CSLA1 Antibody

Shipped with Ice Packs
In Stock

Description

CELA1 Antibody: Therapeutic Potential in Lung Diseases

CELA1 is a protease implicated in elastin degradation, a key pathological process in chronic obstructive pulmonary disease (COPD) and emphysema. A 2024 study published in JCI Insight demonstrated that a novel anti-CELA1 antibody (KF4) reduced elastin remodeling and lung damage in mouse models .

Key Findings:

  • Mechanism: The antibody binds to lung tissue under mechanical stretch, inhibiting CELA1-mediated elastase activity .

  • Therapeutic Efficacy:

    • Reduced alveolar destruction in age-related emphysema models.

    • Mitigated lung damage in three distinct COPD models .

  • Human Relevance: Researchers are developing a humanized version of KF4 for clinical trials, with stability and efficacy testing projected to conclude by 2026 .

Data Table: CELA1 Antibody Study Overview

ParameterDetails
TargetChymotrypsin-like elastase 1 (CELA1)
Antibody NameKF4
Study TypePreclinical (mouse models)
Key OutcomeReduced elastin degradation and alveolar damage
Next StepsHumanization and IND-enabling studies
Funding SourcesNIH (R01HL141229, R01HL142708), Veterans Administration, A1 Foundation

Anti-CS1 (SLAMF7) Antibodies: Applications in Multiple Myeloma

CS1 (CD319/SLAMF7) is a cell surface glycoprotein overexpressed in multiple myeloma cells. Anti-CS1 antibodies, such as HuLuc63 (elotuzumab), have shown therapeutic promise:

Key Findings:

  • Mechanism:

    • Binds CS1 on myeloma cells, inducing antibody-dependent cellular cytotoxicity (ADCC) via NK cells .

    • Synergizes with immunomodulatory drugs (e.g., lenalidomide) to enhance tumor cell killing .

  • Clinical Data:

    • In xenograft models, HuLuc63 reduced tumor burden by >80% .

    • Phase III trials demonstrated improved progression-free survival in relapsed/refractory myeloma .

Data Table: Anti-CS1 Antibody Characteristics

ParameterDetails
TargetCS1 (SLAMF7)
Antibody NameHuLuc63 (elotuzumab)
ApplicationsMultiple myeloma therapy
Binding SpecificityCD138+ myeloma cells, NK cells, CD8+ T cells
FDA Approval StatusApproved in 2015 for combination therapy
Key Clinical BenefitMedian PFS: 19.4 months (vs. 14.9 months in controls)

Cross-Reactivity and Characterization Challenges

Antibody validation remains critical, as highlighted by initiatives like the NIH’s Antibody Characterization Lab and YCharOS:

  • Validation Protocols:

    • Screening via ELISA, Western blot, and immunohistochemistry .

    • Use of knockout cell lines to confirm specificity .

  • Case Study:

    • YCharOS found ~12 publications per protein target included data from non-specific antibodies, underscoring the need for rigorous validation .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CSLA1; At4g16590; dl4320w; FCAALL.402; Probable glucomannan 4-beta-mannosyltransferase 1; Cellulose synthase-like protein A1; AtCslA1; Glucomannan synthase; Mannan synthase 1
Target Names
CSLA1
Uniprot No.

Target Background

Function
CSLA1 Antibody targets a probable mannan synthase enzyme. This enzyme exhibits 4-beta-mannosyltransferase activity on mannan, utilizing GDP-mannose as a substrate. The resulting beta-1,4-mannan product serves as the backbone for galactomannan synthesis through the action of galactomannan galactosyltransferase. Galactomannan, a non-cellulosic polysaccharide, is an integral component of plant cell walls.
Database Links
Protein Families
Glycosyltransferase 2 family, Plant cellulose synthase-like A subfamily
Subcellular Location
Golgi apparatus membrane; Multi-pass membrane protein.

Q&A

What is CELA1 and what is its significance in pulmonary research?

CELA1 (Chymotrypsin-like Elastase 1) is an enzyme that has been identified as playing a significant role in lung tissue remodeling. Recent research has shown that CELA1 actively bonds to lung tissue under physical stretch conditions, enhancing elastase activity - a key disease process in various pulmonary disorders . The significance of CELA1 lies in its involvement in the pathogenesis of chronic obstructive pulmonary disease (COPD) and emphysema, making it an important target for therapeutic intervention. When CELA1 activity is blocked through antibody targeting, researchers have observed decreased elastin remodeling and subsequent protective effects in multiple COPD models . This mechanism represents a novel approach to treating emphysematous lung diseases, which affect millions of people worldwide.

How do researchers distinguish CELA1 antibodies from other elastase-targeting antibodies?

Distinguishing CELA1 antibodies from other elastase-targeting antibodies requires careful attention to specificity testing and validation protocols. Unlike neutrophil elastase antibodies that have been more extensively studied, CELA1 antibodies target a specific elastase subtype with unique tissue distribution and functional characteristics . Researchers typically validate CELA1 antibody specificity through multiple methods including Western blotting, immunohistochemistry, and functional assays that measure elastin remodeling activity. In the development process, scientists must test antibodies against multiple elastase subtypes to confirm selective binding to CELA1 over other elastases. This specificity validation is essential because many elastases share structural similarities that can lead to cross-reactivity if antibodies are not properly characterized. Additionally, researchers often employ knockout models or siRNA techniques to confirm that observed effects are specifically related to CELA1 inhibition rather than other elastase activities.

What experimental models are most effective for studying CELA1 antibody efficacy?

The most effective experimental models for studying CELA1 antibody efficacy include both in vitro and in vivo systems that reflect the physiological conditions of pulmonary diseases. According to recent research, three types of chronic obstructive pulmonary disease models have been successfully used to demonstrate the protective effects of CELA1 antibody blockade . In vitro models typically involve adult lung specimens subjected to physical stretch conditions, which activates CELA1 binding to lung tissue and subsequent elastin remodeling . This approach allows researchers to directly observe the molecular interactions between CELA1 and lung tissue components. For in vivo studies, mouse models of age-related emphysema, cigarette smoke-induced COPD, and genetic models of lung disease provide relevant systems for evaluating antibody efficacy. These models allow for assessment of both biochemical changes (elastin degradation, inflammatory markers) and functional outcomes (breathing parameters, lung compliance). Before advancing to human studies, researchers must validate findings across multiple models to ensure robust efficacy evidence and identify potential model-specific limitations.

How can researchers optimize CELA1 antibody humanization for therapeutic development?

Optimizing CELA1 antibody humanization for therapeutic development is a complex process requiring multiple strategic decisions and technical approaches. As outlined in the research on CELA1 antibodies, the initial step involves humanizing a mouse antibody, which requires careful design to maintain antigen recognition while minimizing immunogenicity . Researchers typically begin by identifying the complementarity-determining regions (CDRs) of the mouse antibody that are essential for CELA1 binding, then grafting these regions onto a human antibody scaffold. This process requires sophisticated computational modeling to predict how the resulting chimeric structure will fold and interact with the target antigen. Multiple humanization approaches may be tested in parallel, including CDR grafting, framework adaptation, and phage display techniques. Development teams often create several candidate antibodies with varying degrees of humanization, which are then screened for binding affinity, stability, and reduced immunogenicity potential. As demonstrated in similar antibody development programs like CH-CSLEX-1, verification of proper IgG formation with predicted isoelectric points provides important quality control checks . Successful humanization strategies must balance maintaining the original specificity and affinity while maximizing human sequence content to reduce anti-drug antibody responses in patients.

What are the critical factors in assessing elastin remodeling inhibition by CELA1 antibodies?

Assessing elastin remodeling inhibition by CELA1 antibodies requires sophisticated analytical approaches that address multiple dimensions of this complex biological process. Critical factors include quantitative measurement of elastin degradation products, visualization of tissue architecture changes, and functional evaluation of mechanical properties. Researchers must establish reliable biomarkers that correlate with disease progression and therapeutic response, as direct measurement of elastin remodeling in situ presents significant technical challenges. According to recent studies, when CELA1 antibodies were used to block CELA1 activity, researchers observed a protective decrease in elastin remodeling across multiple experimental models . This assessment typically involves histological analysis with specialized stains for elastin fibers, immunohistochemistry to detect elastin fragments, and biochemical assays measuring desmosine and isodesmosine levels (specific cross-linking amino acids unique to elastin). Advanced imaging techniques such as second harmonic generation microscopy or atomic force microscopy can provide detailed visualization of extracellular matrix changes. Mechanical testing of tissue samples can determine functional improvements in elasticity following antibody treatment. Researchers must also consider the timing of assessment, as elastin remodeling occurs over extended periods, necessitating both acute and chronic evaluation timepoints to fully characterize antibody efficacy.

How can researchers design validation studies to translate CELA1 antibody findings from mouse models to human applications?

Designing validation studies to translate CELA1 antibody findings from mouse models to human applications requires a systematic, multifaceted approach addressing species differences and clinical relevance. As indicated in the CELA1 research, the next critical step involves clarifying that the remodeling process observed in mouse models also occurs in human tissue . Researchers should begin with ex vivo studies using human lung tissue samples from both healthy donors and COPD patients to validate the presence and activity of CELA1 in human disease contexts. These studies should employ multiple analytical methods including immunohistochemistry, enzymatic activity assays, and protein expression analysis to comprehensively characterize CELA1's role in human pulmonary pathology. Comparative studies examining differences in CELA1 structure, expression patterns, and functional activity between mouse and human samples are essential to identify potential translation challenges. Humanized mouse models, where mouse CELA1 is replaced with the human variant, can provide valuable intermediate data. Prior to clinical trials, researchers should develop and validate biomarkers that correlate with CELA1 activity and can be measured in human samples non-invasively. Additionally, pharmacokinetic/pharmacodynamic (PK/PD) modeling using both mouse data and human tissue samples can help predict effective dosing regimens. Engagement with regulatory agencies early in the translation process ensures appropriate study design that will generate data supportive of eventual investigational new drug (IND) applications.

What assay methods are most reliable for evaluating anti-CELA1 antibody specificity and affinity?

The most reliable assay methods for evaluating anti-CELA1 antibody specificity and affinity combine multiple complementary techniques to provide comprehensive characterization. Surface plasmon resonance (SPR) technology, similar to the Biacore analysis mentioned in research on comparable antibodies, offers real-time, label-free measurement of binding kinetics and affinity constants between CELA1 and candidate antibodies . This provides crucial quantitative data on association and dissociation rates that inform antibody selection. Enzyme-linked immunosorbent assays (ELISAs) remain valuable for high-throughput screening and can be designed in several formats - direct binding, competitive, or sandwich - depending on the specific research question. For instance, the sandwich ELISA approach allows researchers to confirm binding specificity by testing reactivity against related proteins, as demonstrated in the CH-CSLEX-1 antibody studies where multiple antigens were used to confirm specificity . Immunoprecipitation followed by mass spectrometry provides powerful confirmation of antibody specificity by identifying all proteins captured by the antibody from complex biological samples. Flow cytometry using cells expressing CELA1 can assess antibody binding in a cellular context. Additionally, functional assays measuring inhibition of CELA1 enzymatic activity using fluorogenic substrates directly assess the antibody's biological activity beyond simple binding. Researchers should employ at least three independent methods to comprehensively characterize antibody specificity and affinity before progressing to in vivo studies.

What are the optimal immunohistochemistry protocols for detecting CELA1 in tissue samples?

Optimal immunohistochemistry (IHC) protocols for detecting CELA1 in tissue samples require careful optimization across multiple parameters to ensure specific, reproducible, and quantifiable results. Based on antibody research practices, researchers should begin with antigen retrieval optimization, testing both heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) and Tris-EDTA buffer (pH 9.0), as well as enzymatic retrieval methods to determine which best exposes CELA1 epitopes in formalin-fixed, paraffin-embedded tissue. When selecting primary antibodies, both polyclonal and monoclonal options should be compared, with monoclonals often providing higher specificity but potentially lower sensitivity than polyclonals. Antibody validation should include positive controls (tissues known to express CELA1, such as pancreas) and negative controls (tissues known not to express CELA1), as well as technical controls (primary antibody omission, isotype controls) . Signal amplification systems should be selected based on expression levels, with avidin-biotin complexes or polymer-based detection systems offering enhanced sensitivity for potentially low-abundance CELA1. Counterstaining protocols need optimization to provide adequate contrast without obscuring specific CELA1 staining. For quantitative analysis, digital pathology approaches using automated image analysis software can provide standardized scoring of staining intensity and distribution. Multiplex IHC protocols that simultaneously detect CELA1 and markers of cellular stress or inflammation can provide valuable contextual information about CELA1's role in disease processes. Researchers should validate IHC findings with complementary techniques such as in situ hybridization to detect CELA1 mRNA or Western blotting to confirm antibody specificity.

How can researchers overcome the human anti-mouse antibody (HAMA) interference in CELA1 antibody development?

Researchers can overcome human anti-mouse antibody (HAMA) interference in CELA1 antibody development through several strategic approaches that address this common immunological challenge. HAMA interference occurs when human subjects develop antibodies against mouse-derived therapeutic antibodies, potentially neutralizing their efficacy or causing adverse reactions. As demonstrated in the development of CH-CSLEX-1, creating mouse/human chimeric antibodies represents an effective solution, allowing the establishment of highly specific assays that are not affected by HAMA . These chimeric constructs maintain the target-binding regions of the mouse antibody while replacing the remainder with human antibody sequences, significantly reducing immunogenicity. For CELA1 antibody development, researchers should implement an antibody humanization strategy similar to that planned for mouse CELA1 antibodies, where the antibody will be produced by a contract research organization and thoroughly evaluated for stability and efficacy . Additional approaches include using transgenic silkworms or other expression systems that can produce properly folded chimeric antibodies with appropriate post-translational modifications . When developing immunoassays to monitor therapeutic antibody levels or efficacy, researchers should incorporate sample pre-treatment steps to remove or neutralize HAMA, such as the immunoglobulin purification technique that improved specificity and drug tolerance in similar antibody programs . Verification of reduced HAMA interference should include testing assay performance with samples containing known concentrations of human anti-mouse IgG antibodies (HAMA IgG) and HAMA IgM to ensure robustness under realistic clinical conditions .

What strategies address the challenges of distinguishing between endogenous proteins and therapeutic antibodies in immunogenicity testing?

Distinguishing between endogenous proteins and therapeutic antibodies in immunogenicity testing presents significant technical challenges that require sophisticated methodological approaches. This distinction is particularly challenging for plasma-derived therapeutics like CSL112, where differentiating antibodies against the therapeutic from those recognizing endogenous proteins requires specialized techniques . For CELA1 antibody development, researchers should consider implementing a Sample Pre-treatment Electrochemiluminescence (SPECL) assay approach, which incorporates a protein A extraction step before performing a bridging assay . This methodology has demonstrated improved specificity and drug tolerance in similar contexts. Another effective strategy involves using epitope-specific assays that target unique regions present only in the therapeutic antibody, such as the hinge region of chimeric antibodies or engineered sequences not found in endogenous proteins. Competitive inhibition assays can help determine whether detected antibodies preferentially bind to the therapeutic versus endogenous protein by comparing binding in the presence of excess native protein. Researchers should develop assays with sufficient drug tolerance (at least 0.5 mg/mL according to FDA requirements) to ensure accurate detection of anti-drug antibodies even in the presence of circulating therapeutic . Additionally, characterizing pre-existing antibody profiles before treatment is essential for establishing baselines and identifying treatment-emergent responses. Neutralizing antibody assays that specifically measure the inhibition of therapeutic function provide critical information about the clinical relevance of detected antibodies. By combining multiple complementary assay approaches and carefully designing control experiments, researchers can more accurately distinguish between immune responses to therapeutic antibodies versus endogenous proteins.

How should researchers interpret contradictory results between different anti-CELA1 antibody detection methods?

When researchers encounter contradictory results between different anti-CELA1 antibody detection methods, a systematic, analytical approach is essential to resolve these discrepancies and determine the most reliable findings. First, researchers should conduct a thorough comparative analysis of the methodological differences between the conflicting assays, including antibody format (polyclonal vs. monoclonal), detection platforms, and sample processing techniques. Evidence from immunogenicity testing demonstrates that measurement outcomes can be highly dependent on assay format, with pre-existing or treatment-emergent antibodies detected at varying levels across different methodologies . Researchers should evaluate each assay's validation parameters, particularly focusing on specificity, sensitivity, and drug tolerance levels. For example, the SPECL assay demonstrates sensitivity with a lower limit of quantification below 39 ng/mL and drug tolerance of 0.5 mg/mL, which may differ significantly from other methods . Technical variations in sample handling can substantially impact results, as demonstrated by the improved specificity achieved through immunoglobulin purification steps . To systematically address contradictions, researchers should perform head-to-head comparisons using identical sample sets processed in parallel through each method, followed by correlation analysis similar to the approach that showed good concordance (r = 0.98) between conventional and improved assays in related antibody research . When differences persist, orthogonal methods that measure different aspects of antibody binding or function should be employed. Potential biological explanations, such as antibody heterogeneity, epitope masking, or matrix effects, should be systematically investigated through dilution linearity studies and spike-recovery experiments. Ultimately, researchers should prioritize results from methods with the most robust validation data and highest relevance to the biological or clinical question being addressed.

What emerging technologies show promise for enhancing CELA1 antibody development and testing?

Several emerging technologies show significant promise for enhancing CELA1 antibody development and testing, potentially accelerating the path from laboratory discovery to clinical application. Single B-cell antibody discovery platforms represent a revolutionary approach allowing direct isolation of antigen-specific B cells from immunized animals, bypassing traditional hybridoma technology and enabling more efficient identification of high-affinity antibody candidates. For CELA1 antibodies specifically, CRISPR-Cas9 gene editing technology offers unprecedented possibilities for creating humanized mouse models expressing human CELA1, providing more translatable preclinical testing systems than traditional mouse models . Artificial intelligence and machine learning algorithms are increasingly being applied to antibody design, allowing in silico prediction of antibody structures with optimal binding properties and reduced immunogenicity potential. Next-generation sequencing of antibody repertoires enables comprehensive analysis of immune responses to CELA1, potentially identifying naturally occurring high-affinity antibodies that could serve as templates for therapeutic development. Advanced imaging technologies such as high-resolution cryo-electron microscopy now allow visualization of antibody-antigen complexes at near-atomic resolution, providing crucial structural insights to guide optimization of binding properties. Novel expression systems beyond traditional mammalian cell culture, such as the transgenic silkworm system used for CH-CSLEX-1 production, offer potential advantages in glycosylation patterns and production efficiency . Microfluidic and organ-on-chip technologies enable high-throughput screening of antibody candidates in more physiologically relevant contexts than traditional cell culture. These emerging platforms collectively promise to address key challenges in CELA1 antibody development, including humanization, specificity optimization, and translation of findings from preclinical models to human applications.

How might CELA1 antibody research inform therapeutic approaches for other inflammatory lung diseases?

CELA1 antibody research has significant potential to inform therapeutic approaches for a broad spectrum of inflammatory lung diseases beyond COPD and emphysema, particularly through its insights into elastin remodeling mechanisms. The protective effects observed when blocking CELA1 in multiple types of chronic obstructive pulmonary disease models suggests that elastase-mediated tissue remodeling represents a common pathological pathway across diverse lung conditions . This research establishes a conceptual framework for targeting tissue remodeling processes in other inflammatory lung diseases characterized by extracellular matrix degradation, such as idiopathic pulmonary fibrosis, cystic fibrosis, and bronchiectasis. The methodological approaches developed for CELA1 antibody validation, including the evaluation of elastin remodeling under physical stretch conditions, provide valuable experimental paradigms applicable to studying tissue mechanics in various pathological contexts . Furthermore, the humanization strategies being implemented for CELA1 antibodies offer technical blueprints for developing therapeutic antibodies against other promising targets in lung disease . The immunogenicity assessment approaches refined through CELA1 antibody development, particularly methods addressing drug tolerance and distinguishing therapeutic from endogenous protein responses, have broad applicability across biotherepeutic development programs for respiratory conditions . Insights into the spatial and temporal dynamics of CELA1 activity in lung tissue may inform more precise therapeutic targeting strategies for other proteases involved in lung pathology. Additionally, the potential synergies between CELA1 inhibition and other treatment modalities could inspire combination therapy approaches for complex inflammatory lung conditions that involve multiple pathological mechanisms. As researchers further elucidate the downstream molecular consequences of CELA1 inhibition, this knowledge may reveal novel therapeutic targets and biomarkers relevant to a wide range of inflammatory lung diseases.

What clinical trial design considerations are most important for evaluating CELA1 antibody efficacy in human subjects?

Designing clinical trials to evaluate CELA1 antibody efficacy in human subjects requires careful consideration of multiple factors specific to both the therapeutic mechanism and target patient populations. Based on the current understanding of CELA1's role in elastin remodeling and pulmonary disease, researchers should implement adaptive trial designs that allow for dose refinement based on early biomarker responses, maximizing the chance of identifying effective dosing regimens. Patient selection criteria must be meticulously developed to identify those most likely to benefit from CELA1 inhibition, potentially incorporating biomarkers of elastin degradation, pulmonary function parameters, and genetic factors that influence disease progression. Given the chronic nature of COPD and emphysema, trials should be of sufficient duration to capture meaningful clinical outcomes while incorporating interim analyses of surrogate endpoints such as changes in pulmonary function tests, exercise capacity, and quality of life measures. The selection of appropriate endpoints represents a critical decision point - while forced expiratory volume in one second (FEV1) remains the gold standard for pulmonary function, it may not fully capture the therapeutic effects of targeting elastin remodeling. Therefore, trials should incorporate multiple endpoints including imaging biomarkers of lung architecture, functional measures beyond spirometry, exacerbation rates, and patient-reported outcomes. Safety monitoring must be particularly rigorous, with special attention to potential immunogenicity and its clinical consequences, given the challenges previously identified in distinguishing between antibodies to therapeutic versus endogenous proteins . Implementation of the validated immunogenicity assessment approaches, such as the SPECL assay methodology that demonstrated improved specificity and drug tolerance, will be essential for accurate interpretation of safety signals . Stratification strategies should consider factors such as disease severity, exacerbation history, and potentially genetic variations that might influence response to therapy or disease progression rate.

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.