Anti-Human C5 (Ravulizumab) [Clone ALXN-1210]

Anti-Human C5 (Ravulizumab) [Clone ALXN-1210]

Product No.: C3290

- -
- -
Product No.C3290
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
Active
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 as calibration standards and reference controls in pharmacokinetic (PK) bridging ELISA by serving as the reference material for generating the standard curve against which unknown serum concentrations are quantified.

In a PK bridging ELISA, the quantification of Ravulizumab (or its biosimilar) in serum samples relies on comparison to a known reference standard. The typical workflow includes:

  • Preparation of Calibration Standards: Serial dilutions of a research-grade Ravulizumab biosimilar are spiked into a serum matrix to generate calibrators at multiple concentrations (e.g., 50 – 12800 ng/mL). These standards establish the dose-response (standard) curve within the ELISA.

  • Reference Controls (QCs): Additional samples containing known concentrations of the biosimilar (and, often, the originator product) are included as quality control (QC) samples to track assay performance and ensure that the method accurately quantifies both biosimilar and reference products across the entire range of expected concentrations.

  • Analytical Standard Selection: It is now considered best practice to use a single analytical standard—often the biosimilar—for both test (biosimilar) and reference (originator) product quantification, provided bioanalytical equivalence of the two molecules in the assay has been demonstrated. This minimizes variability and eliminates cross-assay inconsistencies.

  • Bridging and Bioanalytical Comparability: Method validation involves demonstrating that both the biosimilar and the originator Ravulizumab produce equivalent signal in the ELISA. This includes testing multiple independent standard and QC preparations with both molecules, statistical comparison of measured values, and setting predefined equivalence criteria (e.g., 90% CI within 0.8–1.25 for signal ratios).

  • Application to Study Samples: The ELISA standard curve, established using the biosimilar, is used to interpolate unknown concentrations of Ravulizumab in patient serum samples, regardless of whether the sample contains biosimilar- or originator-sourced drug. This standardization underpins the PK bridging necessary for biosimilar development.

Key technical points:

  • The research-grade biosimilar must be validated to demonstrate comparable binding and detection characteristics to the originator in the ELISA format before it is adopted as the calibration standard.
  • QC samples using both the biosimilar and the originator can be included to monitor ongoing assay equivalency.

Summary Table:

ComponentUse in PK Bridging ELISA
Research-grade biosimilarAnalytical standard for calibration curve and QCs
Calibration standardsSerial dilutions spiked in serum for curve generation
Reference controls (QC samples)Known concentrations for accuracy/precision, using both products
Originator Ravulizumab (optional)For equivalency assessment during validation
Unknown serum samplesQuantification by interpolation from biosimilar-based curve

This approach ensures robust, reproducible PK quantitation and regulatory-aligned comparability between biosimilar and originator Ravulizumab in clinical bioanalytical studies.

Syngeneic mouse models are the primary in vivo systems where research-grade anti-C5 antibodies are administered to study both tumor growth inhibition and the characterization of tumor-infiltrating lymphocytes (TILs). There is currently little direct evidence for the use of anti-C5 antibodies in fully humanized models for this specific application.

Key details:

  • The BB5.1 monoclonal antibody is a well-established agent for blocking mouse C5 and is mouse-specific; it does not bind human C5, thus making it suitable specifically for murine (syngeneic) models.
  • Commonly used syngeneic tumor models for these studies include RENCA (renal carcinoma), CT26 (colon carcinoma), B16F10 (melanoma), and 4T1 (breast carcinoma); these represent tumors implanted in immunocompetent mice of the same genetic background, allowing for assessment of immunotherapy and immune cell infiltration (including TILs).
  • Anti-C5 antibody treatment in these models has been shown to inhibit C5 cleavage and thereby suppress downstream pro-inflammatory signaling by C5a and membrane attack complex, which is relevant for modulating the tumor immune microenvironment and tumor progression.
  • Immune profiling in these models enables the characterization of TIL subsets—such as CD8+ T cells, myeloid-derived suppressor cells, and other immune populations—before and after experimental manipulation with complement inhibitors.

Humanized mouse models are not commonly utilized for anti-C5 antibody studies targeting tumor growth and TILs because:

  • Most available research-grade anti-C5 antibodies, including BB5.1, have been developed to specifically target mouse C5 and are ineffective against human C5.
  • Humanized models would require an antibody cross-reactive to human C5 or engineered human immune systems, which is more typical for translational studies with clinical-grade antibodies like eculizumab (not research grade and not commonly employed for murine TIL studies).

Summary table:

Model TypeAntibody UsedTarget SpeciesTypical Tumor LinesTIL CharacterizationReference
SyngeneicBB5.1, othersMouseRENCA, CT26, B16F10, 4T1Yes
HumanizedClinical-grade (e.g. eculizumab)HumanPatient-derived xenograftsNot common for anti-C5 research-grade mAbs

In summary: Syngeneic mouse tumor models are the predominant and appropriate in vivo system for studying anti-C5 antibody effects on tumor growth and TIL composition, owing to the specificity of research-grade antibodies for mouse C5 and the ability to assess immune microenvironment changes with fully functional murine immunity.

Researchers explore combination strategies of immune checkpoint inhibitors—including biosimilars of agents such as anti-CTLA-4 and anti-LAG-3—to study synergistic anti-tumor effects, especially in complex immune-oncology models. The use of ravulizumab biosimilar in this context specifically is not directly documented in the search results, as ravulizumab itself is a complement C5 inhibitor used mainly for diseases like paroxysmal nocturnal hemoglobinuria, not as a checkpoint inhibitor. However, general approaches to combination immunotherapy in oncology with biosimilars can be described.

Key Context and Supporting Details:

  • Checkpoint inhibitor combinations (anti-CTLA-4, anti-LAG-3, etc.):

    • Anti-CTLA-4 and anti-PD-1/PD-L1 have different mechanisms, with CTLA-4 blockade primarily augmenting T cell priming and expansion, while PD-1 blockade acts more in the tumor microenvironment.
    • Anti-LAG-3 and related agents are being tested in coordination with PD-1 blockade to overcome resistance mechanisms and further enhance anti-tumor immunity.
    • Combination therapies often produce synergistic effects, with preclinical and clinical studies demonstrating improved progression-free survival and higher rates of durable complete responses over monotherapies—but with a tradeoff in terms of increased toxicity.
  • Role of biosimilars in combination studies:

    • Biosimilars (highly similar, less expensive versions of original biologic drugs) of monoclonal antibodies are increasingly used in research and clinical settings to expand access and lower costs.
    • Combination studies in preclinical models and early-phase clinical trials sometimes employ biosimilars to replicate expected pharmacologic effects while enabling broader study recruitment and cost-effective experimentation.
    • However, published evidence specifically on the use of biosimilars (including hypothetical ravulizumab biosimilars) in combination with checkpoint inhibitors in advanced immune-oncology models remains limited. Much of the current combination research is still conducted with reference/originator biologics rather than biosimilars.
  • Experimental and clinical design:

    • Researchers often use syngeneic mouse tumor models, humanized mouse models, or co-culture systems to test combinations of checkpoint inhibitors for synergy.
    • Endpoints typically include tumor growth inhibition, survival, immune cell infiltration, cytokine production, and biomarker assessment reflecting immune activation.
    • Studies incorporating biosimilars aim to demonstrate pharmacodynamic equivalence when used alone or in combination, ideally mirroring the synergistic effects observed with the original biologics.
  • Unique consideration for ravulizumab:

    • Ravulizumab is a C5 complement inhibitor, not classified as an immune checkpoint inhibitor, and is not commonly used in combination with checkpoint inhibitors in published immune-oncology research.
    • No direct research evidence specifies ravulizumab biosimilar use in these immune-oncology combination models based on the current literature.

Summary Table: Strategies and Evidence

Inhibitor TypeExample Biosimilar UseCombination RationalesMain Study Contexts
CTLA-4Under developmentEnhance T cell primingMelanoma, solid tumors
LAG-3Clinical trials (relatlimab)Overcome PD-1 resistanceAdvanced melanoma
Ravulizumab (C5)No evidence in I-O combinationsNot a checkpoint inhibitorHematology, PNH

Conclusion:
Researchers combine checkpoint inhibitor biosimilars (e.g., anti-CTLA-4, anti-LAG-3) to study synergistic effects in immune-oncology, typically using preclinical models and early trials, but there is no documented use of ravulizumab biosimilar in these synergistic checkpoint inhibitor studies as it serves a different immunological role. Most synergy research and evidence involve combinations of recognized checkpoint inhibitors with demonstrated immune-enhancing effects, sometimes substituting biosimilars for reference products to extend study access.

A Ravulizumab biosimilar can be used as a capture or detection reagent in a bridging ADA ELISA to monitor a patient’s immune response against the therapeutic drug by exploiting the ability of anti-drug antibodies (ADA) to simultaneously bind two drug molecules—one labeled for detection and one immobilized for capture.

In a typical bridging ADA ELISA:

  • The biosimilar Ravulizumab (which closely mimics the structure of the original drug) is either biotinylated (for capture onto streptavidin-coated plates) or labeled (e.g., with HRP for detection).
  • Patient serum is added; if ADAs specific to Ravulizumab are present, they will bind to both the immobilized biosimilar molecule and the labeled biosimilar molecule, forming a "bridge".
  • Detection signal (e.g., colorimetric change) is proportional to the amount of ADA bridging the capture and detection reagents.

Key Points:

  • Using a biosimilar ensures the assay detects antibodies targeting the clinically relevant epitopes of the original drug and maintains sensitivity to immune responses specific to the therapeutic.
  • The assay enables high-throughput and sensitive measurement of immunogenicity, supporting PK/PD and safety evaluations in patients.
  • Standardization with a biosimilar ensures that both preclinical and clinical ADA data can be compared reliably, especially when switching between original and biosimilar therapy.

Advantages:

  • High specificity and sensitivity for clinically relevant ADAs.
  • Allows immunogenicity assessment regardless of whether the patient receives the originator or biosimilar formulation.

Limitations:

  • Assay specificity may be influenced by serum matrix effects, necessitating careful reagent selection and assay validation.
  • The presence of circulating drug may mask ADA detection in patient samples, requiring possible sample acidification or other pre-treatment.

To summarize: In immunogenicity testing, a Ravulizumab biosimilar is used in bridging ADA ELISAs as both the capture and detection reagent to enable sensitive monitoring of anti-Ravulizumab antibody responses in patients treated with either the originator or biosimilar drug.

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.