Anti-Human C5 (Eculizumab) [Clone 5G1.1]

Anti-Human C5 (Eculizumab) [Clone 5G1.1]

Product No.: C3040

- -
- -
Product No.C3040
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
Active
Immunogen
Human C5
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.
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 are commonly used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays due to their demonstrated high structural and functional similarity to the eculizumab reference product. Their biosimilar status allows accurate quantification of drug concentrations in serum samples with validated analytical performance.

Essential context and supporting details:

  • Structural and Functional Equivalence: Eculizumab biosimilars (e.g., SB12) are subject to rigorous analytical and functional testing to ensure high similarity to reference eculizumab in terms of structure, purity, charge heterogeneity, potency, and FcRn binding affinity. Minor differences (such as in N-glycan content) are well-characterized and shown not to impact pharmacokinetics or efficacy.

  • Use as Calibration Standards: In PK bridging ELISAs, biosimilar eculizumab (such as SB12) can be used as the standard to generate calibration curves for quantifying eculizumab concentration in clinical serum samples. This is possible because the biosimilar exhibits comparable binding and detection characteristics to the reference product on analytical platforms.

  • Standard Curve Preparation: The biosimilar is serially diluted in assay buffer or negative human serum to produce standards of known concentration. These standards are run in parallel with serum samples from subjects, allowing drug quantitation based on the resulting dose-response (standard) curve.

  • Assay Validation and Reference Control: Assay parameters such as precision, accuracy, and linearity are validated using these biosimilar-derived standards and quality control (QC) samples. Research-grade biosimilars ensure lot-to-lot consistency and are traceable to characterized reference material, supporting their suitability as controls.

  • Detection Method Example: For eculizumab PK ELISA, a common setup involves acid dissociation to release drug-C5 complexes, then the detection of free drug using a capture reagent (biotinylated C5) and a labeled anti-human IgG4 antibody. The biosimilar is used for both calibration standards and QC material, since it behaves identically within the validated PK assay’s working range.

  • Equivalence Demonstrated in Clinical Populations: PK evaluations, such as cross-over or parallel group studies, use these biosimilars interchangeably with the reference drug to measure serum concentrations and validate assay performance across clinical samples.

Summary Table: Use of Eculizumab Biosimilar in PK Bridging ELISA

Role in ELISADescriptionJustification
Calibration StandardSerial dilutions to create standard curveHigh analytical similarity to reference
Reference ControlValidates precision, accuracy, and linearityConsistent QC and traceability
Analytical EquivalenceComparable epitope/antibody binding in assay matricesConfirmed by head-to-head validation

Key Point: The use of a research-grade eculizumab biosimilar as a standard or control is robustly supported in PK bridging ELISAs because of its validated structural, physicochemical, and functional equivalence to innovator eculizumab, ensuring accurate and reliable serum drug quantitation in both clinical and research contexts.

The primary models used for administering a research-grade anti-C5 antibody in vivo to study tumor growth inhibition and analyze tumor-infiltrating lymphocytes (TILs) are murine syngeneic tumor models.

Syngeneic models involve implanting mouse-derived tumor cell lines into immune-competent mice of the same genetic background. These models preserve the mouse immune system, enabling the study of immunotherapy mechanisms and the characterization of TILs following antibody or drug administration.

Essential context and supporting details:

  • Anti-C5 antibody BB5.1: This is a classic research-grade antibody that specifically blocks mouse C5, used extensively in mouse models for functional and mechanistic studies. BB5.1 does not bind human C5, thus is limited to murine experiments. Its administration in mice inhibits C5 cleavage, thereby blocking pro-inflammatory C5a generation and the membrane attack complex, both relevant for tumor microenvironment modulation.
  • Syngeneic tumor models: Common examples include RENCA (renal cell carcinoma), CT26 (colon carcinoma), EMT6 (mammary carcinoma), and B16F10 (melanoma). These models are widely characterized for baseline TIL populations and tumor-immune signatures, allowing mechanistic studies of how anti-C5 therapy affects immune cell infiltration and tumor progression.
  • Tumor-immune profiling: Syngeneic models allow detailed analysis of TIL composition changes upon treatment. For instance, in RENCA tumors, T cell populations diminish as tumors grow, whereas myeloid cells increase, driving immune suppression—a process potentially targetable by complement inhibition.
  • Why not humanized models? Research-grade anti-mouse C5 antibodies such as BB5.1 are specific to mouse C5 and ineffective in humanized models, which require antibodies cross-reactive with human C5. Humanized mice reconstitute human immune cells, but unless the anti-C5 antibody targets human complement, the approach is not feasible with BB5.1 or similar mouse-specific reagents.

Additional notes:

  • Some studies focus on C5a or its receptor (C5aR1) and blocking antibodies (e.g., IPH5401, anti-C5aR1 agents), testing their efficacy in tumor growth inhibition in syngeneic mouse models (not humanized mice).
  • These models represent the gold standard for preclinical immunotherapy efficacy testing in a functioning immune system, including analysis of TILs via flow cytometry and gene expression.

In summary, mouse syngeneic tumor models are the principal in vivo systems where function-blocking anti-C5 antibodies are administered to study tumor growth and TIL characteristics. Use of these models enables rigorous assessment of immunotherapy mechanisms and provides direct insights translatable to human oncology, though with antibody specificity limitations for humanized systems.

Researchers studying synergistic effects in complex immune-oncology models often combine the eculizumab biosimilar—which inhibits the terminal complement pathway—with checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars in preclinical and early-phase clinical studies. The main goal is to evaluate whether dual targeting of complement-mediated immune suppression and checkpoint-mediated T cell inhibition enhances anti-tumor immunity beyond what is achieved by monotherapies.

Research Strategy:

  • Eculizumab biosimilars block the activation of the complement system, reducing complement-driven immunosuppression in the tumor microenvironment.
  • Checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3 biosimilars) promote T-cell activation by preventing inhibitory signals that would otherwise suppress T-cell function.
  • By co-administering eculizumab biosimilar with checkpoint inhibitors, researchers hypothesize a synergistic effect where the blockade of both suppressive pathways (complement and checkpoint) leads to greater T cell–mediated tumor killing.

Study Designs:

  • Preclinical models: Frequently utilize mouse models of melanoma or other cancers, combining eculizumab biosimilars with checkpoint inhibitors to analyze tumor growth, immune cell activation, and survival outcomes.
  • Immune profiling: Investigators perform flow cytometry and single-cell sequencing to distinguish which immune cells are activated by each combination. For instance, combining anti-PD-1/LAG-3 specifically activates CD4+ T cells, while anti-PD-1/CTLA-4 enhances CD8+ cytotoxic T cell responses; adding complement blockade may further reduce regulatory T cell–mediated suppression and myeloid-derived suppressor cell activity.

Rationale for Combination:

  • The complement system can contribute to immune evasion in cancer by "cooling" the tumor microenvironment. Inhibiting this pathway (with eculizumab biosimilars) may enhance the effectiveness of checkpoint inhibition by reducing local immunosuppression.
  • Combining agents with non-redundant mechanisms of immune activation—such as eculizumab biosimilars and checkpoint inhibitors—allows researchers to investigate additive or synergistic effects, aiming to overcome resistance seen with monotherapies.

Clinical Implications:

  • The long-term safety, bioequivalence, and immunogenicity of eculizumab biosimilars (e.g., SB12) have been validated in settings such as paroxysmal nocturnal hemoglobinuria, supporting their use in combination protocols for immune-oncology research, though direct clinical trial reports in oncology are limited.

In summary, the combination of eculizumab biosimilars and checkpoint inhibitors in immune-oncology models is a methodologically sound strategy to unravel complex immune interactions and to identify potentially more effective therapeutic regimens through synergistic modulation of immune responses. Direct evidence for these combinations in clinical oncology applications is still emerging.

A Eculizumab biosimilar can be used as either the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient’s immune response by enabling the detection of antibodies developed against the therapeutic Eculizumab in patient serum.

In the bridging ADA ELISA format:

  • The Eculizumab biosimilar is immobilized (e.g., via biotin-streptavidin interaction or direct coating) onto a microplate to function as the capture reagent. When patient serum is added, any anti-drug antibodies (ADAs) that have developed against Eculizumab bind to the immobilized biosimilar.

  • The same or a differently labeled Eculizumab biosimilar is used as the detection reagent, typically conjugated to an enzyme (like HRP) or a fluorophore. The bivalent nature of ADAs allows them to bridge the plate-bound (capture) Eculizumab and the labeled (detection) Eculizumab, forming a “bridge”.

  • After washing, a substrate is added for enzymatic conversion by HRP (if used), generating a measurable signal proportional to the amount of ADA present.

This approach exploits the structural and antigenic equivalence between the biosimilar and the reference Eculizumab, ensuring any immune response against the therapeutic is detected, regardless of whether a biosimilar or originator was administered. The assay does not distinguish between antibodies against the biosimilar or originator, which is appropriate since their protein sequences and immunogenic epitopes are identical.

Key points:

  • Eculizumab biosimilar can serve as both capture and detection reagent due to its identical antigenicity to the originator Eculizumab.
  • Bridging ELISA detects bivalent ADAs (mainly IgG) effectively, offering high sensitivity for monitoring immunogenicity during therapy.
  • The signal indicates the presence of anti-Eculizumab antibodies in patient serum, which may impact drug efficacy and safety.

This standard method is widely applied in clinical immunogenicity assessment for therapeutic monoclonal antibodies and their biosimilars.

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.