IL-15 signals via a trimeric receptor complex (IL-15Rα/IL-2Rβ/γc), activating JAK1/3 and downstream STAT3/5 pathways . Key functional features:
Transpresentation: Membrane-bound IL-15/IL-15Rα complexes on dendritic cells (DCs) deliver signals to adjacent NK or T cells expressing IL-2Rβ/γc .
Immune Cell Modulation:
NK Cells: Induces proliferation (10–13.5-fold expansion), cytotoxicity (via TRAIL/perforin upregulation), and IFN-γ production .
CD8+ T Cells: Enhances survival and effector function without promoting activation-induced cell death .
Dendritic Cells: Autocrine IL-15 prevents apoptosis and amplifies antigen presentation .
Phase I Trials (NCT01021059, NCT01727076):
Subcutaneous (SC) Administration: 2 μg/kg/day for 10 days was well tolerated, inducing 10-fold NK cell and 3-fold CD8+ T cell expansion .
Intravenous (IV) Bolus: Limited to 0.3 μg/kg/day due to toxicity (hypotension, troponin elevation) .
Continuous IV Infusion: Achieved 38-fold NK cell and 6-fold CD8+ T cell increases at 2 μg/kg/day .
Administration Route | Max Tolerated Dose | Immune Cell Expansion |
---|---|---|
Subcutaneous | 2 μg/kg/day | NK: 10–13.5×; CD8+ T: 2.8–3.3× |
Continuous IV Infusion | 2 μg/kg/day | NK: 38×; CD8+ T: 6× |
CAR-T/NK Cells: IL-15 augments in vivo persistence and antitumor activity .
Checkpoint Inhibitors: Synergizes with anti-PD-1/PD-L1 to overcome immunosuppression .
Vaccines: Enhances antigen-specific CD8+ T cell responses in HIV and cancer models .
Toxicity: Dose-limiting cardiotoxicity and cytokine release syndrome at ≥3 μg/kg/day .
Delivery Systems:
Multiple GMP-compliant variants are available for clinical use:
FDA Drug Master Files (DMF) and ISO 13485 certifications ensure compliance for cell therapy applications .
Ongoing trials focus on optimizing dosing schedules (e.g., intermittent SC pulses) and combinatorial regimens with IL-7/IL-21 to enhance T cell fitness . Preclinical data support IL-15’s potential in autoimmune diseases and infectious disease vaccines .
Interleukin-15 (IL-15) is a cytokine that stimulates T-lymphocyte proliferation. IL-15 stimulation requires interaction with components of the IL-2 receptor, including IL-2RB and likely IL-2RG, but not IL-2RA. In neutrophils, IL-15 stimulates phagocytosis, likely through signaling via the IL-15 receptor (IL-15RA, IL-2RB, and IL-2RG subunits), leading to SYK kinase activation.
Recombinant human Interleukin-15 (IL-15) is an immune-stimulating cytokine that uses the heterotrimeric receptor IL-2/IL-15Rβ and the γ chain shared with IL-2, along with the cytokine-specific IL-15Rα. Despite sharing receptor components with IL-2, IL-15 demonstrates several distinct functional properties. IL-15 primarily stimulates proliferation, activation, and expansion of natural killer (NK) cells, promotes T-cell proliferation, and generates cytotoxic T lymphocytes. It also plays a crucial role in sustaining long-lasting antitumor immunity by acting as a survival factor for CD8 memory T cells . Unlike IL-2, IL-15 does not significantly affect regulatory T cells (Tregs) that inhibit antitumor immunity, does not cause capillary leak syndrome, and isn't associated with activation-induced cell death . These distinctive characteristics make IL-15 a particularly promising candidate for cancer immunotherapy applications.
IL-15 forms stable complexes with IL-15Rα on cell surfaces, primarily on activated dendritic cells but also on non-hematopoietic cells in lungs and small intestine. These complexes present IL-15 in trans to neighboring NK and CD8 cells that express IL-2/IL-15Rβ (CD122) and the common γc (CD132) but not IL-15Rα . This "trans-presentation" mechanism is crucial for IL-15 function. Furthermore, membrane IL-15/IL-15Rα complexes undergo endosomal internalization but survive lysosomal degradation, allowing the complex to recycle back to the cell surfaces . This recycling phenomenon contributes to the prolonged retention of IL-15 in circulation and tissues. Understanding these interactions is essential for researchers designing IL-15-based therapeutics, as engineered IL-15 complexes with the receptor can significantly enhance half-life and bioactivity compared to the cytokine alone.
The bioactivity of recombinant human IL-15 is commonly assessed through cell proliferation assays using IL-15-responsive cell lines. The CTLL-2 cell proliferation assay is a widely used method to characterize IL-15 bioactivity in vitro . Additionally, researchers evaluate IL-15's capability to stimulate the expansion of memory CD8+ T cells in mouse spleen as a functional readout . For more comprehensive assessment, examining the activation and proliferation of natural killer (NK) cells represents another critical parameter. Cell-based assays can also determine stability under various conditions (37°C for 48 hours, 4°C for 3 months, or after multiple freeze-thaw cycles) . When evaluating GMP-grade IL-15, batch-to-batch consistency is typically verified through comparative cell-based assays, ensuring reliable experimental outcomes across different production lots. These standardized methods provide researchers with reliable metrics to confirm the potency and quality of recombinant IL-15 preparations before use in complex experimental systems.
Recombinant human IL-15 exhibits a notably short half-life in circulation of approximately 0.7 hours when administered as a monomer, significantly limiting its therapeutic potential . This pharmacokinetic limitation stems from rapid clearance mechanisms and strict regulatory pathways governing IL-15 expression. Several strategic approaches have been developed to extend IL-15's half-life. The most successful approach involves complexing IL-15 with the extracellular region of its receptor alpha subunit (sIL-15Rα), which profoundly prolongs the half-life to approximately 13.1 hours - representing an 18-fold increase compared to the monomer . Further enhancements include fusing the receptor component to immunoglobulin G (IgG1) Fc, creating the IL-15·sIL-15Rα/Fc complex. This superagonist not only extends circulation time but also maintains biological activity, stimulating target immune cells more effectively than the native cytokine . The pharmacokinetic profile of IL-15 shows a biphasic pattern with a rapid decline during the first 24 hours (α phase), followed by a β phase where detectable levels persist for extended periods .
Recombinant human IL-15 demonstrates remarkable stability under various storage conditions when properly formulated. Cell-based assays have shown that GMP-grade human IL-15 maintains stability at 37°C for up to 48 hours, indicating reasonable short-term thermostability . For longer storage periods, the protein remains stable at 4°C for at least 3 months in real-time stability experiments . This provides researchers flexibility in handling during experimental protocols. Additionally, IL-15 withstands multiple freeze-thaw cycles (at least three) without significant loss of bioactivity . This freeze-thaw resistance is particularly valuable for laboratories that need to use small aliquots from a single stock over extended periods. When preparing IL-15 for experimental use, researchers should consider using low-protein binding tubes and avoiding excessive agitation that could promote aggregation. While specific buffer formulations may vary between manufacturers, the documented stability profile suggests that properly produced recombinant human IL-15 is a relatively robust protein suitable for various research applications when handled according to manufacturer guidelines.
In direct comparative studies, IL-15 demonstrates a distinctly different pharmacokinetic profile compared to other common gamma chain cytokines such as IL-2 and IL-7. When administered to mice, both IL-2 and IL-7 show rapid clearance with terminal half-times of less than 1 hour . In contrast, IL-15 exhibits a biphasic kinetic profile with an initial rapid decline during the first 24 hours (α phase), followed by a sustained β phase where IL-15 levels remain detectable (>10 pg/mL) even 120 hours after injection of 5 μg of human IL-15 . This extended retention is mediated by a specific mechanism involving IL-15Rα, as demonstrated through studies in IL-15Rα−/− and IL-15Rα transgenic mice . The formation of stable IL-15/IL-15Rα complexes that undergo recycling rather than degradation after endosomal internalization contributes significantly to this prolonged circulation. This unique pharmacokinetic behavior provides a distinct advantage for IL-15 in therapeutic applications, as it allows for less frequent dosing while maintaining biological efficacy. Understanding these comparative pharmacokinetics is essential for researchers designing dosing schedules and developing enhanced cytokine formulations.
The production of biologically active recombinant human IL-15 has been successfully accomplished using various expression systems, with mammalian expression systems demonstrating particular effectiveness. Human embryonic kidney (HEK293) cells have proven to be an efficient platform for IL-15 production through transient gene expression . This approach involves co-transfection of plasmids encoding IL-15 and sIL-15Rα/Fc respectively, yielding significant product levels of approximately 36 mg/L after purification . Mammalian expression systems offer the advantage of proper post-translational modifications, protein folding, and reduced immunogenicity compared to bacterial systems. For GMP-grade IL-15 production, manufacturers typically employ ISO-certified processes under 9001:2015 and 13485:2016 standards, with quality control testing performed under GMP compliance . The animal-free materials used in advanced production systems help minimize contamination risks and ensure consistent biological activity. These production considerations are critical for researchers requiring high-quality IL-15 for preclinical studies or translational research, as expression system selection significantly impacts protein characteristics, yield, and functional properties.
When evaluating recombinant human IL-15 for research applications, several critical quality control parameters warrant careful assessment. Biological activity remains the paramount quality attribute, typically measured through cell-based proliferation assays using IL-15-responsive cell lines or primary immune cells . Consistency across production batches is essential, and comparative assays between GMP and research-grade preparations can verify manufacturing reliability . Stability testing under various conditions (elevated temperature, extended storage, and freeze-thaw cycles) provides crucial insights into protein robustness during experimental handling . Additional quality parameters include purity (typically assessed by SDS-PAGE and HPLC), endotoxin levels (particularly critical for in vivo applications), and protein concentration accuracy. For GMP-grade IL-15 intended for advanced preclinical studies, verification of animal-free manufacturing processes and FDA Drug Master File (DMF) documentation may be relevant considerations . Researchers should also evaluate specific activity (biological activity per unit mass), which can vary between preparations and significantly impact experimental outcomes. By comprehensively assessing these quality parameters, investigators can ensure reliable, reproducible results when working with this potent immunomodulatory cytokine.
Comprehensive characterization of recombinant human IL-15 requires a multi-faceted analytical approach. Protein identity and integrity are typically confirmed through mass spectrometry and N-terminal sequencing, which verify the amino acid sequence and molecular weight. Structural integrity is assessed using circular dichroism and fourier-transform infrared spectroscopy to evaluate secondary and tertiary structure. Size-exclusion chromatography and dynamic light scattering are crucial for detecting aggregates that might affect bioactivity or immunogenicity . Functional characterization relies heavily on cell-based bioassays using IL-15-responsive cell lines like CTLL-2 to confirm biological activity . Endotoxin testing via the Limulus Amebocyte Lysate assay is essential, particularly for in vivo applications, with GMP-grade preparations typically requiring levels below 0.5 EU/mg. Additional characterization may include glycosylation analysis through lectin binding assays or mass spectrometry, though recombinant IL-15 produced in E. coli lacks glycosylation . Immunological characterization to ensure the absence of host cell proteins and DNA contamination is also standard practice. These analytical techniques collectively ensure that recombinant IL-15 preparations meet the rigorous quality standards required for experimental reproducibility and translational research applications.
Determining optimal dosing strategies for IL-15 in preclinical models requires careful consideration of several factors including administration frequency, route, and formulation. Evidence from murine tumor models indicates that more frequent administration (5 times weekly) of lower IL-15 doses (2.5 μg/mouse) produces superior results compared to less frequent (twice weekly) higher doses (5 μg/mouse) in terms of increasing circulating NK, CD8, and CD44hiCD8 T cells . This suggests that maintaining more consistent cytokine levels may be advantageous over intermittent high-dose administration. For rhesus macaque studies, intravenous administration of rhIL-15 at doses ranging from 10-50 μg/kg/day for 12 consecutive days has demonstrated significant expansion of immune cell populations with manageable toxicity . In contrast, subcutaneous administration every 3 days yielded only modest increases in NK and memory T cells .
When designing IL-15 experiments, researchers should consider that different IL-15 formulations (monomer versus receptor complexes) require distinct dosing approaches due to their different pharmacokinetic profiles. The IL-15·sIL-15Rα/Fc complex, with its 18-fold longer half-life (13.1 hours versus 0.7 hours), allows for less frequent administration while maintaining efficacy . These dosing considerations are critical for designing translational studies that accurately predict clinical outcomes and optimize therapeutic efficacy.
Comprehensive assessment of IL-15-induced immune responses requires a multi-parameter approach targeting various immune cell populations and functional readouts. Flow cytometry remains the gold standard for quantifying changes in immune cell populations, particularly focusing on NK cells, CD8+ T cells (especially memory subsets), and the expression of activation markers such as Ki67 . Beyond simple enumeration, functional assays are critical for evaluating IL-15's impact on immune cell capabilities. These include cytotoxicity assays against target cells, intracellular cytokine staining (particularly for IFN-γ, TNF-α), and proliferation assays using CFSE dilution or Ki67 staining . Serum cytokine analysis provides valuable insights into the broader immunological milieu, with particular attention to IL-18 levels, which correlate with IL-15 activity and neutrophil migration .
For in vivo models, especially tumor studies, researchers should track changes in tumor volume, survival rates, and perform immunohistochemical analysis of tumor-infiltrating lymphocytes to assess immune cell trafficking to the tumor microenvironment . Additionally, evaluating memory responses through tumor re-challenge experiments can reveal IL-15's capacity to establish durable anti-tumor immunity . When assessing combination therapies, such as IL-15 with PD-1 antibodies, measuring changes in checkpoint receptor expression and T cell exhaustion markers provides crucial mechanistic insights . These comprehensive immunological assessments enable researchers to fully characterize IL-15's immunomodulatory effects and optimize therapeutic approaches.
Selecting appropriate model systems for IL-15 research requires consideration of both the research question and the translational relevance of the chosen system. For in vitro studies, the CTLL-2 cell line provides a sensitive and reliable system for assessing IL-15 bioactivity through proliferation assays . Primary human peripheral blood mononuclear cells (PBMCs) offer a more physiologically relevant system for studying IL-15's effects on diverse immune cell populations and can demonstrate consistent responses to GMP-grade IL-15 across batches . For cancer immunotherapy research, various preclinical tumor models have demonstrated IL-15's efficacy. The HT-29 xenograft NOD-SCID mouse model has been used to evaluate IL-15·sIL-15Rα/Fc superagonist effects through human immune cell stimulation . The TRAMP-C2 prostatic tumor model in mice has helped optimize dosing frequencies by comparing different administration schedules .
For translational studies with higher predictive value, non-human primate models, particularly rhesus macaques, have proven valuable for toxicology and pharmacokinetic assessments of recombinant human IL-15 . These studies have informed clinical trial designs by establishing safety profiles and effective dosing strategies. When evaluating combination therapies, such as IL-15 with PD-1 antibodies, syngeneic mouse models with intact immune systems provide insights into how IL-15 enhances checkpoint inhibitor efficacy . Each model system offers distinct advantages, and researchers should select models aligned with their specific research objectives while considering species-specific differences in IL-15 biology.
The safety profile of recombinant human IL-15 has been extensively characterized in preclinical models, particularly in non-human primates. The most notable adverse effect is a dose-related, transient grade 3/4 neutropenia observed in rhesus macaques receiving daily intravenous IL-15 administration . Importantly, this neutropenia appears to result from redistribution of neutrophils from circulation to tissues rather than impaired neutrophil production. Bone marrow examinations of treated animals actually demonstrated increased marrow cellularity, including cells of the neutrophil series . Further supporting the redistribution hypothesis, neutrophils were observed in the sinusoids of enlarged livers and spleens of treated animals . This neutrophil redistribution is likely mediated through an IL-15-triggered cytokine cascade involving IL-18, a key finding for monitoring potential toxicities .
Additional laboratory changes included increased platelet counts from baseline levels of approximately 200 × 10³/μL to 333 × 10³/μL . Despite these hematological changes, necropsy studies did not reveal any significant abnormalities in treated animals . Notably, IL-15 administration was not associated with several undesirable effects seen with IL-2, such as capillary leak syndrome or activation-induced cell death . The absence of significant autoimmune manifestations, infections, or animal deaths during toxicology studies supports IL-15's favorable safety profile compared to some other immunotherapeutic agents .
Immunogenicity assessment represents a critical safety consideration for recombinant human IL-15, particularly for repeated administration protocols. In toxicology studies with rhesus macaques receiving daily doses of recombinant human IL-15 for 12 days at doses ranging from 10-50 μg/kg, no antibodies to the administered rhIL-15 were detected, despite the potential immunogenicity risk factors present . These risk factors included the fact that E. coli-produced rhIL-15 is non-glycosylated (which could theoretically yield aggregates) and that human IL-15 has six amino acid differences from rhesus macaque IL-15 . This lack of observed immunogenicity is encouraging for clinical translation.
The immunogenicity assessment utilized an ELISA procedure sensitive to 156 ng/mL of antibody to rhIL-15, providing a reliable detection method . For researchers designing preclinical studies with IL-15, particularly those involving repeated administration, immunogenicity monitoring should be incorporated into study protocols. Additionally, researchers should consider potential differences in immunogenicity between different IL-15 formulations. While basic IL-15 monomers might present certain immunogenicity risks, engineered variants such as IL-15·sIL-15Rα/Fc complexes could present different immunogenic profiles due to their modified structure and potentially different aggregation tendencies . These immunogenicity considerations are particularly important for translational research aimed at clinical applications.
Comprehensive safety monitoring during IL-15 experimental studies should encompass multiple parameters based on established preclinical findings. Hematological parameters warrant particular attention, with complete blood counts focusing on neutrophil levels, as transient neutropenia represents the most common adverse effect . Platelet counts should also be tracked, as increases from baseline have been observed during IL-15 administration . Bone marrow examination may provide valuable insights in cases of persistent cytopenias to distinguish between cell redistribution and production defects .
Serum cytokine profiling provides crucial mechanistic insights into IL-15-induced effects, with IL-18 levels serving as a particularly informative biomarker. IL-18 elevation correlates with neutrophil migration and IL-15 activity, making it a valuable surrogate marker . Additional inflammatory cytokines worth monitoring include MIP-2, MIP-1α, TNF-α, and LTB-4, which form part of the neutrophil migration signaling cascade triggered by IL-15 .
Liver and renal function tests should be included in safety assessments, as enlarged livers were observed in some preclinical studies . For studies evaluating potential autoimmune effects, monitoring autoantibody levels and clinical manifestations of autoimmunity is prudent, though IL-15 has not shown significant autoimmune induction in preclinical models . When evaluating novel IL-15 complexes or formulations, researchers should additionally monitor for hypersensitivity reactions or unexpected toxicities not observed with standard IL-15 preparations. These comprehensive monitoring parameters ensure thorough safety assessment while providing mechanistic insights into IL-15's physiological effects.
Recombinant human IL-15 functions as a cancer immunotherapeutic through multiple mechanisms that collectively enhance anti-tumor immune responses. Its primary mode of action involves stimulating the proliferation, activation, and expansion of natural killer (NK) cells and CD8+ T cells, both critical effector populations in tumor immunity . IL-15 induces cell activation, proliferation, cytolytic activity, and production of cytokines such as interferon-γ (IFN-γ) by these cells . Unlike IL-2, IL-15 does not significantly affect regulatory T cells (Tregs), which can inhibit antitumor immunity and promote tumor development .
A particularly valuable property of IL-15 is its role as a survival factor for CD8 memory T cells, supporting long-term maintenance of high-avidity T cell responses against malignant cells . This memory-sustaining function contributes to durable anti-tumor immunity. In preclinical studies using tumor-bearing mice, administration of recombinant IL-15 led to tumor regression, metastasis reduction, and increased survival . Some mice that received IL-15 treatment completely eliminated tumors and remained tumor-free after subsequent re-challenge, indicating the establishment of long-term immune memory .
The IL-15·sIL-15Rα/Fc superagonist demonstrated tumor growth inhibition in a HT-29 xenograft NOD-SCID mouse model through stimulation of infused human immune cells . Furthermore, combination therapy with IL-15·sIL-15Rα/Fc and programmed death-1 (PD-1) antibody showed stronger inhibitory effects compared to treatment with either agent alone, highlighting its potential in combination immunotherapy approaches .
The clinical development of recombinant human IL-15 has progressed from preclinical studies to early-phase clinical trials based on promising safety and efficacy data. Toxicology studies in rhesus macaques receiving daily intravenous IL-15 administration for 12 days at doses ranging from 10-50 μg/kg established a safety profile with manageable toxicities, primarily transient neutropenia . These studies supported the initiation of human clinical trials, with an FDA-approved phase 1 dose-escalation protocol evaluating doses from 3-25 μg/kg in patients with metastatic malignant melanoma and metastatic renal cell cancer .
The clinical development approach has been shaped by pharmacokinetic and immune response data from preclinical models. While some researchers suggested subcutaneous, intermittent (every 3 days) IL-15 administration to mitigate potential toxicities, comprehensive data demonstrated that daily administration produces more robust expansion of NK and CD8 T cells compared to intermittent dosing . This informed the clinical protocol design featuring 12 daily infusions of rhIL-15 .
The IL-15·sIL-15Rα/Fc complex represents an advanced development candidate with improved pharmacokinetic properties, extending IL-15's half-life approximately 18-fold (from 0.7 to 13.1 hours) . This superagonist has shown promising anti-tumor efficacy both as monotherapy and in combination with PD-1 antibody in preclinical models . Clinical development strategies continue to evolve, with research focusing on optimizing dosing regimens, administration routes, and combination approaches to maximize therapeutic benefit while maintaining acceptable safety profiles.
Combination therapy approaches incorporating IL-15 with other immunotherapeutic agents have demonstrated enhanced efficacy in preclinical models, suggesting promising strategies for clinical development. The combination of IL-15·sIL-15Rα/Fc superagonist with programmed death-1 (PD-1) antibody has shown particularly strong synergistic effects, with greater tumor inhibition compared to treatment with either agent alone . This synergy likely stems from complementary mechanisms of action: IL-15 actively stimulates immune effector cells, while PD-1 blockade removes inhibitory signals that suppress T cell function within the tumor microenvironment.
The mechanistic rationale for this combination includes IL-15's ability to expand NK and CD8+ T cell populations and enhance their cytolytic activity, creating a larger pool of activated effector cells . Simultaneously, PD-1 blockade prevents these effector cells from becoming functionally exhausted upon encountering PD-L1-expressing tumor cells. This dual approach of expanding effector populations while preventing their exhaustion represents a powerful strategy for overcoming tumor immune evasion mechanisms.
While PD-1/PD-L1 blockade represents the most extensively studied combination with IL-15, other potential combination approaches include pairing IL-15 with additional checkpoint inhibitors (e.g., CTLA-4, LAG-3, TIM-3 antagonists), targeted therapies that increase tumor immunogenicity, or adoptive cell therapies like CAR-T cells to enhance their persistence and function. Optimizing dosing schedules and sequences for these combinations remains an active area of investigation, with the potential to significantly advance cancer immunotherapy efficacy through rational combination approaches.
Engineering enhanced IL-15 variants represents a cutting-edge area of research aimed at overcoming the limitations of native IL-15 while preserving or amplifying its beneficial immunostimulatory properties. The development of IL-15·sIL-15Rα/Fc exemplifies this approach, creating a superagonist complex that substantially extends IL-15's half-life from 0.7 hours to 13.1 hours . This complex combines IL-15 with the extracellular region of its receptor alpha subunit fused to immunoglobulin G (IgG1) Fc, yielding a molecule with improved pharmacokinetics while maintaining bioactivity . The production method involves co-transfection of plasmids encoding IL-15 and sIL-15Rα/Fc in HEK293 cells, resulting in yields of approximately 36 mg/L after purification .
Beyond half-life extension, researchers are exploring additional engineering strategies to enhance IL-15's therapeutic profile. These include tissue-targeting approaches to concentrate IL-15 activity in tumor microenvironments while reducing systemic exposure and associated toxicities. Mutational approaches targeting the IL-15 binding interface with receptor components aim to modify signaling strength or duration, potentially enhancing anti-tumor effects while minimizing pro-inflammatory potential associated with autoimmune risks .
The development of bifunctional fusion proteins that combine IL-15 activity with complementary immunomodulatory functions represents another promising direction. These engineered variants undergo rigorous biological characterization, including assessment of receptor binding kinetics, signaling pathway activation, and comparative pharmacokinetics. Such innovative approaches to IL-15 engineering hold significant promise for enhancing the therapeutic window of this potent immunostimulatory cytokine.
Despite IL-15's potent immunostimulatory properties, several mechanisms may limit its efficacy in certain contexts. Understanding these potential resistance pathways is crucial for developing strategies to maximize therapeutic outcomes. One significant limitation involves the tumor microenvironment's immunosuppressive nature, which can counteract IL-15-induced immune activation. Tumors may express elevated levels of immune checkpoint molecules like PD-L1 or secrete immunosuppressive cytokines (TGF-β, IL-10) that dampen IL-15-stimulated effector cell functions . This provides a strong rationale for combination approaches with checkpoint inhibitors, which have shown enhanced efficacy compared to IL-15 monotherapy .
Another potential resistance mechanism involves altered expression or function of IL-15 receptor components on target immune cells. Downregulation of IL-2/IL-15Rβ (CD122) or common γ chain (CD132) on exhausted T cells could limit their responsiveness to IL-15 stimulation. Chronic inflammation within the tumor microenvironment might also induce negative feedback regulators like suppressor of cytokine signaling (SOCS) proteins that inhibit IL-15 signaling pathways.
The short half-life of native IL-15 represents a pharmacokinetic limitation that reduces sustained immune activation. While this has been partially addressed through development of IL-15·sIL-15Rα/Fc complexes , optimization of dosing regimens remains important. Daily administration has shown superior efficacy compared to intermittent dosing in expanding immune cell populations , suggesting that consistent IL-15 exposure may be necessary to overcome resistance mechanisms.
Combination strategies targeting multiple aspects of anti-tumor immunity represent the most promising approach to overcoming these resistance mechanisms, with IL-15 serving as a potent immune activator complemented by agents that address specific resistance pathways.
Identifying predictive biomarkers for IL-15-based therapies represents a critical research direction for patient selection and therapeutic optimization. Several potential biomarker categories warrant investigation based on IL-15's mechanism of action and preclinical findings. Immunological biomarkers include baseline levels and composition of NK and CD8+ T cell populations, particularly memory subsets that respond robustly to IL-15 stimulation . The expression levels of IL-15 receptor components (IL-15Rα, IL-2/IL-15Rβ, common γ chain) on immune effector cells may correlate with response magnitude. Early pharmacodynamic markers during treatment include expansion of NK cells and CD8+ memory T cells, with a 4-fold increase in circulating NK cells and similar expansions of central and effector memory T cells observed in responsive models .
Serum cytokine profiles may offer valuable predictive information, with IL-18 levels serving as a surrogate marker of IL-15 activity . The ratio of effector to suppressor immune cells in the tumor microenvironment (CD8+ T cells vs. regulatory T cells) might predict which tumors will respond to IL-15-mediated immune activation. Additionally, tumor genomic characteristics such as mutational burden, which correlates with neoantigen load and potential immunogenicity, could influence responsiveness to IL-15-enhanced immune recognition.
For combination therapies involving IL-15 and checkpoint inhibitors, PD-L1 expression in tumors and the tumor mutation burden may have particular relevance . Developing multiparameter predictive models incorporating these diverse biomarkers could significantly enhance patient selection for IL-15-based therapies and guide personalized immunotherapy approaches.