Anti-Human CD19 (Tafasitamab) – Fc Muted™

Anti-Human CD19 (Tafasitamab) – Fc Muted™

Product No.: LT3105

- -
- -
Product No.LT3105
Clone
MOR-208
Target
CD19
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Tafasitamab,MOR-00208,MOR-208,CD19
Isotype
Human IgG1κ
Applications
ELISA
,
FA
,
FC
,
IP
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Recommended Isotype Controls
Immunogen
Human CD19
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
WB
IP
FA
FC
IF
IF Microscopy
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 Tafasitamab. This product is for research use only.Tafasitamab activity is directed against human CD19.
Background
CD19 is a B cell surface glycoprotein that enhances B cell receptor signaling and tumor cell proliferation 1. CD19 is an attractive immunotherapy target for cancers of lymphoid origin due to its early and persistent expression throughout B cell maturation 2.

Tafasitamab is a humanized anti-CD19 monoclonal antibody developed by MorphoSys AG under a license from Xencor for the treatment of B cell malignancies 1. The chimeric antibody was engineered by combining the variable region genes of mouse anti-CD19 antibody (clone 4G7) with human light chain κ and heavy chain constant regions 1,2. Light and heavy chain constructs were co-transfected into 293E cells and antibodies were purified using protein A chromatography 2. Additionally, the Fv of 4G7 was humanized, affinity-matured using library design automation, and substitutions S239D/I332E were introduced to increase Fcγ receptor affinity to human 2, mouse 2, and cynomolgus monkey 3 FcγRs, with FcγRIIIa affinity being particularly enhanced 2.

Tafasitamab mediates B cell lysis via apoptosis and immune effector mechanisms including antibody-dependent cellular cytotoxicity (ADCC) 2,4 and antibody-dependent cellular phagocytosis 2. Tafasitamab also increases antiproliferative activity and inhibits lymphoma growth in mouse xenograft models. ADCC is mediated by natural killer cells 5 through a granzyme B-dependent mechanism that is further enhanced by lenalidomide 6,7.

Tafasitamab is also known as XmAb5574 1.
Antigen Distribution
CD19 is a surface antigen present on all B cells (healthy and malignant) except hematopoietic stem cells and plasma cells; it is highly conserved in B-cell malignancies.
Ligand/Receptor
CD21, CD81
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Immuno-Oncology
.
Immunology
.
Oncology

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 Tafasitamab biosimilars are used as calibration standards (also called calibrators or reference controls) in pharmacokinetic (PK) bridging ELISAs to generate the standard curve needed to quantify drug concentrations in serum samples. In a typical PK bridging ELISA, these biosimilars are serially diluted to establish known concentrations, allowing unknown sample concentrations to be determined with reference to this standard curve.

Context and Supporting Details:

  • Biosimilar as Calibrator: In biosimilar PK studies, the consensus best practice is to use a single, well-characterized biosimilar as the assay calibrator for quantification of both the biosimilar and the original (reference) drug in all test samples within a single PK ELISA method.

  • Preparation: The biosimilar Tafasitamab is reconstituted per the supplier's instructions (e.g., Invitrogen guideline: 1 mg/mL in distilled water, gently shaken without vortexing). This reconstituted antibody is then used to prepare a series of standard solutions usually ranging from low to high concentrations (the specific range is method-dependent, e.g., 50‒12,800 ng/mL).

  • Standard Curve Generation: Multiple (typically 5–7) standards covering the expected assay range are run alongside test samples in the ELISA plate to generate a standard curve. The assay signal from each standard is measured, and the curve is fitted using appropriate regression models (such as 4-parameter logistic regression or linear regression).

  • Measurement of Unknowns: The concentrations in unknown serum samples are interpolated from this standard curve. Since both reference and biosimilar products are quantified relative to this biosimilar standard, any differences in assay response between the products are minimized, enabling accurate PK bridging studies.

  • Quality Controls: Additional controls and validation samples—prepared with both biosimilar and reference-sourced Tafasitamab—are included to demonstrate the method’s precision, accuracy, and suitability for detecting both products.

  • Why Use a Biosimilar Standard? Regulatory and industry guidance for bioanalytical comparability studies typically recommend this approach, as it ensures consistency, reduces assay variability, and eliminates the need for cross-reference assays when comparing drug exposure (PK) of biosimilar versus reference products.

In summary: Tafasitamab biosimilars are diluted to prepare ELISA standard curves, serving as the quantitative reference against which serum drug concentrations are measured in PK bridging studies. This practice supports rigorous, reliable pharmacokinetic comparisons in biosimilar drug development.

The primary in vivo models used to study tumor growth inhibition and analyze tumor-infiltrating lymphocytes (TILs) following administration of a research-grade anti-CD19 antibody are xenograft, syngeneic, and humanized mouse models; most published tumor inhibition data use xenografts with human B cell tumors in immunodeficient mice, but studies of TILs are more feasible in syngeneic and humanized settings.

Model Overview:

  • Xenograft Models (Immunodeficient Mice):

    • Tumor type: Mice (e.g., BALB/c nude, scid/scid, NOD-SCID) are injected with human CD19⁺ B cell tumor cell lines such as BJAB or Sultan.
    • Immune context: The host is immunodeficient, limiting the endogenous TIL response to murine innate cells (e.g., NK cells), but allowing the study of direct tumor inhibition and antibody effector functions such as antibody-dependent cellular cytotoxicity (ADCC).
    • TIL analysis: Limited, as adaptive immunity is absent or severely compromised, but evaluation of myeloid and NK cell infiltration is possible.
  • Syngeneic Models (Immunocompetent Mice):

    • Tumor type: Murine B cell tumors are engrafted in immunocompetent mice, and anti-mouse CD19 antibodies are investigated.
    • Immune context: Full murine immune system permits study of TILs, including diverse lymphocyte populations and their function.
    • Limitation: These models use murine antibodies/tumors, not human reagents or tumor cells, so findings may not precisely translate to the activity of human-targeted antibodies.
  • Humanized Mouse Models:

    • Tumor type: Immunodeficient mice are engrafted with human hematopoietic stem cells to reconstitute a human-like immune system, then implanted with human CD19⁺ tumors.
    • Immune context: Enables study of human immune cell (including TIL) infiltration and interplay with therapeutic antibodies or combination therapies (e.g., anti-CD19 plus other agents).
    • Caveats: Complex, resource-intensive, and engraftment variability can affect TIL readouts.
Model TypeTumor SourceHost Immune SystemTypical Use for Anti-CD19 StudyTILs Analyzed
XenograftHuman (e.g., BJAB)Immunodeficient (murine)Tumor growth inhibition, antibody efficacyMostly innate NK/cells, not adaptive TILs
SyngeneicMurineImmunocompetent (murine)Mechanism studies, immune response, TIL profilingFull murine TIL repertoire
HumanizedHuman (CD19⁺ tumor)Human (engrafted in mouse)Tumor inhibition, human immune cell infiltrationHuman TILs

Key Points:

  • Most published anti-CD19 antibody studies demonstrating tumor growth inhibition use xenograft models in immunodeficient mice, which are not suited for full TIL characterization due to a lack of adaptive immune cells.
  • Syngeneic models allow direct assessment of TILs and tumor-immune interactions but require anti-mouse CD19 antibodies and do not capture human antibody biology.
  • Humanized mouse models enable administration of human anti-CD19 antibodies and analysis of human TILs but are more technically challenging and resource-intensive.

In sum, choice of model depends on whether the focus is on tumor growth inhibition efficacy (xenograft) or in-depth TIL characterization (syngeneic or humanized). Most anti-CD19 studies in the literature have used human xenograft models in immunodeficient mice for tumor inhibition endpoints, sometimes with limited TIL analysis constrained by the model’s immune context.

Researchers currently use Tafasitamab biosimilars primarily in combination with other immunomodulatory agents—most notably lenalidomide—to enhance anti-tumor activity via mechanisms like antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP). While direct studies with checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars in synergy with Tafasitamab are relatively limited in published clinical data, there is a strong scientific rationale for exploring such combinations in complex immune-oncology models.

Tafasitamab is an anti-CD19 monoclonal antibody whose action includes direct cytotoxicity against B-cell tumors and robust enhancement of immune-mediated killing by modifying its Fc portion, thus increasing ADCC and ADCP far beyond typical IgG1 analogs. In preclinical and early clinical research, combining multiple immune checkpoint inhibitors (e.g., CTLA-4, PD-1/L1, LAG-3) with antibody therapies is postulated to overcome monotherapy limitations, increase T-cell activation, and broaden immune response against tumors.

Key research approaches for studying synergy:

  • Preclinical and translational models: Researchers test combinations (e.g., Tafasitamab biosimilar + checkpoint inhibitor biosimilars) in vitro and in vivo to evaluate enhanced tumor killing, increased infiltration of effector immune cells, and modulation of the tumor microenvironment.
  • Mechanistic studies: By targeting different immune pathways (e.g., B-cell depletion via Tafasitamab and T-cell activation via checkpoint blockade), scientists assess non-overlapping mechanisms to potentially increase efficacy.
  • Combination strategies: The strategy builds on findings from combinations like CTLA-4/PD-1 blockade, which show increased progression-free survival and complete response rates in tumors not responsive to monotherapies. Combinations with Tafasitamab may harness B-cell targeting alongside T-cell checkpoint inhibition for additive or synergistic effects.
  • Clinical trials: While Tafasitamab combination studies primarily focus on pairing with lenalidomide or rituximab, the promising results—such as improved progression-free survival—provide groundwork for future designs that include checkpoint inhibitor biosimilars.

Important considerations in immune-oncology synergy studies:

  • Researchers must manage increased toxicity, as seen in multi-checkpoint combinations.
  • Biomarker analysis (PD-L1, CD19 expression, etc.) is vital for selecting patient populations most likely to benefit.
  • Sequencing and timing of administration (e.g., whether to administer Tafasitamab before or after checkpoint inhibitors) may impact efficacy and immune cell targeting.
  • Translational research often uses complex immune-oncology animal models and ex vivo patient-derived cell systems to interrogate synergistic effects before clinical adoption.

Summary: The combination of Tafasitamab biosimilars with checkpoint inhibitors in immune-oncology relies on preclinical and early-phase translational research, focusing on leveraging distinct immune mechanisms for enhanced anti-tumor activity. As clinical experience grows, such combinations are likely to be tested in trials evaluating synergistic efficacy and safety in hematologic and possibly solid tumor contexts.

The query asks about using a Tafasitamab biosimilar in a bridging ADA ELISA, but the search results do not contain specific information about Tafasitamab biosimilars being used as capture or detection reagents in anti-drug antibody (ADA) assays. However, I can provide comprehensive information about how bridging ELISA assays work for ADA detection and the principles that would apply to Tafasitamab ADA testing.

Bridging ELISA Principles for ADA Detection

A bridging ELISA is the standard approach for detecting anti-drug antibodies (ADAs) against therapeutic monoclonal antibodies like Tafasitamab. In this assay format, the drug serves dual roles as both capture and detection reagent, creating a "bridge" when ADAs are present in patient samples.

Standard Bridging ELISA Configuration

The typical bridging ELISA setup involves coating microtiter plates with the therapeutic drug (in this case, Tafasitamab or its biosimilar) to capture ADAs from patient serum or plasma samples. After washing away unbound materials, a labeled version of the same drug is added as the detection reagent. If ADAs are present, they form a bridge between the plate-bound drug and the labeled drug in solution.

For Tafasitamab ADA testing, the process would theoretically follow this sequence:

Capture Phase: Tafasitamab or its biosimilar would be immobilized on the ELISA plate surface to capture any anti-Tafasitamab antibodies present in patient samples.

Detection Phase: A labeled version of Tafasitamab (typically conjugated with HRP or biotin) would be added to bind to the captured ADAs, completing the bridge formation.

Signal Detection Methods

The detection system would likely employ enzyme-labeled reagents with chromogenic substrates. Studies have shown successful use of HRP-labeled antibodies combined with TMB (3,3',5,5'-tetramethylbenzidine) substrate, providing sensitive detection limits as low as 0.39 ng/mL in similar antibody drug assays.

Considerations for Tafasitamab ADA Testing

Given that Tafasitamab is an anti-CD19 monoclonal antibody used for treating relapsed or refractory B-cell malignancies, several factors would be important in ADA assay design:

Drug Interference: High concentrations of circulating Tafasitamab could interfere with ADA detection by competing with the assay reagents. Advanced bridging ELISA formats have been developed to address this issue, including acid dissociation pretreatment and solid-phase extraction methods.

Immune Complex Detection: Modern bridging ELISAs can be configured to detect not only free ADAs but also drug-ADA immune complexes that may circulate in treated patients. This would be particularly relevant for monitoring the complete immune response to Tafasitamab therapy.

The information available does not specifically describe Tafasitamab biosimilar use in ADA assays, but the general principles of bridging ELISA methodology would apply to any monoclonal antibody therapeutic, including Tafasitamab and its potential biosimilars.

References & Citations

1. Hoy SM. Tafasitamab: First Approval. Drugs. 80(16):1731-1737. 2020.
2. Horton HM, Bernett MJ, Pong E, et al. Cancer Res. 68(19):8049-8057. 2008.
3. Zalevsky J, Leung IW, Karki S, et al. Blood. 113(16):3735-3743. 2009.
4. Rafiq S, Cheney C, Mo X, et al. Leukemia. 26(7):1720-1722. 2012.
5. Chan WK, Kung Sutherland M, Li Y, et al. Clin Cancer Res. 18(22):6296-6305. 2012.
6. Awan FT, Lapalombella R, Trotta R, et al. Blood. 115(6):1204-1213. 2010.
7. Kellner C, Zhukovsky EA, Pötzke A, et al. Leukemia. 27(7):1595-1598. 2013.
8. Woyach JA, Awan F, Flinn IW, et al. Blood. 124(24):3553-3560. 2014.
Indirect Elisa Protocol
FA
Flow Cytometry
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.