Anti-Human CD47 (Magrolimab) [Clone Hu5F9-G4]

Anti-Human CD47 (Magrolimab) [Clone Hu5F9-G4]

Product No.: C1070

- -
- -
Product No.C1070
Clone
Hu5F9-G4
Target
CD47
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
IAP, Neurophilin, gp42
Isotype
Human IgG4κ
Applications
ELISA
,
FA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Immunogen
Humanized antibody derived from mouse clone 5F9
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.
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.
Regulatory Status
Research Use Only (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8°C Wet Ice
Additional Applications Reported In Literature ?
ELISA,
FA
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.
Ligand/Receptor
SIRPα (CD172α) & thrombospondin
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
Immuno-Oncology
.
Immunology

Leinco Antibody Advisor

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.

Role of Research-Grade Magrolimab Biosimilars in PK Bridging ELISA

Magrolimab is an investigational monoclonal antibody against CD47, and its biosimilars—manufactured versions with similar structure and function—are sometimes used in bioanalytical research, including pharmacokinetic (PK) studies. In such studies, accurate measurement of drug concentration in serum or plasma is essential to understand drug exposure and support regulatory submissions.

Use as Calibration Standards and Reference Controls

Calibration Standards:
In a typical PK ELISA, a series of known concentrations of the drug (calibration standards) is used to generate a standard curve. The concentration of the drug in unknown serum samples is then interpolated from this curve. For novel or proprietary drugs like Magrolimab, reference material may be limited, so research-grade biosimilars—carefully characterized for structural and functional similarity—may be used as surrogate calibration standards when the originator molecule is unavailable or in short supply. These biosimilars must demonstrate bioanalytical comparability to the originator in terms of binding characteristics and performance in the assay system.

Reference Controls:
Reference controls (quality controls, QCs) are samples of known concentration used to monitor assay performance over time. Biosimilars can serve as QCs, provided their concentration-response behavior in the assay is equivalent to the originator. This is validated through precision and accuracy studies, ensuring the biosimilar behaves predictably across the assay range.

Bridging Strategy and Bioanalytical Comparability

A bridging ELISA is designed to measure both the originator and the biosimilar in the same assay, using a single calibration standard—ideally the biosimilar itself, once its equivalence to the originator is established. This approach:

  • Reduces variability by avoiding the need for multiple assays or crossover analyses.
  • Supports PK similarity studies by providing a unified data package for regulatory review.
  • Requires rigorous method validation, including statistical analysis to confirm that the biosimilar and originator yield equivalent concentration estimates within pre-defined acceptance criteria (e.g., 90% confidence interval within [0.8, 1.25]).

Practical Considerations

  • Assay Validation: The PK ELISA must be validated for precision, accuracy, sensitivity, and robustness using both the biosimilar and originator as standards and QCs.
  • Sample Preparation: Serum or plasma samples are diluted to fall within the assay’s linear range (e.g., 62.5–4000 ng/mL for Magrolimab ELISA kits).
  • Stability and Handling: Biosimilars used as standards must be stored and handled according to manufacturer instructions to maintain assay reliability.
  • Regulatory Compliance: While research-grade biosimilars can be used in method development and validation, regulatory submissions require documented evidence of bioanalytical comparability between the biosimilar and originator.

Summary Table: Key Steps in Using Biosimilars as Standards in PK Bridging ELISA

StepDescription
Bioanalytical ComparabilityDemonstrate equivalent binding and concentration-response in the assay for biosimilar/originator.
Calibration StandardUse biosimilar as surrogate standard if originator is unavailable; validate performance.
Reference Control (QC)Prepare QCs from both biosimilar and originator; monitor assay performance.
Method ValidationValidate assay with both molecules; statistical analysis for equivalence.
Sample AnalysisMeasure unknown samples; interpolate concentrations from the standard curve.

Conclusion

Research-grade Magrolimab biosimilars can be used as calibration standards and reference controls in PK bridging ELISAs if they are shown to be bioanalytically equivalent to the originator in the assay system. This strategy streamlines bioanalytical workflows, supports PK similarity assessments, and is contingent upon rigorous method validation and statistical comparability testing. Always ensure that such use complies with regulatory expectations for bioanalytical method validation and documentation.

Primary Models for In Vivo Anti-CD47 Antibody Studies

Syngeneic and humanized (xenograft) mouse models are both used to assess the effects of research-grade anti-CD47 antibodies on tumor growth and tumor-infiltrating lymphocytes (TILs), but they serve different purposes and have distinct advantages and limitations.

Syngeneic Models

  • Description: Syngeneic models use mouse tumor cell lines implanted in immunocompetent mice of the same genetic background. These models preserve the host immune system, allowing for the study of tumor-immune interactions, including TIL dynamics, in a context closer to human physiology.
  • Use in Anti-CD47 Research: While the search results primarily focus on humanized (xenograft) models, syngeneic models are also valuable for studying the impact of anti-CD47 on TILs, as they allow for the full spectrum of immune cell responses, including adaptive immunity.
  • Limitations: These models do not directly use human tumors, so findings may not fully translate to human cancers. However, they are critical for mechanistic studies of immune-mediated tumor clearance.

Humanized (Xenograft) Models

  • Description: Humanized models involve engrafting human tumor cells (either cell line-derived, CDX, or patient-derived, PDX) into immunodeficient mice (e.g., NSG), which lack a functional immune system. To study human immune responses, human immune cells can be co-engrafted, creating a “humanized immune system” model.
  • Use in Anti-CD47 Research: Most of the cited studies use human tumor xenografts in immunodeficient mice (CDX and PDX models) to directly assess the effect of anti-CD47 on human tumor growth and, in some cases, on co-engrafted human immune cells. These models are especially relevant for translational research aiming to predict clinical efficacy in humans.
  • TIL Characterization: In standard xenograft models, endogenous mouse TILs are absent or limited due to the immunodeficient host. However, when human immune cells are co-engrafted, researchers can study the infiltration and function of human TILs in the tumor microenvironment.
  • Examples from Literature:
    • Ovarian cancer: Anti-CD47 was tested in both CDX and PDX models of ovarian cancer, showing potent anti-tumor activity and synergy with PARP inhibitors, but the cited study does not detail TIL analysis in these models.
    • Leiomyosarcoma (LMS): Anti-CD47 mAbs were tested in human LMS xenografts in NSG mice (which lack functional T cells), demonstrating inhibition of primary tumor growth and metastasis, but no TIL analysis was performed due to the absence of functional T cells in the host.
    • Multiple myeloma: Anti-CD47 promoted phagocytosis and tumor regression in xenotransplantation models, but, again, the models used lacked a functional immune system, so TIL dynamics were not characterized.
  • Limitations: Standard xenograft models do not allow for study of the complete tumor-immune microenvironment, including TILs, unless human immune cells are co-engrafted.

Comparison Table

Model TypeTumor SourceImmune SystemTIL Analysis Possible?Key Use Case
SyngeneicMouse tumor cell lineMouse (intact)YesMechanistic immune studies
Xenograft (CDX/PDX)Human tumorMouse (immunodeficient)No (unless humanized)Translational efficacy, human tumor biology
Humanized (with immune cells)Human tumor + human immune cellsHuman (engrafted)YesHuman TIL biology, immunotherapy prediction

Conclusions

  • Most preclinical studies of anti-CD47 antibodies in vivo use human tumor xenograft (CDX/PDX) models in immunodeficient mice to assess tumor growth inhibition, but these models typically lack functional TILs unless human immune cells are co-engrafted.
  • Syngeneic models are essential for studying the full spectrum of immune responses, including TIL dynamics, but are less commonly reported in the anti-CD47 literature referenced here.
  • For comprehensive TIL characterization, especially of human lymphocytes, humanized mouse models with co-engrafted human immune cells are required, though these are more complex and less commonly employed in the cited studies.
  • Current published data emphasize the anti-tumor efficacy of anti-CD47 in xenograft models, with limited direct evidence on TIL modulation in these settings.

In summary, while xenograft models dominate the literature for anti-CD47 efficacy, syngeneic and fully humanized immune-engrafted models are critical for mechanistic and translational studies of TIL responses.

Researchers investigate the use of Magrolimab (a CD47 biosimilar) in combination with other checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) to assess synergistic immune effects in complex oncology models by combining agents targeting distinct immune regulatory pathways and studying their collective impact on tumor control in both preclinical and clinical contexts.

Key Approaches and Supporting Rationale:

  • Mechanism-Based Combinations:

    • Magrolimab blocks the CD47 “don’t eat me” signal, promoting macrophage-mediated phagocytosis of cancer cells.
    • Anti-CTLA-4 and anti-LAG-3 antibodies act mainly on T cells by relieving immune suppression, thus enhancing cytotoxic T cell responses.
    • Combining these agents allows researchers to test if simultaneously promoting innate (macrophage-mediated) and adaptive (T cell-mediated) immunity results in greater antitumor activity than monotherapies.
  • Preclinical Models:

    • Researchers use complex syngeneic mouse tumor models or humanized immune-oncology mouse models to explore the interactions between magrolimab and other checkpoint inhibitors.
    • They monitor for enhanced tumor regression, immune cell infiltration, and changes in the tumor microenvironment when these agents are administered together versus singly.
  • Clinical Synergy:

    • Combination regimens such as nivolumab (PD-1) plus relatlimab (LAG-3) have proven more effective than single agents in some cancer types, illustrating rationale for multi-checkpoint blockade.
    • Translational research bridges preclinical findings to early-phase clinical trials, where safety, tolerability, and response rates are rigorously measured when combining magrolimab biosimilars with other checkpoint inhibitors.
  • Endpoints and Readouts:

    • Endpoints typically include objective response rate, immune cell activation profiles, and detailed safety/tolerability metrics.
    • Molecular and histopathology analyses help determine whether synergistic combinations alter immune cell composition and function within tumors compared to controls.

Notable Insights:

  • Preclinical synergy has been observed when combining multiple checkpoint inhibitors; for example, combining anti–CTLA-4 and anti–PD-1 leads to enhanced T cell activation and tumor regression in mouse models.
  • Multi-arm phase II studies assess magrolimab with other therapies, often including safety run-in phases to define tolerable dosing before expansion cohorts.
  • The overall goal is to overcome the limitations of single-agent therapies by addressing several immune evasion mechanisms simultaneously, potentially leading to durable responses in tumors unresponsive to individual agents.

Current research is heavily focused on translating these findings into clinical settings, with phase II multi-arm studies evaluating magrolimab combinations in specific cancers, sometimes in populations refractory to prior checkpoint inhibitor therapies.

No direct references to clinical trials combining magrolimab with CTLA-4 or LAG-3 biosimilars are provided in these results, but the combination strategies are based on robust preclinical evidence and are being adopted for human studies in related immune-oncology drug development.

A Magrolimab biosimilar can be used as either the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA assay to monitor a patient’s immune response (specifically ADA formation) against Magrolimab treatment.

Context and Mechanism:

  • In a bridging ADA ELISA, the biotherapeutic drug (in this case, Magrolimab or its biosimilar) is used in two forms: one immobilized on the assay plate (capture reagent) and one labeled (e.g., with HRP, biotin, or other tags) as the detection reagent.
  • The patient’s serum is added to the plate, and if the patient has developed ADAs against Magrolimab, these antibodies will bridge the immobilized and labeled (detector) versions of Magrolimab, because ADAs can bind to at least two identical or overlapping epitopes on the drug.
  • The resulting complex (capture-drug : ADA : detection-drug) forms a measurable signal when the detection reagent is developed with a suitable substrate.
  • The use of a Magrolimab biosimilar instead of the clinical drug itself provides a research-grade, cost-effective, and non-clinical reagent. Biosimilars are designed to be structurally and functionally highly similar to the reference drug, ensuring the assay reflects patient response to the therapeutic Magrolimab.

Practical Use in ADA Monitoring:

  • Capture reagent: The Magrolimab biosimilar is coated onto the ELISA plate. Patient serum is then added, and any ADAs present will bind to Magrolimab.
  • Detection reagent: The Magrolimab biosimilar, labeled with a detection molecule (HRP, biotin, etc.), is added. It will bind the other “arm” of any present ADA, creating a bridge.
  • The signal measured correlates with the amount of ADA present in the patient sample.

Summary Table: Bridging ADA ELISA with a Magrolimab Biosimilar

StepReagentPurpose
Plate coatingMagrolimab biosimilar (capture)Binds ADA from patient serum
Sample incubationPatient serumPotential source of anti-Magrolimab ADA
Detector incubationLabeled Magrolimab biosimilar (detection)Binds to ADA, produces bridge and signal
Signal detectionSubstrate for labelQuantifies ADA based on signal intensity

Important Notes:

  • The biosimilar is used only for research and preclinical ADA assay development; clinical immunogenicity testing must comply with regulatory standards.
  • The core assumption is that the biosimilar and the reference Magrolimab are similar enough in their immunogenic epitopes to cross-react with ADAs produced in patients treated with the therapeutic.

Summary: The use of a Magrolimab biosimilar as a capture or detection reagent in a bridging ADA ELISA enables quantification of immune responses against Magrolimab by facilitating the specific detection of anti-Magrolimab antibodies formed in response to therapy.

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.
Indirect Elisa Protocol
FA

Certificate of Analysis

Formats Available

- -
- -
Disclaimer AlertProducts are for research use only. Not for use in diagnostic or therapeutic procedures.