Anti-Human C5 (Ravulizumab) [Clone ALXN-1210] — Fc Muted™

Anti-Human C5 (Ravulizumab) [Clone ALXN-1210] — Fc Muted™

Product No.: C3295

- -
- -
Product No.C3295
Clone
ALXN-1210
Target
C5
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Complement protein C5; C3 and PZP-like alpha-2-macroglobulin domain-containing protein 4
Isotype
Human IgG2/4κ
Applications
ELISA
,
LC-MS/MS

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Hamster
Expression Host
CHO Cells
FC Effector Activity
Muted
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,
LC-MS/MS
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 Ravulizumab. ALXN-1210 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. It 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 the sites of infection. C5b kickstarts the creation of a 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.

ALXN-1210, also known as ravulizumab, is a human monoclonal antibody. This antibody specifically targets and inhibits C5, preventing its cleavage into C5a and C5b, thus blocking (MAC) formation. Ravulizumab serves as a lasting inhibitor of C5 designed for effect with less frequent dosing compared to its precursor eculizumab. Clinical studies have shown that ravulizumab effectively treats conditions like PNH and aHUS offering patients convenience and quality of life due, to its dosing intervals3-5.

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.
Ligand/Receptor
C6
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Immunology
.
Innate Immunity
.
Autoimmune Disease
.
Blood Disorders
.
Complement

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 Ravulizumab biosimilars are used in pharmacokinetic (PK) bridging ELISA assays as calibration standards or reference controls to enable accurate quantification of Ravulizumab concentrations in serum samples from both biosimilar and reference drug studies.

Essential context and application in the assay:

  • The optimal industry approach is to establish a single PK assay using one analytical standard (calibrator)—often the biosimilar itself—for the quantitative measurement of both biosimilar and reference Ravulizumab in serum.
  • A calibration curve is generated by preparing a range of known concentrations of the Ravulizumab biosimilar standard in human serum (e.g., 50–12,800 ng/mL). This standard curve is used to interpolate the unknown concentrations of Ravulizumab in study samples, whether they contain the biosimilar or the reference product.
  • Quality control (QC) samples are independently prepared using both the biosimilar and the licensed reference product at various concentrations. These are run alongside study samples to confirm the assay’s precision (repeatability) and accuracy (closeness to actual concentration), and to ensure that the assay can reliably quantify either product across the expected range.

Bridging and comparability:

  • The PK bridging ELISA is validated to demonstrate that the biosimilar and reference Ravulizumab produce equivalent signals within the assay (demonstrating “bioanalytical equivalence”). This is statistically confirmed, typically by showing that the 90% confidence interval for the ratio of measured concentrations falls within a predefined equivalence range (e.g., 0.8–1.25), signifying the method is suitable for both molecules.
  • This approach reduces variability compared to using separate methods for biosimilar and reference, and aligns with regulatory expectations for biosimilar PK studies.

Summary of key steps in using biosimilars as standards in PK ELISA:

  • Prepare known concentrations of the biosimilar for the calibration curve in the appropriate matrix (usually human serum).
  • Validate the assay with QC samples made from both biosimilar and reference Ravulizumab, quantified against the biosimilar calibration curve.
  • Confirm that measurements of both products are accurate, precise, and comparable within prespecified acceptance criteria.

This strategy ensures the same reference frame for all PK measurements in bridging studies, supporting bioequivalence assessments and regulatory filings.

The primary models for in vivo administration of a research-grade anti-C5 antibody to study tumor growth inhibition and analyze tumor-infiltrating lymphocytes (TILs) are murine syngeneic tumor models. This is chiefly due to the species specificity of available anti-C5 antibodies and the well-characterized immune competence of these models.

Key details:

  • Murine syngeneic models involve implantation of mouse-derived tumor cells into immune-competent mice of the same genetic background, preserving a fully functional mouse immune system.
  • The BB5.1 monoclonal antibody is the classic research-grade anti-C5 antibody, and it specifically targets mouse C5—it does not bind to or inhibit human C5. This means its use is limited to mouse models.
  • Typical syngeneic tumor lines used for immunotherapy studies and TIL characterization include B16F10 (melanoma), CT26 (colon carcinoma), RENCA (renal cell carcinoma), and EMT6 (breast carcinoma).
Model TypeExample TumorsHost SpeciesC5 Antibody CompatibilityMain Use
Syngeneic (mouse)B16F10, CT26, RENCAMouse (e.g., C57BL/6, BALB/c)Yes (mouse anti-C5)Tumor growth, TILs profiling, immune therapy
HumanizedHuman cell linesImmunodeficient mice reconstituted with human immunityNo (mouse anti-C5 ineffective)Human TILs, limited by antibody incompatibility
  • TIL characterization in these models is robust, as they recapitulate the complexity of adaptive and innate immune interactions—critical for studying the impact of complement inhibition on the tumor microenvironment.

  • While humanized models (mice engrafted with human immune cells and tumors) are valuable for translational human immuno-oncology, standard research-grade anti-C5 antibodies like BB5.1 do not function in these models due to lack of cross-reactivity with human C5. Human-specific anti-C5 antibodies (such as those used clinically, e.g., eculizumab) may be used in humanized models, but these are rarely available as non-proprietary research reagents, and published studies with these combinations remain limited.

  • Blockade of C5 or its active fragment C5a in murine syngeneic models has demonstrated effects on tumor growth, response to immunotherapies (e.g., anti–PD-1), and TIL composition, confirming their central use for these mechanistic studies.

Summary of current practice:

  • Murine syngeneic models using mouse-specific anti-C5 monoclonal antibodies (such as BB5.1) represent the primary system for in vivo tumor growth and TIL studies involving complement blockade.
  • Humanized models are theoretically attractive but are not compatible with most research-grade anti-C5 antibodies, limiting their current application in this context.

References to data from the search results:

  • BB5.1 is mouse C5-specific with no human C5 binding.
  • Syngeneic models allow immune profiling and therapy response analysis, including TILs quantification and modulation.
  • Anti–C5/C5a blockade in these models affects tumor growth, immune cell composition, and synergizes with other therapies.

If further clarification is needed on specific tumor lines, dosing, or TIL assessment methods in these models, please specify.

Researchers investigating the synergistic effects of checkpoint inhibitors in complex immune-oncology models often combine therapies targeting distinct immune pathways, such as Ravulizumab biosimilar (an anti-C5 monoclonal antibody) with other checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3 biosimilars. The strategy is designed to block multiple immune regulatory mechanisms simultaneously, thereby enhancing antitumor immune responses beyond what is achievable by monotherapy.

Essential Research Strategies and Context:

  • Ravulizumab's Target (Complement Inhibition):

    • Ravulizumab targets the complement protein C5, inhibiting the terminal complement cascade. This pathway’s blockade is being studied for its potential to improve immunotherapy efficacy by reducing immunosuppressive signals in the tumor microenvironment.
  • Checkpoint Inhibitor Combinations:

    • Checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3 antibodies block T-cell inhibitory pathways, revitalizing antitumor T-cell responses. Combining inhibitors that target different steps of immune regulation (e.g., CTLA-4 mainly in lymph nodes, LAG-3 at both node and periphery, complement at innate immunity level) may lead to superior tumor control by tackling both adaptive and innate immune suppression.
  • Mechanistic Rationale:

    • Anti-CTLA-4 predominantly enhances T-cell priming and activation at the lymph node level.
    • Anti-LAG-3 or anti-TIM-3 are explored to restore T-cell function within the tumor microenvironment, often in combination with PD-1/PD-L1 blockade.
    • Anti-C5 (Ravulizumab) may mitigate complement-mediated immunosuppressive effects and potentially reduce adverse inflammatory reactions associated with robust T-cell activation.
    • Together, such combinations aim to overcome resistance to single-agent immunotherapies and modulate both innate and adaptive immune arms.
  • Preclinical Modeling:

    • Complex models use syngeneic tumor-bearing mice, humanized mouse models, or ex vivo human tumor tissue. Researchers administer biosimilars in defined sequences or together and measure outcomes like tumor growth inhibition, immune cell infiltration, cytokine profiles, and immune memory formation.
    • Researchers monitor synergistic effects, defined immunologically (greater T-cell activation) and therapeutically (enhanced tumor regression) when combinations outperform single agents.
  • Clinical Implications:

    • The combination of checkpoint inhibitors has led to improved clinical responses but also increased toxicity in some trials (e.g., CTLA-4/PD-1 in melanoma); thus, careful dose optimization and mechanistic studies in preclinical models are essential before clinical translation.
  • Current Data Gaps:

    • While the approach is mechanistically compelling and supported by preclinical evidence for combinations involving established checkpoint inhibitors, published data for combinations specifically using Ravulizumab biosimilars with anti-CTLA-4 or anti-LAG-3 biosimilars are limited, and most available evidence is indirect, inferred from similar combination strategies in the literature.

Key Points:

  • Researchers combine Ravulizumab biosimilars with other checkpoint inhibitors to block complementary immune escape mechanisms in tumor models.
  • These studies predominantly occur in preclinical systems to screen for synergy, optimize dosing, and predict potential for clinical translation.
  • Direct published evidence for Ravulizumab biosimilar in combination with anti-CTLA-4 or anti-LAG-3 biosimilars in complex models is currently sparse; most knowledge is extrapolated from combination strategies with other immune-modulating biologics.

References used establish the general research strategies for checkpoint inhibitor combinations and the theoretical basis for combining anti-complement agents like Ravulizumab with other immunotherapies.

A Ravulizumab biosimilar can be used as a key reagent in a bridging ADA (anti-drug antibody) ELISA to detect patient immune responses to therapeutic Ravulizumab. In this assay format, the biosimilar serves either as the capture or detection reagent because it is highly similar in structure and epitope presentation to the original Revulizumab; thus, it reliably binds to any ADAs generated against the therapeutic drug.

Mechanism in Bridging ADA ELISA:

  • Bridging ELISA detects antibodies that "bridge" between two drug molecules: one immobilized on the plate (capture) and one labeled for detection.
  • For Ravulizumab immunogenicity monitoring:
    • The biosimilar Ravulizumab is immobilized on the microtiter plate as the capture reagent.
    • Patient serum, which may contain ADAs to Ravulizumab, is added; these ADAs can bind the immobilized biosimilar.
    • A second, labeled (e.g., HRP-conjugated or biotinylated) biosimilar Ravulizumab is added as the detection reagent.
    • If ADAs are present, they will bind both the capture and detection biosimilar molecules, creating a "bridge" that results in a measurable signal.
    • Detection is typically visualized via a chromogenic reaction.

Why use a biosimilar for this purpose?

  • The biosimilar's structural and functional similarity (primary sequence, post-translational modifications) to the originator ensures it presents the same ADA epitopes, enabling sensitive and specific detection of ADAs that could react with either product.
  • Using a biosimilar for both capture and detection maintains consistency and addresses supply or licensing concerns that can arise with the originator product.

Summary Table: Ravulizumab Biosimilar in Bridging ADA ELISA

StepReagentPurpose/Description
1Plate-bound biosimilarCapture ADAs present in patient sample
2Patient serumSource of suspected anti-Ravulizumab antibodies
3Labeled biosimilar (e.g., HRP)Detects ADAs bridging between two drug molecules
4Substrate (e.g., TMB)Produces color change for quantification

Key Points:

  • Essential for immunogenicity testing during biosimilar clinical development and post-market monitoring.
  • The assay specifically detects ADAs that could neutralize or impact the safety/efficacy of both Ravulizumab and its biosimilars.
  • Methodology parallels immunogenicity testing for other mAb biosimilars, supporting regulatory requirements for biosimilarity and safety.

This approach is standard practice when evaluating the immunogenic potential of both originator and biosimilar therapeutics in clinical and post-marketing studies.

References & Citations

1. Henes JK, Groga-Bada P, Schaeffeler E, et al. Pharmgenomics Pers Med. 2021;14:893-903.
2. Fredslund F, Laursen NS, Roversi P, et al. Nat Immunol. 2008;9(7):753-760.
3. McKeage K. Drugs. 2019;79(3):347-352.
4. Ravulizumab: A Review in Atypical Haemolytic Uraemic Syndrome - PubMed. Accessed August 11, 2024. https://pubmed.ncbi.nlm.nih.gov/33738756/
5. Röth A, Rottinghaus ST, Hill A, et al. Blood Adv. 2018;2(17):2176-2185.
6. RAVMP - Overview: Ravulizumab Monitoring Panel, Serum. Accessed August 11, 2024. https://www.mayocliniclabs.com/test-catalog/Overview/618645
Indirect Elisa Protocol
LC-MS/MS

Certificate of Analysis

Formats Available

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