For what indications are Immune cell therapy being investigated?

17 March 2025
Introduction to Immune Cell Therapy

Immune cell therapy represents a revolutionary treatment modality that harnesses the body's own immune system to target and eliminate disease-causing cells. Over the past few decades, improvements in cell engineering, genetic modification, and a deeper understanding of immunology have paved the way for the development and clinical application of these therapies across a wide spectrum of indications. In the following sections, we detail the definition, basic mechanism of action, historical milestones, and significant research progress that have shaped this emerging field.

Definition and Mechanism of Action

Immune cell therapy involves the isolation, modification, expansion, and re-infusion of immune cells—such as T lymphocytes, natural killer (NK) cells, macrophages, and dendritic cells—into patients to target pathogens, malignant cells, or cells involved in autoimmunity. The fundamental mechanism of action is based on either directly killing diseased cells or modulating the patient’s immune response by overcoming the tumor-induced or pathogen-induced suppression of the immune system. For example, chimeric antigen receptor (CAR)-T cells are engineered to express receptors that recognize specific antigens on tumor cells, triggering cytotoxic activity upon engagement, while tumor infiltrating lymphocytes (TILs) are isolated, expanded, and reinfused to amplify the natural anti-tumor immune response. This process may involve steps such as genetic modification to knock out inhibitory or immunogenic markers (e.g., HLA molecules in allogeneic settings) and integration of costimulatory signals to enhance persistence and functionality.

Historical Development and Milestones

The journey of immune cell therapy began with early observations of the immune system’s ability to recognize and destroy diseased cells. Initial clinical attempts, such as adoptive cell transfer of lymphocytes in melanoma, laid the groundwork for more sophisticated approaches when researchers realized that immune cells could be manipulated ex vivo for more potent therapeutic effects. Milestones include the approval of the first CAR-T cell therapies for B-cell malignancies in 2017, which marked an era where “living drugs” became an integral part of oncology treatment. Over the years, the field expanded from autologous therapies drawn from the patient’s own peripheral blood to allogeneic products derived from healthy donors, as well as products obtained from pluripotent stem cells that can be engineered with precision. These advancements have broadened the potential application of immune cell therapies to a diverse range of conditions, setting the stage for enhanced clinical usability, decreased heterogeneity of manufactured products, and increased scalability.

Current Indications for Immune Cell Therapy

Investigation into immune cell therapies spans multiple areas of disease management. The indications being explored today can generally be grouped into three major categories: cancer treatment, autoimmune diseases, and infectious diseases. Each of these indications involves a distinct underlying pathology and immune dysregulation, and innovative cell therapies are tailored to address these challenges using both traditional and novel engineering approaches.

Cancer Treatment

Cancer remains the most advanced field for immune cell therapy, driven by the remarkable efficacy observed in hematologic malignancies and an emerging focus on solid tumors.

Hematologic Cancers and Lymphoid Malignancies:
CAR-T therapies targeting antigens such as CD19 have demonstrated dramatic clinical responses in B-cell leukemias and lymphomas. In these cases, the engineered T cells (CAR-T cells) are infused into patients with relapsed or refractory disease, leading to rapid and durable tumor regression. Recent approvals of therapies like Tisagenlecleucel and Axicabtagene Ciloleucel underscore the clinical success of immunotherapies in hematologic cancers. These therapies utilize receptor-modified T cells that express costimulatory domains and have shown response rates of up to 80% in selected patient populations.

Solid Tumors:
Solid tumors present unique challenges due to the immunosuppressive tumor microenvironment, heterogeneous antigen expression, and physical barriers that restrict immune cell infiltration. Despite these challenges, immune cell therapies are under investigation for various solid cancers. For instance, tumor infiltrating lymphocyte (TIL) therapy involves harvesting lymphocytes directly from the tumor, expanding them ex vivo, and reintroducing them to mount a strong localized immune response. Novel engineered products such as CAR-T cells targeting antigens expressed on solid tumors, combined with approaches to modulate the tumor microenvironment, are being evaluated in clinical trials. Some studies also highlight the use of innovative platforms like CAR-M cell therapy and CAR-NK cell therapy, which may provide the benefits of both innate and adaptive immunity with reduced toxicity, all aimed at overcoming the resistance mechanisms present in solid tumors.

Specific Tumor Types:
Immune cell therapies have been investigated in melanoma, lung cancer, and hepatocellular carcinoma (HCC), among others. Melanoma was one of the first solid tumors for which adoptive cell therapy was applied, yielding encouraging results even in advanced stages. In HCC, the utilization of immunotherapeutic strategies such as CAR-T and TIL therapy is still in the early phases but holds promise for overcoming the limitations of traditional systemic therapies. Moreover, immune cell therapy is also being explored for other malignancies where there is a high unmet need, including gastrointestinal cancers, cervical cancer, and potentially prostate cancer, through methods that enhance immune cell infiltration and function within the tumor microenvironment.

Autoimmune Diseases

Autoimmune diseases represent another critical area for immune cell therapy investigation. Unlike cancer, where the goal is to eliminate malignant cells, therapies for autoimmune conditions aim to restore immune tolerance and modulate an aberrant immune response without compromising overall host defense.

B-cell and T-cell Directed Therapies:
In autoimmune diseases such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), multiple sclerosis (MS), and neuromyelitis optica spectrum disorder (NMOSD), autoreactive immune cells drive the pathology. Recent developments include the use of CAR-T therapies to selectively target autoreactive B cells (e.g., CD19-directed CAR-T cells), which have shown rapid and sustained depletion of pathological B cells leading to remission in refractory cases. Clinical trials have demonstrated not only depletion of autoreactive cells but also modulation of downstream inflammatory cascades that result in durable responses.

Regulatory Cell Therapies:
Adoptive transfer of regulatory T cells (Tregs) is also under investigation to reinstate immune tolerance. These cells work by countering pro-inflammatory responses and promoting an anti-inflammatory milieu. Investigational treatments have included expanding patient-derived Tregs ex vivo and reinfusing them, as well as engineering Tregs with chimeric autoantibody receptors (CAAR-T cells) to selectively dampen harmful immune responses.

Stem Cell-Based Approaches:
Hematopoietic stem cell transplantation (HSCT) has been studied in several autoimmune conditions to essentially “reset” the immune system. Approaches involving mesenchymal stem cells (MSCs) offer the potential not only to modulate immune responses but also to repair tissue damage caused by chronic inflammation. Early reports in MS-like diseases and other refractory autoimmune conditions have shown promising outcomes in terms of reducing disease activity and improving clinical scores.

Emerging Applications in Neurological Autoimmunity:
More recently, indications such as NMOSD have been targeted with autoantigen-specific cell therapies. For instance, innovative clinical trials using CAR-T therapy directed against specific antigens implicated in the autoimmunity of the central nervous system have yielded encouraging preliminary results, with improvements in clinical status and immune modulation. Additional investigations into myasthenia gravis and immune-mediated necrotizing myopathy further illustrate the broadening scope of immune cell therapies in autoimmunity.

Infectious Diseases

Although historically overshadowed by cancer and autoimmune applications, the use of immune cell therapies in infectious diseases is an area of rapidly emerging interest.

Viral Infections:
Refractory viral infections, especially in immunocompromised hosts, represent a promising area for immune cell therapy. For example, CAR-T cells and other engineered immune cells are being explored for the control of persistent viral infections such as HIV, cytomegalovirus (CMV), and hepatitis. These studies take advantage of engineered cells’ ability to recognize and eliminate virus-infected cells, thereby complementing or even substituting existing antiviral strategies.

Opportunistic and Fungal Infections:
Opportunistic infections, which often complicate the clinical course of immunocompromised patients—such as those with hematologic malignancies or post-transplant—also represent a potential target for immune cell therapies. Investigations into using CAR-modified T cells or NK cells to target fungal pathogens, or to enhance overall immune responsiveness, are underway. Early preclinical data highlight the potential for engineered immune cells to provide targeted protection in these difficult-to-treat infections.

Combination Approaches:
Beyond targeting a single pathogen, there is growing interest in designing multi-specific immune cell products that can be rapidly adapted or combined with small molecule modulators, vaccines, or checkpoint inhibitors to enhance their effect against a broad spectrum of infectious agents. This combinatorial approach could be particularly beneficial in outbreaks where rapid immune enhancement is critical.

Research and Clinical Trials

The active investigation of immune cell therapies is supported by an extensive portfolio of clinical trials and research studies, ranging from early-phase explorations to large, multicenter trials. This global effort is not only validating the efficacy of current therapies but also expanding the list of conditions for which these therapies might be applied.

Ongoing Clinical Trials

The current clinical trial landscape demonstrates a robust pipeline for immune cell therapies across various indications. In oncology, numerous trials are evaluating novel CAR-T therapies targeting new antigens, TIL therapies for solid tumors, and next-generation products such as CAR-NK cells to reduce toxicity and overcome resistance. These trials are being conducted internationally with standardized protocols that monitor both efficacy and safety outcomes such as cytokine release syndrome (CRS) and neurotoxicity.

Similarly, trials investigating immune cell therapies in autoimmune diseases are underway. These include studies on CAR-T cell–mediated depletion of autoreactive B cells in SLE and rheumatoid arthritis, and early-phase trials examining the adoptive transfer of Tregs or engineered regulatory cells in diseases such as multiple sclerosis. Some trials have already shown promising preliminary results in terms of immune modulation and reduction in disease activity, reinforcing the potential of these therapies to provide long-term remission in patients who are refractory to conventional treatments.

In the infectious disease arena, early-stage clinical studies have begun testing the safety and feasibility of immune cell therapies in chronic viral infections and opportunistic infections in immunocompromised patients. Although this is a relatively new application, the success of CAR-T therapies in oncology has spurred interest in applying similar approaches to manage viral load and eradicate infected cells without causing widespread immune suppression.

Emerging Indications

Emerging evidence suggests that immune cell therapies may extend beyond conventional indications. Research is currently exploring the potential application of these therapies in conditions such as chronic inflammatory diseases, graft-versus-host disease (GVHD), and even certain degenerative diseases where immune dysregulation plays a contributory role. Moreover, the evolving field of precision medicine, aided by advanced genetic sequencing and bioinformatics, is enabling the design of personalized immune cell products tailored to an individual’s unique immunological profile.

Another promising avenue is the use of pluripotent stem cells (PSCs) to generate standardized, “off-the-shelf” immune cells that could be deployed rapidly in various clinical settings. The capacity of PSCs to provide a near-unlimited supply of immune effector cells, combined with precise genome editing techniques, holds great promise for democratizing access to cell therapies while minimizing manufacture-related variability. Additionally, combinatorial therapies that integrate cell therapy with checkpoint inhibitors, small molecule drugs, or regenerative medicine approaches are emerging to further enhance therapeutic outcomes, particularly in resistant or poorly responsive disease states.

Challenges and Future Directions

Despite the rapid progress and promising clinical results attained with immune cell therapies, several challenges remain. Addressing these challenges is critical to broadening the therapeutic spectrum and ensuring that these modalities reach their full potential in clinical practice.

Current Challenges in Therapy Development

Safety and Toxicity:
One of the major challenges in immune cell therapy is managing treatment-related toxicity. Conditions such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) have been recurrent complications in CAR-T cell therapy. Although clinical protocols have evolved to mitigate these risks, they still represent significant hurdles, particularly when extending these therapies to solid tumors or autoimmune conditions where the immune environment is already dysregulated.

Manufacturing and Scalability:
Another significant barrier is the complexity, cost, and variability associated with the manufacturing of autologous immune cell therapies. Producing a patient-specific product involves lengthy processes of cell isolation, expansion, and genetic modification, which contribute to high costs and limit patient accessibility. Efforts to develop allogeneic and PSC-derived products are ongoing, but challenges such as host-versus-graft (HvG) reactions, immune rejection, and ensuring long-term stability of engineered cells remain.

Heterogeneity and Persistence:
The heterogeneity of patient-derived immune cells leads to variability in product efficacy. Furthermore, ensuring that the infused cells persist long enough to achieve durable therapeutic responses—and do so without causing off-target effects or uncontrolled proliferation—is a delicate balance. Addressing these issues involves refining gene editing and cell expansion technologies to generate uniform, high-quality cell populations.

Immunosuppressive Microenvironment:
In solid tumors and certain autoimmune diseases, the immunosuppressive microenvironment poses a significant obstacle to the efficacy of immune cell therapies. Tumor cells and autoreactive processes can actively inhibit the function and survival of therapeutic cells. Novel strategies such as combining cell therapy with modulators of the immune microenvironment or using targeted delivery methods are under investigation to overcome these challenges.

Future Prospects and Research Directions

Integration of Advanced Genomic and Bioengineering Techniques:
Future research is likely to be driven by advances in genetic engineering, synthetic biology, and bioinformatics. Precision editing using CRISPR/Cas9 and other genome-modifying tools can improve the safety, persistence, and function of immune cell products. These advancements will help create cells that are not only more effective but also have an improved safety profile.

Development of Universal Donor Cells:
Another promising direction is the creation of “universal” or allogeneic immune cell therapies. The development of PSC-derived immune cells that have been genetically modified to reduce immunogenicity can lead to a standardized, off-the-shelf product that is accessible to a larger patient population. This approach also presents the opportunity to rapidly deploy therapies in acute settings, such as severe infections or rapidly progressing cancers.

Combinatorial Therapeutic Approaches:
Combining immune cell therapy with other modalities such as checkpoint inhibitors, small molecule drugs, or even regenerative medicine technologies is an area of intense research. Such combination therapies may provide synergistic effects that can overcome the limitations of single-agent approaches. For instance, pairing CAR-T cell therapy with targeted modulation of the tumor microenvironment could enhance efficacy in solid tumors where traditional CAR-T cells have struggled.

Personalized and Precision Medicine:
With the advances in patient profiling and biomarker identification, personalized immune cell therapies that are tailored to an individual’s immunological landscape are becoming feasible. By integrating genomic, proteomic, and clinical data, researchers can design more precise therapies that minimize off-target effects while maximally harnessing the immune system’s potential. This personalized approach is particularly promising in autoimmune diseases, where the precise nature of the pathogenic immune cells can be identified and targeted.

Expanding to New Indications:
While cancer, autoimmune diseases, and infectious diseases currently dominate the investigation of immune cell therapies, emerging applications may include treatment of chronic inflammatory conditions, degenerative diseases, and even applications in transplant medicine to reduce rejection risks. The continuous evolution of immune cell technology is likely to uncover novel targets and mechanisms that could be exploited therapeutically.

Conclusion

In summary, immune cell therapy is a rapidly evolving field that is being investigated for a wide array of indications. Starting with foundational principles that harness the immune system to either eradicate tumors or re-establish immune tolerance, the research and clinical application of immune cell therapies have progressed enormously over the past decades.

In the realm of cancer treatment, immune cell therapy has already transformed the therapeutic landscape with highly successful CAR-T cell and TIL therapies for hematologic malignancies, while further research into solid tumors promises to overcome existing limitations. In parallel, the investigation into autoimmune diseases is exploring innovative approaches such as autoreactive B cell depletion, regulatory cell infusions, and hematopoietic stem cell transplantation to restore immune homeostasis. Although the field of infectious diseases remains at an early stage, significant potential exists in applying engineered immune cells to control chronic and opportunistic infections, particularly in immunocompromised patients.

The current pipeline of clinical trials and research efforts reflects both a maturing understanding of immune cell biology and a determination to expand these therapies into new indications. Ongoing studies aim not only to optimize efficacy and safety profiles but also to resolve manufacturing and scalability challenges that currently limit patient access. Furthermore, future directions include the development of universal donor cells, integration of precision medicine strategies, and combinatorial approaches that may provide synergistic benefits.

Overall, while numerous challenges remain—including managing treatment-related toxicities, ensuring long-term cell persistence, overcoming immunosuppressive environments, and reducing manufacturing heterogeneity—the future of immune cell therapy is bright. Advances in genome editing, synthetic biology, and personalized medicine are set to propel the field forward, ultimately transforming the treatment landscape for cancer, autoimmune disorders, infectious diseases, and potentially a host of other conditions. With continued interdisciplinary research and clinical innovation, immune cell therapies may soon provide durable, targeted, and safe treatment options for millions of patients worldwide.

In conclusion, immune cell therapies are being investigated across multiple indications from cancer to autoimmune diseases and infectious challenges, representing a paradigm shift in treatment strategies. The integration of comprehensive translational research, advanced cell engineering techniques, and innovative clinical trial designs is essential to overcoming the current obstacles and realizing the full therapeutic potential of these "living drugs" in a personalized and broadly accessible manner.

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.