CALS10 Antibody

Shipped with Ice Packs
In Stock

Description

CAL10 (Anti-PD-L1 Antibody)

Structure & Function:

  • Target: Programmed Death-Ligand 1 (PD-L1), an immune checkpoint protein overexpressed in many cancers .

  • Format: Rabbit monoclonal IgG antibody .

  • Mechanism: Blocks PD-L1/PD-1 interaction, restoring T-cell-mediated tumor immunity .

Key Applications:

ApplicationValidationSensitivity/Specificity
Immunohistochemistry (IHC)Multi-tissue microarray (TMA) validatedSpecificity confirmed via CD274-knockout cell lines
Western BlottingRecombinant format ensures batch consistencyDetects PD-L1 in human samples
Multiplex ImagingCompatible with Leica BOND® platformsValidated for co-staining with CD68

Research Findings:

  • Demonstrates high specificity for PD-L1 in formalin-fixed paraffin-embedded tissues .

  • Used in >60 studies investigating PD-L1 expression patterns in non-small cell lung cancer (NSCLC) and melanoma .

CAN10 (IL1RAP-Targeting Antibody)

Structure & Function:

  • Target: IL-1 Receptor Accessory Protein (IL1RAP), critical in IL-1, IL-33, and IL-36 signaling .

  • Format: Humanized IgG1 with Fc engineering for prolonged half-life .

Clinical Data (Phase 1 Trial):

ParameterResult
Safety (Single Dose)No severe adverse events in 68 participants
Biomarker EfficacyComplete IL-1/IL-36 signaling blockade for ≥7 days post-dose
Therapeutic PotentialPlanned Phase 2 trials for hidradenitis suppurativa (2025)

Mechanistic Insights:

  • Dual inhibition of IL-1 and IL-36 pathways distinguishes CAN10 from monoclonal antibodies targeting single cytokines .

5C10 (Anti-CaSR Antibody)

Structure & Function:

  • Target: Calcium-Sensing Receptor (CaSR), a G protein-coupled receptor regulating calcium homeostasis .

  • Format: Mouse monoclonal IgG2a .

Experimental Applications:

TechniqueValidationSample Type
Flow Cytometry80% methanol-fixed SH-SY5Y cellsHuman, Mouse
IHC (Frozen Tissue)Stomach, Kidney, BrainSpecies cross-reactivity confirmed

Research Utility:

  • Critical for studying CaSR’s role in parathyroid disorders and neurological calcium signaling .

Nomenclature Analysis

The term "CALS10" may represent:

  1. Typographical error for CAL10, CAN10, or 5C10.

  2. Proprietary compound not yet disclosed in public databases.

  3. Emerging antibody in preclinical development (no published data identified).

Comparative Table: Antibodies with Similar Nomenclature

AntibodyTargetTypeStageKey Feature
CAL10 PD-L1Rabbit IgGResearch UseMultiplex IHC validation
CAN10 IL1RAPHumanized IgG1Phase 2 (2025)Dual IL-1/IL-36 blockade
5C10 CaSRMouse IgG2aPreclinicalCross-species reactivity

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
CALS10 antibody; GSL8 antibody; At2g36850 antibody; T1J8.3Callose synthase 10 antibody; EC 2.4.1.34 antibody; 1,3-beta-glucan synthase antibody; Protein CHORUS antibody; Protein GLUCAN SYNTHASE-LIKE 8 antibody
Target Names
CALS10
Uniprot No.

Target Background

Function
CALS10 antibody plays a crucial role in both sporophytic and gametophytic development. It is essential for normal plant development and the proper accumulation of callose, a polysaccharide, at cell plates, cell walls, and plasmodesmata. During pollen formation, CALS10 is required for the entry of microspores into mitosis. Throughout plant growth and development, callose serves as a temporary component of the cell plate in dividing cells, a major constituent of pollen mother cell walls and pollen tubes, and a structural element of plasmodesmatal canals. This antibody is vital for proper cell division and tissue patterning across all plant organs, including stomatal development.
Gene References Into Functions
  1. GSL8 and SMT2 proteins are essential for maintaining ploidy consistency within the sexual reproduction system of Arabidopsis. PMID: 23404886
  2. GSL8 is indispensable for the completion of plant cytokinesis. PMID: 19067977
  3. Loss-of-function mutations in Arabidopsis CHORUS (GLUCAN SYNTHASE-LIKE 8) disrupt cell-to-cell connectivity and stomatal patterning. PMID: 20430748
  4. GSL8 is required for both patterning and cytokinesis during Arabidopsis development. PMID: 19286936
Database Links

KEGG: ath:AT2G36850

STRING: 3702.AT2G36850.1

UniGene: At.37496

Protein Families
Glycosyltransferase 48 family
Subcellular Location
Cell membrane; Multi-pass membrane protein.

Q&A

What is Kallikrein 10 (KLK10) and why is it significant in cancer research?

Kallikrein 10 (KLK10), also known as Normal epithelial cell-specific 1 (NES1) or Protease serine-like 1 (PRSSL1), is a serine protease that plays a tumor-suppressor role in breast and prostate cancer. Its involvement with proteins like KLK3 (PSA) in prostate cancer highlights its potential utility in therapeutic strategies, especially concerning hormone-dependent tumors . KLK10 is part of the kallikrein family of proteases, which are involved in numerous physiological and pathological processes, making them important targets for cancer research.

What are the optimal experimental conditions for using KLK10 antibodies in Western blot applications?

When using KLK10 antibodies for Western blot applications, researchers should implement the following optimized protocol: use 5% NFDM/TBST as blocking and diluting buffer, with a dilution ratio of 1/1000 for the primary antibody . For secondary antibody, Goat Anti-Rabbit IgG H&L (HRP) at 1/50000 dilution has proven effective. The predicted molecular weight of KLK10 is 30 kDa, which aligns with the observed band size in Western blot experiments . Validation studies showing positive expression in NIH:OVCAR-3 (human ovary adenocarcinoma epithelial cell) and negative expression in MCF7 (human breast adenocarcinoma epithelial cell) provide useful positive and negative controls.

How do you differentiate between non-specific binding and true KLK10 detection in immunoassays?

To differentiate between non-specific binding and true KLK10 detection, researchers should implement several validation strategies. First, include appropriate negative controls such as MCF7 cells, which have been documented to lack KLK10 expression (PMID: 11691827) . Second, verify that the observed band matches the expected molecular weight of 30 kDa. Third, perform antibody validation using knockdown or knockout models where available. Finally, use multiple antibodies against different epitopes of KLK10 to confirm specificity. Cross-validation with complementary methods such as mass spectrometry or quantitative PCR provides additional confidence in antibody specificity.

What are the critical considerations when designing experiments to study KLK10's tumor suppressor function?

When designing experiments to study KLK10's tumor suppressor function, researchers should consider several critical factors. First, select appropriate cell models that reflect the tissue-specific context where KLK10 functions as a tumor suppressor, particularly in breast and prostate cancer contexts . Second, implement both gain-of-function (overexpression) and loss-of-function (knockdown/knockout) approaches to establish causality. Third, assess multiple cancer hallmarks including proliferation, apoptosis, migration, invasion, and anchorage-independent growth. Fourth, investigate interactions with known cancer pathways, including potential interactions with other kallikrein family members. Finally, validate findings in patient-derived samples and consider the impact of KLK10 isoforms and post-translational modifications on its tumor suppressor activity.

How can researchers effectively compare different anti-KLK10 monoclonal antibodies for therapeutic development?

Effective comparison of anti-KLK10 monoclonal antibodies for therapeutic development requires systematic evaluation of multiple parameters. Researchers should assess:

  • Binding affinity using surface plasmon resonance or biolayer interferometry

  • Epitope specificity through epitope mapping techniques

  • Functional effects on KLK10 enzymatic activity

  • Ability to recognize KLK10 in different conformational states

  • Cross-reactivity with other kallikrein family members

  • Pharmacokinetic properties including half-life and clearance rates

  • Ability to induce antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC)

Drawing from experience with other therapeutic antibodies, researchers should consider the importance of antibody format (e.g., full IgG vs. fragments), isotype selection, and potential glycoengineering to optimize effector functions .

What pharmacokinetic parameters should be evaluated when developing KLK10-targeting antibodies for cancer therapy?

When developing KLK10-targeting antibodies for cancer therapy, researchers should evaluate several key pharmacokinetic parameters:

ParameterSignificanceEvaluation Methods
Binding affinityDetermines potencySurface plasmon resonance, ELISA
Half-lifeDuration of effectIn vivo clearance studies
Volume of distributionTissue penetrationPK modeling, imaging studies
Target-mediated drug dispositionAffects dosingDose-dependent PK studies
ImmunogenicityLong-term efficacyAnti-drug antibody assays

Researchers should be aware that target-mediated clearance can significantly affect antibody pharmacokinetics, as demonstrated with other therapeutic antibodies . Dose-dependent non-linear pharmacokinetics may be observed if significant target-mediated clearance occurs, requiring careful dose selection and scheduling.

How might circulating versus tissue-bound KLK10 affect antibody efficacy in vivo?

The distribution between circulating and tissue-bound KLK10 can significantly impact antibody efficacy in vivo. Drawing parallels from studies with other therapeutic antibodies, researchers should consider:

  • Soluble target proteins can act as decoys, reducing antibody availability to reach tissue-bound targets

  • Antibodies with different binding properties may exhibit vastly different in vivo efficacy despite similar in vitro potency

  • Target accessibility in different tissue compartments affects therapeutic outcomes

Researchers should quantify both soluble and tissue-bound KLK10 in disease models and consider developing antibodies that preferentially bind to the disease-relevant form. Similar to findings with anti-CXCL10 antibodies, an antibody's ability to recognize KLK10 in its biologically active context (potentially bound to interacting proteins or cell surfaces) may be more important than its absolute binding affinity measured in solution .

What are the most effective strategies for optimizing immunohistochemical detection of KLK10 in clinical samples?

Optimizing immunohistochemical (IHC) detection of KLK10 in clinical samples requires careful attention to several factors:

  • Tissue fixation and processing: Standardize fixation times and buffer compositions to maintain KLK10 antigenicity

  • Antigen retrieval: Test multiple methods (heat-induced vs. enzymatic) and pH conditions

  • Antibody validation: Use positive and negative control tissues with known KLK10 expression patterns

  • Signal amplification: Compare different detection systems for optimal signal-to-noise ratio

  • Quantification: Implement digital pathology tools for objective scoring

Researchers should validate IHC findings with orthogonal methods such as in situ hybridization or laser capture microdissection followed by proteomic analysis. For clinical samples, correlation with patient outcomes is essential to establish the biological and clinical relevance of KLK10 expression patterns.

How can researchers address batch-to-batch variability in KLK10 antibody performance?

To address batch-to-batch variability in KLK10 antibody performance, researchers should implement a systematic quality control workflow:

  • Maintain reference samples with known KLK10 expression for comparative testing of new antibody batches

  • Perform side-by-side testing of old and new batches on identical samples

  • Document lot-specific optimal working dilutions and conditions

  • Quantify binding affinity of each batch using ELISA or surface plasmon resonance

  • Maintain detailed records of antibody performance metrics for longitudinal comparison

When possible, use recombinant monoclonal antibodies like EPR23573-107 , which offer greater consistency compared to polyclonal antibodies. For critical long-term studies, consider purchasing sufficient quantities of a single lot or developing in-house monoclonal antibody production capabilities.

What are the potential approaches for developing KLK10-targeting therapeutic antibodies for cancer treatment?

Several approaches show promise for developing KLK10-targeting therapeutic antibodies:

  • Naked monoclonal antibodies that modulate KLK10 activity or induce ADCC/CDC

  • Antibody-drug conjugates (ADCs) linking anti-KLK10 antibodies to cytotoxic payloads

  • Bispecific antibodies targeting both KLK10 and immune effector cells

  • CAR-T cell therapy utilizing KLK10-specific recognition domains

Drawing from experiences with other therapeutic antibodies in cancer, researchers should consider engineering antibodies with optimal Fc domain properties for enhanced immune effector functions . Additionally, understanding the expression pattern of KLK10 in normal tissues is crucial for predicting potential on-target, off-tumor toxicities.

How can researchers integrate KLK10 antibody-based assays with other biomarkers for improved cancer diagnostic applications?

To integrate KLK10 antibody-based assays with other biomarkers for improved cancer diagnostics, researchers should:

  • Develop multiplexed assays combining KLK10 with established cancer biomarkers

  • Implement machine learning algorithms to identify optimal biomarker combinations

  • Validate diagnostic panels in large, diverse patient cohorts

  • Correlate biomarker expression with treatment response and clinical outcomes

  • Consider the biological context of KLK10 in relation to other kallikreins and proteases

Since KLK10's involvement with proteins like KLK3 (PSA) in prostate cancer has been documented , researchers should explore the added diagnostic value of KLK10 in combination with established biomarkers. Longitudinal studies tracking changes in KLK10 levels during disease progression and treatment response would provide valuable insights into its utility as a dynamic biomarker.

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.