Fusion protein of the extra cellular part of IL-1RAP (Pro100-Lys330)
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
≤ 1.0 EU/mg as determined by the LAL method
Purity
≥95% by SDS Page
⋅
≥95% monomer by analytical SEC
Formulation
This biosimilar antibody is aseptically packaged and formulated in 0.01 M phosphate buffered saline (150 mM NaCl) PBS pH 7.2 - 7.4 with no carrier protein, potassium, calcium or preservatives added. Due to inherent biochemical properties of antibodies, certain products may be prone to precipitation over time. Precipitation may be removed by aseptic centrifugation and/or filtration.
State of Matter
Liquid
Product Preparation
Recombinant biosimilar antibodies are manufactured in an animal free facility using only in vitro protein free cell culture techniques and are purified by a multi-step process including the use of protein A or G to assure extremely low levels of endotoxins, leachable protein A or aggregates.
Pathogen Testing
To protect mouse colonies from infection by pathogens and to assure that experimental preclinical data is not affected by such pathogens, all of Leinco’s recombinant biosimilar antibodies are tested and guaranteed to be negative for all pathogens in the IDEXX IMPACT I Mouse Profile.
Storage and Handling
Functional grade preclinical antibodies may be stored sterile as received at 2-8°C for up to one month. For longer term storage, aseptically aliquot in working volumes without diluting and store at ≤ -70°C. Avoid Repeated Freeze Thaw Cycles.
Each investigator should determine their own optimal working dilution for specific applications. See directions on lot specific datasheets, as information may periodically change.
Description
Description
Specificity
This non-therapeutic biosimilar antibody uses the same variable region sequence as
the therapeutic antibody Nadunolimab. Nadunolimab (CAN04) activity is directed against IL-
1RAP.
Background
IL-1RAP (interleukin-1 receptor accessory protein) is a co-receptor that works with the IL-36 subfamily of the IL-1 interleukin family1. Members of the IL-1 family play key signaling roles in innate and adaptive immunity. IL-1RAP is particularly involved in promoting inflammatory responses via the IL-1, IL-33, and IL-36 signaling pathways. IL-1RAP also plays a role in cancer promotion, progression, and metastasis. Additionally, IL-1RAP dimerizes with IL-1R1 to initiate signaling of IL-1α and IL-1β, both of which promote chemotherapy resistance2. As such, IL-1RAP is a target for cancer therapy1. Two different therapies targeting IL-1RAP are under development: 1) chimeric antigen receptor T-cells (CAR-T) therapy and 2) antibody immunotherapy for either direct blockade or activation of antibody-dependent cell-mediated cytotoxicity (ADCC).
Nadunolimab was generated by grafting the complementarity-determining regions of a murine monoclonal antibody into a human IgG1 isotype3. BALB/c mice were immunized with a fusion protein of the extracellular part of IL-1RAP (Pro100-Lys330) and the Fc-part of human IgG1. Splenocytes were fused with mouse myeloma cell line Sp2/0 and screened by ELISA for binding to the IL-1RAP extracellular domain3,4.
Nadunolimab binds IL-1RAP in a manner that blocks IL-1α and IL-1β as well as partially inhibits IL-33 signaling. Nadunolimab binds to the top of the IL-1RAP D2 domain, specifically to residues 105-114, 145-158, and 169-1765. Residues Thr154 to Ile171 are necessary for binding, with Q165 being critical. Additionally, binding is directly coincident with portions of the IL-1β and IL-33 signaling complexes.
Alternative names for nadunolimab are CAN04 and mAb3F8.
Antigen Distribution
IL-1RAP is ubiquitously expressed. On normal blood cells, IL-1RAP is
expressed mainly on monocytes. IL-1RAP is overexpressed on tumor cells of several
hematological and solid cancers. IL-1RAP has both membrane-bound and soluble forms.
Powered by AI: AI is experimental and still learning how to provide the best assistance. It may occasionally generate incorrect or incomplete responses. Please do not rely solely on its recommendations when making purchasing decisions or designing experiments.
Research-grade Nadunolimab biosimilars are commonly used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays to accurately measure drug concentrations in serum samples during biosimilar development and PK studies.
In practice:
The biosimilar (such as Nadunolimab) is selected as the assay calibrator—i.e., the reference standard used to generate the ELISA calibration curve that relates optical density (OD) to drug concentration in serum samples.
The calibrator is serially diluted into the relevant matrix (such as human serum), covering a wide range of concentrations to ensure the assay's dynamic range fits expected sample values.
Quality controls (QCs) and test samples—including both the biosimilar and reference product (e.g., the branded Nadunolimab)—are quantified by comparison against the calibration curve made from the biosimilar standard.
Why use the biosimilar as the calibration standard?
Regulatory and industry consensus indicates that a single PK assay with a single analytical standard (the biosimilar) for both biosimilar and reference product quantification minimizes assay variability, eliminates the need for multiple methods, and streamlines blinded studies.
This strategy requires rigorous qualification: method qualification studies must demonstrate the assay's precision, accuracy, and bioanalytical comparability for both the biosimilar and reference products, ensuring both are measured equivalently within the same method.
Key procedures in an ELISA for PK bridging:
A robust method is developed and validated, often analyzing multiple biosimilar standards prepared in serum at different concentrations.
Validation samples of both biosimilar and reference compounds are assayed, confirming the accuracy and comparability of drug concentration measurements regardless of product source.
Serial dilutions and internal standards may be included on each plate to control for variation across assays, with both negative and positive controls recommended for reproducibility.
Additional notes:
While Nadunolimab and its biosimilars have been studied for safety and efficacy as drugs, the use of research-grade biosimilars as ELISA calibrators is focused on generating reliable, reproducible PK data during biosimilar development and regulatory submissions.
This approach aligns with guidance from regulatory agencies regarding ligand binding assays (LBAs) and method validation for therapeutic protein bioanalysis.
In summary, research-grade Nadunolimab biosimilars are used as the analytical standard in PK bridging ELISAs to establish accurate calibration curves, allowing comparable, robust quantification of both biosimilar and reference product concentrations in serum samples for PK analysis.
The primary in vivo models for administering a research-grade anti-IL-1RAP antibody to study tumor growth inhibition and analyze tumor-infiltrating lymphocytes (TILs) are:
Syngeneic mouse tumor models, notably using a murine-specific anti-IL-1RAP antibody (such as 3A9 in mice).
Human xenograft models in immunodeficient mice, using anti-human IL-1RAP antibodies (including fully humanized or ADCC-enhanced forms).
Humanized mouse models are not explicitly described in the provided data, though such platforms are valuable for detailed TIL characterization with a human immune system.
Essential context and specifics:
Syngeneic models (such as the 4T1 breast cancer model in Balb/c mice) use a surrogate antibody targeting murine IL-1RAP to study effects on both tumor cells and the immune system in an immune-competent setting. This approach enables characterization of the tumor microenvironment and immune cell infiltration, critical for understanding TILs. In these models, anti-IL-1RAP treatment inhibited metastasis more than primary tumor growth and affected myeloid suppressor cell populations, reflecting changes in the TIL landscape.
Human xenograft models involve transplantation of human tumor cells (e.g., pancreatic cancer, NSCLC, leukemias) into immunodeficient mice, followed by administration of anti-human IL-1RAP antibodies (e.g., CAN04/nadunolimab). This setup predominantly evaluates direct anti-tumor effects (like ADCC) and, to a lesser extent due to limited immune components, some aspects of TIL characterization.
TIL characterization: In the syngeneic setting with a functional immune system, blockade of IL-1RAP influences the density and subsets of myeloid-derived suppressor cells and potentially T/NK cells within the tumor. In clinical and preclinical studies using nadunolimab (CAN04) in humans or xenografts, changes in NK cells, CD8+ T cells, and myeloid cell populations have been tied to anti-tumor activity and immune response potential.
Primary referenced models for TIL studies:
4T1 syngeneic breast cancer in Balb/c mice with murine-specific anti-IL-1RAP antibody—best for TIL and microenvironment analysis in vivo.
NSCLC, pancreatic, triple-negative breast cancer human xenografts in immunodeficient mice with human(ized) anti-IL-1RAP—good for tumor cell, some effector cell studies; limited for full TIL complexity.
There is no direct evidence from provided results for classic fully "humanized" immune system mouse models being used in published anti-IL-1RAP antibody preclinical testing for TIL analysis; most immune-competent studies rely on murine syngeneic models with mouse-specific antibodies.
Summary Table: Models for Anti-IL-1RAP Tumor & TIL Studies
Model Type
Species/Cell Line
Antibody Type
Immune Context
TIL Analysis Feasibility
Syngeneic (e.g., 4T1, Balb/c)
Mouse/mouse tumor
Murine anti-IL-1RAP
Fully immune-competent
Optimal
Human xenograft
Human tumor/NSG mouse
Anti-human IL-1RAP
Immunodeficient
Limited
Humanized mouse (not explicit in search)
Human tumor/humanized mouse
Anti-human IL-1RAP
Human immune system
High (but not detailed here)
Key references: , ,
Conclusion: The most robust preclinical TIL and tumor growth inhibition data for anti-IL-1RAP antibodies come from syngeneic mouse models using murine-specific antibodies in immune-competent hosts, with additional mechanistic insight available from human xenografts for tumor-intrinsic effects. Humanized mouse models are likely valuable for future research, but no direct evidence of their use for anti-IL-1RAP/TIL studies appears in these sources.
Researchers studying Nadunolimab biosimilars in combination with other checkpoint inhibitors—like anti-CTLA-4 or anti-LAG-3 biosimilars—aim to elucidate potential synergistic effects on anti-tumor immune responses within complex immune-oncology models. While published clinical data on direct combinations of Nadunolimab biosimilars with anti-CTLA-4 or anti-LAG-3 biosimilars are limited, established methodologies and early-phase studies provide insight into the framework used for such research.
Key Approaches in Studying Synergy:
Model Selection: Researchers use preclinical in vivo models (e.g., murine syngeneic or humanized mouse models with complex tumor microenvironments) and in vitro co-culture systems to assess how dual or triple checkpoint blockade impacts tumor growth, immune cell infiltration, and cytokine profiles.
Mechanistic Rationale:
Nadunolimab targets IL1RAP, blocking both IL-1α and IL-1β signaling and promoting tumor cell killing via enhanced antibody-dependent cell-mediated cytotoxicity (ADCC), mainly through NK cells and macrophages.
Checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3) disrupt negative immune regulation, restoring and enhancing T-cell activation and proliferation.
The central hypothesis for synergy is that Nadunolimab may counteract myeloid-driven immunosuppression in the tumor microenvironment—potentially augmenting T-cell–mediated anti-tumor effects induced by checkpoint inhibitors.
Experimental Design:
Combination therapy arms: Multiple arms are set up, ranging from monotherapies to double and triple combinations (e.g., Nadunolimab biosimilar alone, anti-CTLA-4 biosimilar alone, and the combination).
Endpoints measured:
Tumor growth inhibition and survival
Immune cell profiling (presence and activation state of NK, CD8+ T cells, regulatory T cells, myeloid cells, etc.)
Cytokine and chemokine levels (e.g., monitoring IL-6, IL-8, IFN-γ as pharmacodynamic markers)
Tissue analysis: Quantification of tumor-infiltrating lymphocytes and myeloid-derived suppressor cells in treated tumors.
Readouts and Integration:
Enhanced infiltration and activation of cytotoxic immune cells (e.g., NK, CD8+ T cells) in combination arms versus monotherapy suggests synergy.
Reductions in immunosuppressive cell types (such as M2 macrophages and granulocytic myeloid cells) and systemic markers (like IL-6 or NLR) give mechanistic support for additive or synergistic effects.
Researchers frequently use gene expression and proteomics to further dissect pathway activation and immune cell dynamics following treatment.
Context from Related Combination Studies:
Research with PD-1/PD-L1 and CTLA-4 or LAG-3 inhibitors demonstrates that targeting multiple non-redundant immune checkpoints—in parallel with stromal or myeloid targets like IL1RAP (blocked by Nadunolimab)—can overcome resistance and augment therapeutic responses, especially in models with highly immunosuppressive tumor microenvironments. Although Nadunolimab is most often reported in combination with chemotherapy or anti–PD-1/PD-L1 agents, the same principles and experimental structure would apply to studies using biosimilars of anti-CTLA-4 or anti-LAG-3 antibodies.
Summary Table: Combination Approaches
Component
Mechanism of Action
Model Systems Used
Key Readouts
Nadunolimab
Blocks IL1RAP; ADCC
In vitro, murine xenograft
Tumor volume, immune profiling
Anti-CTLA-4/LAG-3
T-cell checkpoint blockade
Same as above
T cell activation, cytokines
Combination
Multi-faceted (above)
Same as above
Synergy: additive tumor inhibition
Expert Commentary:
The most promising synergy is hypothesized when Nadunolimab relieves myeloid-mediated immune suppression, thus enabling checkpoint inhibitors to maximize T cell–dependent anti-tumor immunity, as shown by increased infiltration and function of effector T cells and decreased immunosuppressive cell signatures.
Limitations: There are presently no widely published studies combining Nadunolimab biosimilars specifically with anti-CTLA-4 or anti-LAG-3 biosimilars; available information is often extrapolated from related checkpoint inhibitor combination research and mechanistic insights from preclinical and translational studies with Nadunolimab and other agents.
A Nadunolimab biosimilar can be used as the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient’s immune response against the therapeutic Nadunolimab by specifically binding to ADAs in patient serum.
Essential context and details:
In a bridging ADA ELISA, the assay leverages the bivalency of ADAs (which have two antigen-binding sites) and the structure of the therapeutic monoclonal antibody (here, Nadunolimab or its biosimilar). In this format:
The Nadunolimab biosimilar is typically immobilized (captured) on the solid phase (e.g., microplate).
Patient serum is then added. If ADAs are present, their two binding sites will attach to two Nadunolimab molecules—one bound on the plate, and the other that is labeled for detection (e.g., with biotin or HRP).
A labeled Nadunolimab biosimilar (distinctly marked, e.g., with dye, biotin, or HRP) is used as the detection reagent. The presence of signal after adding detection substrate indicates the formation of this "bridge" by patient ADAs.
Why use a biosimilar?
A Nadunolimab biosimilar is structurally and functionally very close to the original and is especially important if the reference drug supply is limited, expensive, or one wants to compare responses between biosimilar and reference product, as is performed in biosimilar immunogenicity validation studies.
Studies show consistent immunogenicity profiles between reference therapeutics and their biosimilars, validating this approach for immunogenicity assessment.
Interpretation of results:
The assay detects total ADA reactive to Nadunolimab, whether induced by the original or biosimilar product.
Results can inform clinicians about ADA development in patients, which may affect therapeutic efficacy, drug levels, and risk of adverse events.
Additional points:
Assay sensitivity and specificity depend on the quality and equivalence of the biosimilar used as a reagent, and cross-reactivity or matrix effects should be checked to avoid false positives or negatives.
This technique is widely used for clinical immunogenicity monitoring of monoclonal antibody therapeutics, including Nadunolimab, by research labs and in regulatory submissions.
In summary, Nadunolimab biosimilar serves as both capture and detection reagent in the bridging ADA ELISA to quantitatively monitor anti-Nadunolimab immune responses in patients.
References & Citations
1 Frenay J, Bellaye PS, Oudot A, et al. Int J Mol Sci. 23(23):14918. 2022.
2 Rydberg Millrud C, Deronic A, Grönberg C, et al. Cancer Immunol Immunother. 72(3):667-678. 2023.
3 Ågerstam H, Karlsson C, Hansen N, et al. Proc Natl Acad Sci U S A. 112(34):10786-10791.2015.
4 Askmyr M, Ågerstam H, Hansen N, et al. Blood. 121(18):3709-3713. 2013.
5 Fields JK, Kihn K, Birkedal GS, et al. Front Immunol. 12:779100. 2021.
6 Robbrecht D, Jungels C, Sorensen MM, et al. Br J Cancer. 126(7):1010-1017. 2022.
7 Ågerstam H, Hansen N, von Palffy S, et al. Blood. 128(23):2683-2693. 2016.