Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Gastroenterology ; 162(2): 590-603.e14, 2022 02.
Article in English | MEDLINE | ID: mdl-34627860

ABSTRACT

BACKGROUND AND AIMS: Patients with pancreatic ductal adenocarcinoma (PDA) have not yet benefitted from the revolution in cancer immunotherapy due in large part to a dominantly immunosuppressive tumor microenvironment. MEK inhibition combined with autophagy inhibition leads to transient tumor responses in some patients with PDA. We examined the functional effects of combined MEK and autophagy inhibition on the PDA immune microenvironment and the synergy of combined inhibition of MEK and autophagy with CD40 agonism (aCD40) against PDA using immunocompetent model systems. METHODS: We implanted immunologically "cold" murine PDA cells orthotopically in wide type C57BL/6J mice. We administered combinations of inhibitors of MEK1/2, inhibitors of autophagy, and aCD40 and measured anticancer efficacy and immune sequelae using mass cytometry and multiplexed immunofluorescence imaging analysis to characterize the tumor microenvironment. We also used human and mouse PDA cell lines and human macrophages in vitro to perform functional assays to elucidate the cellular effects induced by the treatments. RESULTS: We find that coinhibition of MEK (using cobimetinib) and autophagy (using mefloquine), but not either treatment alone, activates the STING/type I interferon pathway in tumor cells that in turn activates paracrine tumor associated macrophages toward an immunogenic M1-like phenotype. This switch is further augmented by aCD40. Triple therapy (cobimetinib + mefloquine + aCD40) achieved cytotoxic T-cell activation in an immunologically "cold" mouse PDA model, leading to enhanced antitumor immunity. CONCLUSIONS: MEK and autophagy coinhibition coupled with aCD40 invokes immune repolarization and is an attractive therapeutic approach for PDA immunotherapy development.


Subject(s)
Autophagy/immunology , Azetidines/pharmacology , CD40 Antigens/agonists , Carcinoma, Pancreatic Ductal/immunology , Mefloquine/pharmacology , Pancreatic Neoplasms/immunology , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Animals , Autophagy/drug effects , Cell Line, Tumor , Drug Synergism , Humans , Hydroxychloroquine/pharmacology , Immunotherapy , Interferon Type I/drug effects , Interferon Type I/immunology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Macrophages , Membrane Proteins/drug effects , Membrane Proteins/immunology , Mice , Paracrine Communication/drug effects , Paracrine Communication/immunology , Tumor Escape , Tumor Microenvironment/drug effects , Tumor-Associated Macrophages/drug effects
2.
J Immunol ; 207(8): 2060-2076, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34551965

ABSTRACT

CD40 is a potent activating receptor within the TNFR family expressed on APCs of the immune system, and it regulates many aspects of B and T cell immunity via interaction with CD40 ligand (CD40L; CD154) expressed on the surface of activated T cells. Soluble CD40L and agonistic mAbs directed to CD40 are being explored as adjuvants in therapeutic or vaccination settings. Some anti-CD40 Abs can synergize with soluble monomeric CD40L. We show that direct fusion of CD40L to certain agonistic anti-CD40 Abs confers superagonist properties, reducing the dose required for efficacy, notably greatly increasing total cytokine secretion by human dendritic cells. The tetravalent configuration of anti-CD40-CD40L Abs promotes CD40 cell surface clustering and internalization and is the likely mechanism of increased receptor activation. CD40L fused to either the L or H chain C termini, with or without flexible linkers, were all superagonists with greater potency than CD40L trimer. The increased anti-CD40-CD40L Ab potency was independent of higher order aggregation. Moreover, the anti-CD40-CD40L Ab showed higher potency in vivo in human CD40 transgenic mice compared with the parental anti-CD40 Ab. To broaden the concept of fusing agonistic Ab to natural ligand, we fused OX40L to an agonistic OX40 Ab, and this resulted in dramatically increased efficacy for proliferation and cytokine production of activated human CD4+ T cells as well as releasing the Ab from dependency on cross-linking. This work shows that directly fusing antireceptor Abs to ligand is a useful strategy to dramatically increase agonist potency.


Subject(s)
Antibodies, Monoclonal/metabolism , B-Lymphocytes/immunology , CD40 Antigens/agonists , CD40 Ligand/metabolism , Dendritic Cells/immunology , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/genetics , CD40 Antigens/immunology , CD40 Ligand/genetics , CHO Cells , Cell Differentiation , Cricetulus , Cytokines/metabolism , Humans , Lymphocyte Activation , Receptor Aggregation , Recombinant Fusion Proteins/genetics
3.
J Immunol ; 206(6): 1372-1384, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33558374

ABSTRACT

Pancreatic cancer is a particularly lethal malignancy that resists immunotherapy. In this study, using a preclinical pancreatic cancer murine model, we demonstrate a progressive decrease in IFN-γ and granzyme B and a concomitant increase in Tox and IL-10 in intratumoral tumor-specific T cells. Intratumoral myeloid cells produced elevated IL-27, a cytokine that correlates with poor patient outcome. Abrogating IL-27 signaling significantly decreased intratumoral Tox+ T cells and delayed tumor growth yet was not curative. Agonistic αCD40 decreased intratumoral IL-27-producing myeloid cells, decreased IL-10-producing intratumoral T cells, and promoted intratumoral Klrg1+Gzmb+ short-lived effector T cells. Combination agonistic αCD40+αPD-L1 cured 63% of tumor-bearing animals, promoted rejection following tumor rechallenge, and correlated with a 2-log increase in pancreas-residing tumor-specific T cells. Interfering with Ifngr1 expression in nontumor/host cells abrogated agonistic αCD40+αPD-L1 efficacy. In contrast, interfering with nontumor/host cell Tnfrsf1a led to cure in 100% of animals following agonistic αCD40+αPD-L1 and promoted the formation of circulating central memory T cells rather than long-lived effector T cells. In summary, we identify a mechanistic basis for T cell exhaustion in pancreatic cancer and a feasible clinical strategy to overcome it.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , CD40 Antigens/agonists , Carcinoma, Pancreatic Ductal/drug therapy , Myeloid Cells/drug effects , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Disease Models, Animal , Drug Screening Assays, Antitumor , Female , Humans , Interleukins/metabolism , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Mice, Transgenic , Myeloid Cells/immunology , Myeloid Cells/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Primary Cell Culture , Tumor Cells, Cultured/transplantation , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
4.
Proc Natl Acad Sci U S A ; 117(1): 541-551, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31889004

ABSTRACT

Cancer immunotherapies are increasingly combined with targeted therapies to improve therapeutic outcomes. We show that combination of agonistic anti-CD40 with antiangiogenic antibodies targeting 2 proangiogenic factors, vascular endothelial growth factor A (VEGFA) and angiopoietin 2 (Ang2/ANGPT2), induces pleiotropic immune mechanisms that facilitate tumor rejection in several tumor models. On the one hand, VEGFA/Ang2 blockade induced regression of the tumor microvasculature while decreasing the proportion of nonperfused vessels and reducing leakiness of the remaining vessels. On the other hand, both anti-VEGFA/Ang2 and anti-CD40 independently promoted proinflammatory macrophage skewing and increased dendritic cell activation in the tumor microenvironment, which were further amplified upon combination of the 2 treatments. Finally, combined therapy provoked brisk infiltration and intratumoral redistribution of cytotoxic CD8+ T cells in the tumors, which was mainly driven by Ang2 blockade. Overall, these nonredundant synergistic mechanisms endowed T cells with improved effector functions that were conducive to more efficient tumor control, underscoring the therapeutic potential of antiangiogenic immunotherapy in cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , CD40 Antigens/agonists , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Tumor Microenvironment/drug effects , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Angiopoietin-2/antagonists & inhibitors , Angiopoietin-2/metabolism , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD40 Antigens/immunology , Cell Line, Tumor/transplantation , Disease Models, Animal , Drug Synergism , Female , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Neoplasms/blood supply , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
5.
Proc Natl Acad Sci U S A ; 117(14): 8022-8031, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32213589

ABSTRACT

Innate immune receptors such as toll-like receptors (TLRs) provide critical molecular links between innate cells and adaptive immune responses. Here, we studied the CD40 pathway as an alternative bridge between dendritic cells (DCs) and adaptive immunity in cancer. Using an experimental design free of chemo- or radiotherapy, we found CD40 activation with agonistic antibodies (⍺CD40) produced complete tumor regressions in a therapy-resistant pancreas cancer model, but only when combined with immune checkpoint blockade (ICB). This effect, unachievable with ICB alone, was independent of TLR, STING, or IFNAR pathways. Mechanistically, αCD40/ICB primed durable T cell responses, and efficacy required DCs and host expression of CD40. Moreover, ICB drove optimal generation of polyfunctional T cells in this "cold" tumor model, instead of rescuing T cell exhaustion. Thus, immunostimulation via αCD40 is sufficient to synergize with ICB for priming. Clinically, combination αCD40/ICB may extend efficacy in patients with "cold" and checkpoint-refractory tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , CD40 Antigens/agonists , Carcinoma, Pancreatic Ductal/drug therapy , Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD40 Antigens/immunology , CD40 Antigens/metabolism , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Drug Screening Assays, Antitumor , Drug Synergism , Female , Humans , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
6.
J Hepatol ; 74(5): 1145-1154, 2021 05.
Article in English | MEDLINE | ID: mdl-33276030

ABSTRACT

BACKGROUND & AIMS: While cholangiocarcinomas (CCAs) commonly express programmed cell death 1 (PD-1) and its ligand (PD-L1), they respond poorly to immune checkpoint inhibitors (ICIs). We aimed to determine whether stimulating antigen-presenting cells, including macrophages and dendritic cells, using a CD40 agonist could improve this response. METHODS: We compared treatment responses in subcutaneous, orthotopic, and 2 plasmid-based murine intrahepatic CCA (iCCA) models. Mice were treated for 4 weeks with weekly IgG control, a CD40 agonistic antibody, anti-PD-1, or the combination of both (anti-CD40/PD-1). Flow cytometric (FACS) analysis of lymphocytes and myeloid cell populations (including activation status) was performed. We used dendritic cell knockout mice, and macrophage, CD4+ and CD8+ T cell depletion models to identify effector cells. Anti-CD40/PD-1 was combined with chemotherapy (gemcitabine/cisplatin) to test for improved therapeutic efficacy. RESULTS: In all 4 models, anti-PD-1 alone was minimally efficacious. Mice exhibited a moderate response to CD40 agonist monotherapy. Combination anti-CD40/PD-1 therapy led to a significantly greater reduction in tumor burden. FACS demonstrated increased number and activation of CD4+ and CD8+ T cells, natural killer cells, and myeloid cells in tumor and non-tumor liver tissue of tumor-bearing mice treated with anti-CD40/PD-1. Depletion of macrophages, dendritic cells, CD4+ T cells, or CD8+ T cells abrogated treatment efficacy. Combining anti-CD40/PD-1 with gemcitabine/cisplatin resulted in a significant survival benefit compared to gemcitabine/cisplatin alone. CONCLUSION: CD40-mediated activation of macrophages and dendritic cells in iCCA significantly enhances response to anti-PD-1 therapy. This regimen may enhance the efficacy of first-line chemotherapy. LAY SUMMARY: Checkpoint inhibition, a common form of immune therapy, is generally ineffective for the treatment of cholangiocarcinoma. These tumors suppress the infiltration and function of surrounding immune cells. Stimulating immune cells such as macrophages and dendritic cells via the CD40 receptor activates downstream immune cells and enhances the response to checkpoint inhibitors.


Subject(s)
CD40 Antigens/agonists , Cholangiocarcinoma , Immune Checkpoint Inhibitors/pharmacology , Liver Neoplasms , Macrophage Activation/immunology , Tumor Microenvironment , Animals , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Cholangiocarcinoma/immunology , Cholangiocarcinoma/pathology , Cisplatin/pharmacology , Dendritic Cells/immunology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Collateral Sensitivity , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/drug effects , Macrophage-Activating Factors/immunology , Mice , Mice, Knockout , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Gemcitabine
7.
Cancer Immunol Immunother ; 70(7): 1853-1865, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33392713

ABSTRACT

Targeting CD40 with agonist antibodies is a promising approach to cancer immunotherapy. CD40 acts as a master regulator of immunity by mobilizing multiple arms of the immune system to initiate highly effective CD8 + T-cell-mediated responses against foreign pathogens and tumors. The clinical development of CD40 agonist antibodies requires careful optimization of the antibody to maximize therapeutic efficacy while minimizing adverse effects. Both epitope specificity and isotype are critical for CD40 agonist antibody mechanism of action and potency. We developed a novel antibody, APX005M, which binds with high affinity to the CD40 ligand-binding site on CD40 and is optimized for selective interaction with Fcγ receptors to enhance agonistic potency while limiting less desirable Fc-effector functions like antibody-dependent cellular cytotoxicity of CD40-expressing immune cells. APX005M is a highly potent inducer of innate and adaptive immune effector responses and represents a promising CD40 agonist antibody for induction of an effective anti-tumor immune response with a favorable safety profile.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity , B-Lymphocytes/immunology , CD40 Antigens/agonists , Epitopes/immunology , Immunoglobulin G/immunology , Receptors, Fc/metabolism , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes , Epitopes/metabolism , Female , Humans , Macaca fascicularis , Male
8.
J Cardiovasc Pharmacol ; 78(2): 228-234, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34029270

ABSTRACT

ABSTRACT: Epicardial adipose tissue (EAT) dysfunction mediates chronic inflammation by regulating inflammation-related adipokines and cytokines, and it further promotes coronary artery disease (CAD) development. CD40L/CD40 is involved in multiple inflammatory pathways that contribute to various pathophysiological processes. However, the function of CD40L/CD40 in the expression and production of adipokines and cytokines in epicardial adipocytes remains unclear. The purpose of the present study was to explore the role and underlying mechanisms of CD40L/CD40 in adipokine and cytokine expression and production. We isolated adipocytes from EAT tissues of CAD and non-CAD patients. We noticed that CD40 was dramatically increased in EAT tissues of CAD patients. Loss-of-function and gain-of-function studies were performed. The results showed that CD40 silencing reduced recombinant CD40 ligand (rCD40L)-induced upregulation of plasminogen activator inhibitor-1, leptin, interleukin-6, and monocyte chemotactic protein-1 messenger RNA levels and secretion. Overexpression of CD40 displayed the opposite results. In addition, rCD40L triggered mixed lineage leukemia protein-1 (MLL1) expression both in messenger RNA and protein levels. CD40 depletion apparently blocked MLL1 expression, whereas gain of function of CD40 resulted in augmentation of MLL1 levels. Interestingly, chromatin immunoprecipitation-quantitative real-time polymerase chain reaction analysis revealed that CD40 elimination dampened histone H3 lysine 4 trimethylation enrichment at plasminogen activator inhibitor-1, leptin, interleukin-6, and monocyte chemotactic protein-1 promoter regions in the presence of rCD40L. The reverse pattern was observed upon ectopic expression of CD40. Most important, MLL1 silencing effectively reversed the promotive effects of CD40 on adipokine and cytokine secretion. Taken together, our findings suggest that CD40L/CD40 regulates adipokine and cytokine expression by H3 lysine 4 trimethylation modification in adipocytes.


Subject(s)
Adipocytes/drug effects , Adipokines/metabolism , CD40 Antigens/agonists , CD40 Ligand/pharmacology , Coronary Artery Disease/metabolism , Cytokines/metabolism , Histones/metabolism , Pericardium/drug effects , Adipocytes/metabolism , Adipokines/genetics , Aged , CD40 Antigens/genetics , CD40 Antigens/metabolism , Case-Control Studies , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cytokines/genetics , Female , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Leptin/genetics , Leptin/metabolism , Male , Methylation , Middle Aged , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Pericardium/metabolism , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Promoter Regions, Genetic , Protein Processing, Post-Translational
9.
Cancer Immunol Immunother ; 68(2): 233-245, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30382327

ABSTRACT

Limitations of immunotherapy include poorly functioning events early in the immune response cycle, such as efficient antigen presentation and T cell priming. CD40 signaling in dendritic cells leads to upregulation of cell surface costimulatory and MHC molecules and the generation of cytokines, which promotes effective priming of CD8+ effector T cells while minimizing T cell anergy and the generation of regulatory T cells. This naturally occurs through interaction with CD40 ligand (CD40L) expressed on CD4+ T-helper cells. CD40 signaling can also be achieved using specific antibodies, leading to several agonist CD40 antibodies entering clinical development. Our approach to select a CD40 agonist antibody was to define a balanced profile between sufficiently strong immune stimulation and the untoward effects of systemic immune activation. CDX-1140 is a human IgG2 antibody that activates DCs and B cells and drives NFkB stimulation in a CD40-expressing reporter cell line. These activities are Fc-independent and are maintained using an F(ab')2 fragment of the antibody. CDX-1140 binds outside of the CD40L binding site, and addition of recombinant CD40L greatly enhances DC and B activation by CDX-1140, suggesting that CDX-1140 may act synergistically with naturally expressed CD40L. CDX-1140 also has both direct and immune-mediated anti-tumor activity in xenograft models. CDX-1140 does not promote cytokine production in whole blood assays and has good pharmacodynamic and safety profiles in cynomolgus macaques. These data support the potential of CDX-1140 as part of a cancer therapy regimen, and a phase 1 trial has recently commenced.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD40 Antigens/agonists , Immunotherapy/methods , Neoplasms/therapy , Animals , Antibodies, Monoclonal/immunology , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD40 Ligand/immunology , CD40 Ligand/metabolism , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , HEK293 Cells , Humans , Macaca fascicularis , Mice, SCID , Mice, Transgenic , Neoplasms/immunology , Neoplasms/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
10.
J Immunol ; 197(1): 179-87, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27217585

ABSTRACT

Cancer immunotherapies are increasingly effective in the clinic, especially immune checkpoint blockade delivered to patients who have T cell-infiltrated tumors. Agonistic CD40 mAb promotes stromal degradation and, in combination with chemotherapy, drives T cell infiltration and de novo responses against tumors, rendering resistant tumors susceptible to current immunotherapies. Partnering anti-CD40 with different treatments is an attractive approach for the next phase of cancer immunotherapies, with a number of clinical trials using anti-CD40 combinations ongoing, but the optimal therapeutic regimens with anti-CD40 are not well understood. Pancreatic ductal adenocarcinoma (PDA) is classically resistant to immunotherapy and lacks baseline T cell infiltration. In this study, we used a tumor cell line derived from a genetically engineered mouse model of PDA to investigate alterations in the sequence of anti-CD40 and chemotherapy as an approach to enhance pharmacological delivery of chemotherapy. Unexpectedly, despite our previous studies showing anti-CD40 treatment after chemotherapy is safe in both mice and patients with PDA, we report in this article that anti-CD40 administration <3 d in advance of chemotherapy is lethal in more than half of treated C57BL/6 mice. Anti-CD40 treatment 2 or 3 d before chemotherapy resulted in significantly increased populations of both activated myeloid cells and macrophages and lethal hepatotoxicity. Liver damage was fully abrogated when macrophage activation was blocked using anti-CSF-1R mAb. These studies highlight the dual nature of CD40 in activating both macrophages and T cell responses, and the need for preclinical investigation of optimal anti-CD40 treatment regimens for safe design of clinical trials.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD40 Antigens/agonists , Carcinoma, Ductal/therapy , Drug-Related Side Effects and Adverse Reactions/prevention & control , Immunotherapy/methods , Liver Failure/prevention & control , Pancreatic Neoplasms/therapy , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Animals , Carcinoma, Ductal/complications , Carcinoma, Ductal/immunology , Cell Line, Tumor , Clinical Protocols , Drug Interactions , Drug Therapy , Genetic Engineering , Humans , Liver Failure/etiology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/immunology , Receptor, Macrophage Colony-Stimulating Factor/immunology
11.
Biochem Biophys Res Commun ; 484(3): 662-667, 2017 03 11.
Article in English | MEDLINE | ID: mdl-28153727

ABSTRACT

Expansion of PD-1-expressing CD8+ cytotoxic T lymphocytes (CTLs) and associated CTL exhaustion are chief issues for ineffective virus-elimination in chronic infectious diseases. PD-1 blockade using antagonistic anti-PD-L1 antibodies results in a moderate conversion of CTL exhaustion. We previously demonstrated that CD40L signaling of ovalbumin (OVA)-specific vaccine, OVA-Texo, converts CTL exhaustion via the activation of the mTORC1 pathway in OVA-expressing adenovirus (AdVova)-infected B6 mice showing CTL inflation and exhaustion. Here, we developed AdVova-infected B6 and transgenic CD11c-DTR (termed AdVova-B6 and AdVova-CD11c-DTR) mice with chronic infection, and assessed a potential effect of CD40 agonist on the conversion of CTL exhaustion and on a potential enhancement of PD-1 antagonist action in rescuing exhausted CTLs in our chronic infection models. We demonstrate that a single dose of anti-CD40 alone can effectively convert CTL exhaustion by activating the mTORC1 pathway, leading to CTL proliferation, up-regulation of an effector-cytokine IFN-γ and the cytolytic effect in AdVova-B6 mice. Using anti-CD4 antibody and diphtheria toxin (DT) to deplete CD4+ T-cells and dendritic cells (DCs), we discovered that the CD40 agonist-induced conversion in AdVova-B6 and AdVova-CD11c-DTR mice is dependent upon host CD4+ T-cell and DC involvements. Moreover, CD40 agonist significantly enhances PD-1 antagonist effectiveness in rescuing exhausted CTLs in chronic infection. Taken together, our data demonstrate the importance of CD40 signaling in the conversion of CTL exhaustion and its ability to enhance PD-1 antagonist action in rescuing exhausted CTLs in chronic infection. Therefore, our findings may positively impact the design of new therapeutic strategies for chronic infectious diseases.


Subject(s)
CD40 Antigens/immunology , Infections/immunology , Infections/pathology , Multiprotein Complexes/metabolism , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Cytotoxic/immunology , TOR Serine-Threonine Kinases/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , CD40 Antigens/agonists , Cells, Cultured , Chronic Disease , Lymphocyte Activation/immunology , Mechanistic Target of Rapamycin Complex 1 , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/immunology
12.
Gastroenterology ; 149(1): 201-10, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25888329

ABSTRACT

BACKGROUND & AIMS: Immunotherapies that induce T-cell responses have shown efficacy against some solid malignancies in patients and mice, but these have little effect on pancreatic ductal adenocarcinoma (PDAC). We investigated whether the ability of PDAC to evade T-cell responses induced by immunotherapies results from the low level of immunogenicity of tumor cells, the tumor's immunosuppressive mechanisms, or both. METHODS: Kras(G12D/+);Trp53(R172H/+);Pdx-1-Cre (KPC) mice, which develop spontaneous PDAC, or their littermates (controls) were given subcutaneous injections of a syngeneic KPC-derived PDAC cell line. Mice were then given gemcitabine and an agonist of CD40 to induce tumor-specific immunity mediated by T cells. Some mice were also given clodronate-encapsulated liposomes to deplete macrophages. Tumor growth was monitored. Tumor and spleen tissues were collected and analyzed by histology, flow cytometry, and immunohistochemistry. RESULTS: Gemcitabine in combination with a CD40 agonist induced T-cell-dependent regression of subcutaneous PDAC in KPC and control mice. In KPC mice given gemcitabine and a CD40 agonist, CD4(+) and CD8(+) T cells infiltrated subcutaneous tumors, but only CD4(+) T cells infiltrated spontaneous pancreatic tumors (not CD8(+) T cells). In mice depleted of Ly6C(low) F4/80(+) extratumoral macrophages, the combination of gemcitabine and a CD40 agonist stimulated infiltration of spontaneous tumors by CD8(+) T cells and induced tumor regression, mediated by CD8(+) T cells. CONCLUSIONS: Ly6C(low) F4/80(+) macrophages that reside outside of the tumor microenvironment regulate infiltration of T cells into PDAC and establish a site of immune privilege. Strategies to reverse the immune privilege of PDAC, which is regulated by extratumoral macrophages, might increase the efficacy of T-cell immunotherapy for patients with PDAC.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/drug therapy , Immunotherapy/methods , Macrophages/cytology , Macrophages/immunology , Pancreatic Neoplasms/drug therapy , Animals , Antimetabolites, Antineoplastic/pharmacology , CD40 Antigens/agonists , CD8-Positive T-Lymphocytes/drug effects , Carcinoma, Pancreatic Ductal/immunology , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Models, Animal , Immunohistochemistry , Macrophages/drug effects , Mice , Pancreatic Neoplasms/immunology , Gemcitabine , Pancreatic Neoplasms
13.
Ann Oncol ; 26(12): 2483-90, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26386124

ABSTRACT

BACKGROUND: Data from murine models suggest that CD40 activation may synergize with cytotoxic chemotherapy. We aimed to determine the maximum tolerated dose (MTD) and toxicity profile and to explore immunological biomarkers of the CD40-activating antibody CP-870,893 with cisplatin and pemetrexed in patients with malignant pleural mesothelioma (MPM). PATIENTS AND METHODS: Eligible patients had confirmed MPM, ECOG performance status 0-1, and measurable disease. Patients received cisplatin 75 mg/m(2) and pemetrexed 500 mg/m(2) on day 1 and CP-870,893 on day 8 of a 21-day cycle for maximum 6 cycles with up to 6 subsequent cycles single-agent CP-870,893. Immune cell subset changes were examined weekly by flow cytometry. RESULTS: Fifteen patients were treated at three dose levels. The MTD of CP-870,893 was 0.15 mg/kg, and was exceeded at 0.2 mg/kg with one grade 4 splenic infarction and one grade 3 confusion and hyponatraemia. Cytokine release syndrome (CRS) occurred in most patients (80%) following CP-870,893. Haematological toxicities were consistent with cisplatin and pemetrexed chemotherapy. Six partial responses (40%) and 9 stable disease (53%) as best response were observed. The median overall survival was 16.5 months; the median progression-free survival was 6.3 months. Three patients survived beyond 30 months. CD19+ B cells decreased over 6 cycles of chemoimmunotherapy (P < 0.001) with a concomitant increase in the proportion of CD27+ memory B cells (P < 0.001) and activated CD86+CD27+ memory B cells (P < 0.001), as an immunopharmacodynamic marker of CD40 activation. CONCLUSIONS: CP-870,893 with cisplatin and pemetrexed is safe and tolerable at 0.15 mg/kg, although most patients experience CRS. While objective response rates are similar to chemotherapy alone, three patients achieved long-term survival. AUSTRALIA NEW ZEALAND CLINICAL TRIALS REGISTRY NUMBER: ACTRN12609000294257.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , CD40 Antigens/metabolism , Cisplatin/administration & dosage , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Pemetrexed/administration & dosage , Pleural Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal, Humanized , CD40 Antigens/agonists , Cohort Studies , Female , Follow-Up Studies , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Male , Mesothelioma/diagnosis , Mesothelioma/metabolism , Mesothelioma, Malignant , Middle Aged , Pleural Neoplasms/diagnosis , Pleural Neoplasms/metabolism , Prospective Studies
14.
PLoS Pathog ; 9(7): e1003490, 2013.
Article in English | MEDLINE | ID: mdl-23853599

ABSTRACT

The intrahepatic immune environment is normally biased towards tolerance. Nonetheless, effective antiviral immune responses can be induced against hepatotropic pathogens. To examine the immunological basis of this paradox we studied the ability of hepatocellularly expressed hepatitis B virus (HBV) to activate immunologically naïve HBV-specific CD8⁺ T cell receptor (TCR) transgenic T cells after adoptive transfer to HBV transgenic mice. Intrahepatic priming triggered vigorous in situ T cell proliferation but failed to induce interferon gamma production or cytolytic effector function. In contrast, the same T cells differentiated into cytolytic effector T cells in HBV transgenic mice if Programmed Death 1 (PD-1) expression was genetically ablated, suggesting that intrahepatic antigen presentation per se triggers negative regulatory signals that prevent the functional differentiation of naïve CD8⁺ T cells. Surprisingly, coadministration of an agonistic anti-CD40 antibody (αCD40) inhibited PD-1 induction and restored T cell effector function, thereby inhibiting viral gene expression and causing a necroinflammatory liver disease. Importantly, the depletion of myeloid dendritic cells (mDCs) strongly diminished the αCD40 mediated functional differentiation of HBV-specific CD8⁺ T cells, suggesting that activation of mDCs was responsible for the functional differentiation of HBV-specific CD8⁺ T cells in αCD40 treated animals. These results demonstrate that antigen-specific, PD-1-mediated CD8⁺ T cell exhaustion can be rescued by CD40-mediated mDC-activation.


Subject(s)
Adaptive Immunity , CD40 Antigens/agonists , CD8-Positive T-Lymphocytes/immunology , Hepatitis B virus/physiology , Hepatitis B/immunology , Host-Pathogen Interactions , Programmed Cell Death 1 Receptor/metabolism , Animals , Antigen Presentation , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Antigens, Viral/metabolism , CD40 Antigens/genetics , CD40 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation , Cell Proliferation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Dendritic Cells/virology , Gene Expression Regulation, Viral , Hepatitis B/metabolism , Hepatitis B/pathology , Hepatitis B/virology , Hepatitis B virus/immunology , Liver/immunology , Liver/metabolism , Liver/pathology , Liver/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Programmed Cell Death 1 Receptor/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/virology
15.
Brain Behav Immun ; 50: 125-140, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26173174

ABSTRACT

The similarity between sickness behavior syndrome (SBS) in infection and autoimmune disorders and certain symptoms in major depressive disorder (MDD), and the high co-morbidity of autoimmune disorders and MDD, constitutes some of the major evidence for the immune-inflammation hypothesis of MDD. CD40 ligand-CD40 immune-activation is important in host response to infection and in development of autoimmunity. Mice given a single intra-peritoneal injection of CD40 agonist antibody (CD40AB) develop SBS for 2-3days characterized by weight loss and increased sleep, effects that are dependent on the cytokine, tumor necrosis factor (TNF). Here we report that CD40AB also induces behavioral effects that extend beyond acute SBS and co-occur with but are not mediated by kynurenine pathway activation and recovery. CD40AB led to decreased saccharin drinking (days 1-7) and decreased Pavlovian fear conditioning (days 5-6), and was without effect on physical fatigue (day 5). These behavioral effects co-occurred with increased plasma and brain levels of kynurenine and its metabolites (days 1-7/8). Co-injection of TNF blocker etanercept with CD40AB prevented each of SBS, reduced saccharin drinking, and kynurenine pathway activation in plasma and brain. Repeated oral administration of a selective indoleamine 2,3-dioxygenase (IDO) inhibitor blocked activation of the kynurenine pathway but was without effect on SBS and saccharin drinking. This study provides novel evidence that CD40-TNF activation induces deficits in saccharin drinking and Pavlovian fear learning and activates the kynurenine pathway, and that CD40-TNF activation of the kynurenine pathway is not necessary for induction of the acute or extended SBS effects.


Subject(s)
CD40 Antigens/immunology , CD40 Ligand/immunology , Illness Behavior/physiology , Kynurenine/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Behavior, Animal/drug effects , CD40 Antigens/agonists , CD40 Ligand/metabolism , Conditioning, Psychological/drug effects , Depressive Disorder, Major/immunology , Depressive Disorder, Major/metabolism , Drinking Behavior/drug effects , Fear/drug effects , Illness Behavior/drug effects , Kynurenine/metabolism , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/immunology , Signal Transduction/drug effects , Syndrome , Tumor Necrosis Factor-alpha/metabolism
16.
J Immunol ; 190(8): 3928-38, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23509363

ABSTRACT

CD40 is expressed on cells of the immune system and in some tissues that are targets for autoimmune-mediated damage. It is not known if CD40 expression in target tissues plays a role in the pathology of autoimmune diseases. This study shows that agonistic anti-CD40 induces strong and sustained proliferation of thyroid epithelial cells (TECs), or thyrocytes, in IFN-γ(-/-) autoimmune-prone NOD and NOD.H-2h4 mice. TEC proliferation is accompanied by greatly increased expression of CD40 on TECs, development of fibrosis and hypothyroidism, and increased expression of proinflammatory molecules in thyroids. Bone marrow chimera experiments indicate that TEC expression of CD40 is required for anti-CD40-induced TEC proliferation, but lymphoid cells do not have to express CD40. TEC proliferation is reduced in wild-type mice given anti-CD40, presumably because they produce IFN-γ, which inhibits TEC proliferation. CD40 also increases on TECs during development of an autoimmune thyroid disease characterized by TEC hyperproliferation that develops spontaneously in IFN-γ(-/-) NOD.H-2h4 mice. TEC hyperproliferation development is accelerated in mice given agonistic anti-CD40. These studies provide new information regarding the role of target tissue expression of CD40 in development of autoimmunity and suggest that use of agonistic anti-CD40 for tumor therapy could result in autoimmune disease.


Subject(s)
Antibodies/physiology , Autoimmunity , CD40 Antigens/agonists , CD40 Antigens/immunology , Cell Proliferation , Epithelial Cells/immunology , Thyroid Gland/immunology , Thyroid Gland/metabolism , Animals , CD40 Antigens/biosynthesis , Epithelial Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred DBA , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Primary Cell Culture , Thyroid Gland/cytology
17.
Cancer Immunol Immunother ; 63(8): 847-57, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24878890

ABSTRACT

Malignant gliomas are heavily infiltrated by immature myeloid cells that mediate immunosuppression. Agonistic CD40 monoclonal antibody (mAb) has been shown to activate myeloid cells and promote antitumor immunity. Our previous study has also demonstrated blockade of cyclooxygenase-2 (COX-2) reduces immunosuppressive myeloid cells, thereby suppressing glioma development in mice. We therefore hypothesized that a combinatory strategy to modulate myeloid cells via two distinct pathways, i.e., CD40/CD40L stimulation and COX-2 blockade, would enhance anti-glioma immunity. We used three different mouse glioma models to evaluate therapeutic effects and underlying mechanisms of a combination regimen with an agonist CD40 mAb and the COX-2 inhibitor celecoxib. Treatment of glioma-bearing mice with the combination therapy significantly prolonged survival compared with either anti-CD40 mAb or celecoxib alone. The combination regimen promoted maturation of CD11b(+) cells in both spleen and brain, and enhanced Cxcl10 while suppressing Arg1 in CD11b(+)Gr-1(+) cells in the brain. Anti-glioma activity of the combination regimen was T-cell dependent because depletion of CD4(+) and CD8(+) cells in vivo abrogated the anti-glioma effects. Furthermore, the combination therapy significantly increased the frequency of CD8(+) T-cells, enhanced IFN-γ-production and reduced CD4(+)CD25(+)Foxp3(+) T regulatory cells in the brain, and induced tumor-antigen-specific T-cell responses in lymph nodes. Our findings suggest that the combination therapy of anti-CD40 mAb with celecoxib enhances anti-glioma activities via promotion of type-1 immunity both in myeloid cells and T-cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , CD40 Antigens/agonists , Glioma/drug therapy , Glioma/immunology , Myeloid Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , CD40 Antigens/immunology , Celecoxib , Cyclooxygenase 2 Inhibitors/administration & dosage , Female , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Pyrazoles/administration & dosage , Rats , Sulfonamides/administration & dosage , Transfection
18.
J Leukoc Biol ; 115(6): 1084-1093, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38372596

ABSTRACT

The cell surface molecule CD40 is a member of the tumor necrosis factor receptor superfamily and is broadly expressed by immune cells including B cells, dendritic cells, and monocytes, as well as other normal cells and some malignant cells. CD40 is constitutively expressed on antigen-presenting cells, and ligation promotes functional maturation, leading to an increase in antigen presentation and cytokine production, and a subsequent increase in the activation of antigen-specific T cells. It is postulated that CD40 agonists can mediate both T cell-dependent and T cell-independent immune mechanisms of tumor regression in mice and patients. In addition, it is believed that CD40 activation also promotes apoptotic death of tumor cells and that the presence of the molecule on the surface of cancer cells is an important factor in the generation of tumor-specific T cell responses that contribute to tumor cell elimination. Notably, CD40 agonistic therapies were evaluated in patients with solid tumors and hematologic malignancies with reported success as a single agent. Preclinical studies have shown that subcutaneous administration of CD40 agonistic antibodies reduces systemic toxicity and elicits a stronger and localized pharmacodynamic response. Two independent studies in cynomolgus macaque (Macaca fascicularis) were performed to further evaluate potentially immunotoxicological effects associated with drug-induced adverse events seen in human subjects. Studies conducted in monkeys showed that when selicrelumab is administered at doses currently used in clinical trial patients, via subcutaneous injection, it is safe and effective at stimulating a systemic immune response.


Subject(s)
CD40 Antigens , Macaca fascicularis , Animals , CD40 Antigens/agonists , CD40 Antigens/immunology , Humans , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal/pharmacology , Neoplasms/immunology , Neoplasms/drug therapy
19.
Expert Opin Biol Ther ; 24(5): 351-363, 2024 May.
Article in English | MEDLINE | ID: mdl-38764393

ABSTRACT

INTRODUCTION: There is a need for new therapies that can enhance response rates and broaden the number of cancer indications where immunotherapies provide clinical benefit. CD40 targeting therapies provide an opportunity to meet this need by promoting priming of tumor-specific T cells and reverting the suppressive tumor microenvironment. This is supported by emerging clinical evidence demonstrating the benefits of immunotherapy with CD40 antibodies in combination with standard of care chemotherapy. AREAS COVERED: This review is focused on the coming wave of next-generation CD40 agonists aiming to improve efficacy and safety, using new approaches and formats beyond monospecific antibodies. Further, the current understanding of the role of different CD40 expressing immune cell populations in the tumor microenvironment is reviewed. EXPERT OPINION: There are multiple promising next-generation approaches beyond monospecific antibodies targeting CD40 in immuno-oncology. Enhancing efficacy is the most important driver for this development, and approaches that maximize the ability of CD40 to both remodel the tumor microenvironment and boost the anti-tumor T cell response provide great opportunities to benefit cancer patients. Enhanced understanding of the role of different CD40 expressing immune cells in the tumor microenvironment may facilitate more efficient clinical development of these compounds.


Subject(s)
CD40 Antigens , Immunotherapy , Neoplasms , Tumor Microenvironment , Humans , CD40 Antigens/agonists , CD40 Antigens/immunology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy , Animals , Tumor Microenvironment/immunology
20.
Biochem Biophys Res Commun ; 436(3): 467-72, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23751348

ABSTRACT

Activation of CD40 by CD40L results in diverse effects on different malignant cells, causing either promotion of survival, growth and resistance to chemotherapy, or induction of cytostasis and apoptosis. The molecular mechanisms underlying CD40-mediated growth regulation and apoptosis induction in cancer cell are not fully understood. In this study, we investigated the role of NF-κB activation in CD40-mediated cytotoxicity in cancer cells. The results show that activation of CD40 by recombinant soluble CD40 ligand (rsCD40L) readily induced NF-κB activation and blocking NF-κB significantly enhanced rsCD40L-induced apoptosis in cancer cells. Importantly, autocrine of TNF-α induced by rsCD40L was indispensable for both NF-κB activation and cytotoxicity induction, establishing a dual role of autocrine TNF-α that constitutes both pro-apoptotic and anti-apoptotic arms of CD40 signaling. Our results indicate that autocrine TNF-α-mediated NF-κB activation is a determinant for cancer cells' evasion of CD40L-induced cytotoxicity and blocking NF-κB may have potential for improve the value of CD40 as an anticancer agent.


Subject(s)
Apoptosis , CD40 Antigens/metabolism , NF-kappa B/immunology , Tumor Escape , Tumor Necrosis Factor-alpha/immunology , Antibodies, Neutralizing/immunology , Antineoplastic Agents/pharmacology , Autocrine Communication , Benzoquinones/pharmacology , CD40 Antigens/agonists , CD40 Antigens/immunology , CD40 Ligand/metabolism , CD40 Ligand/pharmacology , Cell Line, Tumor , Humans , I-kappa B Kinase/antagonists & inhibitors , Lactams, Macrocyclic/pharmacology , Luteolin/pharmacology , NF-kappa B/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL