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.
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 Magrolimab. This product is for research use only. Magrolimab activity is directed against CD47.
Background
In healthy cells, signal molecules stimulate programmed cell removal via various proteins, phospholipids, and abnormal glycosylation1. However, cancer cells are able to evade phagocytic elimination, the normal method of cell removal by the innate immune system1, due to the inhibitory antiphagocytic “don’t eat me” signal generated by CD472. The CD47 signal, which is overexpressed on cancer cells3, enables immune evasion from macrophages and other phagocytes2. Since CD47 overexpression has been found on all known solid tumors and leukemias, it is a universal blocking target for cancer immunotherapy1.
Magrolimab was generated by immunizing Balb/c mice with a recombinant human-mouse CD47/mFC fusion protein composed of a cDNA fragment of human CD47 encoding the extracellular domain fused to mouse Fc1. Hybridomas were created by fusing spleen cells with SP2/0 cells, and screening resulted in clone 5F9. Humanization of mouse anti-CD47 5F9 was performed by CDR grafting onto a human IgG4 scaffold to minimize recruitment of antibody Fc-dependent effector functions. Magrolimab does not induce antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, or apoptosis. Additionally, residue optimization was performed and the human IgG4 heavy chain constant region was modified by a Ser228Pro substitution to reduce the rate of Fab arm exchange, which can occur in human IgG4 molecules.
Magrolimab blocks the interaction between CD47 and one of its ligands, signal regulatory protein alpha (SIRPα)1. As a result, magrolimab is able to induce potent macrophage-mediated phagocytosis of primary human acute myeloid leukemia cells1, HER2+ breast cancer cells4, and lymphoma cells5, either on its own or in combination with other antibodies.
Antigen Distribution
CD47 is a cell-surface protein with ubiquitous expression that is also overexpressed on cancer cells.
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 Magrolimab biosimilars are used as calibration standards and reference controls in pharmacokinetic (PK) bridging ELISA to quantitate drug concentration in serum by serving as the single analytical standard against which all serum samples—regardless of whether they contain biosimilar or reference Magrolimab—are measured.
Essential context and methodology:
Calibration Standard Use: The biosimilar form of Magrolimab is serially diluted and used to generate a standard curve within each ELISA plate, covering the expected range of drug concentrations in serum (for example, 62.5 ng/mL to 4000 ng/mL in a research ELISA). All unknown serum samples are then interpolated against this curve to determine drug concentration.
Bridging ELISA Principle: In a PK bridging ELISA, the assay uses the same detection reagents to measure both the biosimilar and the originator (reference) Magrolimab, ensuring comparability. The biosimilar standard is validated to behave identically to the reference product in the assay, supporting regulatory and scientific requirements for demonstrating similarity in PK measurements.
Analytical Standard Justification: Using a single, validated standard (often the biosimilar itself) as the calibrator is considered best practice because it minimizes inter-assay variability, simplifies assay logistics, and eliminates the need for method bridging or additional comparative analyses. Before using the biosimilar as the calibration standard, bioanalytical comparability with the reference product is thoroughly demonstrated, including parallelism, accuracy, and precision across several assays.
Reference Control Use: Alongside the calibration curve, sets of quality control (QC) samples prepared using both biosimilar and reference material are run to validate assay performance and confirm equivalence in detection, further supporting the interchangeability of the analytical standard.
Key requirements and practical notes:
Validation: The PK ELISA must be fully validated according to regulatory guidelines (e.g., FDA) for sensitivity, specificity, accuracy, and precision across a clinically relevant concentration range.
Format: In commercial research kits, the standard is usually supplied lyophilized for reconstitution. Typical assay formats are competitive or sandwich ELISA, often using colorimetric or chemiluminescent detection.
Matrix Consideration: Standards and QC samples are prepared in serum to mirror the sample matrix, reducing matrix effects on assay accuracy.
Summary table: Use of research-grade Magrolimab biosimilars as calibration standards and controls
Function
How Magrolimab Biosimilar Is Used
Calibration Standard
Serial dilutions provide standard curve for quantifying drug in serum samples
Reference Control
QC samples contain both biosimilar and reference; both measured to confirm comparability
PK Bridging
Single biosimilar standard used to quantify both biosimilar and reference drug
Validation Requirement
Demonstrated analytical equivalence with reference product before use as sole standard
This approach provides reliable, reproducible measurements essential for bioequivalence and PK bridging studies, which are foundational for regulatory submissions for biosimilars.
Syngeneic and Humanized Models for In Vivo Anti-CD47 Antibody Studies
Anti-CD47 antibodies have been investigated in several preclinical models to assess their impact on tumor growth inhibition and the tumor immune microenvironment, particularly the roles of tumor-infiltrating lymphocytes (TILs) and macrophages.
Key Animal Models
Model Type
Tumor Type
Administration Route
Immune Context
TIL Characterization
References
Humanized Xenograft (NSG)
Human leiomyosarcoma (LMS04, LMS05)
Intraperitoneal
Lacks B, T, NK cells; retains macrophages
No data on TILs (model lacks functional T cells)
Humanized Xenograft
Human multiple myeloma (RPMI 8226)
Not specified
Immunodeficient (likely NSG/SCID)
Not assessed (model lacks adaptive immunity)
Details of the Primary Models
Humanized Xenograft Models (Immunodeficient Mice)
Tumor Growth Inhibition: In these models, human tumor cell lines (e.g., LMS04, LMS05 leiomyosarcoma, RPMI 8226 myeloma) are engrafted into immunodeficient mice such as NOD/SCID/IL-2Rγ null (NSG) mice, which lack adaptive immune cells (B, T, NK cells) but possess functional macrophages. Anti-CD47 monoclonal antibodies (mAbs) are administered systemically (e.g., intraperitoneally), leading to significant inhibition of primary tumor growth and, in metastatic models, reduction in the size and number of metastases.
Mechanism: The primary mechanism in these models is macrophage-mediated phagocytosis of tumor cells, enabled by blocking the CD47–SIRPα "don't eat me" signal. This results in increased tumor cell clearance by macrophages.
TILs: Because NSG mice lack functional T and B cells, these models cannot be used to study the effects of anti-CD47 therapy on tumor-infiltrating lymphocytes (TILs) or adaptive immune responses. The focus is exclusively on innate immunity (macrophages).
Relevance: These models are valuable for demonstrating proof-of-concept that CD47 blockade can engage macrophages to phagocytose and eliminate human tumor cells in vivo, but they do not recapitulate the full complexity of the human tumor immune microenvironment, especially regarding TILs.
Syngeneic (Immunocompetent) Models
Current Evidence: The provided search results do not describe syngeneic (mouse tumor in immunocompetent mouse) models where anti-CD47 antibodies are used to study tumor growth inhibition and TIL characterization. Such models would allow assessment of how CD47 blockade affects not only macrophages but also T cells, B cells, and other immune populations within the tumor.
Literature Gap: To fully characterize the impact of anti-CD47 therapy on TILs—including changes in T cell activation, exhaustion, and subset composition—syngeneic models are required. However, these are not highlighted in the provided sources.
Summary
Humanized xenograft models (e.g., NSG mice with human tumor cell lines) are the primary in vivo systems where research-grade anti-CD47 antibodies have been administered to study tumor growth inhibition, with a focus on macrophage-mediated phagocytosis.
TIL characterization is not possible in these models due to the lack of functional adaptive immunity; thus, the effects on lymphocytes within the tumor cannot be assessed.
Syngeneic models (immunocompetent mice with mouse tumors) are needed to study the full spectrum of anti-CD47 effects on the tumor immune microenvironment, including TILs, but are not described in the provided results.
Clinical Implications: While humanized xenograft models demonstrate therapeutic potential, syngeneic models are essential for understanding how CD47 blockade may modulate adaptive immune responses and TIL dynamics in tumors.
Researchers combine Magrolimab biosimilars with other checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) to study synergistic effects in complex immune-oncology models by leveraging the distinct mechanisms of each inhibitor, validating combinations in preclinical and early-phase clinical studies, and assessing immune responses and tumor control.
Magrolimab is a monoclonal antibody that blocks CD47, a "don't eat me" signal on tumor cells, thereby enabling macrophage-mediated phagocytosis of cancer cells. Unlike traditional checkpoint inhibitors (which typically target T cell regulation), Magrolimab targets the interaction between tumor cells and macrophages, offering an orthogonal pathway to modulate anti-tumor immunity.
Rationale for Combinations:
Checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 antibodies work primarily by modulating T cell activation and exhaustion, enhancing cytotoxic lymphocyte function.
Magrolimab acts by enhancing innate immune clearance via macrophages, which can complement the adaptive immune stimulation by other checkpoint inhibitors.
Preclinical findings show that combining therapies targeting different immune pathways (macrophage activation by Magrolimab, T-cell activation/exhaustion modulation by anti-CTLA-4 or anti-LAG-3) can result in increased tumor regression compared to monotherapy, as each approach addresses distinct mechanisms of immune evasion.
Experimental and Translational Strategies:
Researchers typically use advanced preclinical models (e.g., humanized mouse models, syngeneic tumor models) to evaluate the combinatorial effect of Magrolimab plus various checkpoint inhibitors on tumor growth, immune cell infiltration, and cytokine profiles.
Mechanistic studies often involve flow cytometry, gene profiling, and immunohistochemistry to assess:
Enhanced macrophage phagocytosis
T-cell infiltration and activation
Changes in the tumor microenvironment
Clinical studies are then designed to further evaluate efficacy and safety (often using dose-escalation and expansion phases) by measuring objective response rates, progression-free survival, and adverse event profiles.
Examples and Progress:
Opdualag (nivolumab plus relatlimab, a PD-1 and LAG-3 combination) has shown that multi-checkpoint inhibition improves anti-tumor immunity and provides a clinical proof of principle for such combinations.
While published data on Magrolimab combined specifically with other checkpoint inhibitor biosimilars (like anti-CTLA-4 or anti-LAG-3) is limited, the mechanistic rationale and early clinical reports support exploration of these combinations in immune-oncology, with ongoing research focusing on identifying which tumor types and immune contexts benefit most from synergistic effects.
In summary, researchers use Magrolimab biosimilars with other checkpoint inhibitors to exploit complementary mechanisms—macrophage engagement and T cell reactivation—to maximize anti-tumor immune responses. These strategies are validated through a combination of sophisticated preclinical models and translational studies, paving the way for clinical trials targeting cancers with high unmet needs.
A Magrolimab biosimilar is used as either the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA by mimicking the structure and binding properties of the therapeutic Magrolimab to enable detection of patient antibodies that form against the drug.
In a typical bridging ADA ELISA:
Magrolimab biosimilar, which closely resembles the original therapeutic in structure and antigenicity, is immobilized onto a microtiter plate as the capture agent.
Patient serum is added; if anti-Magrolimab antibodies (ADAs) are present in the serum, they will bind to the immobilized biosimilar.
A second, labeled Magrolimab biosimilar (e.g., biotinylated or HRP-conjugated) is then added as the detection reagent. This labeled biosimilar will bind to a different epitope of any ADA that has already bound to the plate, forming a "bridge".
The resulting complex is detected via a signal (such as colorimetric change using HRP substrate), which proportionally reflects ADA levels.
This approach is used because:
ADAs are typically bivalent (can bind two antigens), enabling them to bridge between capture and detection molecules that are the same or similar (i.e., Magrolimab biosimilar).
Using a biosimilar in place of the reference Magrolimab enables cost-effective, preclinical, and high-throughput ADA monitoring while maintaining immunological specificity.
Key points:
Magrolimab biosimilars are research-only tools and are used outside clinical diagnostics but are ideal for assay development and preclinical immunogenicity studies.
This bridging format is widely accepted for ADA detection against monoclonal antibodies and biosimilars, as it detects a wide range of ADA isotypes and avoids the need for species-specific secondary antibodies.
Thus, in monitoring immune response, Magrolimab biosimilars serve as functional stand-ins for the clinical drug, enabling detection and quantification of ADAs that may neutralize drug efficacy or impact safety, via the bridging ELISA format.
References & Citations
1. Liu J, Wang L, Zhao F, et al. PLoS One. 10(9):e0137345. 2015.
2. Advani R, Flinn I, Popplewell L, et al. N Engl J Med. 379(18):1711-1721. 2018.
3. Maute R, Xu J, Weissman IL. Immunooncol Technol. 13:100070. 2022.
4. Upton R, Banuelos A, Feng D, et al. Proc Natl Acad Sci U S A. 118(29):e2026849118. 2021.
5. Zeller T, Lutz S, Münnich IA, et al. Front Immunol. 13:929339. 2022.
6. Sikic BI, Lakhani N, Patnaik A, et al. J Clin Oncol. 37(12):946-953. 2019.