IL2 Canine Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 160 amino acids (21-155 a.a) and having a molecular mass of 18.1 kDa.
IL2 Canine is fused to a 25 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.
Interleukin-2, IL-2, T-cell growth factor, TCGF.
MGSSHHHHHH SSGLVPRGSH MGSHMAPITS SSTKETEQQM EQLLLDLQLL LNGVNNYENP QLSRMLTFKF YTPKKATEFT HLQCLAEELK NLEEVLGLPQ SKNVHLTDTK ELISNMNVTL LKLKGSETSY NCEYDDETAT ITEFLNKWIT FSQSIFSTLT.
Canine interleukin-2 (IL-2) is a cytokine predominantly produced by activated CD4-positive helper T-cells, with lesser production by CD8-positive T-cells and natural killer (NK) cells. This protein serves as a pivotal regulator in both immune response and tolerance maintenance in dogs . The biological significance of IL-2 extends across multiple immune pathways, functioning primarily as a T-cell growth factor while simultaneously enhancing NK-cell cytolytic activity . In the canine immune system, IL-2 strongly promotes the proliferation of activated B-cells, consequently increasing immunoglobulin production and supporting humoral immunity . Additionally, this cytokine plays a critical role in regulating adaptive immunity by controlling the survival and proliferation of regulatory T-cells, which are essential for maintaining immune tolerance .
IL-2 operates through binding to receptor complexes that exist in either high-affinity trimeric (IL2RA/CD25, IL2RB/CD122, and IL2RG/CD132) or low-affinity dimeric (IL2RB and IL2RG) configurations on target cells . This receptor binding triggers a cascade of intracellular signaling events, beginning with the phosphorylation of Janus kinases JAK1 and JAK3, which subsequently phosphorylate the receptor to create docking sites for downstream substrates including STAT5 . The activation of these pathways - particularly STAT, phosphoinositide-3-kinase (PI3K), and mitogen-activated protein kinase (MAPK) - drives the cellular responses that underpin IL-2's immunomodulatory functions . Beyond its effects on T-cells, NK cells, and B-cells, canine IL-2 contributes significantly to the differentiation and homeostasis of various effector T-cell subsets, including Th1, Th2, Th17, and memory CD8-positive T-cells .
Histidine tags (His-tags) in recombinant canine IL-2 represent a strategic molecular modification that significantly enhances research capabilities with this important immune cytokine. The His-tag typically consists of six consecutive histidine residues (HHHHHH) engineered into the protein sequence, as evidenced in commercial recombinant dog IL-2 proteins . This modification serves multiple critical research functions without substantially altering the protein's biological activity. The primary advantage of His-tagged canine IL-2 lies in simplified purification protocols utilizing immobilized metal affinity chromatography (IMAC), where the histidine residues selectively bind to metal ions like nickel or cobalt, enabling efficient one-step purification from complex biological mixtures .
For researchers working with canine IL-2, His-tagged versions provide enhanced detection capabilities through anti-His antibodies, eliminating the need for protein-specific antibodies that may be less readily available for canine proteins. The recombinant dog IL-2 protein with His-tag available commercially spans amino acids 21 to 155 of the native sequence and is expressed in Escherichia coli expression systems . This expression system generates high-purity preparations (>95%) with minimal endotoxin contamination (<1 EU/μg), making the product suitable for various analytical techniques including SDS-PAGE and mass spectrometry . The incorporation of the His-tag preserves the cytokine's ability to bind its receptor complex and activate downstream signaling pathways including JAK/STAT, PI3K, and MAPK cascades, maintaining its functional activity in experimental systems studying canine immune responses .
Researchers studying canine IL-2 receptor (IL-2R) expression have developed reliable flow cytometry-based detection methods that provide quantitative assessment of receptor levels. A well-established technique involves using phycoerythrin-labeled human recombinant IL-2 (IL-2-PE) as a detection reagent . This approach begins with isolation of peripheral blood mononuclear cells (PBMCs) from canine blood samples, followed by washing steps to remove serum proteins that might interfere with binding . The cleaned cells are then incubated with IL-2-PE, allowing the labeled cytokine to bind to IL-2 receptors expressed on cell surfaces. After removing unbound IL-2-PE through additional washing steps, flow cytometric analysis can be performed using a 488 nm argon laser while applying appropriate lymphocyte gating strategies .
The specificity of this detection method has been validated through competitive binding experiments, where the addition of unlabeled human recombinant IL-2 to phytohemagglutinin (PHA)-stimulated cells reduces fluorescence intensity approximately four-fold, confirming that IL-2-PE specifically binds to canine IL-2 receptors . Using this technique, researchers have determined that approximately 21% of resting canine lymphocytes express IL-2R . When applying this methodology to track receptor dynamics, investigators have observed that following PHA stimulation, both the percentage of IL-2R-expressing cells and receptor density increase significantly, with peak expression (76.4% positive cells) occurring around day 3 post-stimulation . The receptor density, measured by mean fluorescence intensity, reaches maximum levels on days 2-3 at approximately twenty-fold greater than resting cells . This one-step direct method provides researchers with a valuable tool for quantitatively assessing IL-2R expression in canine samples without requiring species-specific antibodies.
Administration protocols for IL-2 in canine experimental models have been thoroughly established in the scientific literature, with continuous infusion representing a well-documented approach. In seminal studies, normal adult dogs received human recombinant IL-2 through continuous infusion regimens consisting of two consecutive weekly cycles, with IL-2 administered for 4 days per week . The dosage employed in these protocols was 3 × 10^6 units per m^2 of body surface area per day, providing standardized dosing across animals of different sizes . This administration route ensures stable blood levels of the cytokine throughout the treatment period, which is particularly advantageous for maintaining consistent immune stimulation.
More recent experimental approaches have explored intratumoral (IT) administration of IL-2, either as recombinant protein or as part of fusion proteins such as the GD2-reactive hu14.18-IL2 immunocytokine (IC) . In these protocols, dogs with locally advanced or metastatic melanoma received intratumoral injections at doses of 12 mg/m^2 on three consecutive days, often following radiation therapy to enhance immune responses . The intratumoral delivery method represents a strategic approach to increase local cytokine concentration within the tumor microenvironment while potentially reducing systemic toxicity. Researchers monitoring dogs receiving IL-2 via either method should implement comprehensive observation protocols for potential side effects, including assessment of gastrointestinal symptoms (vomiting, diarrhea), activity levels, body temperature, respiratory rate, and weight . These administration protocols provide researchers with established methodologies for investigating IL-2-mediated immune responses in canine models, with options for either systemic or localized delivery depending on the specific research objectives.
Administration of IL-2 in canine models produces multifaceted immunological and physiological effects that researchers should carefully monitor. At the hematological level, IL-2 treatment induces a marked lymphocytosis and eosinophilia in dogs, with cell counts increasing more than sevenfold above baseline levels following completion of each treatment course . This expanded lymphocyte population can persist for extended periods, in some cases remaining elevated for more than one month post-treatment . Functionally, peripheral blood lymphocytes (PBL) obtained during this lymphocytosis period demonstrate enhanced cytotoxic capacity, with increased in vitro lysis activity against natural-killer-cell-sensitive canine tumor cell lines such as CTAC .
In canine melanoma studies combining radiation therapy with intratumoral IL-2-containing immunocytokines, researchers observed increases in intratumoral lymphocytic inflammation through this methodology . Serial biopsy analysis revealed that the timing and magnitude of this immune infiltration varied between treatment protocols, with some dogs showing rapid increases in intratumoral lymphoid cells while others exhibited predominantly tumor necrosis . IHC characterization demonstrated that the majority of infiltrating cells were CD3-positive T lymphocytes, providing critical phenotypic information about the nature of the immune response . For optimal results, researchers should collect biopsies at consistent timepoints (e.g., pre-treatment, day 1, day 10, day 17, day 24) and ensure standardized tissue processing and staining protocols . This systematic approach to immunohistochemical analysis allows investigators to correlate treatment variables with specific patterns of immune infiltration, providing mechanistic insights into how IL-2-based therapies modulate the tumor microenvironment in canine models.
A notable difference emerges in receptor expression patterns, with approximately 21% of resting canine lymphocytes expressing IL-2R compared to typically lower percentages in humans . Following mitogenic stimulation with phytohemagglutinin (PHA), canine lymphocytes demonstrate rapid upregulation of IL-2R, reaching 76.4% positive cells by day 3, with receptor density increasing twenty-fold above baseline . The kinetics of this response may differ from human lymphocytes, potentially reflecting evolutionary adaptations in immune responsiveness. Additionally, studies demonstrate that canine peripheral blood lymphocytes from IL-2-treated dogs exhibit enhanced in vitro proliferative responses to IL-2 restimulation, with responses detectable earlier and progressing faster than in pre-treatment samples . This suggests potential differences in receptor sensitization or downstream signaling amplification mechanisms between species. These comparative aspects of IL-2 receptor biology highlight the importance of species-specific research rather than simple extrapolation from human studies, particularly when developing targeted immunotherapeutics for veterinary applications or using canine models to inform human medicine.
Storage stability represents another important consideration, as His-tagged proteins may exhibit different aggregation tendencies compared to native cytokines. Researchers should establish optimal buffer conditions (typically PBS with low concentrations of carrier protein) and validate protein activity after freeze-thaw cycles. For applications involving cell culture, potential metal ion leaching from the His-tag should be considered, particularly in low-serum conditions where chelating effects may be less buffered . When using His-tagged IL-2 for receptor binding studies, competitive binding assays with both tagged and untagged ligands should be employed to distinguish potential tag-mediated artifacts from genuine receptor-ligand interactions. Finally, researchers should consider the potential for anti-His antibody responses in longitudinal in vivo studies, which could neutralize the protein or cause hypersensitivity reactions. Including appropriate controls and potentially monitoring anti-His antibody development through ELISA would strengthen experimental design. These methodological considerations ensure that research findings accurately reflect IL-2 biology rather than artifacts introduced by the His-tag modification.
Interpreting the clinical relevance of IL-2 studies in canine cancer models requires sophisticated analysis frameworks that bridge veterinary and human oncology while acknowledging species-specific differences. Researchers should first evaluate antitumor responses using standardized criteria analogous to RECIST (Response Evaluation Criteria In Solid Tumors) guidelines, categorizing outcomes as complete response, partial response, stable disease, or progressive disease . In canine melanoma studies combining radiation therapy with intratumoral IL-2-containing immunocytokines, clinical outcomes included partial responses in approximately 25% of dogs (3/12) at day 30 post-treatment, with an additional 33% (4/12) showing mixed responses . These response rates provide benchmarks for evaluating treatment efficacy, though researchers should contextualize them against historical controls receiving standard-of-care therapies.
Beyond tumor measurements, comprehensive immune monitoring through sequential tumor biopsies offers mechanistic insights into treatment effects. Histological analysis revealing increased intratumoral lymphocytic infiltration and tumor necrosis following therapy provides evidence of immune activation . Correlating these histological changes with clinical outcomes helps identify potential biomarkers of response, such as early CD8+ T cell infiltration or reduction in FOXP3+ regulatory T cells. Researchers should also consider the translational significance of canine studies, noting that spontaneous canine cancers often better recapitulate human disease biology than induced rodent models, particularly regarding tumor heterogeneity, immune microenvironment, and metastatic behavior . The observation that IL-2-based immunotherapies demonstrate activity in canine melanoma suggests potential applicability to human melanomas with similar immunological profiles . Finally, toxicity profiles must be carefully documented and compared with human experience; the finding that intratumoral IL-2 administration in dogs produces manageable side effects suggests this approach might provide a favorable therapeutic window in human patients . This multifaceted interpretative framework maximizes the translational value of canine IL-2 studies for both veterinary and human oncology.
Escherichia coli represents a well-established expression system for producing His-tagged canine IL-2, offering distinct advantages for research applications. Commercial recombinant dog IL-2 protein with His-tag is successfully expressed in E. coli with high purity (>95%) and low endotoxin levels (<1 EU/μg) . The bacterial expression platform provides cost-effective production with high yield potential, making it accessible for laboratory-scale research. For optimal expression, the mature canine IL-2 sequence (amino acids 21-155) should be cloned into expression vectors containing strong inducible promoters such as T7 or tac, with the His-tag typically positioned at the N-terminus to minimize interference with the protein's C-terminal functional regions . Induction protocols typically employ IPTG (isopropyl β-d-1-thiogalactopyranoside) at concentrations of 0.5-1.0 mM during the logarithmic growth phase, with expression at lower temperatures (16-25°C) often improving protein solubility.
Alternative expression systems offer complementary advantages for specific research applications. Mammalian expression systems (CHO or HEK293 cells) provide proper post-translational modifications, particularly glycosylation, which may affect cytokine stability and activity in vivo. For studies requiring glycosylated canine IL-2, these systems merit consideration despite higher production costs. Baculovirus-insect cell systems represent an intermediate option, offering some eukaryotic processing capabilities with higher protein yields than mammalian systems. Yeast expression platforms (Pichia pastoris or Saccharomyces cerevisiae) provide another alternative with moderate glycosylation capacity and potential for high-density fermentation. Regardless of the expression system chosen, researchers should validate the biological activity of their His-tagged canine IL-2 through functional assays such as T-cell proliferation tests or phospho-STAT5 induction in IL-2-responsive cell lines. Each expression system presents distinct trade-offs between yield, cost, endotoxin contamination, and post-translational modifications, with the optimal choice depending on specific experimental requirements and available resources.
Purification of His-tagged canine IL-2 requires a strategic multi-step approach to achieve high purity while preserving biological activity. The cornerstone technique leverages the affinity of the His-tag for immobilized metal ions in immobilized metal affinity chromatography (IMAC) . This process typically begins with cell lysis under conditions that maximize protein solubility, often using mild detergents (0.1% Triton X-100) and protease inhibitors to prevent degradation. For E. coli-expressed His-tagged canine IL-2, inclusion body formation may occur, necessitating solubilization with chaotropic agents (6-8M urea or 4-6M guanidine hydrochloride) followed by refolding through controlled dilution or dialysis against decreasing concentrations of denaturant .
The IMAC purification typically employs nickel or cobalt-charged resins, with cobalt often providing higher specificity despite lower capacity. Buffer optimization is critical, with imidazole concentrations carefully tiered: low concentrations (5-10 mM) in binding buffers to minimize non-specific binding, intermediate levels (20-50 mM) in wash buffers to remove weakly bound contaminants, and high concentrations (250-500 mM) for elution of the target protein . Following IMAC, secondary purification steps such as size exclusion chromatography separate aggregates and provide buffer exchange into physiological conditions. For applications requiring endotoxin removal, additional steps such as anion exchange chromatography or specialized endotoxin removal resins should be employed to achieve levels below 0.1 EU/μg .
Quality control assessment should include SDS-PAGE analysis under both reducing and non-reducing conditions to verify purity and evaluate potential disulfide-mediated aggregation. Western blotting using anti-His antibodies confirms tag presence, while mass spectrometry verifies protein identity and integrity . Activity assays measuring proliferation of IL-2-dependent cell lines provide the critical final validation step, with specific activity (units of activity per mg protein) compared against reference standards. This comprehensive purification strategy typically yields His-tagged canine IL-2 with >95% purity and fully preserved biological activity suitable for sophisticated immunological investigations.
Accurate quantification of IL-2 receptor expression and binding kinetics in canine samples requires sophisticated methodological approaches combining flow cytometry with biochemical binding assays. Flow cytometry using phycoerythrin-labeled IL-2 (IL-2-PE) provides a direct method for detecting and quantifying IL-2 receptors on canine lymphocytes . This technique allows researchers to determine both the percentage of receptor-positive cells and the receptor density through mean fluorescence intensity measurements . For optimal results, researchers should include appropriate controls, including unstained cells and cells stained with irrelevant fluorochrome-conjugated proteins (such as anti-mouse IgG-PE) to establish background fluorescence levels . Additionally, competitive binding experiments using unlabeled IL-2 confirm binding specificity, with a significant reduction in fluorescence intensity (approximately four-fold) validating that IL-2-PE genuinely detects IL-2 receptors rather than binding non-specifically .
For detailed characterization of binding kinetics, surface plasmon resonance (SPR) or biolayer interferometry (BLI) provides quantitative binding parameters including association rate (ka), dissociation rate (kd), and equilibrium dissociation constant (KD). These techniques typically employ recombinant canine IL-2 receptors (either as soluble extracellular domains or as membrane preparations) immobilized on sensor surfaces, with varying concentrations of His-tagged IL-2 as the analyte. Scatchard analysis using radiolabeled IL-2 in equilibrium binding assays offers an alternative approach, particularly useful for distinguishing high-affinity (trimeric) from low-affinity (dimeric) receptor complexes through biphasic binding curves. Researchers can also employ real-time cellular assays measuring downstream signaling events, such as phospho-STAT5 induction using phospho-flow cytometry or Western blotting, to assess functional receptor activity. These methods allow time-course and dose-response analyses that complement binding data with functional outcomes. Together, these techniques provide researchers with comprehensive tools to characterize IL-2 receptor biology in canine samples, supporting sophisticated investigations of cytokine-receptor interactions in both health and disease states.
Canine models of IL-2 immunotherapy provide valuable translational insights for human clinical approaches due to several key advantages over traditional rodent models. Dogs develop spontaneous cancers that share many molecular and immunological features with human malignancies, including similar tumor heterogeneity, metastatic patterns, and immune evasion mechanisms . This natural disease development in the context of an intact immune system more accurately reflects the challenges faced in human cancer immunotherapy compared to xenograft or induced tumor models in immunocompromised mice. Studies of intratumoral IL-2 administration in combination with radiation therapy in canine melanoma have demonstrated the feasibility of converting tumors into in situ vaccines, with observable increases in intratumoral lymphocytic infiltration and clinical responses including partial tumor regression in some animals . These findings provide proof-of-concept for similar approaches in human patients, particularly for accessible tumors amenable to intratumoral injection.
The toxicity profile observed in dogs receiving IL-2 therapy offers critical information for human trial design. Canine studies have shown that continuous infusion of IL-2 at doses of 3 × 10^6 units/m^2/day produces manageable toxicities including mild vomiting, diarrhea, and lethargy without severe adverse events like fever, tachypnea, or weight gain . This suggests potential dosing strategies that might balance efficacy and safety in human patients. Additionally, the observed immunological effects, including marked lymphocytosis, eosinophilia, and enhanced natural killer cell activity, provide biomarkers that could be monitored in human trials to assess biological activity . The finding that human recombinant IL-2 can effectively bind canine IL-2 receptors and elicit biological responses suggests structural conservation across species that supports translational relevance . As larger animals with body size and physiology more similar to humans than rodents, dogs also provide better pharmacokinetic and pharmacodynamic models for dose translation to human studies. These multiple advantages position canine IL-2 studies as valuable bridges between preclinical rodent work and human clinical trials, potentially accelerating the development of effective immunotherapy approaches.
Combination strategies pairing IL-2 with complementary therapeutic modalities have demonstrated significant promise in canine immunotherapy research. The integration of radiation therapy with intratumoral IL-2 administration represents one of the most thoroughly investigated approaches . In canine melanoma studies, dogs received either a single 8 Gy radiation fraction or three 8 Gy fractions delivered over one week, followed by intratumoral injection of IL-2-containing immunocytokine (hu14.18-IL2) at 12 mg/m^2 for three consecutive days . This protocol generated measurable clinical responses, with 25% of dogs (3/12) achieving partial responses and an additional 33% (4/12) showing mixed responses . The radiation component is believed to enhance immunogenicity through tumor cell death and release of danger-associated molecular patterns (DAMPs), while the localized IL-2 delivery stimulates immune cell proliferation and activation within the tumor microenvironment .
Additional promising combinations include IL-2 with immune checkpoint inhibitors, although data in canine models remains more limited compared to human studies. The rationale for this approach stems from the complementary mechanisms of action: checkpoint inhibitors (targeting PD-1/PD-L1 or CTLA-4 pathways) remove brakes on immune responses, while IL-2 provides positive stimulation to expand tumor-reactive T-cells. Early research suggests that timing and sequencing of these agents significantly impacts efficacy, with IL-2 potentially most effective following checkpoint inhibitor priming. Combinations of IL-2 with tumor-targeting antibodies represent another strategy leveraging the ability of IL-2 to enhance antibody-dependent cellular cytotoxicity through NK cell activation . The fusion of these approaches in the form of immunocytokines (antibodies linked to IL-2) allows targeted delivery of the cytokine to the tumor microenvironment, as demonstrated with the GD2-reactive hu14.18-IL2 construct used in canine melanoma . These combination approaches highlight the versatility of IL-2 as an immunotherapeutic agent and its potential synergy with diverse treatment modalities, providing a rich pipeline for translational development in both veterinary and human oncology.
Designing optimal dose-finding studies for canine IL-2 therapy requires sophisticated methodological approaches that balance efficacy, toxicity, and practical considerations. A scientifically rigorous design begins with clear definition of the dose-limiting toxicities (DLTs) based on established veterinary adverse event criteria, typically including gastrointestinal, hematological, and constitutional symptoms . Researchers should employ a modified 3+3 design or a Bayesian optimal interval design, starting at approximately 30% of the maximum tolerated dose established in previous animal studies . For continuous infusion protocols, initial dosing at 1 × 10^6 units/m^2/day with escalation to 3 × 10^6 units/m^2/day and beyond provides a rational framework based on published experience . For intratumoral administration, starting doses around 4 mg/m^2 with escalation to 12 mg/m^2 follow established precedents .
Comprehensive pharmacokinetic and pharmacodynamic assessments strengthen dose-finding studies. Serial blood sampling should measure IL-2 levels using validated ELISA methods with appropriate sensitivity for canine samples . Concurrently, researchers should monitor immune biomarkers including absolute lymphocyte counts, NK cell activity, and cytokine profiles to establish exposure-response relationships . Flow cytometric analysis of peripheral blood using IL-2-PE can quantify IL-2 receptor upregulation as a pharmacodynamic endpoint . Tumor biopsies at baseline and post-treatment timepoints provide critical information about immune infiltration and activation within the target tissue . Researchers should incorporate quality-of-life assessments using validated canine instruments to capture subjective treatment effects beyond laboratory parameters.
The recombinant canine IL-2 protein consists of amino acids 21-155 of the canine IL-2 sequence and includes an N-terminal His-tag . The amino acid sequence of the recombinant protein is as follows :
MGSSHHHHHH SSGLVPRGSH MGSHMAPITS SSTKETEQQM EQLLLDLQLL LNGVNNYENP QLSRMLTFKF YTPKKATEFT HLQCLAEELK NLEEVLGLPQ SKNVHLTDTK ELISNMNVTL LKLKGSETSY NCEYDDETAT ITEFLNKWIT FSQSIFSTLT
The predicted molecular mass of the recombinant canine IL-2 protein is approximately 18.1 kDa . The protein is highly pure, with a purity greater than 95% as determined by SDS-PAGE . The endotoxin level is less than 1.0 EU per microgram of protein, as determined by the Limulus Amebocyte Lysate (LAL) method .
Recombinant canine IL-2 has been shown to stimulate cell cytotoxicity in the CTLL-2 mouse cytotoxic T cell line . The effective dose (ED50) range for this activity is less than or equal to 0.3 ng/mL . This demonstrates the protein’s ability to promote immune cell activation and proliferation, which is consistent with the known functions of natural IL-2 .
Recombinant canine IL-2 is used in various research applications to study the immune response and to develop potential therapeutic strategies for diseases involving the immune system . It may also be of use as an anti-tumor agent in cancer therapy, given its role in promoting the activation and proliferation of immune cells .
The recombinant canine IL-2 protein is typically formulated in phosphate-buffered saline (PBS) at pH 7.4 and is provided without preservatives . For short-term storage, the protein should be kept at 4°C, while for long-term storage, it should be aliquoted and stored at -20°C to avoid freeze-thaw cycles .