HCK Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Stored at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method or location. Please consult your local distributor for specific delivery times.
Synonyms
Bmk antibody; Hck 1 antibody; Hck antibody; HCK_HUMAN antibody; Hemopoietic cell kinase antibody; JTK9 antibody; p59-HCK/p60-HCK antibody; p59Hck antibody; p59HCK/p60HCK antibody; p61Hck antibody; Tyrosine protein kinase HCK antibody; Tyrosine-protein kinase HCK antibody
Target Names
HCK
Uniprot No.

Target Background

Function
Hck (Hematopoietic cell kinase) is a non-receptor tyrosine-protein kinase primarily found in hematopoietic cells. It acts as a signal transducer from cell surface receptors, playing a critical role in regulating innate immune responses. This kinase is involved in various cellular functions, including neutrophil, monocyte, macrophage, and mast cell functions, phagocytosis, cell survival and proliferation, cell adhesion, and migration. Hck acts downstream of various receptors, including those that bind the Fc region of immunoglobulins (e.g., FCGR1A and FCGR2A), CSF3R, PLAUR, receptors for IFNG, IL2, IL6, and IL8, and integrins like ITGB1 and ITGB2. During phagocytosis, Hck mediates the mobilization of secretory lysosomes, degranulation, and activation of NADPH oxidase, leading to the respiratory burst. It also participates in the release of inflammatory molecules. Additionally, Hck promotes the reorganization of the actin cytoskeleton and actin polymerization, formation of podosomes and cell protrusions. It inhibits TP73-mediated transcription activation and apoptosis. Hck phosphorylates CBL in response to activation of immunoglobulin gamma Fc region receptors and also phosphorylates ADAM15, BCR, ELMO1, FCGR2A, GAB1, GAB2, RAPGEF1, STAT5B, TP73, VAV1, and WAS.
Gene References Into Functions
  1. HIV infection induces Hck activation. Myeloid cells require Hck for ADAM17 activation and secretion PMID: 29331674
  2. Lysophosphatidylcholines prime polymorphonuclear neutrophil through Hck-dependent activation of PKCdelta, which stimulates PKCgamma, resulting in translocation of phosphorylated p47(phox). PMID: 27531930
  3. HCK represents a novel therapeutic target for MYD88-mutated Waldenstrom macroglobulinemia and activated-B cell diffuse large B-cell lymphoma, potentially extending to other diseases driven by mutated MYD88. PMID: 27143257
  4. Elevated HCK levels correlate with reduced survival in colorectal cancer patients and are associated with an activated macrophages gene signature. PMID: 28399411
  5. Three-dimensional (3D) QSAR pharmacophore models for Hck, based on experimentally known inhibitors, have been generated. A prominent model, Hypo1, displays high correlation coefficient (0.975), low RMS deviation (0.60), and large cost difference (49.31), containing three ring aromatic and one hydrophobic aliphatic feature. PMID: 27485399
  6. Interaction with the Src homology (SH3-SH2) region of Hck structures the HIV-1 Nef dimer, enabling kinase activation and effector recruitment. PMID: 25122770
  7. The interaction between Nef and Hck is crucial for Nef-dependent modulation of viral infectivity. PMID: 24051604
  8. The SRC family tyrosine kinase HCK and the ETS family transcription factors SPIB and EHF regulate transcytosis across a human follicle-associated epithelium model. PMID: 23439650
  9. This specific binding mode allows Hck SH3 to sense a specific non-canonical residue situated in the SH3 RT-loop of the kinase. PMID: 22641034
  10. The activation of Hck, Lyn, and c-Src by Nef is highly conserved among all major clades of HIV-1. PMID: 22393415
  11. Significant differences in the genotype and allele distribution of the -627 G/T polymorphism in the Hck gene were observed between cases and controls. PMID: 22185326
  12. Hck activation at the Golgi apparatus causes the HIV-1 Nef-induced c-Fms proto-oncogene N-glycosylation defect. PMID: 21567396
  13. Loss of HCK is associated with acute promyelocytic leukemia. PMID: 21993313
  14. Hck plays a key role in LPS/TLR4-induced TNF and IL-6 production. PMID: 22021612
  15. Hck acts as a key regulator controlling gene expression in alternatively activated monocytes/macrophages. PMID: 21878628
  16. Data indicate that the structures and relative orientations of the SH2 and SH3 domains down-regulate Hck. PMID: 20810664
  17. Molecular recognition mechanisms between the human haemopoietic cell kinase Src homology domain 3 and ALG-2-interacting protein X have been identified and biophysically assessed. PMID: 20670214
  18. BSS-SAXS reconstruction is used to reveal the structural organization of Hck in solution and the different shifts in the equilibrium population of assembly states upon binding of different signaling peptides. PMID: 20798061
  19. PKR and Hck are critical for DON-induced ribosomal recruitment of p38, its subsequent phosphorylation, and ultimately, p38-driven proinflammatory cytokine expression. PMID: 20181660
  20. Nef participates in HIV-1-induced multinucleated giant cells formation via a p61Hck- and lysosomal enzyme-dependent pathway. PMID: 20488787
  21. SH3-dependent stimulation of Src-family kinase autophosphorylation without tail release from the SH2 domain occurs in vivo. PMID: 11976726
  22. The interaction of the Bcr-Abl tyrosine kinase with this protein is mediated by multiple binding domains. PMID: 12592324
  23. CSF-induced and HIV-1-mediated regulation of Hck and C/EBPbeta explain the heterogeneous susceptibility of tissue macrophages to HIV-1 infection. PMID: 12900520
  24. SRC kinases LYN & HCK enable engaged b2 integrins to form focal-adhesion-like structures, essential for stable, shear-resistant PMN adhesion. SRC-dependent outside-in signaling is crucial for integrin adhesiveness triggered by classical chemoattractants like IL-8. PMID: 14969582
  25. Gab2 docking proteins play a role in IL-6-induced proliferation and survival of multiple myeloma cells. PMID: 15010462
  26. In humans, the cytoplasmic domain of ADAM15v2 interacts strongly with Lck and Hck and regulates leukocyte function. PMID: 15263807
  27. HIV-1 Nef interferes with M-CSF receptor signaling through Hck activation, thus inhibiting M-CSF functions in monocytes/macrophages. PMID: 15626739
  28. Data supports the existence of multiple active conformations of Src family member Hck kinase that potentially generate unique downstream signals. PMID: 16210316
  29. The free energy surface reveals that the N-terminal end of HCK acts as a reversible two-state conformational switch, coupling the catalytic domain to the regulatory modules. PMID: 16271895
  30. Data suggests that the insertion/deletion polymorphism could be a functional polymorphism of the Hck gene, potentially contributing to COPD pathogenesis and modifying COPD-related phenotypes. PMID: 17024369
  31. The structure of the HckSH3:PD1 complex reveals novel features of SH3 ligand binding and provides new insights into the structural basics of SH3-ligand interactions. PMID: 17141806
  32. p73 has been identified as a novel substrate and interacting partner of Hck. It regulates p73 through mechanisms dependent on either catalytic activity or protein interaction domains. PMID: 17535448
  33. Hematopoietic cell kinase (hct) phosphorylates fems-like tyrosine kinase 3(FLT3) in the JM region, inhibiting its maturation. PMID: 17668209
  34. Nef disrupts the intracellular maturation and trafficking of nascent Fms through a unique mechanism that requires both the activation of Hck and the aberrant spatial regulation of the active Hck. PMID: 17893228
  35. Hck plays a non-redundant function as a key downstream signaling partner for Bcr-Abl and may represent a potential drug target in CML. PMID: 18794796
  36. HCK and BIN1 play critical roles in AHI-1-mediated leukemic transformation of cutaneous T-Cell Lymphoma. PMID: 19211505
  37. This finding establishes an intriguing link between the pathogenesis of Nef and the emerging concept that Golgi-localized Src kinases regulate Golgi function. PMID: 19585521
  38. Alternate use of a non-AUG (CUG), and an in-frame, downstream AUG translation initiation codon, results in the production of 2 isoforms in mouse and human. PMID: 1875927
  39. Alternate use of a non-AUG (CUG), and an in-frame, downstream AUG translation initiation codon, results in the production of two isoforms with different subcellular localization. PMID: 10967098
  40. A dominant negative form of Hck, in an interaction that is SH3 domain dependent, blocks HIV-1 Nef induced MHC class I downregulation. PMID: 11500821

Show More

Hide All

Database Links

HGNC: 4840

OMIM: 142370

KEGG: hsa:3055

STRING: 9606.ENSP00000365012

UniGene: Hs.655210

Involvement In Disease
Aberrant activation of HCK by HIV-1 protein Nef enhances HIV-1 replication and contributes to HIV-1 pathogenicity.; DISEASE: Note=Aberrant activation of HCK, e.g. by the BCR-ABL fusion protein, promotes cancer cell proliferation.
Protein Families
Protein kinase superfamily, Tyr protein kinase family, SRC subfamily
Subcellular Location
[Isoform 1]: Lysosome. Membrane; Lipid-anchor. Cell projection, podosome membrane; Lipid-anchor. Cytoplasm, cytosol. Note=Associated with specialized secretory lysosomes called azurophil granules. At least half of this isoform is found in the cytoplasm, some of this fraction is myristoylated.; [Isoform 2]: Cell membrane; Lipid-anchor. Membrane, caveola; Lipid-anchor. Cell junction, focal adhesion. Cytoplasm, cytoskeleton. Golgi apparatus. Cytoplasmic vesicle. Lysosome. Nucleus. Note=20% of this isoform is associated with caveolae. Localization at the cell membrane and at caveolae requires palmitoylation at Cys-3. Colocalizes with the actin cytoskeleton at focal adhesions.; Cytoplasmic vesicle, secretory vesicle. Cytoplasm, cytosol.
Tissue Specificity
Detected in monocytes and neutrophils (at protein level). Expressed predominantly in cells of the myeloid and B-lymphoid lineages. Highly expressed in granulocytes. Detected in tonsil.

Q&A

What is HCK and why is it important in research?

HCK (hematopoietic cell kinase) is a member of the SRC family of cytoplasmic tyrosine kinases expressed primarily in hematopoietic cells of myeloid and B-lymphocyte lineages. It plays crucial roles in cell signaling pathways related to immune responses, inflammation, and cancer development. HCK has emerged as an important research target because excessive HCK activation is associated with several types of leukemia and solid malignancies, including breast and colon cancer, where it correlates with decreased patient survival rates . Furthermore, HCK enhances secretion of growth factors and pro-inflammatory cytokines from myeloid cells and promotes macrophage polarization toward a tumor-promoting phenotype, making it a valuable target for both basic research and therapeutic development .

What are the structural characteristics of the HCK protein that researchers should be aware of?

HCK exists in multiple isoforms, with the primary protein reported to be approximately 59.6 kilodaltons in mass . Researchers should be aware that alternate translation initiation site usage, including a non-AUG (CUG) codon, results in the production of two distinct isoforms: p59Hck and p61Hck . These isoforms demonstrate different subcellular localization patterns, which is critical knowledge when designing immunofluorescence or subcellular fractionation experiments. The protein contains a catalytic domain and regulatory regions that control its activation state. Most importantly for antibody-based research, HCK contains specific phosphorylation sites, particularly Tyr411 (pTyr411), which serves as a critical marker of HCK activation and can be detected using phospho-specific antibodies in techniques such as Phosflow analysis .

How should researchers select the most appropriate HCK antibody for their specific experimental applications?

When selecting an HCK antibody, researchers should consider several critical factors:

  • Target epitope: Determine whether you need an antibody targeting the N-terminal region (for detecting all isoforms), C-terminal region, or a specific phosphorylation site like pTyr411 for activation studies .

  • Application compatibility: Verify the antibody has been validated for your specific application. For example, search results show antibodies validated for Western blot (WB), immunohistochemistry (IHC-p), immunofluorescence (IF), immunoprecipitation (IP), ELISA, and flow cytometry .

  • Species reactivity: Confirm cross-reactivity with your experimental species. Available antibodies show reactivity with human, mouse, rat, and sometimes additional species like rabbit and dog .

  • Clonality: Choose between monoclonal antibodies for high specificity to a single epitope (like the mouse monoclonal IgG1 from Santa Cruz Biotechnology) or polyclonal antibodies for broader epitope recognition .

  • Validation data: Review the supplier's validation data, including published citations, figures showing expected bands in Western blots, or appropriate staining patterns in IHC/IF applications .

What validation steps should be performed to ensure HCK antibody specificity?

To ensure antibody specificity, researchers should implement the following validation steps:

  • Positive and negative controls: Use cell lines with known HCK expression levels (high in myeloid lineage cells) versus those with minimal expression.

  • Knockdown verification: Implement HCK-specific siRNA or antisense oligonucleotides to confirm signal reduction. As reported in the literature, antisense ODN sequences like 5′-GAA CTT GGA CTT CAT GCA CCC-3′ have been successfully used to inhibit endogenous HCK expression .

  • Phospho-specificity testing: For phospho-specific antibodies, treat samples with phosphatase to verify signal loss.

  • Cross-reactivity assessment: Test against related SRC family kinases to ensure specificity for HCK.

  • Comparison across techniques: Validate findings using multiple methods (e.g., Western blot plus immunofluorescence).

  • Recombinant protein: Use purified recombinant HCK protein as a positive control for antibody testing.

What are the optimal protocols for using HCK antibodies in flow cytometry?

For optimal flow cytometry with HCK antibodies, follow these methodological guidelines:

  • Cell preparation and fixation:

    • Harvest 1 × 10^6 cells per sample

    • Fix cells with BD Phosflow Fix Buffer I at 37°C for 10 minutes

    • Centrifuge at 300g for 5 minutes and wash twice with Phosflow Perm/Wash Buffer I

  • Staining procedure:

    • For cell lines: Stain with HCK (pTyr 411) antibody (or other HCK antibodies)

    • For primary cells: Co-stain with lineage markers (e.g., CD20–APC-Cy7 for B cells)

    • Incubate in the dark for 30 minutes at room temperature

    • Wash three times with BD Phosflow Perm/Wash Buffer I

    • Apply secondary antibody (e.g., anti-rabbit IgG DyLight-649) if using unconjugated primary antibodies

    • Incubate for an additional 20 minutes and wash twice

  • Data acquisition and analysis:

    • Acquire minimum of 10,000 events using a flow cytometer (e.g., BD FACSCanto II)

    • For primary samples, gate on the appropriate lineage marker before analyzing HCK expression

    • Compare to appropriate isotype controls to determine positive staining

How can researchers effectively use HCK antibodies in the context of studying cancer pathways?

To effectively study HCK in cancer pathways, researchers should implement these methodological approaches:

  • Multi-parameter signaling analysis:

    • Use phospho-specific HCK antibodies (pTyr411) alongside other pathway markers (e.g., pBTK, pERK, pAKT) to map signaling networks

    • Implement Phosflow or multiplex Western blotting to assess pathway activation following treatment with inhibitors

  • HCK activity assessment in patient samples:

    • For blood cancers like Waldenström macroglobulinemia (WM) or acute myeloid leukemia, use CD19 or other lineage markers with HCK antibodies to analyze malignant populations

    • Compare HCK activation levels between healthy donor cells and patient samples to establish baseline differences

  • Inhibitor efficacy studies:

    • Use HCK antibodies in cell viability assays following HCK inhibitor treatment

    • Perform target engagement studies using pull-down experiments with biotinylated inhibitors to confirm binding to HCK

    • Assess downstream signaling changes with multi-parameter analysis

  • Genetic manipulation approaches:

    • Combine HCK knockdown (using shRNA or CRISPR) with antibody-based detection to confirm specificity

    • Overexpress wild-type or mutant HCK (e.g., gatekeeper mutant HCK T333M) to study resistance mechanisms

What are the best practices for using HCK antibodies in immunohistochemistry?

For optimal immunohistochemistry results with HCK antibodies:

  • Tissue preparation:

    • Use formalin-fixed, paraffin-embedded (FFPE) tissue sections (4-6 μm thickness)

    • Include positive control tissues with known HCK expression (e.g., spleen, which shows strong HCK expression)

  • Antigen retrieval:

    • Perform heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)

    • Optimize retrieval time (typically 15-20 minutes) for your specific tissue and antibody

  • Blocking and antibody incubation:

    • Block endogenous peroxidase activity with 3% H₂O₂

    • Apply protein block to reduce background

    • Use optimized antibody dilution (typically 1:100 to 1:500 for commercial antibodies)

    • Incubate primary antibody overnight at 4°C or 1-2 hours at room temperature

  • Detection system:

    • Use a sensitive detection system compatible with your primary antibody species (e.g., HRP-polymer)

    • Develop with DAB and counterstain with hematoxylin

    • Include negative controls (primary antibody omission and isotype control)

  • Analysis considerations:

    • Evaluate both intensity and distribution of staining

    • Consider subcellular localization (cytoplasmic, membrane, nuclear)

    • Quantify using appropriate scoring systems (H-score, Allred score) for comparative studies

How can researchers effectively study HCK's role in the tumor microenvironment?

To investigate HCK's role in the tumor microenvironment, implement these advanced methodologies:

  • Multiplex immunofluorescence:

    • Co-stain for HCK along with markers of different immune cell populations (CD68+ macrophages, CD11b+ myeloid cells)

    • Use phospho-specific HCK antibodies to identify activated cells within the microenvironment

    • Quantify spatial relationships between HCK-expressing cells and tumor cells

  • 3D co-culture systems:

    • Establish co-cultures of tumor cells with macrophages or other immune cells

    • Monitor HCK activation status using phospho-specific antibodies during co-culture

    • Assess podosome formation and ECM degradation, which HCK has been shown to regulate in macrophages

  • In vivo models:

    • Use tissue from mouse models (like the HCK^CA mice that develop lung adenocarcinomas as a consequence of lung inflammation)

    • Compare HCK expression and activation in tumor-associated macrophages versus normal tissue macrophages

    • Correlate HCK expression with markers of tumor progression

  • Functional readouts:

    • Measure cytokine production (particularly TNFα) in relation to HCK activation status

    • Assess the impact of HCK inhibition on immune cell recruitment and polarization

    • Evaluate changes in extracellular matrix degradation, which HCK-expressing macrophages facilitate

What are the methodological considerations when using HCK antibodies to study resistance mechanisms to targeted therapies?

When investigating HCK-related resistance mechanisms to targeted therapies:

  • Target engagement studies:

    • Use biotinylated versions of inhibitors (as done with ibrutinib and CC-292) to pull down HCK and confirm direct binding

    • Perform KiNativ profiling to determine inhibitor engagement in living cells by measuring protection from ATP-biotin probe labeling

  • Resistance model development:

    • Generate cell lines expressing inhibitor-resistant HCK mutants (e.g., HCK T333M gatekeeper mutant)

    • Compare antibody-detected phosphorylation patterns between wild-type and mutant HCK-expressing cells

    • Monitor changes in downstream signaling networks

  • Combination therapy assessment:

    • Use phospho-specific HCK antibodies to monitor HCK activation status during treatment with multiple inhibitors

    • Track correlation between HCK phosphorylation and cell survival markers

  • Patient sample analysis:

    • Compare HCK activation in samples from treatment-naïve versus relapsed/refractory patients

    • Correlate HCK activation with clinical outcomes and treatment response

  • Pathway crosstalk investigation:

    • Study HCK's role in facilitating TLR/BCR crosstalk through SYK activation in response to mutated MYD88

    • Use multi-parameter antibody panels to delineate compensatory signaling pathways

How can HCK antibodies be used to understand the role of HCK in normal versus malignant hematopoiesis?

To investigate HCK's differential roles in normal versus malignant hematopoiesis:

  • Cell subset analysis:

    • Compare HCK expression and activation across different hematopoietic cell lineages using flow cytometry

    • Create a baseline expression map in healthy donor B cells, memory B cells, and plasma cells

    • Compare to malignant counterparts in diseases like Waldenström macroglobulinemia

  • Signaling network analysis:

    • Use phospho-specific HCK antibodies to map activation patterns in response to specific stimuli (e.g., LPS, IFNγ)

    • Compare signaling responses between normal and malignant cells

    • Investigate the relationship between HCK activation and downstream effects on AKT, ERK, and BTK

  • Transcriptional regulation studies:

    • Combine HCK protein analysis with transcriptional assessment (e.g., qRT-PCR)

    • Study how HCK transcription is influenced by factors like IL-6 or MYD88 mutations

    • Compare regulatory mechanisms between normal and malignant cells

  • Differentiation studies:

    • Monitor HCK expression and activation during normal hematopoietic differentiation

    • Assess changes in cellular localization of HCK during differentiation using immunofluorescence

    • Evaluate the impact of HCK inhibition or knockdown on differentiation potential

What are common challenges when working with HCK antibodies and how can they be overcome?

Common challenges with HCK antibodies and their solutions include:

ChallengePotential Solutions
Non-specific binding- Optimize antibody concentration
- Use additional blocking steps (e.g., with 5% BSA)
- Try different antibody clones
- Include proper negative controls (isotype, secondary-only)
Weak signal in Western blots- Increase protein loading (50-100 μg total protein)
- Optimize antibody concentration and incubation time
- Try enhanced chemiluminescence (ECL) detection systems
- Consider different extraction buffers to improve HCK solubilization
Poor reproducibility- Standardize cell culture conditions and treatments
- Use consistent protein extraction methods
- Aliquot antibodies to avoid freeze-thaw cycles
- Include internal loading controls
Cross-reactivity with other SFK members- Use antibodies targeting unique regions of HCK
- Validate with HCK knockdown experiments
- Consider immunoprecipitation followed by Western blotting
Inconsistent flow cytometry results- Optimize fixation and permeabilization protocols
- Use fresh samples when possible
- Include fluorescence-minus-one (FMO) controls
- Establish standardized gating strategies

How can researchers optimize experimental conditions for detecting HCK activation in primary patient samples?

For optimal detection of HCK activation in primary patient samples:

  • Sample handling optimization:

    • Process samples immediately after collection to preserve phosphorylation status

    • If immediate processing is not possible, use phosphorylation preservation reagents

    • For bone marrow samples, use gentle RBC lysis buffers to maintain cell integrity

  • Phosflow protocol refinement:

    • Optimize fixation time (8-12 minutes at 37°C) with BD Phosflow Fix Buffer I

    • Use gentle permeabilization conditions to maintain epitope accessibility

    • For bone marrow mononuclear cells, include CD19–APC-cy7 antibody for identifying B cells

  • Multi-parameter analysis:

    • Include viability dyes to exclude dead cells

    • Use lineage markers to identify specific populations of interest

    • Incorporate markers of activation status (e.g., CD86) to correlate with HCK activation

  • Signal amplification strategies:

    • Consider signal amplification systems for samples with low HCK expression

    • Optimize secondary antibody concentration and incubation time

    • Use high-sensitivity detection reagents

  • Standardization across samples:

    • Include internal controls (healthy donor samples) in each experimental run

    • Use reference standards for flow cytometry calibration

    • Normalize data to account for inter-sample variability

How might single-cell approaches advance our understanding of HCK biology?

Single-cell approaches offer transformative potential for HCK research:

  • Single-cell phospho-profiling:

    • Apply mass cytometry (CyTOF) with HCK antibodies to simultaneously analyze multiple phosphorylation events at single-cell resolution

    • Identify rare subpopulations with unique HCK activation signatures

    • Map signaling heterogeneity within seemingly homogeneous cell populations

  • Spatial transcriptomics integration:

    • Combine HCK protein detection with spatial transcriptomics

    • Correlate HCK protein levels with transcriptional networks in individual cells

    • Map HCK-expressing cells within tissue microenvironments

  • Live-cell imaging approaches:

    • Develop HCK biosensors for real-time activation monitoring

    • Track HCK translocation and activation dynamics in response to stimuli

    • Correlate temporal activation patterns with functional outcomes

  • Single-cell multi-omics:

    • Integrate HCK protein detection with single-cell RNA-seq and ATAC-seq

    • Identify transcriptional and epigenetic regulators of HCK in individual cells

    • Discover novel regulatory mechanisms specific to cell subsets

What are the emerging applications of HCK antibodies in precision medicine approaches?

Emerging precision medicine applications for HCK antibodies include:

  • Patient stratification biomarkers:

    • Develop standardized protocols for measuring HCK activation in patient samples

    • Correlate HCK activation profiles with treatment response in clinical trials

    • Identify patient subgroups that might benefit from HCK-targeted therapies

  • Resistance mechanism identification:

    • Monitor changes in HCK activation during treatment to detect emerging resistance

    • Identify compensatory signaling pathways activated upon HCK inhibition

    • Develop rational combination therapy approaches based on HCK activation status

  • Therapeutic antibody development:

    • Design antibodies that specifically target HCK for therapeutic applications

    • Develop antibody-drug conjugates targeting HCK-expressing cells

    • Create bispecific antibodies linking HCK-expressing cells to immune effectors

  • Ex vivo drug sensitivity testing:

    • Use HCK activation status as a readout for ex vivo drug sensitivity testing

    • Develop standardized assays for clinical implementation

    • Correlate ex vivo findings with in vivo treatment responses

  • Liquid biopsy applications:

    • Detect HCK in circulating tumor cells or exosomes as biomarkers

    • Monitor treatment response through changes in HCK activation in accessible samples

    • Develop minimally invasive methods for longitudinal patient monitoring

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.