LCR48 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to CD48 Antibodies

CD48, also known as BLAST-1, BCM-1, and SLAMF2, is a 65 kDa GPI-linked protein in the CD2 family of immunoglobulin superfamily molecules . It plays a crucial role in the immune system by interacting with other molecules like CD2 and CD244 (2B4), influencing the activation of T cells and natural killer (NK) cells .

Applications and Research Findings

CD48 antibodies have been explored for their therapeutic potential, particularly in cancer treatment. For example, a chimeric antibody against CD48, known as cHuLym3, has shown potent antitumor activity in preclinical studies against lymphoid leukemia and lymphoma .

Table 1: Characteristics of CD48 Antibodies

Antibody TypeTargetApplicationKey Findings
Murine Anti-CD48 (mHuLym3)CD48Leukemia and Lymphoma TreatmentPotent antitumor activity in preclinical studies .
Chimeric Anti-CD48 (cHuLym3)CD48Leukemia and Lymphoma TreatmentEnhanced antibody-dependent cellular cytotoxicity (ADCC) compared to murine version .

Mechanism of Action

CD48 interacts with CD244 (2B4) on NK cells and T cells, which can either enhance or inhibit immune cell activation depending on the context . This complex interaction highlights the potential for CD48-targeting antibodies to modulate immune responses.

Table 2: CD48 Interactions and Their Effects

InteractionCell TypeEffect
CD48-CD244NK CellsInhibitory or Coactivating .
CD48-CD2T Cells, APCsActivatory .

Therapeutic Potential

The therapeutic potential of CD48 antibodies lies in their ability to target hematopoietic cells, which are involved in various immune-related diseases. For instance, anti-CD48 antibodies have been explored for treating autoimmune diseases and cancers .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
LCR48 antibody; At3g48231 antibody; T24C20Defensin-like protein 51 antibody; Low-molecular-weight cysteine-rich protein 48 antibody; Protein LCR48 antibody
Target Names
LCR48
Uniprot No.

Target Background

Database Links

KEGG: ath:AT3G48231

STRING: 3702.AT3G48231.1

UniGene: At.63257

Protein Families
DEFL family
Subcellular Location
Secreted.

Q&A

What is the molecular structure of RC48 antibody and how does it function?

RC48 (disitamab vedotin) is a novel humanized anti-HER2 antibody conjugated with monomethyl auristatin E (MMAE) . The antibody component targets HER2-expressing cells, while the MMAE payload acts as a cytotoxic agent by disrupting the microtubule network in dividing cells. The antibody binds to HER2 receptors on cancer cells, leading to internalization of the antibody-drug conjugate, followed by release of MMAE within the cell, resulting in cell cycle arrest and apoptosis .

Structurally, RC48 consists of:

  • A humanized monoclonal antibody targeting the HER2 receptor

  • A linker system that connects the antibody to the drug payload

  • MMAE, a potent antimitotic agent

The drug-to-antibody ratio is optimized to deliver sufficient cytotoxic payload while maintaining the binding properties of the antibody component .

What experimental models are used to evaluate RC48 efficacy?

Researchers evaluate RC48 efficacy through multiple experimental approaches:

Methodologically, researchers must carefully characterize HER2 expression levels (using IHC scoring 0, 1+, 2+, 3+) in their experimental systems to ensure relevant clinical translation .

How does the binding site specificity of RC48 compare to other therapeutic antibodies?

The binding site specificity of therapeutic antibodies is critical for their efficacy and safety profile. Studies examining other therapeutic antibodies, such as the anti-LcrV monoclonal antibody 7.3, demonstrate that binding site specificity rather than just binding affinity may be responsible for therapeutic efficacy .

  • Epitope mapping: Determining the precise binding region on HER2 can be accomplished through techniques such as:

    • X-ray crystallography of antibody-antigen complexes

    • Hydrogen-deuterium exchange mass spectrometry

    • Site-directed mutagenesis of potential binding residues

    • Peptide array analysis

  • Binding kinetics: Beyond simple affinity measurements, association and dissociation rates (kon and koff) provide insights into binding dynamics that may correlate with therapeutic efficacy .

  • Conformational effects: Understanding whether RC48 binds to and stabilizes specific HER2 conformations that affect downstream signaling is important for elucidating its mechanism of action .

Research comparing RC48 binding characteristics with those of established anti-HER2 antibodies like trastuzumab would provide valuable insights into its therapeutic potential and potential synergies or redundancies with existing therapies.

What are the current methodologies for analyzing RC48 biodistribution and tumor penetration?

Advanced techniques for tracking RC48 biodistribution and tumor penetration include:

  • Radiolabeling approaches:

    • Conjugation with radioisotopes (e.g., 89Zr, 124I, 111In) for PET or SPECT imaging

    • Allows for quantitative, whole-body biodistribution assessment over time

    • Can be validated with ex vivo gamma counting of tissues

  • Fluorescence-based methods:

    • Near-infrared fluorophore conjugation for optical imaging

    • Confocal microscopy of tumor sections to assess tissue penetration depth

    • Intravital microscopy in window chamber models for real-time visualization

  • Mass spectrometry imaging:

    • MALDI-MSI for spatial localization of antibody and released MMAE payload

    • Provides simultaneous visualization of drug distribution and molecular changes in the tumor microenvironment

  • Computational modeling:

    • Pharmacokinetic/pharmacodynamic modeling integrating physiological parameters

    • Tumor penetration simulations based on vascular architecture and interstitial pressure

When designing biodistribution studies, researchers should consider timing of assessments, as the pharmacokinetics of antibody-drug conjugates differ significantly from small molecule drugs, with longer circulation half-lives and slower tumor accumulation .

What mechanisms contribute to RC48 resistance in clinical settings?

Several mechanisms may contribute to RC48 resistance, similar to those observed with other anti-HER2 therapies:

  • Target-related mechanisms:

    • Downregulation of HER2 expression

    • Expression of truncated HER2 isoforms lacking the antibody binding domain

    • Mutations in the HER2 extracellular domain affecting antibody binding

    • Altered HER2 trafficking and internalization pathways

  • Drug payload-related mechanisms:

    • Upregulation of drug efflux pumps (e.g., P-glycoprotein)

    • Changes in lysosomal processing affecting MMAE release

    • Alterations in microtubule composition reducing MMAE sensitivity

    • Expression of drug-metabolizing enzymes

  • Compensatory signaling:

    • Activation of alternative receptor tyrosine kinases (EGFR, HER3)

    • Upregulation of downstream signaling pathways (PI3K/AKT, MAPK)

    • Changes in tumor microenvironment signaling

Research methodologies to study resistance include:

  • Developing resistant cell lines through prolonged exposure

  • Comparative proteomics and transcriptomics of sensitive versus resistant models

  • Patient-derived xenografts from responders versus non-responders

  • Analysis of paired biopsies (pre-treatment and at progression)

How can we optimize RC48 antibody-dependent cellular cytotoxicity (ADCC) in experimental models?

ADCC is an important mechanism for many therapeutic antibodies. For RC48 and similar antibodies, optimization strategies include:

  • Antibody engineering approaches:

    • Fc region glycoengineering to enhance FcγR binding

    • Amino acid substitutions in the Fc region to improve ADCC

    • Isotype selection (IgG1 demonstrates superior ADCC compared to other isotypes)

  • Experimental design considerations:

    • Selection of appropriate effector cells (NK cells, monocytes, or PBMCs)

    • Effector-to-target ratio optimization

    • Incubation time standardization

    • Cytotoxicity detection method validation

  • Combination strategies:

    • Addition of cytokines to activate effector cells (IL-2, IL-15)

    • Combination with immune checkpoint inhibitors

    • Co-treatment with agents that upregulate HER2 expression

Research shows that chimeric antibodies with human constant regions (like cHuLym3) demonstrate more potent ADCC compared to their murine counterparts when human peripheral blood mononuclear cells are used as effectors. This suggests that proper humanization of RC48 is critical for optimizing its ADCC potential .

What are the optimal biomarkers for patient selection in RC48 clinical trials?

Identifying appropriate biomarkers for patient selection is critical for optimizing clinical outcomes with RC48. Based on the search results, several biomarker approaches should be considered:

  • HER2 expression levels:

    • Immunohistochemistry (IHC) scoring (0, 1+, 2+, 3+)

    • In situ hybridization (ISH) for HER2 gene amplification

    • The data shows differential response rates based on HER2 expression levels, with higher response rates in HER2 IHC 2+/3+ patients (92.3-100%) compared to HER2 IHC 0/1+ patients (50%)

  • PD-L1 expression:

    • Combined Positive Score (CPS) assessment

    • RC48 combination studies with immunotherapy show differential responses based on PD-L1 status

    • PD-L1 positive (CPS ≥ 10) status appears to correlate with enhanced response in some combination settings

  • Multiparametric biomarker approaches:

    • Combined analysis of HER2 and PD-L1 status

    • Gene expression signatures related to HER2 signaling

    • Immune infiltration patterns in the tumor microenvironment

The following table summarizes response rates based on HER2 and PD-L1 biomarker status from a clinical trial combining RC48-ADC with toripalimab:

StratificationConfirmed Objective Response Rate
HER2 IHC (2+/3+) PD-L1 (+)100% (5/5)
HER2 IHC (2+/3+) PD-L1 (-)92.3% (12/13)
HER2 IHC (1+) PD-L1 (+)50% (2/4)
HER2 IHC (1+) PD-L1 (-)50% (3/6)
HER2 IHC (0) PD-L1 (-)50% (1/2)
Total76.7% (23/30)

These data suggest that a combined biomarker strategy using both HER2 and PD-L1 status may provide the most robust patient selection approach .

How can we develop standardized methods for measuring cytokine release syndrome risk with RC48?

Cytokine release syndrome (CRS) is a potential concern with antibody therapeutics. Developing standardized methods for assessing CRS risk with RC48 should involve:

  • In vitro cytokine release assays (CRAs):

    • Whole blood assays: Using fresh human whole blood from multiple donors

    • PBMC assays: Isolating peripheral blood mononuclear cells for more controlled testing

    • Key methodological considerations:

      • Anticoagulant selection (heparin vs. EDTA)

      • Incubation conditions (time, temperature, agitation)

      • Antibody concentration range

      • Controls (positive and negative reference antibodies)

  • Reference antibody panel:
    The international collaborative study reported in search result developed a reference antibody panel for qualification and validation of cytokine release assay platforms. Similar reference standards should be established for testing RC48, including:

Antibody specificityFunction in CRA validation
Anti-CD28SAStrong positive control
Anti-CD3Positive control
Anti-CD52Moderate positive control
IgG1K isotypeNegative control
  • Cytokine readout standardization:

    • Core cytokine panel: IL-6, TNF-α, IL-1β, IFN-γ

    • Extended panel: IL-2, IL-8, IL-10, IL-12

    • Standardized assay platforms (e.g., multiplex bead arrays, ELISA)

  • Correlation with clinical data:

    • Retrospective analysis of cytokine levels in patients experiencing adverse events

    • Establishment of threshold values predictive of clinical CRS

    • Integration with pharmacokinetic data

What are the mechanistic synergies between RC48 and immune checkpoint inhibitors?

Research demonstrates promising results when combining RC48 with immune checkpoint inhibitors such as toripalimab (anti-PD-1) and cadonilimab (anti-PD-1/CTLA-4 bispecific). The mechanistic synergies may include:

  • Complementary cell death mechanisms:

    • RC48-ADC: Direct cytotoxicity via MMAE-mediated microtubule disruption

    • Checkpoint inhibitors: Enhanced T-cell-mediated tumor killing

    • Combined effect: Multiple parallel cell death pathways

  • Immunogenic cell death promotion:

    • RC48-induced tumor cell death may release tumor antigens

    • Checkpoint blockade prevents T-cell exhaustion/inhibition

    • Result: Amplified anti-tumor immune response

  • Modulation of tumor microenvironment:

    • ADC-mediated depletion of HER2+ immunosuppressive cells

    • Checkpoint inhibition reverses T-cell dysfunction

    • Combined: Conversion from "cold" to "hot" tumor microenvironment

  • Biomarker interactions:

    • Clinical data shows particularly high response rates (100%) in patients with both HER2 positivity (IHC 2+/3+) and PD-L1 positivity, suggesting a biological interaction between these pathways

Research approaches to study these synergies include:

  • Immune profiling of tumor biopsies before and after treatment

  • Analysis of circulating immune cell populations and phenotypes

  • Multiplex immunohistochemistry to assess spatial relationships between tumor cells, HER2 expression, and immune infiltrates

  • Single-cell RNA sequencing to characterize cell type-specific responses

How can we design preclinical models to predict optimal sequencing of RC48 with other therapies?

Designing effective preclinical models for therapy sequencing requires sophisticated approaches:

  • Advanced animal models:

    • Humanized immune system mouse models that recapitulate human immune interactions

    • Patient-derived xenografts that maintain tumor heterogeneity

    • Syngeneic mouse models with murine versions of the therapies to preserve intact immunity

  • Sequential treatment schedules:

    • RC48 followed by immunotherapy

    • Immunotherapy followed by RC48

    • Concurrent administration

    • Intermittent scheduling options

  • Readout parameters:

    • Tumor growth inhibition

    • Survival analysis

    • Immune infiltration patterns

    • Pharmacodynamic biomarkers

    • Resistance development monitoring

  • In vitro 3D models:

    • Tumor spheroids with immune components

    • Microfluidic tumor-on-a-chip systems

    • Organoid co-cultures with immune cells

  • Computational approaches:

    • Systems pharmacology modeling

    • Machine learning algorithms integrating multiple parameters

    • Mathematical modeling of tumor growth under different treatment sequences

When designing these studies, researchers should consider the unique pharmacokinetics of antibody-drug conjugates versus checkpoint inhibitors, as well as potential overlapping toxicities that may limit certain combination regimens in clinical applications .

What methodologies are most effective for evaluating RC48 off-target effects?

Evaluating off-target effects of RC48 requires comprehensive approaches:

  • Cross-reactivity screening:

    • Tissue cross-reactivity studies using immunohistochemistry across multiple human tissues

    • Protein microarray screening for binding to non-HER2 proteins

    • Surface plasmon resonance with potential off-target proteins

  • Advanced safety models:

    • Humanized mouse models expressing human HER2

    • Non-human primates with high HER2 homology

    • Ex vivo human tissue slice cultures

  • Mechanistic toxicology studies:

    • Evaluation of MMAE-mediated vs. antibody-mediated toxicities

    • Investigation of bystander effects on HER2-negative cells

    • Immune-mediated toxicity assessment

  • Specialized assays for common ADC toxicities:

    • Peripheral neuropathy models (DRG neuron cultures)

    • Hepatotoxicity assessment (3D liver spheroids, hepatocyte cultures)

    • Hematological toxicity (colony formation assays, bone marrow cultures)

Clinical data shows that RC48-ADC is associated with specific toxicities that require monitoring, including hypoesthesia (60.5%), alopecia (55.8%), and leukopenia (55.8%). More severe grade 3 toxicities include hypoesthesia (23.3%) and neutropenia (14.0%) .

How can we develop predictive biomarkers for RC48-associated peripheral neuropathy?

Peripheral neuropathy is a significant adverse event observed with RC48 treatment. Developing predictive biomarkers requires:

  • Patient-derived biospecimen analysis:

    • Baseline peripheral blood biomarkers

    • Genetic polymorphism screening (e.g., genes involved in MMAE metabolism)

    • Nerve conduction studies pre- and post-treatment

    • Skin biopsy for small fiber assessment

  • In vitro neurotoxicity models:

    • Dorsal root ganglion (DRG) neuron cultures

    • Human iPSC-derived sensory neurons

    • Microfluidic neuron-Schwann cell co-cultures

    • High-content imaging for neurite outgrowth and degeneration

  • Mechanistic studies:

    • Disruption of axonal transport by MMAE

    • Mitochondrial dysfunction in neurons

    • Neuroinflammatory markers

    • Blood-nerve barrier integrity assessment

  • Clinical correlation studies:

    • Cumulative dose analysis

    • Time course of symptom development

    • Recovery patterns post-treatment

    • Correlation with pharmacokinetic parameters

Research suggests that peripheral sensory neuropathy affects approximately 56-58% of patients receiving RC48-based therapies, with severe (grade ≥3) cases occurring in a subset of patients. Developing predictive biomarkers would enable better patient selection and proactive management strategies .

How might next-generation RC48 derivatives overcome current limitations?

Future RC48 derivatives could address current limitations through several approaches:

  • Linker technology optimization:

    • Site-specific conjugation methods for homogeneous drug loading

    • Linkers with tailored stability profiles for specific tumor types

    • Tumor microenvironment-triggered release mechanisms

    • Self-immolative linkers with enhanced release kinetics

  • Alternative payload strategies:

    • DNA-damaging agents for MMAE-resistant tumors

    • Immunomodulatory payloads to enhance immune responses

    • Dual-payload ADCs targeting multiple cell death pathways

    • Non-cytotoxic payloads (e.g., signaling inhibitors)

  • Antibody engineering:

    • Affinity modulation for improved tumor penetration

    • Fc engineering for enhanced ADCC or extended half-life

    • Bispecific formats targeting HER2 plus complementary targets

    • pH-sensitive binding to improve tumor selectivity

  • Novel formulations and delivery systems:

    • Nanoparticle encapsulation for altered biodistribution

    • Targeted delivery to specific anatomical sites

    • Extended-release formulations for reduced dosing frequency

    • Local delivery systems for specific cancer types

These approaches should be guided by detailed understanding of current RC48 limitations, including resistance mechanisms, toxicity profiles, and pharmacokinetic/pharmacodynamic relationships .

What methodological approaches can optimize RC48 for difficult-to-treat cancers with low HER2 expression?

Optimizing RC48 for cancers with low HER2 expression requires innovative approaches:

  • Enhancement of HER2 expression:

    • Epigenetic modifiers to upregulate HER2 transcription

    • Proteasome inhibitors to reduce HER2 degradation

    • Heat shock protein inhibitors to increase membrane HER2

    • HDAC inhibitors as HER2 expression modulators

  • ADC design modifications:

    • Higher drug-to-antibody ratios for increased potency

    • More potent payloads requiring fewer HER2 receptors

    • Bystander effect-enhanced linker-payload systems

    • Cleavable linkers optimized for low-receptor density conditions

  • Novel targeting strategies:

    • Bispecific antibodies targeting HER2 plus a highly expressed secondary target

    • Avidity enhancement through multivalent binding

    • Pan-HER family targeting to increase binding sites

    • Peptide-drug conjugates with broader HER binding profiles

  • Combination approaches:

    • Synergistic drug combinations that enhance ADC efficacy

    • Sequential therapy to induce HER2 expression

    • Combining with immune therapies that work independently of HER2 levels

    • Rational combinations addressing resistance pathways

Clinical data shows that RC48 can achieve moderate activity even in patients with low HER2 expression (HER2 IHC 0 or 1+), with response rates of approximately 50% when combined with checkpoint inhibitors. This suggests potential approaches to extend the utility of RC48 beyond HER2-high cancers .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.