IL-2 Canine

Interleukin-2 Canine Recombinant
Shipped with Ice Packs
In Stock

Description

Definition and Biochemical Characteristics

IL-2 Canine is a recombinant cytokine protein derived from Canis lupus familiaris (domestic dog) that plays a critical role in immune regulation. It belongs to the interleukin family and functions as a T-cell growth factor, stimulating lymphocyte proliferation and differentiation .

Key Biochemical Features

PropertyDetailsSource
Molecular Weight15.6 kDa (non-glycosylated monomer)
Amino Acid Sequence136 residues (Ala21-Thr155)
MutationsCys147Ser variant for enhanced stability
Sequence Similarity90% feline, 86% human, 76% murine

IL-2 Canine binds to the IL-2 receptor complex (αβγ subunits), shared with IL-4, IL-7, and IL-9, driving T-cell activation and survival . Its recombinant forms are produced in E. coli, with >95% purity confirmed via SDS-PAGE .

Biological Activity and Mechanism

IL-2 Canine exhibits dose-dependent bioactivity, with an ED₅₀ of 0.15–0.8 ng/mL in CTLL-2 cell proliferation assays . Key functions include:

  • T-cell proliferation: Supports long-term culture of canine lymphocytes (>10 passages) with PHA-P stimulation .

  • Immune modulation: Enhances NK cell cytolytic activity and B-cell differentiation .

  • Antitumor effects: Induces cytostasis in transmissible venereal sarcoma cells and pulmonary metastases .

Experimental Data

ParameterResultSource
Max IL-2 Production2×10⁶ cells/mL + 10 µg/mL PHA-P for 48h
StabilitySensitive to pH <4 or >10; trypsin-resistant
Lymphocytosis7-fold increase post-treatment

Therapeutic Applications in Canine Oncology

IL-2 Canine has been tested in diverse cancer models, with varying efficacy:

Intratumoral Injection

  • Mast Cell Tumors (MCT):

    • Dose: 4.5×10⁶ IU per tumor .

    • Responses:

      • Complete Regression (CR): 3/10 dogs (30%); duration: 6–21 months .

      • Partial Regression (PR): 4/10 dogs (40%); tumor size reduction by 50–90% .

  • Soft Tissue Sarcomas (STS):

    • Collagen-bound IL-2 + IL-12: Transient T-cell infiltration; Grade 1/2 toxicity (fever, thrombocytopenia) .

Inhalation Therapy

  • Pulmonary Metastases:

    • Regimen: Nebulized IL-2 liposomes for 30 days .

    • Outcomes:

      • CR: 2/7 dogs (osteosarcoma metastases; stable >12 months) .

      • Stabilization: 1/2 dogs (lung carcinoma; >8 months) .

Systemic IL-2/CD40 Immunotherapy

  • Phase I Trial:

    • Dosing: 9 cohorts (3 dogs each) with escalating IL-2 + anti-CD40 antibodies .

    • Responses:

      • Clinical Benefit Rate: 68.4% at 1 month (13/19 dogs) .

      • Durable SD: 7 dogs (>50 days); 1 dog achieved PR with 90% tumor reduction .

Future Directions

  • Combination Therapies: IL-2 + CD40 agonists or checkpoint inhibitors to counter immune counterregulation .

  • Collagen-anchoring: Enhanced tumor retention of IL-2/IL-12 for sustained T-cell activation .

  • Translational Models: Comparative oncology trials in dogs as surrogates for human cancers .

Product Specs

Introduction
Interleukin-2 (IL-2) is a cytokine crucial for the growth and differentiation of T and B lymphocytes. Its receptor is a heterotrimeric complex, with the gamma chain shared by IL-4 and IL-7 receptors. Interestingly, IL-2 expression in mature thymocytes is monoallelic, representing a unique regulatory mechanism for precise gene expression control.
Description
Recombinant Canine Interleukin-2, produced in E. coli, is a non-glycosylated monomeric protein with a molecular weight of 15.6 kDa. It comprises 136 amino acids and is purified using proprietary chromatographic methods.
Physical Appearance
White, lyophilized powder, sterile-filtered.
Formulation
The protein is lyophilized from a sterile solution containing 20 mM sodium bicarbonate at pH 8.5. The solution is filtered through a 0.2 μm filter before lyophilization.
Solubility
Reconstitute the lyophilized IL-2 in sterile 10 mM sodium bicarbonate (pH 8.5) at a concentration of 0.1 mg/mL. This solution can be further diluted in other aqueous solutions as needed.
Stability
Lyophilized IL-2 remains stable at room temperature for up to 3 weeks. However, for long-term storage, store desiccated below -18°C. After reconstitution, store IL-2 at 4°C for 2-7 days. For longer storage, freeze below -18°C, avoiding repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 95% by SDS-PAGE analysis.
Biological Activity
The ED50, determined by CTLL-2 cell proliferation assay, is less than 5 ng/ml. This corresponds to a specific activity greater than or equal to 2.0 x 10^5 units/mg.
Synonyms

Interleukin-2, IL-2, T-cell growth factor, TCGF.

Source

Escherichia Coli.

Amino Acid Sequence

MAPITSSSTK ETEQQMEQLL LDLQLLLNGV NNYENPQLSR MLTFKFYTPK KATEFTHLQC LAEELKNLEE VLGLPQSKNV HLTDTKELIS NMNVTLLKLK GSETSYNCEY DDETATITEF LNKWITFSQS IFSTLT.

Q&A

What are the fundamental characteristics of canine interleukin-2?

Canine IL-2 is a cytokine produced by stimulated peripheral blood lymphocytes with key immunomodulatory functions. Research characterization has revealed that optimal production occurs when canine peripheral blood lymphocytes (PBL) at a concentration of 2 × 10^6 cells/ml are stimulated with phytohemagglutinin-P (PHA-P) at 10 μg/ml and cultured at 38°C for 48 hours. The produced IL-2 has a molecular weight of approximately 31,000 as determined by gel filtration . Canine IL-2 displays sensitivity to environmental conditions, with significant activity inhibition occurring when exposed to temperatures above 65°C, pH conditions below 4 or above 10, or trypsin treatment . These properties must be considered when designing experimental protocols involving canine IL-2 isolation or application.

How can researchers validate the bioactivity of recombinant canine IL-2 preparations?

Validation of canine IL-2 bioactivity requires specific proliferation assays using IL-2-dependent cell lines. The standard approach involves culturing 10,000 CTLL-2 cells (ATCC Cat# TIB-214, RRID:CVCL_0227) per well in incomplete T-cell media (RPMI 1640 supplemented with 10% fetal bovine serum, 2mM L-glutamine, and 1mM sodium pyruvate) . These cells are then incubated for 48 hours at 37°C with serial dilutions of the IL-2 preparation being tested. Cell viability is subsequently assessed using the CellTiter-Glo 2.0 assay or comparable methodologies . It's important to note that while murine CTLL-2 cells respond to canine IL-2, researchers should be aware that canine IL-2 does not cross-react efficiently with the high-affinity murine IL-2Rα, which may influence comparative potency assessments .

What detection systems are available for quantifying canine IL-2 in research samples?

Several validated detection systems exist for canine IL-2 quantification:

  • Sandwich ELISA Development Kits: Commercial DuoSet ELISA systems contain optimized capture and detection antibody pairings that can measure both natural and recombinant canine IL-2 in cell culture supernatants . These systems typically include capture antibodies, detection antibodies, recombinant standards, and streptavidin-HRP conjugates.

  • Complete ELISA Kits: Pre-optimized for canine IL-2 detection in serum, plasma, and supernatant samples .

When selecting a detection system, researchers should consider the sample type being analyzed, as the diluents for complex matrices such as serum and plasma may require validation prior to use . For cell culture supernatants, standard diluents included in commercial kits are generally suitable without additional optimization.

How have collagen-anchored IL-2 formulations been engineered for intratumoral applications in canine cancer models?

Collagen-anchored IL-2 formulations represent an advanced approach to overcome the narrow therapeutic window of conventional IL-2 therapy. These engineered cytokines are created by fusing canine IL-2 to the collagen-binding domain LAIR1, resulting in molecules that can bind to and associate with tumor collagen following intratumoral injection . The molecular engineering process involves:

  • Cloning the canine IL-2 gene sequence

  • Fusion with the LAIR1 collagen-binding domain

  • Expression in appropriate systems

  • Confirmation of both biological activity and collagen-binding capacity

Functional validation of these fusion proteins requires a multi-step approach:

  • Bioactivity testing using CTLL-2 cell proliferation assays

  • Collagen-binding confirmation via ELISA methodologies

  • In vivo assessment of tissue retention

This engineering approach addresses the fundamental challenge of systemic cytokine toxicity by restricting cytokine activity to the tumor microenvironment, thereby reducing off-target effects while maintaining localized immunostimulatory function .

What are the observed immunological changes in the tumor microenvironment following intratumoral administration of canine IL-2?

Intratumoral administration of canine IL-2 induces significant immunological remodeling of the tumor microenvironment. Research findings demonstrate:

  • Enhanced T-cell infiltration: Immunohistochemistry analyses reveal increased T-cell presence within treated tumors, representing a key mechanism of antitumor activity .

  • Upregulation of cytotoxic gene expression: NanoString RNA profiling shows enhanced expression of genes associated with cytotoxic immune function following IL-2 treatment .

  • Concurrent counter-regulatory gene induction: Treatment also triggers upregulation of immunosuppressive pathways, suggesting a complex balance between pro-inflammatory and regulatory responses .

  • Spatiotemporal dynamics: The immunological changes show time-dependent patterns, with observations that these effects may be transient in nature without additional intervention .

These findings provide critical insights for researchers designing immunotherapeutic protocols, suggesting that additional targeting of counter-regulatory pathways might be necessary to achieve sustained antitumor responses.

What factors influence the production and stability of canine IL-2 in experimental systems?

Multiple factors significantly impact canine IL-2 production and stability in research settings:

  • Cell density optimization: Maximum IL-2 production occurs at specific peripheral blood lymphocyte concentrations (2 × 10^6 cells/ml), with deviations resulting in suboptimal yields .

  • Stimulation conditions: PHA-P concentration (optimal at 10 μg/ml) and stimulation duration (48 hours) critically determine IL-2 production efficiency .

  • Temperature sensitivity: Canine IL-2 activity demonstrates significant reduction when exposed to temperatures above 65°C, requiring careful handling during purification and storage protocols .

  • pH stability threshold: Activity is maintained within a pH range of 4-10, with significant loss of function outside this range .

  • Proteolytic susceptibility: Trypsin exposure rapidly degrades IL-2 activity, necessitating protease inhibition during isolation procedures .

  • Long-term culture considerations: For maintaining IL-2-dependent canine lymphocyte cultures, periodic restimulation with PHA-P (approximately every 3 passages) is necessary to sustain IL-2 responsiveness and cellular viability .

These parameters must be carefully controlled when designing experiments involving canine IL-2 production, purification, or application to ensure reproducible outcomes.

How should dose-finding studies for canine IL-2 be designed to maximize safety and efficacy?

Designing dose-finding studies for canine IL-2 requires a structured approach that prioritizes both safety and biological activity. Based on successful translational studies, the following methodological framework is recommended:

  • Initial dose determination: Begin with allometric scaling from human IL-2 maximum tolerated doses, adjusted for canine physiology .

  • Dose escalation design: Implement a 3+3 dose escalation design, where cohorts of three animals receive increasing doses, with careful monitoring for dose-limiting toxicities before advancing to the next level .

  • Toxicity monitoring parameters: Include comprehensive assessments of:

    • Vital signs (temperature, heart rate, respiratory rate)

    • Complete blood counts with particular attention to neutrophil and platelet counts

    • Serum chemistry panels

    • Clinical symptom evaluation (lethargy, appetite, vomiting)

  • Biomarker sampling: Incorporate serial sampling of relevant biomarkers to correlate dose with biological activity, including:

    • Serum cytokine levels

    • Immune cell phenotyping

    • When feasible, tumor biopsies for assessing local immune infiltration

  • Healthy animal validation: Consider initial studies in healthy research animals (e.g., beagles) before advancing to client-owned pets with spontaneous disease .

This approach has successfully identified well-tolerated yet biologically active dosing regimens for intratumoral IL-2 in dogs with soft tissue sarcomas, where only Grade 1/2 adverse events (mild fever, thrombocytopenia, neutropenia) were observed despite robust local immune activation .

What are the methodological approaches for analyzing IL-2-induced changes in canine tumor microenvironments?

Comprehensive analysis of IL-2-induced changes in canine tumor microenvironments requires multiple complementary techniques:

  • Immunohistochemistry (IHC):

    • Enables spatial visualization of immune cell infiltrates

    • Quantifies treatment-associated changes in T-cell populations

    • Assesses changes in regulatory cell populations

    • Documents alterations in tumor cell phenotype

  • Transcriptomic profiling:

    • NanoString RNA profiling permits multiplexed quantification of immune-related gene expression

    • Allows for comprehensive pathway analysis of both stimulatory and counter-regulatory mechanisms

    • Can identify predictive biomarkers of response (e.g., B2m loss as a predictor of poor response)

    • Enables comparison with untreated control samples to isolate treatment effects

  • Functional immune assays:

    • Ex vivo stimulation of tumor-infiltrating lymphocytes

    • Cytotoxicity assays against autologous tumor cells

    • Cytokine production profiling

  • Imaging assessments:

    • CT measurement of tumor regression at treated lesions

    • Evaluation of untreated metastatic sites to assess systemic immune activation

These complementary approaches provide a comprehensive understanding of both the magnitude and mechanism of IL-2-induced antitumor immune responses, critical for optimizing therapeutic protocols.

How can researchers distinguish between local and systemic effects of intratumoral IL-2 administration in canine models?

Distinguishing between local and systemic effects following intratumoral IL-2 administration requires a methodical assessment strategy:

  • Multi-site tumor evaluation:

    • In dogs with metastatic disease, monitor both treated and untreated tumor sites

    • Document regression at untreated sites as evidence of systemic immune activation

    • Comparative advanced imaging (CT/MRI) to quantify differential responses

  • Pharmacokinetic mapping:

    • Serial blood sampling to detect IL-2 in circulation

    • Tissue sampling from tumor and non-tumor sites when possible

    • Comparison of standard IL-2 versus collagen-anchored IL-2 biodistribution

  • Immune cell trafficking studies:

    • Phenotyping of tumor-infiltrating lymphocytes at treatment site

    • Concurrent evaluation of peripheral blood immune composition

    • Assessment of lymph nodes draining both treated and untreated tumor sites

  • Adverse event profiling:

    • Systematic documentation of local reactions (injection site inflammation)

    • Monitoring for systemic cytokine-associated toxicities (fever, hypotension, capillary leak)

    • Correlation between systemic cytokine levels and observed adverse events

  • Serum biomarker analysis:

    • Longitudinal assessment of serum immune markers

    • Correlation with treatment timing and clinical outcomes

These approaches collectively enable researchers to determine the extent to which intratumoral IL-2 remains localized versus generating systemic immunomodulatory effects, a critical distinction for therapeutic application.

What are the outcomes of intratumoral IL-2 therapy across different canine cancer models?

Intratumoral IL-2 therapy has demonstrated variable efficacy across different canine cancer models, with response patterns that provide valuable insights for translational research:

These findings suggest that intratumoral IL-2 has meaningful clinical activity across multiple canine cancer types, with response rates varying by tumor histology, treatment combination, and molecular features. The particular success in combining IL-2 with radiation therapy for melanoma highlights the potential synergy between immunotherapy and standard-of-care treatments.

How does the combination of IL-2 with IL-12 affect therapeutic outcomes in canine oncology research?

The combination of IL-2 with IL-12 represents a strategic approach to enhance antitumor immunity through complementary mechanisms of action. Research findings demonstrate:

  • Mechanistic synergy:

    • IL-2 primarily expands and activates T cells and NK cells

    • IL-12 promotes Th1 polarization and enhances IFN-γ production

    • Together, they establish a more robust cytotoxic immune environment

  • Safety profile:

    • Collagen-anchored formulations of both cytokines administered intratumorally demonstrate favorable safety

    • Only Grade 1/2 adverse events observed (mild fever, thrombocytopenia, neutropenia)

    • Lack of dose-limiting toxicities typically associated with systemic administration

  • Clinical efficacy in advanced cancers:

    • In malignant melanoma (combined with radiation therapy):

      • 76.9% of dogs showed tumor regression at treated sites

      • 61.5% of dogs with metastatic disease showed partial responses across combined lesions

      • Median survival of 256 days across all tumor stages

  • Immune microenvironment remodeling:

    • Enhanced T-cell infiltration into tumors

    • Increased expression of cytotoxic function genes

    • Concurrent upregulation of counter-regulatory pathways

These findings suggest that the IL-2/IL-12 combination provides enhanced therapeutic potential over either cytokine alone, while the collagen-anchoring approach successfully mitigates the systemic toxicity that has historically limited cytokine therapy in both veterinary and human oncology.

What predictive biomarkers have been identified for response to canine IL-2 therapy?

Research has identified several potential predictive biomarkers for response to canine IL-2 therapy:

  • B2m (Beta-2-microglobulin) expression:

    • Loss of B2m has been identified as predictive of poor response to IL-2/IL-12 combination therapy

    • NanoString profiling revealed this correlation in dogs with malignant melanoma

    • Mechanistically consistent with the role of B2m in MHC class I assembly and antigen presentation

  • Pre-treatment immune infiltration:

    • Baseline presence of tumor-infiltrating lymphocytes may correlate with response

    • Suggests importance of pre-existing immune recognition of tumor

  • Counter-regulatory pathway activation:

    • Expression of immune checkpoint molecules (e.g., PD-L1)

    • Upregulation of immunosuppressive cytokines

    • These pathways may limit duration of response to IL-2 therapy

  • Tumor mutation burden:

    • Higher mutation burden may correlate with improved response to immunotherapy

    • Generates more neoantigens for immune recognition

These biomarkers highlight the importance of personalized approaches to canine immunotherapy, suggesting that patient selection based on tumor molecular characteristics may optimize treatment outcomes. The identification of B2m loss as a resistance mechanism particularly emphasizes the critical role of intact antigen presentation machinery for effective IL-2-mediated immune responses.

What combinatorial approaches might enhance the efficacy of canine IL-2 therapy?

Several promising combinatorial approaches warrant investigation to enhance canine IL-2 therapy efficacy:

  • Radiation therapy combinations:

    • Radiation has demonstrated synergy with collagen-anchored IL-2/IL-12

    • Median survival of 256 days in dogs with malignant melanoma

    • Likely mechanisms include increased tumor antigen release and enhanced immune recognition

  • Immune checkpoint inhibition:

    • Targeting counter-regulatory pathways induced by IL-2 therapy

    • Mouse models confirm improved responses when combining cytokine therapy with checkpoint inhibition

    • Could potentially convert transient responses into durable remissions

  • Antigen-presenting cell activation:

    • Combining with toll-like receptor agonists

    • Addition of GM-CSF to enhance dendritic cell function

    • Strategies to compensate for B2m loss in resistant tumors

  • Tumor microenvironment modulation:

    • Targeting immunosuppressive myeloid populations

    • Matrix-modifying agents to enhance immune cell infiltration

    • Angiogenesis inhibitors to normalize tumor vasculature

  • Novel cytokine engineering approaches:

    • Development of multi-specific cytokine fusion proteins

    • Exploration of alternative collagen-binding domains with optimized affinity

    • Controlled-release formulations for sustained local activity

These approaches address the identified limitations of IL-2 monotherapy, particularly the transient nature of responses and the activation of counter-regulatory pathways that may limit therapeutic efficacy.

How might findings from canine IL-2 studies translate to human clinical applications?

Canine IL-2 studies offer several translational insights with significant implications for human clinical applications:

  • Validation of localized cytokine delivery:

    • Collagen-anchored cytokines demonstrated safety and efficacy in spontaneous canine cancers

    • Addresses the fundamental limitation of systemic cytokine toxicity

    • Provides rationale for similar approaches in human trials

  • Identification of resistance mechanisms:

    • B2m loss as a predictor of poor response has direct parallels in human cancer

    • Suggests patient selection strategies and potential combination approaches

  • Optimization of radiation-immunotherapy sequencing:

    • Successful combination of radiation with IL-2/IL-12 in canine melanoma

    • Informs timing and dosing for human protocols

  • Counter-regulatory pathway targeting:

    • Identification of immune feedback mechanisms limiting cytokine efficacy

    • Directs rational combination strategies with checkpoint inhibitors

  • Comparative assessment of tumor microenvironment changes:

    • Similar patterns of immune infiltration and activation

    • Shared mechanisms of both response and resistance

The significant advantages of canine studies include evaluation in immunocompetent hosts with spontaneous tumors that mirror the genetic heterogeneity, microenvironmental complexity, and metastatic patterns observed in human cancer, providing more predictive models than traditional mouse studies.

Product Science Overview

Introduction

Interleukin-2 (IL-2) is a cytokine that plays a crucial role in the immune system by promoting the growth, differentiation, and survival of certain immune cells. It was initially identified as a T cell growth factor, but its functions extend to other cell types, including B cells, natural killer (NK) cells, and monocytes/macrophages . The recombinant form of canine IL-2 is produced using genetic engineering techniques, primarily in Escherichia coli (E. coli) bacteria .

Production and Structure

Recombinant canine IL-2 is typically produced in E. coli as a non-glycosylated polypeptide chain. The production process involves inserting the gene encoding canine IL-2 into E. coli, which then expresses the protein. The recombinant protein is purified using proprietary chromatographic techniques to ensure its purity and functionality .

The recombinant canine IL-2 produced in E. coli is a single, non-glycosylated polypeptide chain containing 136 to 160 amino acids, depending on the specific production method. It has a molecular mass ranging from 15.6 kDa to 18.1 kDa . Some versions of the recombinant protein may include additional tags, such as a His-tag, to facilitate purification .

Biological Activity

Recombinant canine IL-2 retains the biological activity of the native protein. It can induce the proliferation of T cells and other immune cells, demonstrating its functional activity . This makes it a valuable tool for research and therapeutic applications, particularly in the field of veterinary medicine.

Applications

The primary applications of recombinant canine IL-2 include:

  1. Research: It is used in various immunological studies to understand the role of IL-2 in the canine immune system. Researchers use it to investigate the mechanisms of T cell activation, proliferation, and differentiation.
  2. Therapeutics: Recombinant canine IL-2 has potential therapeutic applications in treating immune-related disorders in dogs. It can be used to enhance the immune response in cases of infections, cancer, and other diseases where immune modulation is beneficial.
  3. Diagnostics: It can be used in diagnostic assays to measure the levels of IL-2 or to assess the functional status of the immune system in dogs.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.