CKL4 Antibody

Shipped with Ice Packs
In Stock

Description

Target Overview: CKAP4 in Cancer Biology

Cytoskeleton-associated protein 4 (CKAP4) is a transmembrane receptor that binds Dickkopf 1 (DKK1), a secreted glycoprotein. The DKK1-CKAP4 signaling axis promotes tumor growth and metastasis in pancreatic, lung, esophageal, and liver cancers . Elevated serum levels of CKAP4 and DKK1 correlate with poor prognosis in these cancers .

Development of Anti-CKAP4 Antibodies

Researchers engineered humanized monoclonal antibodies against CKAP4 to block DKK1-CKAP4 signaling. Key milestones include:

Preclinical Development

  • Mouse-derived antibody (3F11-2B10):

    • Inhibited DKK1 binding to CKAP4 .

    • Reduced AKT pathway activation and tumor growth in xenograft models .

Humanization Process

  • Hv1Lt1 and Hv2Lt1:

    • Derived from 3F11-2B10 via CDR grafting onto human IgG frameworks .

    • Demonstrated superior binding affinity compared to the murine parent antibody:

AntibodyK<sub>D</sub> (nM)Target Specificity
3F11-2B1014.5Human, canine, murine CKAP4
Hv1Lt10.76Cross-reactive with multiple species
Hv2Lt10.05High affinity for CKAP4 ECD

Source: Biacore assays and immunoprecipitation data .

Mechanism of Action

Anti-CKAP4 antibodies exert antitumor effects through:

  1. Blockade of DKK1-CKAP4 Interaction:

    • Prevents activation of AKT/mTOR signaling, reducing cancer cell proliferation .

  2. Immune Modulation:

    • Increases cytotoxic T-cell infiltration into tumors .

  3. Synergy with Chemotherapy:

    • Enhanced tumor suppression when combined with gemcitabine or paclitaxel .

Pancreatic Cancer

  • Orthotopic mouse models:

    • Hv1Lt1 monotherapy reduced tumor volume by 65% vs. controls .

    • Combination with gemcitabine achieved 85% tumor suppression .

Immune Profiling

  • Post-treatment tumors showed:

    • ↑ CD8<sup>+</sup> T cells

    • ↓ Regulatory T cells (Tregs)

Clinical Implications

  • Diagnostic Potential: Serum CKAP4 levels may serve as a biomarker for patient stratification .

  • Therapeutic Advantages:

    • Targets both tumor cells and immunosuppressive microenvironments.

    • Minimal off-target effects due to CKAP4’s restricted expression in healthy tissues .

Ongoing Challenges

  • Humanization Trade-offs: While Hv1Lt1 retained high affinity, Hv2Lt1’s ultra-high affinity may increase immunogenicity risks .

  • Combination Strategies: Optimal dosing schedules with chemotherapy require further validation .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (Made-to-order)
Synonyms
CKL4 antibody; At4g28860 antibody; F16A16.30Casein kinase 1-like protein 4 antibody; EC 2.7.11.1 antibody; Protein CASEIN KINASE I-LIKE 4 antibody; CK1.4 antibody
Target Names
CKL4
Uniprot No.

Target Background

Function
This antibody targets a protein kinase implicated in blue light-mediated responses, such as hypocotyl elongation and flowering. Its mechanism involves the phosphorylation of CRY2, thereby reducing CRY2 stability.
Gene References Into Functions

Research indicates that CK1.3 (At4g28880) and CK1.4 (At4g28860) directly phosphorylate CRY2 at Ser-587 and Thr-603 in vitro. This phosphorylation negatively regulates CRY2 stability, a process enhanced by blue light. PMID: 23897926

Database Links

KEGG: ath:AT4G28860

STRING: 3702.AT4G28860.1

UniGene: At.31980

Protein Families
Protein kinase superfamily, CK1 Ser/Thr protein kinase family, Casein kinase I subfamily
Subcellular Location
Cytoplasm. Nucleus.
Tissue Specificity
Expressed in seedlings, stems, leaves and flowers.

Q&A

What is CKAP4 and why is it a target for antibody development?

CKAP4, also known as Climp-63 or p63, is a cytoskeleton-associated protein that mediates the anchoring of the endoplasmic reticulum to microtubules. It functions as a high-affinity cell surface receptor for antiproliferative factor (APF) and is involved in the DKK1-CKAP4 signaling pathway that promotes tumor growth. CKAP4 is targeted for antibody development because elevated DKK1 and CKAP4 levels in patients typically indicate malignant transformation and poor prognosis, making the DKK1-CKAP4 pathway a promising target for cancer therapeutics .

How does CKAP4 contribute to cancer progression?

CKAP4 contributes to cancer progression primarily through its role as a cell receptor that can be activated by Dickkopf 1 (DKK1). When activated, the DKK1-CKAP4 pathway stimulates cancer cell growth and proliferation. This pathway is particularly important in several cancers, including pancreatic cancer. Research indicates that elevated levels of both DKK1 and CKAP4 in cancer patients correlate with malignant transformation and poor clinical outcomes, suggesting that this signaling pathway plays a critical role in tumor development and progression .

What are the key characteristics of commercially available CKAP4 antibodies?

Currently available CKAP4 antibodies include rabbit polyclonal antibodies suitable for various applications including immunohistochemistry on paraffin-embedded tissues (IHC-P), Western blotting (WB), and immunocytochemistry/immunofluorescence (ICC/IF). These antibodies typically recognize human CKAP4 samples and are generated using immunogens corresponding to recombinant fragment proteins within human CKAP4, often spanning amino acids 200-600. They function by mediating the anchoring of the endoplasmic reticulum to microtubules and can recognize CKAP4's role as a high-affinity epithelial cell surface receptor .

What are the recommended protocols for validating anti-CKAP4 antibody specificity?

To validate anti-CKAP4 antibody specificity, researchers should implement a multi-method approach. Begin with Western blotting to confirm the antibody detects a protein of the correct molecular weight (~63 kDa). Next, perform immunocytochemistry/immunofluorescence to verify appropriate subcellular localization, focusing on the endoplasmic reticulum-microtubule interface. For definitive validation, include negative controls using CKAP4 knockout cell lines or CKAP4-deficient tissues. Additionally, competitive binding assays with the immunogen peptide can demonstrate specificity. For humanized antibodies like Hv1Lt1, researchers should compare binding affinity and functionality to the original recombinant mouse antibody to ensure comparable or enhanced target recognition, as demonstrated in the Osaka University research .

How can researchers effectively employ CKAP4 antibodies in tumor microenvironment studies?

For effective tumor microenvironment studies using CKAP4 antibodies, researchers should adopt a comprehensive approach starting with multiplex immunofluorescence staining to visualize CKAP4 expression in relation to other cell types within the tumor ecosystem. Co-staining with markers for cancer cells, immune cells, and stromal components will reveal spatial relationships. Flow cytometry can quantify CKAP4 expression across different cell populations. For functional studies, researchers should assess how anti-CKAP4 antibodies modulate immune cell recruitment and activation, as demonstrated by the Osaka University team who found that humanized anti-CKAP4 antibodies help modulate anti-tumor immune reactions. Additionally, researchers should evaluate antibody penetration within tumor spheroids or organoids to simulate in vivo drug delivery. Finally, combining anti-CKAP4 treatment with immune checkpoint inhibitors may provide insights into potential synergistic therapeutic strategies .

What techniques are recommended for measuring the inhibition of DKK1-CKAP4 signaling by anti-CKAP4 antibodies?

To measure the inhibition of DKK1-CKAP4 signaling by anti-CKAP4 antibodies, researchers should employ multiple complementary approaches. First, implement phosphorylation assays to monitor downstream proteins in the pathway, particularly focusing on AKT (Protein kinase B) activation levels using phospho-specific antibodies in Western blotting or ELISA formats. Second, conduct reporter gene assays using luciferase constructs under the control of DKK1-CKAP4 responsive elements to quantitatively measure pathway inhibition. Third, analyze sphere formation assays, which directly measure the ability of cancer stem cells to multiply into sphere-shaped colonies when treated with the antibody – a method successfully used with the humanized Hv1Lt1 antibody. Fourth, measure changes in cancer cell proliferation, migration, and invasion using standard assays like MTT, wound healing, and transwell assays respectively. Finally, examine gene expression profiles of DKK1-CKAP4 target genes using qRT-PCR or RNA-seq to comprehensively assess pathway suppression .

How do humanized anti-CKAP4 antibodies compare to mouse-derived antibodies in efficacy and immunogenicity?

Humanized anti-CKAP4 antibodies demonstrate several advantages over mouse-derived antibodies in both efficacy and immunogenicity profiles. The humanized anti-CKAP4 antibody Hv1Lt1, developed by researchers at Osaka University, exhibited enhanced binding affinity to CKAP4 compared to the original recombinant mouse antibody. This improved target recognition translated to greater efficacy in inhibiting sphere formation by cancer stem cells. From an immunogenicity perspective, humanized antibodies significantly reduce the human anti-mouse antibody (HAMA) response that typically limits the therapeutic potential of mouse-derived antibodies in human patients. Similar to other humanized therapeutic antibodies, this modification enables repeated administration without diminishing efficacy or triggering adverse immune reactions. The structural modifications made during the humanization process preserved or enhanced the functional domains responsible for blocking the DKK1-CKAP4 interaction while minimizing the recognition of the antibody as foreign by the human immune system .

What is the current evidence for combining anti-CKAP4 antibodies with conventional chemotherapy?

Current evidence strongly supports the synergistic potential of combining anti-CKAP4 antibodies with conventional chemotherapy. Research conducted by Osaka University demonstrated that the humanized anti-CKAP4 antibody Hv1Lt1, when administered in combination with standard chemotherapy drugs, produced superior tumor suppression compared to chemotherapy alone in pancreatic cancer mouse models. This combination approach offers multiple advantages: first, it simultaneously targets different cancer cell survival mechanisms; second, anti-CKAP4 antibodies may help overcome chemoresistance by inhibiting the AKT pathway, which is typically activated during chemotherapy and contributes to treatment resistance; and third, this combination potentially allows for reduced chemotherapy dosages, thereby decreasing toxicity while maintaining therapeutic efficacy. The mechanistic basis for this synergy appears to involve the complementary targeting of both proliferative pathways and cell survival mechanisms, creating a more comprehensive anti-cancer strategy .

How does antibody-dependent cellular cytotoxicity (ADCC) contribute to anti-CKAP4 antibody efficacy?

Antibody-dependent cellular cytotoxicity (ADCC) significantly contributes to anti-CKAP4 antibody efficacy through a multi-step immune-mediated mechanism. When anti-CKAP4 antibodies bind to CKAP4 expressed on cancer cell surfaces, their Fc regions become available to interact with Fcγ receptors on immune effector cells, particularly natural killer (NK) cells. This interaction triggers NK cell activation, resulting in the release of cytotoxic granules containing perforin and granzymes that directly kill the antibody-tagged cancer cells. The efficacy of ADCC is influenced by several factors, including the density of CKAP4 on target cells, the quantity and activity of NK cells in the tumor microenvironment, and the affinity of the antibody Fc region for Fcγ receptors. Research with other antibodies like KW-0761 (an anti-CCR4 antibody) has shown that the degree of ADCC is primarily determined by the amount of effector NK cells present rather than the amount of target molecules on cancer cell surfaces. This insight suggests that combining anti-CKAP4 therapy with NK cell-stimulating agents could significantly enhance therapeutic outcomes .

What strategies can overcome low CKAP4 detection sensitivity in certain tissue types?

To overcome low CKAP4 detection sensitivity in challenging tissue types, researchers should implement a comprehensive optimization strategy. First, evaluate multiple antigen retrieval methods, comparing heat-induced epitope retrieval at varying pH levels (citrate buffer pH 6.0 versus EDTA buffer pH 9.0) to determine optimal conditions for CKAP4 epitope exposure. Second, employ signal amplification systems such as tyramide signal amplification (TSA) or polymer-based detection systems that can enhance sensitivity by 10-100 fold without increasing background signal. Third, consider using higher antibody concentrations with extended incubation periods (overnight at 4°C) to improve target binding in tissues with low CKAP4 expression. Fourth, reduce background interference by incorporating appropriate blocking steps using bovine serum albumin, normal serum, or commercial blocking solutions specific to the tissue type. Finally, compare multiple anti-CKAP4 antibodies targeting different epitopes, as accessibility of certain epitopes may vary across tissue types. For particularly challenging samples, consider using fresh-frozen tissues instead of formalin-fixed paraffin-embedded specimens to better preserve CKAP4 antigenicity .

How can researchers address non-specific binding when using anti-CKAP4 antibodies?

To address non-specific binding when using anti-CKAP4 antibodies, researchers should implement a systematic troubleshooting approach. Begin by optimizing blocking protocols using a combination of serum from the same species as the secondary antibody (5-10%) and BSA (1-3%) to effectively block Fc receptors and other non-specific binding sites. Next, titrate the primary antibody concentration to determine the optimal dilution that maintains specific signal while minimizing background. For immunohistochemistry applications, incorporate an avidin-biotin blocking step if biotin-based detection systems are used, as endogenous biotin can be abundant in certain tissues. When performing Western blotting, increase washing duration and detergent concentration (0.1-0.3% Tween-20) in wash buffers to remove weakly bound antibodies. Include appropriate negative controls in each experiment, such as isotype controls matched to the primary antibody's class and concentration. For challenging samples, consider pre-absorbing the antibody with the immunogen peptide to confirm binding specificity. Finally, if non-specific binding persists, try alternative anti-CKAP4 antibodies that target different epitopes, as some regions may be more prone to cross-reactivity than others .

What are the best practices for long-term storage and handling of anti-CKAP4 antibodies to maintain activity?

For optimal long-term storage and handling of anti-CKAP4 antibodies to maintain activity, researchers should adhere to several critical practices. Store antibody stocks at -20°C to -80°C in small aliquots (10-50 µl) to minimize freeze-thaw cycles, as repeated freezing and thawing can lead to antibody denaturation and reduced activity. For working solutions, maintain at 4°C with the addition of sodium azide (0.02-0.05%) as a preservative to prevent microbial contamination, but ensure this preservative is removed before cell-based assays as it can affect cell viability. Avoid exposing antibodies to direct light, particularly if conjugated to fluorophores, to prevent photobleaching and loss of signal intensity. When handling, minimize protein denaturation by avoiding vigorous shaking or vortexing; instead, gently invert or tap tubes to mix. Monitor solution clarity regularly, as cloudiness or precipitates may indicate protein aggregation and loss of activity. For long-term storage exceeding one year, consider lyophilization or the addition of stabilizing proteins like BSA (1-5%) to maintain antibody conformation. Finally, validate antibody performance periodically with positive controls to ensure continued specific reactivity, particularly before critical experiments .

How might anti-CKAP4 antibodies be engineered for improved tumor penetration?

Engineering anti-CKAP4 antibodies for improved tumor penetration requires a multifaceted approach addressing size, charge, and binding dynamics. First, researchers should develop smaller antibody formats such as Fab fragments, single-chain variable fragments (scFvs), or nanobodies derived from the original anti-CKAP4 antibody, reducing molecular weight from ~150 kDa to 25-50 kDa to enhance tissue penetration. Second, optimizing the isoelectric point through targeted amino acid substitutions in the framework regions can reduce non-specific interactions with extracellular matrix components. Third, implementing site-specific modifications to increase hydrophilicity while maintaining specificity would improve diffusion through the tumor interstitium. Fourth, engineering bispecific antibodies with one binding domain targeting CKAP4 and another targeting tumor vasculature markers would facilitate active transport across endothelial barriers. Finally, applying computational modeling to optimize antibody affinity is crucial—moderately reducing affinity may counter the "binding site barrier" phenomenon where high-affinity antibodies bind strongly to the first antigens they encounter, preventing deeper penetration. These approaches should be validated in three-dimensional tumor spheroids and patient-derived xenografts before advancing to clinical development .

What potential exists for developing anti-CKAP4 antibody-drug conjugates (ADCs) for targeted therapy?

The development of anti-CKAP4 antibody-drug conjugates (ADCs) represents a promising frontier in targeted cancer therapy with significant potential for treating pancreatic and other cancers. Anti-CKAP4 antibodies are particularly well-suited for ADC development due to CKAP4's overexpression in cancer cells and its ability to internalize upon antibody binding, facilitating intracellular drug delivery. For optimal ADC design, researchers should consider using site-specific conjugation methods like ThioBridge technology to control the drug-to-antibody ratio and conjugation sites, ensuring consistent drug loading and preserved antibody functionality. The selection of appropriate cytotoxic payloads should be guided by the specific cancer type, with highly potent agents like auristatins, maytansinoids, or pyrrolobenzodiazepines for pancreatic cancer's typically resistant phenotype. Linker chemistry must balance stability in circulation with efficient drug release in the tumor microenvironment, potentially utilizing acid-sensitive hydrazone linkers responsive to the tumor's acidic environment or protease-cleavable linkers activated by cathepsins overexpressed in cancer cells. Prior to clinical translation, comprehensive testing in patient-derived xenograft models would help predict efficacy and optimize dosing strategies for these promising therapeutics .

How can high-throughput specificity profiling improve next-generation anti-CKAP4 antibodies?

High-throughput specificity profiling can significantly enhance next-generation anti-CKAP4 antibody development through comprehensive characterization of binding properties and cross-reactivity. Implementing Drop-seq-based microfluidic analysis, similar to the approach used for SARS-CoV-2 antibodies, allows simultaneous evaluation of antibody binding to multiple CKAP4 variants and structurally similar proteins. This technology enables the generation of detailed "PolyMap scores" that quantify binding specificity across thousands of individual cells, providing unprecedented resolution of antibody cross-reactivity profiles. By testing antibody candidates against diverse CKAP4 epitopes and potential off-target proteins, researchers can identify candidates with optimal specificity. Additionally, this approach facilitates the identification of conserved epitopes across species, enabling the development of antibodies with cross-species reactivity—valuable for translational research from animal models to human applications. The high-throughput nature of this profiling allows screening of much larger antibody libraries, increasing the probability of identifying rare clones with exceptional properties. Furthermore, this methodology provides critical data on epitope accessibility in different cell types and tissue environments, helping predict in vivo efficacy more accurately than traditional binding assays. Integration of this approach into the antibody development pipeline would significantly accelerate the optimization process and reduce late-stage development failures .

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.