NK cell sources: NK cells can be obtained from peripheral blood, umbilical cord blood, induced pluripotent stem cells (iPSCs), and established NK cell lines. Off-the-shelf NK cell products overcome the limitations of subject-specific therapies and offer streamlined manufacturing processes.
- Enhancement of cytotoxicity and persistence: Efforts include genetic engineering to improve antibody-dependent cell-mediated cytotoxicity (ADCC) through modifications of CD16 receptors and deletion of inhibitory receptors or molecules that limit NK cell persistence. Approaches such as CRISPR/Cas9 gene editing have been successfully employed to increase NK cell efficiency.
- Combination with cytokine therapies: Administering cytokines such as IL-15 can help stimulate NK cell expansion and maintenance, an approach also reflected in some current clinical investigations.

Recent patents emphasize methods for producing CAR-NK cells as well as combinations that incorporate universal chimeric antigen receptors to target multiple antigens across tumor types. These design improvements and manufacturing strategies suggest that NK cell therapy is on a path to become a mainstream component of future immunotherapeutic regimens.

Clinical Trials and Research

In parallel with the development of the three main immune cell therapy approaches, extensive clinical research has been undertaken to evaluate safety, efficacy, and optimal dosing regimens. Both large-scale, multi-center trials and early-phase clinical studies are exploring these new modalities.

Ongoing Clinical Trials

Numerous clinical trials are registered worldwide focusing on the application of immune cell therapies. For example:

- CAR-T Cell Therapy Trials: Clinical trials for CAR-T cell therapies have rapidly expanded in recent years. Early-phase and pivotal trials targeting CD19, BCMA, and other tumor-associated antigens have achieved high response rates in hematologic malignancies. Recently, trials have extended to solid tumors using dual-targeting and combinatorial approaches. Safety improvements such as engineered safety switches and optimized manufacturing processes are being evaluated continuously as well as approaches to reduce adverse events such as CRS.

- TCR-T Cell Therapy Trials: Several phase I and II trials are examining the safety and efficacy of TCR-T cell therapies in patients with cancers that express tumor-associated antigens such as NY-ESO-1, WT-1, and MAGE-A4. Some trials are focusing on targeting neoantigens with personalized TCRs while others assess strategies to improve receptor affinity, persistence, and safety.

- NK Cell Therapy Trials: Clinical trials for NK cell therapies, particularly for CAR-NK cells, are being initiated, especially for patients with both hematologic and solid tumors. Off-the-shelf approaches and the use of NK cells derived from iPSCs or cord blood have been registered to test safety, persistence, and antitumor activity. The early phase data suggest that NK cell-based therapies might have fewer side effects than T-cell therapies and could complement other modalities in combination studies.

Overall, these trials are addressing key issues such as the durability of responses in patients, strategies for reducing toxicities, optimal dosing and preconditioning regimens, and manufacturing challenges that must be overcome for broader clinical adoption.

Recent Research Findings

Recent research from the Synapse database has provided several new insights into immune cell therapies:

- Mechanistic Insights: A range of studies have examined how modifications to CAR structure, costimulatory signals, and the inclusion of safety switches improve both activation and persistence of CAR-T cells. Similarly, new approaches for TCR-T cell design have focused on enhancing antigen specificity while minimizing off-target toxicity.

- Genetic Engineering Innovations: Advanced gene-editing techniques, including CRISPR/Cas9, have enabled researchers to knock out endogenous receptors that might lead to mispairing (a concern particularly with TCR-T cell therapies) and to insert genes that enhance cell function and survival. Such innovations also assist in constructing allogeneic cell products that reduce production times and costs.

- Biomaterials and Manufacturing: Innovative biomaterial platforms have been designed to aid in the ex vivo expansion, delivery, and persistence of immune cell therapies. Nanoparticle-based systems and scaffold technologies, for instance, are being explored to improve the in vivo kinetics of CAR-T cell products, offering more controlled release and targeting capabilities.

- Combination Approaches: Researchers are increasingly looking at combination therapies where immune cell therapies are paired with cytokines, checkpoint inhibitors, or even other cell therapy modalities. Such combinations have been shown to enhance efficacy while potentially reducing the dosage of each therapeutic component, which might reduce adverse events.

- Universal and Multi-antigen Targeting Strategies: The development of universal CAR constructs and reverse universal chimeric antigen receptors allow for a broader targeting spectrum. This approach is particularly important in dealing with tumor heterogeneity and potential antigen escape, and innovative patents have been filed describing such methods.

These findings underscore the multifaceted nature of immune cell therapy research, combining deep mechanistic studies with translational research to push toward more effective and safer treatments.

Challenges and Future Directions

Despite the remarkable progress in immune cell therapy, several technical, biological, and regulatory challenges remain. Researchers are continuously striving to improve the safety profile, efficacy, and scalability of these therapies while mitigating their potential risks.

Technical and Biological Challenges

Immune cell therapies face numerous hurdles:

- Manufacturing and Scalability: The production of autologous therapies, particularly CAR-T cells, is technically complex and patient-specific, leading to variability and high costs. The development of “off-the-shelf” solutions such as allogeneic CAR-T or CAR-NK cells is one strategy to overcome these challenges, but issues with immune rejection and persistence remain.

- Safety Concerns: Off-target activity, cytokine release syndrome (CRS), neurotoxicity, and other adverse events are significant concerns, especially with CAR-T cell therapies. Although safety switches and modified costimulatory domains help mitigate these issues, balancing potent antitumor activity with safety is an ongoing challenge. TCR-T therapies also have inherent risks related to MHC-restricted recognition, which can lead to on-target, off-tumor toxicities if antigens are not strictly tumor-specific.

- Tumor Microenvironment (TME): Solid tumors in particular present a hostile microenvironment that can impair immune cell homing, persistence, and cytotoxic function. Factors such as immunosuppressive cytokines, regulatory cells, and physical barriers all impact the efficacy of infused immune cells. Strategies incorporating checkpoint blockade or localized cytokine release are being explored to overcome these obstacles.

- Antigen Heterogeneity and Escape: The phenomenon of antigen loss or downregulation in tumor cells can lead to relapse after immune cell therapy. Multi-antigen targeting approaches (such as dual or trivalent CAR constructs) and the use of TCRs against shared intracellular antigens are methods being used to minimize this risk.

- Persistence and Exhaustion: Ensuring long-term persistence and preventing exhaustion of adoptively transferred cells is critical for durable responses. Research is focusing on integrating genes that improve cell survival and using biomarkers to monitor exhaustion status, thereby optimizing the dosing and re-administration strategies.

- Immunogenicity and Alloreactivity: For allogeneic approaches, the risk of graft-versus-host disease (GvHD) is lower in NK cell therapies compared to T cell–based therapies, yet precise gene editing must be carefully performed to avoid unintended immune reactions. Overcoming these challenges requires innovative genetic engineering techniques and better preclinical models.

Future Prospects and Innovations

Despite these daunting challenges, the field holds enormous promise and is poised for further breakthroughs:

- Gene Editing and Precision Engineering: Innovations in CRISPR/Cas9 and other gene-editing tools are expected to further refine immune cell therapies. These techniques can be used to insert safety switches, enhance specificity, and downregulate inhibitory receptors, thus optimizing immune cell function while minimizing adverse effects. The incorporation of universal CAR constructs and TCR-like CARs represent additional advances that may broaden the scope of patients who can benefit from these therapies.

- Universal/Allogeneic Cell Platforms: Off-the-shelf products, including allogeneic CAR-T and CAR-NK cells, are under active investigation. These products use cells derived from healthy donors or engineered stem cells, providing a more consistent and scalable solution. Universal immune cell products can potentially reduce the time to treatment and lower production costs – a major step forward in making therapies widely accessible.

- Combination Therapies: Future clinical strategies are likely to incorporate immune cell therapy in combination with other therapeutic modalities such as immune checkpoint inhibitors, targeted small molecules, or even conventional chemotherapy. This combinatorial approach might simultaneously attack multiple pathways involved in tumor survival or immune suppression, leading to improved clinical outcomes.

- Biomaterials and Drug Delivery Systems: The application of biomaterials to improve the delivery and persistence of immune cells in vivo is a promising area. Nanoparticle carriers and scaffolds that can release cytokines or create immunomodulatory niches around the tumor are under investigation. These engineering solutions can protect immune cells from the harsh tumor microenvironment and improve their homing to target sites.

- Personalized Neoantigen-Based Therapies: TCR-T cell therapies targeting patient-specific neoantigens have shown early promise in clinical trials. As sequencing and computational methods improve, generating personalized cell therapies that target unique mutational profiles of a patient’s tumor could lead to highly effective and individualized treatments.

- Next-Generation Immune Cell Modifications: Ongoing work in synthetic biology and systems immunology promises to deliver smart cell therapies that can sense multiple inputs in the tumor microenvironment and adjust their behavior accordingly. For instance, engineered immune cells that secrete specific cytokines only when encountering tumor cells could minimize systemic toxicities while maintaining robust local antitumor activity.

- Expanding Applications Beyond Cancer: Although cancer remains the primary focus, cell therapies are also being developed for infectious diseases, autoimmune disorders, and even regenerative medicine. For example, therapies based on abdominal cavity cells for autoimmune conditions and immune cell sequencing approaches for the treatment of antibody-mediated diseases are being explored.

- Regulatory and Quality Control Innovations: With the rapid evolution of these complex therapies, the regulatory landscape is also evolving. Efforts to standardize manufacturing, improve quality control, and develop predictive biomarkers for response and toxicity will help ensure that next-generation immune cell therapies are safe, effective, and accessible.

In summary, each of these innovations is making immune cell therapies more potent, versatile, and safer. They offer hope for improved treatment outcomes not only in hematological malignancies but also increasingly in solid tumors and immune-mediated diseases.

Conclusion

In conclusion, a diverse spectrum of immune cell therapies is currently being developed across multiple platforms. Immune cell therapies—ranging from CAR-T and TCR-T to NK cell-based approaches—represent a new generation of “living drugs” that harness both the innate and adaptive branches of the immune system to combat complex diseases such as cancer, autoimmune disorders, and infections. The broad definition of immune cell therapies covers genetically engineered T cells (CAR-T and TCR-T), natural killer cells (including CAR-NK cells), as well as other emerging immunomodulatory strategies that involve engineered cell populations.

Current developments emphasize sophisticated genetic reprogramming techniques, improvements in manufacturing through “off-the-shelf” allogeneic platforms, and innovative safety features. Significant clinical trials have already established the efficacy of these therapies in hematologic malignancies, while ongoing trials and preclinical studies are extending their application to solid tumors and other disease indications. Researchers are addressing critical challenges such as immune cell persistence, toxicity management, antigen heterogeneity, and tumor microenvironment interference by combining precision gene-editing, biomaterials, and combination therapy strategies.

Despite technical and biological challenges—ranging from manufacturing complexities, immunogenicity, and safety concerns to the hostile tumor microenvironment—the future prospects for immune cell therapy remain promising. Advances in personalized medicine, next-generation gene editing, and universal cell platforms signal a future where immune cell therapies are more accessible, effective, and versatile. As research continues to integrate clinical insights with cutting-edge technology, immune cell therapies will likely become a mainstay in the treatment of cancers, autoimmune diseases, and beyond.

This detailed examination illustrates that immune cell therapy is not a single approach but an evolving constellation of technologies. By combining robust preclinical data, carefully designed clinical trials, and ongoing innovations in cell engineering, the field is moving steadily towards personalized, scalable, and safer therapeutic solutions. Continued interdisciplinary research and collaboration between biotechnology, clinical medicine, and regulatory bodies will be essential to translate these promising advances into widely available treatments that can bring long-term clinical benefits to patients worldwide.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.