LCR30 Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Structure and Function

Antibodies are Y-shaped glycoproteins with two functional regions:

  • Fab (Fragment antigen-binding): Binds antigens via hypervariable complementarity-determining regions (CDRs) .

  • Fc (Fragment crystallizable): Mediates immune effector functions (e.g., phagocytosis, complement activation) .

FeatureDescription
Heavy ChainsDetermine antibody class (IgG, IgM, IgA, etc.); ~50 kDa .
Light Chainsκ or λ subtypes; ~25 kDa .
GlycosylationCritical for Fc stability and effector functions; occurs at N297 in IgG .

Therapeutic Antibody Development Trends

  • Clinical Pipeline: Over 500 antibodies are in early research phases, with >50 in late-stage trials for cancer and autoimmune diseases .

  • Engineering Advances: Fc modifications (e.g., afucosylation) and humanization improve efficacy and safety .

  • Targets: Immune checkpoints (e.g., PD-1, GARP:TGF-β1) dominate oncology pipelines .

Methodological Frameworks for Antibody Research

Key methodologies from the search results applicable to novel antibodies like LCR30:

  • Avidity Index (AI) Testing: Measures antibody-antigen binding strength using chaotropic agents (e.g., thiocyanate) .

  • Pharmacodynamic Biomarkers:

    • T-cell activation (Ki67+ CD8+ cells) .

    • Receptor occupancy assays (e.g., GARP:TGF-β1 saturation) .

Recommendations for Further Investigation

  1. Database Searches:

    • UniProtKB or PDB for structural/functional annotations.

    • ClinicalTrials.gov for ongoing trials involving LCR30.

  2. Nomenclature Verification:

    • Cross-reference with synonyms (e.g., "LCR-30," "CD30 ligand inhibitors").

  3. Proprietary Sources:

    • Contact biotech firms (e.g., AbbVie, GenScript) for internal data disclosures .

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
LCR30 antibody; At3g43505 antibody; T18D12 antibody; Defensin-like protein 122 antibody; Low-molecular-weight cysteine-rich protein 30 antibody; Protein LCR30 antibody
Target Names
LCR30
Uniprot No.

Target Background

Database Links
Protein Families
DEFL family
Subcellular Location
Secreted.
Tissue Specificity
Expressed in flower buds, but not in stems, roots or rosette leaves.

Q&A

What are the key considerations when selecting antibodies for research applications?

When selecting antibodies for research applications, several critical factors must be considered to ensure experimental success. First, determine whether monoclonal or polyclonal antibodies are more appropriate for your specific application; monoclonals offer higher specificity to a single epitope, while polyclonals provide broader antigen recognition . Second, validate antibody specificity through multiple methods including Western blotting, immunoprecipitation, or ELISA to confirm target binding . Third, consider species cross-reactivity if your research involves multiple model organisms, as demonstrated in the IL1RL1 study where only 5.7% of antibodies showed cross-reactivity with murine IL1RL1 . Fourth, evaluate functional capabilities if blocking or neutralizing properties are required; in some cases, as with the IL1RL1-specific antibodies, only 16.4% demonstrated functional blocking of ligand-receptor interactions . Fifth, assess antibody concentration requirements, as sufficient quantities are essential for comprehensive characterization assays. A systematic approach to antibody selection significantly increases experimental reproducibility and reliability.

What methods are available for antibody generation from peripheral B cells?

Several robust methods exist for generating antibodies from peripheral B cells, with recent technological advances significantly enhancing efficiency and throughput. The B-cell cloning approach involves isolating antigen-specific B cells from peripheral blood, typically using fluorescence-activated cell sorting (FACS) with labeled antigens as demonstrated in the rabbit IL1RL1 antibody generation study . Single B cells are then cultured in conditions supporting antibody secretion, often achieving concentrations around 0.9 μg/ml for rabbit-derived cells . Alternatively, the B-cell PCR workflow allows for direct amplification of variable heavy (VH) and light (VL) chain genes from isolated B cells, followed by recombinant expression in suitable host cells like HEK293 . This approach yielded a remarkable 90% amplification efficiency in the IL1RL1 study . Sequencing-informed antibody engineering can further optimize antibodies for specific applications. The development of semi-automated platforms has dramatically increased throughput, with the described rabbit B-cell platform maintaining approximately 75% unique antibody sequences among 227 IL1RL1-specific B cell clones, demonstrating exceptional diversity . Each method offers distinct advantages in terms of speed, diversity, and antibody characteristics.

How can researchers effectively characterize antibody binding epitopes?

Effective characterization of antibody binding epitopes requires a multi-method approach to generate comprehensive binding profiles. Cross-competition ELISA represents a powerful initial technique, involving a two-dimensional binding matrix where antibodies compete against each other for antigen binding . This method successfully identified six major epitope groups among IL1RL1-specific antibodies, revealing distinct competition patterns that correlated with cross-reactivity properties . Hydrogen-deuterium exchange mass spectrometry (HDX-MS) provides more detailed epitope mapping by measuring changes in hydrogen-deuterium exchange rates upon antibody binding. X-ray crystallography and cryo-electron microscopy offer atomic-level resolution of antibody-antigen complexes, though they require significant expertise and resources. Alanine scanning mutagenesis, where individual amino acids are systematically replaced with alanine, helps identify critical binding residues. Analysis of CDR sequences contributes valuable complementary information, as demonstrated in the IL1RL1 study where CDR-H3 length varied between 4-19 amino acid residues with a mean of 11.4±2.7 . This comprehensive epitope characterization enables rational selection of antibodies targeting functionally important epitopes and facilitates the development of antibody panels covering diverse recognition sites.

What strategies exist for enhancing antibody cross-species reactivity for translational research?

Enhancing antibody cross-species reactivity presents a significant challenge in translational research but can be approached through several sophisticated strategies. Epitope-focused immunization targets highly conserved regions between species, increasing the probability of generating cross-reactive antibodies; this approach proved valuable in the IL1RL1 study where 20.6% of human IL1RL1-binding antibodies cross-reacted with cynomolgus IL1RL1, although only 5.7% cross-reacted with murine IL1RL1 . Sequential immunization with orthologous antigens from different species can educate the immune system to recognize conserved epitopes. Antibody engineering approaches include grafting cross-reactive complementarity-determining regions (CDRs) onto stable frameworks or introducing specific mutations that enhance binding to orthologous proteins. Rational design using structural bioinformatics identifies conserved surface-exposed regions as immunization targets. High-throughput screening remains crucial, as exemplified in the IL1RL1 study where researchers screened 978 IgG-positive supernatants to identify the small percentage with desired cross-reactivity . The complexity of enhancing cross-reactivity underscores the value of combination approaches, potentially implementing multiple strategies to achieve optimal cross-species recognition while maintaining therapeutic efficacy.

How do CDR sequence variations impact antibody specificity and affinity?

Complementarity-determining region (CDR) sequence variations fundamentally influence antibody specificity and affinity through complex structure-function relationships. CDR-H3, showing the greatest variability among CDRs, plays a particularly crucial role in antigen recognition; in the IL1RL1 study, CDR-H3 lengths ranged from 4-19 amino acids with a mean of 11.4±2.7 residues, following a Poisson distribution typical for rabbit antibodies . This length variation significantly impacts the antibody binding pocket architecture, with longer CDR-H3 loops potentially forming more complex interactions with antigens. Amino acid composition within CDRs critically affects binding characteristics through electrostatic interactions, hydrogen bonding, and hydrophobic contacts. The IL1RL1 study revealed 2-36 amino acid replacements per VH with a mean value of 13±5.3 replacements compared to germline sequences, indicating substantial somatic hypermutation . Clustering analysis of CDR-H3 and CDR-L3 sequences demonstrated that approximately 75% of 227 B-cell clones produced unique antibody chains, highlighting remarkable diversity even within antigen-specific responses . This diversity explains the identification of at least six distinct epitope groups among IL1RL1-binding antibodies, each with unique functional properties including differential blocking of ligand-receptor interactions and species cross-reactivity patterns .

What methodologies can assess antibody neutralization capacity in receptor-ligand interactions?

Assessment of antibody neutralization capacity in receptor-ligand interactions requires a multi-tiered approach combining biochemical and cellular assays to establish functional relevance. Biochemical inhibition assays provide initial screening data by quantifying antibody-mediated disruption of purified receptor-ligand binding; in the IL1RL1 study, researchers identified antibodies inhibiting IL33-IL1RL1 interaction with varying efficacies, applying a 40% inhibition threshold for initial selection . Cell-based functional assays offer more physiologically relevant assessments by measuring downstream signaling or cellular responses to receptor-ligand engagement. The IL1RL1 study employed a human IL33-activated NK cell assay, demonstrating that selected antibodies inhibited IL-33 dependent NK-cell activation with 40-100% efficacy . Importantly, researchers observed strong correlation between biochemical and cellular inhibition assays (RSq = 0.90) at high inhibition thresholds (>90%), validating the predictive value of biochemical screening . Competition binding studies using surface plasmon resonance or bio-layer interferometry provide kinetic insights into inhibition mechanisms. Flow cytometry-based assays can evaluate antibody-mediated blocking of ligand binding to cell-surface receptors. This comprehensive assessment ensures selection of antibodies with consistent neutralizing activity across different experimental contexts, crucial for therapeutic applications.

What are the key considerations in transitioning from rabbit-derived to humanized therapeutic antibodies?

Transitioning from rabbit-derived to humanized therapeutic antibodies involves sophisticated engineering approaches balanced against the need to preserve critical binding properties. Sequence analysis must first identify the minimal rabbit-specific regions essential for antigen recognition, typically focusing on the complementarity-determining regions (CDRs) . These regions are then grafted onto human antibody framework regions, maintaining proper structural alignment while minimizing immunogenicity. Computational modeling guides selection of human framework regions that best accommodate rabbit CDRs with minimal conformational distortion. Post-grafting optimization frequently requires back-mutations of specific framework residues that support CDR conformation, identified through structural analysis or regression techniques. The IL1RL1 study noted that "for therapy of human diseases a humanization of the functional rabbit antibody is required to reduce possible immunogenicity issues in the clinic" . Critical quality attributes including affinity, specificity, and functional activity must be systematically compared between original and humanized versions through binding kinetics measurements, cross-reactivity profiling, and functional assays. Stability and manufacturability assessments ensure the humanized antibody maintains pharmaceutical viability. This comprehensive approach preserves the exceptional specificity and affinity of rabbit-derived antibodies while creating molecules suitable for human therapeutic applications.

What strategies can overcome antibody expression and purification challenges?

Optimization of antibody expression and purification involves systematic refinement of multiple parameters to maximize yield while maintaining functional integrity. For mammalian expression systems, transfection optimization is critical; the IL1RL1 antibody study achieved high efficiency using the 293-free transfection reagent with HEK293 cells, resulting in an average IgG productivity of 32 μg/ml after transient co-transfection . Cell culture conditions significantly impact productivity—researchers cultured HEK293 cells at 37°C with 8% CO2 in F17-medium with continuous shaking at 180 rpm for seven days to maximize antibody yields . Vector design considerations include optimized promoters, signal peptides, and codon usage; the IL1RL1 study employed the CMV-promoter and human CD33-signal peptide for efficient expression . For purification, the researchers implemented a two-step approach: initial Protein A affinity chromatography followed by size exclusion chromatography to separate monomeric antibodies from aggregates . This process was performed in 20 mM Histidine, 140 mM NaCl pH 6.0 buffer to maintain stability . Quality control measures included SDS-PAGE, size exclusion chromatography, mass spectrometry, and endotoxin determination . Scaling considerations for research versus production purposes may require balancing between transient systems for rapid generation of multiple candidates and stable cell line development for larger-scale production of selected antibodies.

How can researchers address potential cross-reactivity and off-target binding concerns?

Addressing cross-reactivity and off-target binding concerns requires comprehensive validation strategies applied throughout antibody development. Negative selection screens during early development help eliminate cross-reactive antibodies; the IL1RL1 study implemented a critical counter-screen against the human Fc part (used as an antigen conjugate), identifying and removing 14.2% of antibodies with undesired Fc binding . Multi-platform validation comparing binding profiles across different assay formats (ELISA, Western blot, immunoprecipitation) can reveal format-dependent cross-reactivity. Tissue cross-reactivity panels using immunohistochemistry on diverse tissue arrays help identify unexpected binding to related epitopes. Competitive binding assays with structurally similar proteins assess specificity against closely related targets. For therapeutic antibodies, in silico analysis identifying proteins with sequence or structural homology to the target epitope guides selection of potential cross-reactants for testing. The IL1RL1 researchers meticulously characterized antibody diversity through CDR sequence analysis, finding remarkably high diversity with approximately 75% of B-cell clones producing unique antibody chains . This diversity enabled selection of highly specific antibodies from the large candidate pool. Importantly, cross-reactivity assessment should be conducted under conditions mimicking the intended application environment, including appropriate buffer systems, protein concentrations, and temperature considerations.

What approaches can determine antibody stability and shelf-life for research applications?

Determining antibody stability and shelf-life for research applications requires multi-parameter analysis focusing on structural integrity and functional retention over time. Accelerated stability studies expose antibodies to elevated temperatures (25°C, 37°C, 40°C) for defined periods, followed by comparative analysis against refrigerated controls (2-8°C) to predict long-term stability. Techniques including size exclusion chromatography effectively monitor aggregation tendencies—a primary degradation pathway for antibodies . The IL1RL1 study implemented this approach to separate monomeric antibodies from aggregates during purification . Differential scanning calorimetry and circular dichroism spectroscopy provide thermal stability data by measuring antibody unfolding temperatures and conformational changes. Binding activity assays performed at regular intervals during storage assess functional stability, with retained antigen binding serving as the critical quality attribute. Freeze-thaw studies determine resistance to structural damage during laboratory handling, typically testing 3-5 cycles. Buffer optimization significantly impacts stability; the IL1RL1 researchers used 20 mM Histidine, 140 mM NaCl pH 6.0 for their antibody preparations, a formulation known to enhance stability . For longer-term storage, many researchers divide antibody preparations into single-use aliquots stored at -80°C to minimize repeated freeze-thaw cycles . Implementation of these comprehensive stability assessments ensures experimental reproducibility across extended research timelines.

How are high-throughput platforms revolutionizing antibody discovery and characterization?

High-throughput platforms have fundamentally transformed antibody discovery through integration of automation, microfluidics, and computational approaches. Semi-automated B-cell isolation platforms significantly increase throughput while maintaining single-cell resolution; the rabbit IL1RL1 antibody study employed such a platform, noting it was "highly effective in isolating a large number of rabbit B-cell clones secreting sufficient monoclonal antigen specific IgG" . Sequence-based characterization utilizing next-generation sequencing enables rapid analysis of antibody repertoire diversity; researchers analyzing IL1RL1-specific antibodies found remarkable diversity with approximately 75% of 227 B-cell clones producing unique antibody chains . Automated expression systems efficiently convert identified sequences into testable proteins through robotics-assisted cloning and transfection; the study implemented SLIC cloning with 94% efficiency, dramatically accelerating the process . Multiplexed functional screening assays simultaneously evaluate multiple parameters including binding specificity, affinity, and functional activity; researchers performed six different primary screening assays to comprehensively characterize their antibody candidates . Computational epitope mapping and antibody modeling increasingly complement experimental approaches for candidate prioritization. These integrated platforms substantially reduce discovery timelines while expanding the diversity of candidates evaluated, as demonstrated by the IL1RL1 researchers who noted their workflow "will be significantly reduced, because exclusively the most promising antibody candidates from the primary screening will be processed" .

What roles are dual-antibody therapies playing in addressing variant escape in viral diseases?

Dual-antibody therapies are emerging as pivotal solutions to the challenge of variant escape in viral diseases through multiple complementary mechanisms. Research at Washington University School of Medicine demonstrated that dual-antibody combinations maintain effectiveness against emerging SARS-CoV-2 variants even when individual antibodies show reduced neutralization capacity in vitro . This retained efficacy stems from complementary epitope targeting, where simultaneous binding to distinct viral regions creates redundant neutralization mechanisms less susceptible to individual escape mutations. The Nature publication highlighted that "combination therapy is likely needed for treating infections with this virus as more variants emerge" . Critically, these dual therapies create a significantly higher genetic barrier to resistance development; the study found "no drug resistance to combinations whatsoever, across all of the different variants" . This contrasts with monotherapy approaches where single viral mutations can substantially reduce efficacy. Animal model validation further confirmed these advantages; when tested in mice and hamsters challenged with various SARS-CoV-2 variants, dual-antibody therapies consistently outperformed expectations based on in vitro data . Similar principles apply to other viral diseases, including MERS-CoV, where the REGN3048 and REGN3051 monoclonal antibody combination targets multiple epitopes on the spike glycoprotein . These findings collectively establish dual-antibody approaches as a robust strategy against viral evolution.

How do animal model selection and humanization strategies influence therapeutic antibody development?

Animal model selection and humanization strategies profoundly influence therapeutic antibody development through their impact on translatability and immunogenicity. Humanized mice expressing human target proteins provide critical models for antibody validation; the MERS-CoV study utilized mice humanized for dipeptidyl peptidase-4 (huDPP4), the viral receptor, to evaluate antibody efficacy against viral challenge . Similarly, Washington University researchers tested antibody combinations in animal models to validate in vitro findings, noting that "some of the combinations performed better than we thought they would, based on in vitro data" . This highlights the importance of in vivo validation beyond cell-based assays. For antibody generation, rabbits offer advantages including robust immune responses and antibodies with naturally human-like characteristics; the IL1RL1 study successfully generated diverse, high-affinity antibodies from rabbits, with 22.5% of antibodies binding specifically to human IL1RL1 . Humanization strategies must balance immunogenicity reduction against maintaining critical binding properties. The IL1RL1 researchers noted that "for therapy of human diseases a humanization of the functional rabbit antibody is required to reduce possible immunogenicity issues in the clinic" . Modern approaches include CDR grafting onto human frameworks, veneering (surface residue modification), and deimmunization through computational epitope prediction. Careful validation throughout the humanization process ensures preservation of binding affinity, specificity, and functional activity while minimizing potential human anti-drug antibody responses.

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 2024 Thebiotek. All Rights Reserved.