What are the different types of drugs available for TIL therapy?

17 March 2025
Introduction to TIL Therapy

Definition and Mechanism
Tumor-infiltrating lymphocyte (TIL) therapy is an adoptive cell transfer (ACT) immunotherapy that exploits a patient’s own immune cells harvested directly from the tumor microenvironment, expanded ex vivo, and re-infused into the patient to elicit an anti-tumor response. TILs naturally encompass a heterogeneous mixture of T cells that have recognized tumor-specific or neoantigenic determinants. Their inherent recognition of mutated self-antigens makes them particularly attractive agents for fighting cancers that are refractory to standard therapies. The mechanism of TIL therapy involves isolating lymphocytes from tumor fragments, stimulating and expansively culturing these lymphocytes—often in a cytokine-enriched environment—and then reintroducing the enhanced, tumor-reactive cells into the patient following a preparative lymphodepleting regimen. The success of such therapy is based on the intrinsic anti-tumor specificity of TILs coupled with external modulation by cytokines, costimulatory signals, and checkpoint blockade agents.

Historical Development and Milestones
Since the pioneering work in the 1980s and the landmark clinical studies led by Rosenberg and colleagues, utilization of TIL therapy has risen from small-scale experimental procedures to more standardized clinical protocols. Early clinical trials using high-dose interleukin-2 (IL-2) as an adjunct post-TIL infusion have demonstrated objective response rates in metastatic melanoma, laying the groundwork for improvements in TIL expansion and manufacturing techniques. Over time, modifications—such as the incorporation of lymphodepletion prior to TIL infusion, the refinement of cytokine dosages, and the combination with checkpoint inhibitors—have been introduced to enhance the persistence and anti-tumor efficacy of these cells. The evolution has also encompassed the engineering of TILs to incorporate surface-associated immunomodulatory agents and even streamlined expansion protocols that reduce culture time and mitigate cell exhaustion. Today, TIL therapy is viewed as an important personalized treatment modality with the potential to treat a broad range of solid tumors, exemplifying not only a clinical milestone but also a platform for integrating immunomodulatory drugs into cellular therapy.

Classification of Drugs in TIL Therapy

Immunomodulatory Drugs
In the context of TIL therapy, immunomodulatory drugs are employed both as adjunct agents to boost the in vivo efficacy of the expanded lymphocytes and, in some cases, to be physically associated with the TIL product.
- Surface-Associated Immunomodulators: One innovative approach involves the attachment of cytokines or immunomodulatory agents directly onto the TILs’ surface. For instance, certain compositions incorporate cytokine molecules on the cell membrane of TILs to provide localized, sustained immunostimulation once re-infused into the patient. This strategy aims to improve T cell survival, proliferation, and anti-tumor activity without systemic exposure to high-dose cytokines.
- Checkpoint Inhibitors: Immunomodulatory drugs such as anti-PD-1, anti-PD-L1, and anti-CTLA-4 antibodies have been widely studied for their ability to “release the brakes” on T cells, thereby enhancing the anti-tumor response of TILs. Combining checkpoint inhibitors with TIL therapy has shown promise, with studies demonstrating improved and even shortened TIL expansion protocols that increase therapeutic efficacy. The use of checkpoint inhibitors is especially significant given that TILs often express inhibitory receptors (e.g., PD-1, CTLA-4) as a consequence of chronic antigen exposure; these drugs counteract depletion and functional exhaustion.
- Targeted Signaling Modulators: Drugs that target intracellular signaling pathways, such as AKT inhibitors, are sometimes used during the TIL expansion process. These pharmacologic agents can modulate T cell differentiation and help maintain a less-differentiated, more “juvenile” T cell phenotype that correlates with better in vivo persistence and anti-tumor effect.
- Targeted Small Molecule Inhibitors: Additionally, targeted therapies such as BRAF inhibitors, MEK inhibitors, and KRAS inhibitors have been combined with TIL therapy, especially in tumors harboring specific mutations. For example, treatment strategies involving TILs have been combined with BRAF and MEK inhibitors in the context of V600 mutation-induced cancers to further potentiate the tumoricidal response. Similarly, emerging strategies are assessing the combination of TIL therapy with KRAS inhibitors, particularly for tumors with KRAS mutations, to overcome potential resistance pathways.
- Engineered Cytokine Receptor Agonists: Some recent patent disclosures highlight the use of IL-15 receptor agonists and engineered cytokine receptor pairs to endow TILs with enhanced metabolic health and sustained anti-tumor function. This approach combines both the targeted delivery of cytokine signals along with precision engineering to optimize TIL efficacy, serving as an immunomodulatory modality that is both biological and chemical in nature.

Cytokines and Growth Factors
Cytokines play a central role in the ex vivo expansion and in vivo sustenance of TILs. They are critical to promote proliferation, activation, and persistence while combating the immunosuppressive tumor microenvironment.
- Interleukin-2 (IL-2): Historically, IL-2 remains one of the most important cytokines in TIL therapy. High-dose IL-2 has been used to support TIL expansion and reactivation post-infusion; however, its administration is also associated with significant systemic toxicity. Modified regimens using a lower IL-2 dosage or alternative schedules have been explored to balance efficacy and adverse effects.
- Interleukin-15 (IL-15): IL-15 is a cytokine known to enhance the survival and cytotoxic function of CD8+ T cells. Studies have demonstrated that IL-15 not only supports T cell proliferation but also improves metabolic fitness. IL-15-based treatments are increasingly combined with other immunotherapeutic agents, such as TIGIT blockade, to further boost anti-tumor activity in solid tumors like lung adenocarcinoma.
- Interleukin-18 (IL-18): IL-18, often used in synergy with IL-15 or IL-12, has been reported to upregulate interferon-gamma (IFN-γ) secretion and enhance the cytotoxic function of TILs. In vitro co-stimulation studies with IL-15 and IL-18 have revealed synergistic activation of TILs, leading to enhanced cytotoxicity and improved anti-tumor responses.
- Interleukin-12 (IL-12): IL-12 is a potent Th1 cytokine that can pivot the T cell response toward cytotoxicity. When used in combination with IL-18, it exhibits synergistic effects on activating TILs by boosting pro-inflammatory cytokine secretion (such as IFN-γ and TNF-α) and downregulating inhibitory cytokines like IL-10.
- Combination and Engineered Cytokines: Recent developments involve the simultaneous utilization or even engineered versions of cytokines (e.g., PEGylated IL-10 or tailored IL-15 receptor agonists) to create a more favorable cytokine milieu that supports TIL anti-tumor responses while minimizing systemic adverse events. These approaches aim to uncouple the need for high systemic cytokine doses by enriching the TIL infusion product with locally acting cytokine signals.

Mechanism of Action

How Drugs Enhance TIL Efficacy
The various drugs available for TIL therapy serve to enhance the efficacy of the adoptively transferred lymphocytes through several well‐documented mechanisms:
- Local Immunostimulation: The attachment of cytokines directly onto the TIL surface (as in formulations discussed in patent) allows for a sustained, localized delivery of immunostimulatory signals. This not only aids in maintaining TIL viability and proliferation upon infusion but also helps to recruit other effector immune cells to the tumor microenvironment in a paracrine manner.
- Checkpoint Blockade: By targeting inhibitory molecules such as PD-1, CTLA-4, and PD-L1, checkpoint inhibitors reinvigorate exhausted TILs. This enhances their cytotoxic function and in vivo persistence. When used in combination with TIL therapy, checkpoint inhibitors have the potential to convert otherwise hypofunctional T cells into highly active effector cells capable of mediating durable tumor regression.
- Prevention of Terminal Differentiation: Agents such as AKT inhibitors are employed during the ex vivo expansion process to help preserve a less-differentiated, memory-like phenotype of T cells. This is important because less-differentiated T cells tend to persist longer and are more effective upon re-infusion.
- Synergistic Cytokine Co-stimulation: The combination of cytokines (e.g., IL-15 with IL-18 or IL-12) has been shown to synergistically boost the secretion of cytotoxic cytokines such as IFN-γ while suppressing inhibitory pathways like TGF-β and IL-4 production. This dual modulation enables TILs to exert more potent lytic activity against tumor cells.
- Targeted Combination Strategies: In addition to the cytokine and checkpoint strategies, the integration of targeted small molecule inhibitors (e.g., BRAF, MEK, or KRAS inhibitors) with TIL therapy helps in modulating the tumor cells and microenvironment directly. Such drugs can inhibit growth factor signaling within the tumor, thereby sensitizing cancer cells to T cell-mediated killing and helping overcome intrinsic resistance mechanisms.
- Enhanced Cellular Metabolism: Newer approaches involving engineered cytokine receptor agonists work to improve the metabolic health of TILs. By ensuring improved mitochondrial function and cellular fitness, these strategies help maintain TIL proliferation and functionality over extended periods in the hostile tumor microenvironment.

Interaction with TILs and Tumor Environment
The interaction of these drugs with TILs occurs both at the cellular and molecular levels, impacting the tumor microenvironment (TME) as well:
- Modulation of the TME: Immunomodulatory drugs can shift the TME from an immunosuppressive milieu to one that facilitates immune cell infiltration and activation. For example, the localized release of cytokines at the tumor site helps counteract the effects of suppressive factors, such as regulatory T cells and myeloid-derived suppressor cells (MDSCs), thereby improving TIL infiltration and function.
- Checkpoint Reversal and TIL Activation: Under chronic antigen stimulation in the TME, TILs tend to express high levels of inhibitory receptors. The administration of checkpoint inhibitors directly reverses this exhaustion, restoring the cytotoxic potential of TILs and promoting their cytokine production.
- Enhanced Trafficking and Persistence: Cytokines such as IL-15 not only drive TIL proliferation but also promote the expression of adhesion molecules and chemokine receptors that facilitate homing to the tumor site. This is crucial because effective TIL therapy relies on a sustained presence of activated effector cells in the tumor.
- Combination with Targeted Therapy: Small molecular inhibitors like BRAF and MEK inhibitors can modulate cancer cell survival pathways while simultaneously reducing the immunosuppressive signaling within the TME. This dual effect makes tumor cells more vulnerable to TIL-mediated cytotoxicity, thus predisposing tumors to better responses following TIL infusion.
- Synergistic Co-stimulation: The cross-talk between various cytokine signals (IL-2, IL-15, IL-18, and IL-12) and costimulatory receptors ensures that TILs retain their effector functions even after multiple rounds of stimulation. This is essential in counteracting ongoing suppression by the TME and achieving a robust anti-tumor effect.

Clinical Applications and Case Studies

Current Clinical Trials
A large number of clinical trials are underway to assess the efficacy and safety of TIL therapy when combined with various drug regimens. Recent studies have explored the use of TILs in combination with checkpoint inhibitors, targeted kinase inhibitors, and novel cytokine formulations.
- Checkpoint Blockade Combinations: Clinical trials have investigated TIL therapy in conjunction with PD-1 inhibitors such as pembrolizumab and nivolumab, leading to response rates that suggest a synergistic benefit from the dual modality approach. For instance, studies have reported an 86% response rate when TIL therapy was followed by checkpoint blockade, compared to historical monotherapy response rates.
- Targeted Therapy Combinations: Trials combining TILs with targeted drugs, including BRAF inhibitors, MEK inhibitors, and, more recently, KRAS inhibitors, are currently being evaluated in advanced solid tumors. These studies aim to assess whether targeted inhibition of oncogenic pathways can sensitize tumors to TIL-mediated killing and overcome mechanisms of treatment resistance.
- Cytokine-Driven Protocols: Numerous clinical trials have also focused on optimizing the cytokine support provided during TIL expansion and post-infusion. Early-phase studies are investigating modifications to IL-2 regimens, the use of IL-15 receptor agonists, and combination cytokine therapies (e.g., IL-15 plus IL-18) to improve TIL persistence and efficacy, while minimizing toxicities associated with high-dose cytokine therapies.
- Manufacturing and Novel Expansion Methods: In addition to clinical efficacy studies, several trials are assessing novel manufacturing techniques that incorporate drugs during the expansion phase. These methods typically aim at shortening expansion time, reducing microbial contamination, and maintaining a favorable T cell phenotype (e.g., central memory-like) through the use of engineered cytokine receptor agonists and immunomodulatory drug combinations.

Successful Case Studies
Successful clinical case studies provide strong evidence supporting the use of drugs in combination with TIL therapy:
- Melanoma Treatment: Landmark studies in metastatic melanoma have demonstrated that the combined administration of TILs with a high-dose IL-2 regimen can lead to durable complete responses in heavily pretreated patients, particularly when checkpoint inhibitors are subsequently applied to reverse T cell exhaustion.
- Combination Therapy with Targeted Agents: Reports exist of patients with BRAF-mutant tumors who have benefited from TIL therapy combined with BRAF and MEK inhibitors. These case reports indicate that the concurrent inhibition of oncogenic pathways alongside TIL-driven immune activity results in better clinical outcomes compared to either modality alone.
- Synergistic Cytokine Approaches: Clinical observations and preclinical experiments have shown that co-administration of IL-15 with IL-18 can significantly enhance TIL cytotoxicity against tumor cells. Such studies provide a rationale for clinical trials that merge these cytokine environments with TIL infusion to boost anti-tumor responses.
- Adjunctive Checkpoint Blockade: There are also encouraging reports where patients treated with TIL therapy alongside anti-PD-1 or anti-CTLA-4 antibodies have experienced substantial tumor regression, highlighting the importance of addressing TIL exhaustion and the inhibitory tumor microenvironment with immunomodulatory drugs.

Challenges and Future Directions

Current Limitations
Despite the exciting prospects, several challenges remain:
- Cytokine Toxicity: High doses of IL-2, traditionally used to support TIL therapy, can result in severe systemic toxicities such as hypotension, capillary leak syndrome, and organ dysfunction. These adverse effects have stimulated research into alternative cytokine regimens and the development of modified cytokine formulations (e.g., PEGylated cytokines) that provide equivalent support with minimized side effects.
- TIL Exhaustion and Tumor Microenvironment: Chronic antigen exposure in the tumor microenvironment leads TILs to express inhibitory receptors (e.g., PD-1, CTLA-4), which compromise their functionality. Although checkpoint inhibitors help to counter this exhaustion, there is a continuous need to optimize the combination regimens and determine the best sequencing strategies for maximal efficacy.
- Manufacturing Complexities: The individualized nature of TIL therapy necessitates patient-specific cell culture, resulting in high costs and variable product quality. Innovative approaches such as closed-system expansion and the use of immunomodulatory drugs during culture (e.g., AKT inhibitors) are being investigated to streamline the process and achieve consistent TIL products.
- Patient Selection and Biomarkers: A significant hurdle is the identification of robust predictive biomarkers that can stratify patients likely to benefit from TIL therapy. With heterogeneous response rates across different tumor histologies, there remains a critical need to develop companion diagnostics and surrogate markers that guide patient selection and predict long-term outcomes.

Future Research and Drug Development
Looking ahead, future endeavors in TIL therapy drug development are expected to focus on several key directions:
- Optimization of Cytokine Combinations: Further research will likely refine the use of cytokine cocktails—or engineered cytokine receptor agonists—that promote TIL expansion and functionality while mitigating systemic toxicity. Studies are planned to explore combinatorial use of IL-15, IL-18, and IL-12, and to evaluate next-generation formulations that allow more precise control of cytokine delivery.
- Integration of Novel Immunomodulatory Agents: The development of new checkpoint blockers and costimulatory agonists is poised to enhance TIL therapy even further. New agents targeting additional inhibitory receptors, such as LAG-3, TIM-3, and TIGIT, are in various stages of clinical development, and their potential integration with TIL therapy could address the multifaceted immune resistance mechanisms within tumors.
- Combination with Targeted Therapies and Small Molecules: Future clinical trials will focus on rationally designed combination therapies that include TILs with targeted kinase inhibitors (BRAF, MEK, KRAS inhibitors) and other small molecules aimed at modulating the tumor microenvironment. This approach is expected to offer a dual mechanism of direct tumor growth inhibition along with enhanced immune surveillance.
- Advances in Manufacturing Technology: Innovations in cell-culture techniques, including the development of automated closed systems and improvements in expansion protocols using supportive drugs (like AKT inhibitors), will be essential. These strategies are designed to not only reduce production time and cost but also to preserve the “youthful” central memory phenotype of TILs, which correlates with improved clinical responses.
- Biomarker Discovery and Personalized Approaches: There is an increasing emphasis on the deployment of high-throughput sequencing, flow cytometry, and other omics approaches to comprehensively profile TIL products. Such analyses will help in identifying robust biomarkers that can predict response and guide treatment decisions, ultimately leading to more personalized and effective TIL therapy regimens.
- Regulatory and Safety Enhancements: As TIL therapy continues to evolve, a crucial aspect will be the implementation of strategies to mitigate adverse events while preserving therapeutic efficacy. This includes refining dosing strategies for immunomodulatory drugs and exploring novel formulations that limit systemic exposure. Regulatory pathways will also need to adapt to the complexity of these personalized therapies, ensuring that safety monitoring keeps pace with innovative drug combinations.

Conclusion
In summary, the different types of drugs available for TIL therapy span a broad spectrum of immunomodulatory agents and cytokines designed to enhance the intrinsic anti-tumor capabilities of patient-derived lymphocytes. Immunomodulatory drugs encompass surface-associated cytokine agents, checkpoint inhibitors (such as anti-PD-1, anti-PD-L1, and anti-CTLA-4), targeted signaling modulators, and designer small molecules that interfere with oncogenic pathways. These agents work synergistically to reverse TIL exhaustion, promote a less differentiated memory phenotype, counteract the suppressive tumor microenvironment, and improve TIL trafficking, persistence, and cytotoxicity. Cytokines and growth factors, particularly IL-2, IL-15, IL-18, and IL-12 in various combinations, serve as critical supportive agents both in the ex vivo expansion of TILs and the maintenance of their anti-tumor function post-infusion.

Clinical applications of these drug classes have been explored in a wide array of trials and case studies, demonstrating that TIL therapy—especially when combined with novel immunomodulatory agents and targeted therapies—can result in significant tumor regression and durable responses. However, challenges remain in terms of systemic toxicity (notably from high-dose IL-2), manufacturing complexity, and the need for reliable predictive biomarkers. Future endeavors in drug development for TIL therapy will likely focus on optimizing cytokine combinations, integrating novel checkpoint and costimulatory inhibitors, refining manufacturing protocols, and enhancing personalized approaches through biomarker discovery.

Overall, the current landscape of drugs available for TIL therapy is robust and multifaceted, embodying a synergy of immunology, molecular biology, and pharmaceutical innovation. With continuous refinements and strategic combination therapies, TIL-based treatments are poised to play an increasingly pivotal role in the management of solid tumors and potentially other malignancies. The integration of immunomodulatory drugs and cytokines not only elevates the therapeutic potential of TILs but also paves the way for more efficient, tailored, and less toxic treatment regimens in the future.

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.