Anti-Human C5 (Eculizumab) [Clone 5G1.1] — Fc Muted™

Anti-Human C5 (Eculizumab) [Clone 5G1.1] — Fc Muted™

Product No.: C3045

- -
- -
Product No.C3045
Clone
5G1.1
Target
C5
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Complement protein C5, C5D, C5a, C5b, ECLZB, CPAMD4
Isotype
Human IgG4κ
Applications
ELISA
,
FA
,
IF
,
IP
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Immunogen
Human C5
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
≤ 1.0 EU/mg as determined by the LAL method
Purity
≥95% monomer by analytical SEC
>95% by SDS Page
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.
Regulatory Status
Research Use Only
Country of Origin
USA
Shipping
2 – 8° C Wet Ice
Additional Applications Reported In Literature ?
ELISA,
FA,
IF,
IP,
WB
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 Eculizumab. This product is research use only.Clone 5G1.1 binds with high affinity and specificity to the complement protein C5.
Background
Complement component 5 (C5) is a crucial protein in the complement system, part of the body’s immune response. It is synthesized in the liver and circulates in the blood as an inactive precursor. When triggered, C5 splits into C5a and C5b. C5a acts as an inflammation promoter and attracts immune cells to infection sites. C5b kickstarts the creation of the membrane attack complex (MAC), which creates pores in the membranes of pathogens, leading to their destruction. Irregularities in C5 activation are linked to illnesses such as paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS)1,2.

The clone 5G1.1, also known as Eculizumab, is a humanized monoclonal antibody designed to target the complement protein C5. Binding to C5 prevents its cleavage into C5a and C5b, thereby blocking the formation of the membrane attack complex (MAC). This inhibition is essential for reducing the inflammatory and cell-lysing effects that come with complement activation. Eculizumab has received approval for treating paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS), both of which are rare, life-threatening conditions resulting from uncontrolled complement activation. Its effectiveness in these disorders has established it as a pioneering treatment in immunology3-9.
Antigen Distribution
The complement component C5 is primarily found in the plasma and extracellular matrix. It is synthesized in the liver and then secreted into the bloodstream.
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Complement Pathway
.
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.

Research-grade Eculizumab biosimilars, such as SB12, are used as calibration standards and reference controls in pharmacokinetic (PK) bridging ELISA assays to measure drug concentration in serum because they are analytically and functionally highly similar to the originator/reference Eculizumab, enabling accurate quantification without clinically meaningful differences in measurement outcomes.

Essential context and details:

  • Calibration Standards: In a PK-bridging ELISA, biosimilar material (e.g., SB12) is used to generate the calibration standard curve because its structural, physicochemical, and functional properties are nearly identical to the reference product (US/EU Soliris), thus its detection characteristics in ELISA are essentially equivalent.
  • Validated Assay: A validated assay (such as an electrochemiluminescent ELISA or similar ligand-binding assay) can use the biosimilar as the "standard" for constructing the standard curve. Serum samples containing unknown concentrations of Eculizumab are interpolated on this curve for quantification.
  • Reference Controls: The same biosimilar batch may also be used as a reference control to confirm assay performance (accuracy and precision during each run), ensuring comparability across experiments and batches.
  • Assay Compatibility: Because the biosimilar and originator have highly similar binding affinities to target antigens (C5) and consistent immunological properties, there is no clinically meaningful difference in how the ELISA will detect them.

Example Application:

  • In a published PK study, SB12 was used to prepare calibration standards and quality control (QC) samples for measuring serum Eculizumab concentrations in human samples. All serum samples (both SB12-treated and Soliris-treated) were subjected to the same acid dissociation and neutralization steps to ensure target-drug complexes were dissociated and available for quantification.
  • The detection involved incubation with excess biotinylated C5, capturing on streptavidin-coated plates, and use of an anti-human IgG4 detection antibody, suitable for both reference and biosimilar forms.
  • The assay was validated for range (lower and upper limit of quantification), accuracy, and precision, demonstrating suitability for both biosimilar and reference product quantification.

Additional notes:

  • Reference standards may be chosen from either the biosimilar or the originator reference product, provided analytical equivalence has been robustly demonstrated through comparative studies.
  • The described approach enables PK bridging (comparing the PK profiles of biosimilar and reference product) and facilitates regulatory comparability exercises.

Summary Table: Calibration Standard Role of Research-Grade Eculizumab Biosimilars in PK Bridging ELISA

PurposeHow Biosimilar Is UsedRationale
Calibration curveSerial dilutions as stdAllows accurate quantification; high similarity
Reference control/QCControl sampleVerifies assay performance equivalence
PK comparability assessmentCommon standardStandardizes across biosimilar/reference arms

References:
: Demonstrates the analytical and functional similarity of SB12 to US/EU Eculizumab, supporting its use as a standard.
: Describes the explicit use of SB12 as calibration and QC material in serum PK assay for both biosimilar and reference arms.
: Protocols detail the workflow for performing Eculizumab ELISA with serum samples.

The primary models used to evaluate in vivo anti-tumor efficacy and to characterize tumor-infiltrating lymphocytes (TILs) following administration of a research-grade anti-C5 antibody are murine syngeneic models. These models involve implanting mouse tumor cell lines into immunocompetent mice of the same genetic background, thereby preserving a functional mouse immune system that is necessary for assessing both tumor growth inhibition and immune cell infiltration, including TIL analysis.

Supporting details:

  • Syngeneic models (such as TC-1, RENCA, CT26, B16F10, and EMT6 in various mouse strains) are widely used to study novel immunotherapies, including complement pathway inhibitors like anti-C5 antibodies. These models enable measurement of shifts in immune populations—specifically TILs (such as CD8^+ T cells)—and allow for mechanistic studies on how complement blockade influences the immune microenvironment.

  • Blocking C5a signaling or C5 function (using antagonists or blocking antibodies like BB5.1) in syngeneic models has been shown to:

    • Inhibit tumor growth in vivo, sometimes synergistically with immune checkpoint blockade.
    • Increase CD8^+ T cell infiltration in tumors, as assessed by immunofluorescence or flow cytometry of tumor-infiltrating cells.
  • Humanized mouse models (mice engrafted with a human immune system or patient-derived xenografts) are not typically the primary models for anti-C5 antibodies when the focus is on immune cell infiltration and TILs, because most anti-C5 antibodies used for preclinical research (such as BB5.1) are specific to mouse C5 and do not cross-react with human C5. Humanized settings may be used for translational studies with cross-reactive or human-specific reagents, but published in vivo mechanistic dissection of TIL changes following anti-C5 therapy remains primarily in syngeneic models.

Key murine syngeneic tumor models used for these purposes:

  • TC-1 (lung epithelial cells transformed with HPV oncogenes; C57BL/6 mice)
  • RENCA (renal cell carcinoma; BALB/c mice)
  • CT26 (colorectal carcinoma; BALB/c mice)
  • B16F10 (melanoma; C57BL/6 mice)
  • EMT6 (mammary carcinoma; BALB/c mice)

Summary table:

Model TypeImmune SystemAnti-C5 Antibody UseTIL CharacterizationTypical Tumor Lines
Syngeneic (mouse)Mouse (intact)Yes (mouse C5)YesTC-1, RENCA, CT26, B16F10, EMT6
Humanized (mouse)Human (reconstituted)Rare (needs cross-reactivity)Limited (depends on antibody/host)Not primary for C5 studies

Conclusion:
Murine syngeneic models are the standard for in vivo testing of anti-C5 antibodies to study both tumor growth inhibition and the resulting composition of TILs, due to antibody specificity and the requirement for a fully functional immune system. Humanized models are rarely used for mouse-specific anti-C5 research targeting tumor-immune interactions unless the antibody or system is specifically engineered for cross-species compatibility.

Researchers studying the synergistic effects of eculizumab biosimilars with other checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) in complex immune-oncology models typically design experiments that combine these agents to observe whether their distinct mechanisms of immune modulation can enhance antitumor responses beyond what is seen with monotherapies.

Eculizumab biosimilars (e.g., SB12, ABP 959, Elizaria) are meticulously developed to ensure functional and structural equivalence to reference eculizumab; their primary mechanism is the inhibition of the terminal complement pathway via blocking C5, thereby reducing complement-mediated inflammation and cytotoxicity. While eculizumab and its biosimilars are primarily indicated for diseases like paroxysmal nocturnal hemoglobinuria and other complement-mediated disorders, the rationale for combining them with checkpoint inhibitors in oncology arises from the interplay of the complement system and tumor immunity.

Checkpoint inhibitors such as anti-CTLA-4, anti-PD-1, and anti-LAG-3 antibodies act by blocking key inhibitory pathways that suppress T cell function, thereby enhancing the immune system’s ability to attack cancer cells. Each checkpoint inhibitor targets a different phase or compartment of the immune response:

  • Anti-CTLA-4: Acts mainly in lymph nodes, boosting the induction and proliferation of T cells.
  • Anti-PD-1/PD-L1: Functions mainly at tumor sites, preventing T cell exhaustion and tumor-mediated immunosuppression.
  • Anti-LAG-3: Modulates a distinct T cell regulatory pathway and may particularly enhance CD4+ T helper and CD8+ cytotoxic responses.

Combination Strategies in Preclinical Models

Researchers use mouse tumor models or advanced humanized in vitro systems to explore:

  • Whether blocking the complement pathway (with eculizumab biosimilars) reduces immunosuppressive tumor microenvironment (TME) components, such as myeloid-derived suppressor cells or regulatory T cells, which may otherwise blunt the efficacy of checkpoint inhibitors.
  • The effects of simultaneous immune checkpoint blockade (e.g., anti-CTLA-4 or anti-LAG-3) on T cell activation, proliferation, and infiltration in tumors, measuring tumor growth, survival, and immune cell composition.
  • The potential to overcome resistance to single-agent checkpoint inhibitors by modifying multiple suppressive pathways in the TME.

Mechanistic Insights

  • Anti-PD-1/LAG-3 combinations: Depend mainly on the presence and activation of CD4+ T cells, with effects such as reduced regulatory T cell (Treg) activity and enhanced CD8+ cytotoxic T cell function.
  • Anti-PD-1/CTLA-4 combinations: More directly boost cytotoxic CD8+ T cells, sometimes independently of CD4+ T cell help.

The idea of synergy is based on the hypothesis that:

  • Inhibiting complement with eculizumab biosimilars could reduce certain suppressive signals in the TME, “unmasking” the tumor to immune attack.
  • Concurrent checkpoint inhibition could then enable a broader and stronger T cell–mediated antitumor response.

Clinical and Translational Implications

While data on direct use of eculizumab biosimilars with checkpoint inhibitors in oncology is limited, such combination approaches are an active area of early-phase investigation in immune-oncology. The design is often based on comprehensive preclinical studies establishing pharmacokinetic equivalence and safety for biosimilars, and the mechanistic rationale is extrapolated from the distinct but potentially complementary actions of complement inhibition and T cell checkpoint blockade.

In summary, researchers hypothesize that targeting both the complement system (with eculizumab biosimilars) and immune checkpoints (with anti-CTLA-4, anti-LAG-3, etc.) could yield greater antitumor effects by overcoming resistant mechanisms in the tumor microenvironment, and they test these hypotheses using immunologically complex preclinical models.

In a bridging ADA ELISA designed to monitor patient immune responses to eculizumab, a biosimilar version can be used as either the capture or detection reagent due to its close structural and functional similarity to the reference drug—meaning both will bind ADAs generated against eculizumab equally well.

Essential context and supporting details:

  • Assay format: The bridging ADA ELISA relies on the bivalent nature of ADAs: one arm of the ADA binds to capture eculizumab (often immobilized or biotinylated and attached to streptavidin-coated plates), while the other binds to the detection eculizumab (often labeled with HRP or a fluorophore).
  • Using a biosimilar as reagent: Because a biosimilar has identical amino acid sequence and similar post-translational modifications to the reference product, it can replace the originator eculizumab for assay construction:
    • As capture reagent: Biotinylated eculizumab biosimilar is immobilized on the ELISA plate. Serum ADAs (if present) bind this molecule.
    • As detection reagent: After ADAs are captured, a labeled eculizumab biosimilar is added to bind the other ADA arm, forming a “bridge”. Signal is generated from the label upon substrate addition.
  • Advantages: The biosimilar’s analytical equivalence—proven by identical peptide maps, sequence, and molecular weight—assures that ADAs against either the biosimilar or original eculizumab will be detected, allowing monitoring for immunogenicity regardless of which drug was administered.

Additional relevant information:

  • Validation: Regulatory guidelines require head-to-head immunogenicity testing between biosimilars and reference drugs in clinical studies, and results confirm that biosimilars are suitable for use as reagents in ADA assays.
  • Interference and specificity: High-quality, well-characterized biosimilars must be used to minimize assay interference from endogenous matrix components, soluble targets, or drug remnants in serum samples.
  • Flexibility: Either the biosimilar or reference drug can be used in the bridging ELISA reagent roles, provided their structural identity has been demonstrated. This approach is supported by regulatory and scientific consensus for biosimilar immunogenicity assessment.

In summary, an eculizumab biosimilar can confidently be used as the capture or detection reagent in bridging ADA ELISA, enabling sensitive monitoring of a patient's immune response to eculizumab-based therapies.

References & Citations

1. Fredslund F, Laursen NS, Roversi P, et al. Nat Immunol. 2008;9(7):753-760.
2. C5 Variant rs10985126 is Associated with Mortality in Patients with Symptomatic Coronary Artery Disease - PubMed. Accessed August 11, 2024. https://pubmed.ncbi.nlm.nih.gov/34321906/
3. Wong EKS, Kavanagh D. Transl Res. 2015;165(2):306-320.
4. Sarno L, Tufano A, Maruotti GM, Martinelli P, Balletta MM, Russo D. J Nephrol. 2019;32(1):17-25.
5. Merrill SA, Brodsky RA.Hematology Am Soc Hematol Educ Program. 2018;2018(1):371-376.
6. McKeage K. Drugs. 2011;71(17):2327-2345.
7. Keating GM. Drugs. 2013;73(18):2053-2066.
8. Diamante Chiodini B, Davin JC, Corazza F, et al. Pediatrics. 2014;133(6):e1764-1768.
9. Wong E, Challis R, Sheerin N, Johnson S, Kavanagh D, Goodship THJ. Immunobiology.2016;221(6):715-718.
10. Eculizumab Monoclonal Antibody (4F6), GenScript - Antibodies, Primary Antibodies. Accessed August 8, 2024. https://www.fishersci.com/shop/products/eculizumab-monoclonal-antibody-4f6-genscript/502530049
11. Mouse Anti-C5 Recombinant Antibody (clone m5G1.1) - Creative Biolabs. Accessed August 8, 2024. https://www.creativebiolabs.net/Anti-C5-Recombinant-Antibody-clone-m5G1-1-75875.htm
12. ECULI - Overview: Eculizumab, Serum. Accessed August 11, 2024. https://www.mayocliniclabs.com/test-catalog/Overview/65676
Indirect Elisa Protocol
FA
IF
Immunoprecipitation Protocol
General Western Blot Protocol

Certificate of Analysis

Formats Available

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