The 4CLL4 Antibody (Product Code: CSB-PA608890XA01OFG) is an affinity-purified polyclonal antibody produced in rabbits. It is designed to recognize the recombinant 4CLL4 protein in rice, facilitating studies on its expression, localization, and function in plant systems.
ELISA: Used for quantitative detection of 4CLL4 protein in rice lysates.
Western Blot: Validates the presence and molecular weight (~[specific kDa; data not provided in sources]) of 4CLL4 in plant tissue extracts.
Immunogen Design: The antibody was generated using a recombinant protein immunogen, ensuring specificity for the target epitope.
Purification Method: Antigen-affinity purification enhances antibody specificity by removing non-IgG proteins.
Validation: Confirmed reactivity via ELISA and WB, though exact validation data (e.g., dilution ranges, cross-reactivity tests) are not publicly disclosed .
Reconstitution: No reconstitution required; supplied as a liquid.
Shelf Life: Long-term stability at -80°C; short-term use (≤2 weeks) at 4°C.
Recommended Dilutions:
Species Reactivity: Limited to rice; no cross-reactivity data for other plant species are available.
Research Use Only: Not validated for diagnostic or therapeutic applications.
Lead Time: Custom orders require 14–16 weeks for production .
Distributed by manufacturers specializing in research antibodies (e.g., Cusabio).
Pricing and bulk-order inquiries are typically directed to commercial providers.
While the 4CLL4 Antibody is a critical tool for rice proteomics, its role in plant stress responses, growth regulation, or pathogen interactions remains unexplored. Further studies could employ techniques like immunoprecipitation or immunohistochemistry to elucidate 4CLL4’s biological functions.
Anti-CTLA-4 antibodies function by blocking the inhibitory signals that suppress T cell activation. CTLA-4 normally acts as a negative regulator of T cell activity by competing with CD28 for binding to B7 ligands on antigen-presenting cells. When anti-CTLA-4 antibodies bind to CTLA-4, they prevent this inhibitory interaction, thereby enhancing T cell activation and proliferation. This enhanced T cell response can lead to improved tumor recognition and elimination. Clinical trials have demonstrated that anti-CTLA-4 antibody treatment produces significant clinical responses in melanoma patients, though often accompanied by autoimmune manifestations .
While anti-CTLA-4 antibodies work by removing inhibitory signals (releasing the brakes), anti-4-1BB antibodies function by providing costimulatory signals (pressing the accelerator) in the immune response. The 4-1BB receptor (CD137) is expressed on activated T cells, and binding of anti-4-1BB antibodies delivers activating signals that enhance CD8+ T cell proliferation, survival, and effector functions. Interestingly, anti-4-1BB antibodies can both stimulate antitumor responses and decrease certain autoimmune manifestations, creating a distinctive immunological profile compared to anti-CTLA-4 antibodies . This contrasting mechanism creates the foundation for their complementary effects when used in combination.
Successful engagement of CTLA-4 by therapeutic antibodies can be monitored through several cellular markers. Researchers should assess the increased ratio of effector T cells to regulatory T cells (Tregs) in both peripheral blood and tumor microenvironment. Additional markers include elevated expression of activation markers such as CD25 and CD69 on T cells, increased production of pro-inflammatory cytokines like IFN-γ and TNF-α, and enhanced T cell receptor diversity in the tumor-infiltrating lymphocyte population. Flow cytometry and immunohistochemistry techniques provide quantitative measures of these parameters, while functional assays measuring T cell proliferation and cytokine production offer insights into the functional consequences of CTLA-4 blockade .
Combination therapy with anti-CTLA-4 and anti-4-1BB antibodies represents a novel approach that simultaneously enhances cancer immunity while reducing autoimmune side effects. Research shows that when used together, these antibodies led to CD8+ T-cell-mediated rejection of large established MC38 tumors and long-lasting immunity in mouse models. Statistical analysis suggests the combination could have more than an additive effect in tumor rejection .
The reduction of autoimmune effects appears to correlate with increased function of regulatory T cells. Specifically, combination therapy significantly lowered anti-dsDNA antibody production that was observed with anti-CTLA-4 treatment alone. The estimated anti-dsDNA levels in mice treated with the combination were comparable to levels in control groups, indicating substantial suppression of this autoimmune marker .
Additionally, while anti-4-1BB antibody alone enhanced inflammation in the liver, this inflammation was significantly decreased when combined with anti-CTLA-4. This mutual suppression of side effects suggests distinct mechanisms that can be manipulated to optimize therapeutic outcomes .
The differential response of tumor types to combination therapy reflects complex molecular mechanisms. In studies with MC38 colon carcinoma models, combination therapy was highly effective, while the same regimen showed limited efficacy against B16 melanoma . This difference likely stems from:
Tumor-specific antigen landscape and immunogenicity
Distinct tumor microenvironments affecting T cell infiltration and function
Differential expression of costimulatory and inhibitory molecules on tumor-infiltrating immune cells
Variations in tumor-associated regulatory T cell populations
Research suggests that tumors with higher mutational burden may respond better to combination immunotherapy due to increased neoantigen presentation. Advanced researchers should consider analyzing the tumor mutational burden, immune infiltrate composition, and cytokine profiles when studying differential responses across tumor types .
Anti-CTLA-4 antibodies have complex effects on regulatory T cell (Treg) populations that vary significantly across tissue contexts. While generally thought to inhibit Treg function, the actual impact depends on:
The specific antibody isotype and its ability to engage Fc receptors
The tissue microenvironment and local cytokine milieu
The activation state of the Treg population
Interestingly, combination therapy with anti-4-1BB antibodies appears to increase the regulatory function of CD4+CD25+ T cells compared to anti-CTLA-4 monotherapy. This enhanced regulatory capacity may explain the reduced autoimmune manifestations observed with combination therapy . Researchers investigating Treg dynamics should employ multi-parameter flow cytometry, FOXP3 demethylation analysis, and functional suppression assays to comprehensively characterize Treg populations across different tissues.
When selecting experimental models for testing anti-CTLA-4 and anti-4-1BB combination therapy, researchers should consider multiple factors:
Syngeneic mouse models: MC38 colon carcinoma models have demonstrated significant responses to combination therapy and should be prioritized for initial studies. Research has shown this model is particularly responsive, with combination therapy leading to rejection of large established tumors .
Human CTLA-4 knock-in mice: These models, where mouse CTLA-4 is replaced with its human counterpart, are valuable for testing humanized antibodies. Studies demonstrate that in these mice, a combination of anti-mouse 4-1BB and anti-human CTLA-4 antibodies induces tumor rejection and long-lasting cancer immunity .
Genetically engineered mouse models: For specific cancer types, genetically engineered models that better recapitulate human disease progression may provide more clinically relevant insights.
Treatment timing: Establish models with large, pre-existing tumors (>100mm³) rather than prophylactic models to better reflect clinical scenarios.
The experimental design should include careful monitoring of both tumor regression and potential autoimmune manifestations, with tissue sampling for immunohistochemistry and flow cytometry analysis .
Comprehensive evaluation of autoimmune side effects in CTLA-4 antibody studies requires a multi-modal approach:
Serum analysis:
Histopathological examination:
Immune complex deposition:
Flow cytometric analysis:
Statistical analysis should include ANOVA to compare autoimmune manifestations across treatment groups, particularly focusing on interactions between anti-CTLA-4 and anti-4-1BB effects .
Optimizing dosing strategies for combination immunotherapy requires systematic evaluation of multiple parameters:
Dose-ranging studies:
Test multiple doses of each antibody individually to establish dose-response relationships
Evaluate combination therapies using factorial design to identify potential synergistic or antagonistic interactions
Consider staggered administration schedules to potentially reduce toxicity
Pharmacokinetic/pharmacodynamic (PK/PD) analysis:
Measure serum antibody concentrations at multiple timepoints
Correlate antibody exposure with biomarkers of immune activation and clinical outcomes
Develop PK/PD models to predict optimal dosing regimens
Biomarker monitoring:
Track peripheral blood T cell activation markers
Monitor tumor-infiltrating lymphocyte populations when possible
Assess cytokine profiles to gauge immune activation and potential cytokine release syndrome
Anti-antibody response consideration:
Researchers should employ statistical modeling approaches like response surface methodology to identify the optimal combination of doses that maximize tumor rejection while minimizing autoimmune toxicity.
The efficacy of CTLA-4 blockade therapy is significantly influenced by tumor antigen expression profiles through multiple mechanisms:
Minor histocompatibility antigens: Novel tumor-associated minor histocompatibility antigens, such as those encoded by C19orf48, can serve as targets for T cell recognition following CTLA-4 blockade. These antigens arise from nonsynonymous single nucleotide polymorphisms (SNPs) in coding sequences that cause amino acid changes within CTL epitope sequences .
Antigen processing pathways: Some tumor-associated antigens can be processed through both TAP-dependent and TAP-independent pathways, expanding the repertoire of targetable antigens. Understanding these processing mechanisms helps predict which tumors might respond better to CTLA-4 blockade .
Expression breadth across tissues: Antigens with limited expression in normal tissues but broad expression in tumors represent ideal targets. For instance, C19orf48-encoded peptides have been demonstrated to be widely expressed in renal tumors and solid tumors of other histologies, making them potential targets following immune checkpoint inhibition .
Neoantigen burden: Tumors with higher mutational load and thus more potential neoantigens typically respond better to CTLA-4 blockade. Researchers should quantify neoantigen load through whole-exome sequencing and HLA binding prediction algorithms when evaluating potential response to therapy.
Predicting successful response to combination therapy requires consideration of multiple biomarker categories:
Immune infiltrate composition:
Higher baseline CD8+ T cell infiltration correlates with better responses
Lower regulatory T cell to effector T cell ratios suggest improved outcomes
Presence of dendritic cells expressing both B7 and 4-1BBL indicates potential for strong response
Genetic biomarkers:
Expression levels of CTLA-4 and 4-1BB in tumor-infiltrating lymphocytes
Polymorphisms in genes encoding CTLA-4, 4-1BB, and related signaling molecules
Tumor mutational burden, with higher burden typically predicting better response
Peripheral blood biomarkers:
Expansion of activated CD8+ T cells following initial treatment
Emergence of tumor antigen-specific T cell populations
Changes in cytokine profiles, particularly IFN-γ and TNF-α
Tumor microenvironment factors:
Expression of immunosuppressive molecules like PD-L1, IDO, and TGF-β
Metabolic characteristics of the tumor microenvironment
Vascular normalization status affecting immune cell trafficking
Multi-parameter analysis combining these biomarkers using machine learning approaches may provide the most accurate prediction of treatment response .
Development of host immune responses against therapeutic antibodies presents a significant challenge in repeated administration regimens. Researchers can address this issue through several approaches:
Co-administration strategies: Anti-4-1BB antibodies can reduce host antibody responses to anti-CTLA-4 antibodies by >30-fold. This approach leverages the ability of anti-4-1BB to reduce antibody responses to proteins while enhancing cellular immunity .
Antibody engineering:
Humanization or fully human antibodies to reduce xenotypic responses
Modification of allotypic determinants
Deimmunization approaches to remove T cell epitopes within the antibody sequence
Alternative administration protocols:
Intermittent dosing schedules with drug holidays
Local rather than systemic administration where appropriate
Dose escalation protocols to induce tolerance
Companion immunomodulation:
Transient immunosuppression during antibody administration
Induction of B cell tolerization to the therapeutic antibody
Use of nanoparticle delivery systems to shield antibodies from immune recognition
Researchers should implement comprehensive monitoring of anti-antibody responses using sensitive ELISA techniques and correlate these with pharmacokinetic profiles and clinical outcomes .
The differential effects of combination therapy with anti-CTLA-4 and anti-4-1BB antibodies on cancer immunity versus autoimmunity represent a fascinating paradox with several potential mechanisms:
Distinct T cell population targeting: The combination therapy may preferentially activate tumor-specific CD8+ T cells while enhancing regulatory function on autoreactive T cells. This selective modulation could explain the uncoupling of tumor immunity from autoimmunity .
Differential effector mechanisms: Studies have shown that tumor rejection and autoimmune destruction may employ distinct effector mechanisms. For instance, in melanoma models, antibody-mediated tumor rejection and autoimmune depigmentation require different levels of antibodies and can be distinguished by requirements for FcR and complement .
Context-dependent costimulation: The tumor microenvironment and sites of potential autoimmunity present different cytokine milieus and antigen-presenting cell populations, potentially leading to contextual differences in how the combination therapy modulates immune responses.
Regulatory T cell modulation: Combination therapy increases the regulatory function of CD4+CD25+ T cells, which may preferentially suppress autoimmune responses while preserving tumor immunity due to differences in antigen affinity and local microenvironments .
Understanding these mechanisms requires careful experimental designs that can distinguish tumor immunity from autoimmunity, potentially through parallel assessment of tumor rejection and autoimmune manifestations in the same experimental system .