Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Res ; 19(8): 1422-1436, 2021 08.
Article in English | MEDLINE | ID: mdl-33888600

ABSTRACT

Only a subset of patients responds to immune checkpoint blockade (ICB) in melanoma. A preclinical model recapitulating the clinical activity of ICB would provide a valuable platform for mechanistic studies. We used melanoma tumors arising from an Hgftg;Cdk4R24C/R24C genetically engineered mouse (GEM) model to evaluate the efficacy of an anti-mouse PD-L1 antibody similar to the anti-human PD-L1 antibodies durvalumab and atezolizumab. Consistent with clinical observations for ICB in melanoma, anti-PD-L1 treatment elicited complete and durable response in a subset of melanoma-bearing mice. We also observed tumor growth delay or regression followed by recurrence. For early treatment assessment, we analyzed gene expression profiles, T-cell infiltration, and T-cell receptor (TCR) signatures in regressing tumors compared with tumors exhibiting no response to anti-PD-L1 treatment. We found that CD8+ T-cell tumor infiltration corresponded to response to treatment, and that anti-PD-L1 gene signature response indicated an increase in antigen processing and presentation, cytokine-cytokine receptor interaction, and natural killer cell-mediated cytotoxicity. TCR sequence data suggest that an anti-PD-L1-mediated melanoma regression response requires not only an expansion of the TCR repertoire that is unique to individual mice, but also tumor access to the appropriate TCRs. Thus, this melanoma model recapitulated the variable response to ICB observed in patients and exhibited biomarkers that differentiate between early response and resistance to treatment, providing a valuable platform for prediction of successful immunotherapy. IMPLICATIONS: Our melanoma model recapitulates the variable response to anti-PD-L1 observed in patients and exhibits biomarkers that characterize early antibody response, including expansion of the TCR repertoire.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Biomarkers/metabolism , Melanoma/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , Antibodies, Monoclonal/pharmacology , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Melanoma/drug therapy , Mice
2.
J Immunol ; 204(11): 2931-2939, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32321754

ABSTRACT

During human pregnancy, proinflammatory responses in the placenta can cause severe fetal complications, including growth restriction, preterm birth, and stillbirth. Villitis of unknown etiology (VUE), an inflammatory condition characterized by the infiltration of maternal CD8+ T cells into the placenta, is hypothesized to be secondary to either a tissue rejection response to the haploidentical fetus or from an undiagnosed infection. In this study, we characterized the global TCR ß-chain profile in human T cells isolated from placentae diagnosed with VUE compared with control and infectious villitis-placentae by immunoSEQ. Immunosequencing demonstrated that VUE is driven predominantly by maternal T cell infiltration, which is significantly different from controls and infectious cases; however, these T cell clones show very little overlap between subjects. Mapping TCR clones to common viral epitopes (CMV, EBV, and influenza A) demonstrated that Ag specificity in VUE was equal to controls and significantly lower than CMV-specific clones in infectious villitis. Our data indicate VUE represents an allograft response, not an undetected infection. These observations support the development of screening methods to predict those at risk for VUE and the use of specific immunomodulatory therapies during gestation to improve outcomes in affected fetuses.


Subject(s)
Chorionic Villi/immunology , Genes, T-Cell Receptor beta/genetics , Graft Rejection/immunology , Inflammation/immunology , Placenta Diseases/immunology , Pregnancy/immunology , T-Lymphocytes/immunology , Adult , Allografts/immunology , Antigens, Viral/immunology , Cell Movement , Cohort Studies , Epitopes, T-Lymphocyte/immunology , Female , Fetus , HLA Antigens/immunology , Humans , Young Adult
3.
Clin Cancer Res ; 26(1): 71-81, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31694832

ABSTRACT

PURPOSE: Pelareorep is an intravenously delivered oncolytic reovirus that can induce a T-cell-inflamed phenotype in pancreatic ductal adenocarcinoma (PDAC). Tumor tissues from patients treated with pelareorep have shown reovirus replication, T-cell infiltration, and upregulation of PD-L1. We hypothesized that pelareorep in combination with pembrolizumab and chemotherapy in patients with PDAC would be safe and effective. PATIENTS AND METHODS: A phase Ib single-arm study enrolled patients with PDAC who progressed after first-line treatment. Patients received pelareorep, pembrolizumab, and either 5-fluorouracil, gemcitabine, or irinotecan until disease progression or unacceptable toxicity. Study objectives included safety and dose-limiting toxicities, tumor response, evaluation for reovirus replication, and immune analysis in peripheral blood and tumor biopsies. RESULTS: Eleven patients were enrolled. Disease control was achieved in three of the 10 efficacy-evaluable patients. One patient achieved partial response for 17.4 months. Two additional patients achieved stable disease, lasting 9 and 4 months, respectively. Treatment was well tolerated, with mostly grade 1 or 2 treatment-related adverse events, including flu-like symptoms. Viral replication was observed in on-treatment tumor biopsies. T-cell receptor sequencing from peripheral blood revealed the creation of new T-cell clones during treatment. High peripheral clonality and changes in the expression of immune genes were observed in patients with clinical benefit. CONCLUSIONS: Pelareorep and pembrolizumab added to chemotherapy did not add significant toxicity and showed encouraging efficacy. Further evaluation of pelareorep and anti-PD-1 therapy is ongoing in follow-up studies. This research highlights the potential utility of several pretreatment and on-treatment biomarkers for pelareorep therapy warranting further investigation.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Oncolytic Virotherapy/methods , Oncolytic Viruses , Pancreatic Neoplasms/therapy , Adenocarcinoma/pathology , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Combined Modality Therapy , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Female , Fluorouracil/administration & dosage , Humans , Irinotecan/administration & dosage , Leucovorin/administration & dosage , Male , Maximum Tolerated Dose , Middle Aged , Pancreatic Neoplasms/pathology , Patient Safety , Treatment Outcome , Gemcitabine
4.
Oncotarget ; 7(14): 17565-78, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27007050

ABSTRACT

The success of adoptive T cell-based immunotherapy (ACT) in cancer is limited in part by the accumulation of myeloid-derived suppressor cells (MDSC), which block several T cell functions, including T cell proliferation and the expression of various cytotoxic mediators. Paradoxically, the inhibition of CD8+ T cell differentiation into cytotoxic populations increased their efficacy after ACT into tumor-bearing hosts. Therefore, we aimed to test the impact of conditioning CD8+ T cells with MDSC on their differentiation potential and ACT efficacy. Our results indicate that MDSC impaired the progression of CD8+ T cells into effector populations, without altering their activation status, production of IL-2, or signaling through the T cell receptor. In addition, culture of CD8+ T cells with MDSC resulted in an increased ACT anti-tumor efficacy, which correlated with a higher frequency of the transferred T cells and elevated IFNγ production. Interestingly, activated CD62L+ CD8+ T cells were responsible for the enhanced anti-tumor activity showed by MDSC-exposed T cells. Additional results showed a decreased protein synthesis rate and lower activity of the mammalian/mechanistic target of rapamycin (mTOR) in T cells conditioned with MDSC. Silencing of the negative mTOR regulator tuberous sclerosis complex-2 in T cells co-cultured with MDSC restored mTOR activity, but resulted in T cell apoptosis. These results indicate that conditioning of T cells with MDSC induces stress survival pathways mediated by a blunted mTOR signaling, which regulated T cell differentiation and ACT efficacy. Continuation of this research will enable the development of better strategies to increase ACT responses in cancer.


Subject(s)
Immunotherapy, Adoptive/methods , Myeloid Cells/immunology , T-Lymphocytes/immunology , Animals , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/therapy , Cell Communication/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Female , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Thymoma/immunology , Thymoma/therapy , Thymus Neoplasms/immunology , Thymus Neoplasms/therapy
5.
Cancer Res ; 75(2): 275-83, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25406192

ABSTRACT

Enzymatic depletion of the nonessential amino acid l-Arginine (l-Arg) in patients with cancer by the administration of a pegylated form of the catabolic enzyme arginase I (peg-Arg I) has shown some promise as a therapeutic approach. However, l-Arg deprivation also suppresses T-cell responses in tumors. In this study, we sought to reconcile these observations by conducting a detailed analysis of the effects of peg-Arg I on normal T cells. Strikingly, we found that peg-Arg I blocked proliferation and cell-cycle progression in normal activated T cells without triggering apoptosis or blunting T-cell activation. These effects were associated with an inhibition of aerobic glycolysis in activated T cells, but not with significant alterations in mitochondrial oxidative respiration, which thereby regulated survival of T cells exposed to peg-Arg I. Further mechanistic investigations showed that the addition of citrulline, a metabolic precursor for l-Arg, rescued the antiproliferative effects of peg-Arg I on T cells in vitro. Moreover, serum levels of citrulline increased after in vivo administration of peg-Arg I. In support of the hypothesis that peg-Arg I acted indirectly to block T-cell responses in vivo, peg-Arg I inhibited T-cell proliferation in mice by inducing accumulation of myeloid-derived suppressor cells (MDSC). MDSC induction by peg-Arg I occurred through the general control nonrepressed-2 eIF2α kinase. Moreover, we found that peg-Arg I enhanced the growth of tumors in mice in a manner that correlated with higher MDSC numbers. Taken together, our results highlight the risks of the l-Arg-depleting therapy for cancer treatment and suggest a need for cotargeting MDSC in such therapeutic settings.


Subject(s)
Arginine/immunology , Carcinoma, Lewis Lung/immunology , Myeloid Cells/immunology , T-Lymphocytes/immunology , Animals , Arginine/administration & dosage , Arginine/deficiency , Arginine/metabolism , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/therapy , Energy Metabolism/drug effects , Immunotherapy, Adoptive , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Polyethylene Glycols/administration & dosage , T-Lymphocytes/drug effects
6.
Immunity ; 41(3): 389-401, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25238096

ABSTRACT

Adaptation of malignant cells to the hostile milieu present in tumors is an important determinant of their survival and growth. However, the interaction between tumor-linked stress and antitumor immunity remains poorly characterized. Here, we show the critical role of the cellular stress sensor C/EBP-homologous protein (Chop) in the accumulation and immune inhibitory activity of tumor-infiltrating myeloid-derived suppressor cells (MDSCs). MDSCs lacking Chop had decreased immune-regulatory functions and showed the ability to prime T cell function and induce antitumor responses. Chop expression in MDSCs was induced by tumor-linked reactive oxygen and nitrogen species and regulated by the activating-transcription factor-4. Chop-deficient MDSCs displayed reduced signaling through CCAAT/enhancer-binding protein-ß, leading to a decreased production of interleukin-6 (IL-6) and low expression of phospho-STAT3. IL-6 overexpression restored immune-suppressive activity of Chop-deficient MDSCs. These findings suggest the role of Chop in tumor-induced tolerance and the therapeutic potential of targeting Chop in MDSCs for cancer immunotherapy.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/immunology , T-Lymphocytes/immunology , Transcription Factor CHOP/genetics , Tumor Escape/immunology , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Cell Line, Tumor , Cell Proliferation , Endothelial Cells/metabolism , Female , Interleukin-6/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Neoplasms , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Reactive Nitrogen Species/immunology , Reactive Oxygen Species/immunology , STAT3 Transcription Factor/biosynthesis , Transcription Factor CHOP/biosynthesis
7.
Cancer Immunol Res ; 2(8): 800-11, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24830414

ABSTRACT

An impaired antitumor immunity is found in patients with cancer and represents a major obstacle in the successful development of different forms of immunotherapy. Signaling through Notch receptors regulates the differentiation and function of many cell types, including immune cells. However, the effect of Notch in CD8(+) T-cell responses in tumors remains unclear. Thus, we aimed to determine the role of Notch signaling in CD8(+) T cells in the induction of tumor-induced suppression. Our results using conditional knockout mice show that Notch-1 and Notch-2 were critical for the proliferation and IFNγ production of activated CD8(+) T cells and were significantly decreased in tumor-infiltrating T cells. Conditional transgenic expression of Notch-1 intracellular domain (N1IC) in antigen-specific CD8(+) T cells did not affect activation or proliferation of CD8(+) T cells, but induced a central memory phenotype and increased cytotoxicity effects and granzyme B levels. Consequently, a higher antitumor response and resistance to tumor-induced tolerance were found after adoptive transfer of N1IC-transgenic CD8(+) T cells into tumor-bearing mice. Additional results showed that myeloid-derived suppressor cells (MDSC) blocked the expression of Notch-1 and Notch-2 in T cells through nitric oxide-dependent mechanisms. Interestingly, N1IC overexpression rendered CD8(+) T cells resistant to the tolerogenic effect induced by MDSC in vivo. Together, the results suggest the key role of Notch in the suppression of CD8(+) T-cell responses in tumors and the therapeutic potential of N1IC in antigen-specific CD8(+) T cells to reverse T-cell suppression and increase the efficacy of T cell-based immunotherapies in cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive , Neoplasms/therapy , Receptor, Notch1/immunology , Animals , Antigens/immunology , Cell Line, Tumor , Immune Tolerance , Mice, Transgenic , Neoplasms/immunology , Neoplasms/pathology , Ovalbumin/immunology , Receptor, Notch2/immunology , Signal Transduction , Tumor Burden
8.
Int J Cancer ; 134(12): 2853-64, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24259296

ABSTRACT

The accumulation of myeloid-derived suppressor cells (MDSC) in tumor-bearing hosts is a hallmark of malignancy-associated inflammation and a major mediator for the induction of T cell suppression in cancer. MDSC can be divided phenotypically into granulocytic (G-MDSC) and monocytic (Mo-MDSC) subgroups. Several mechanisms mediate the induction of T cell anergy by MDSC; however, the specific role of these pathways in the inhibitory activity of MDSC subpopulations remains unclear. Therefore, we aimed to determine the effector mechanisms by which subsets of tumor-infiltrating MDSC block T cell function. We found that G-MDSC had a higher ability to impair proliferation and expression of effector molecules in activated T cells, as compared to Mo-MDSC. Interestingly, both MDSC subgroups inhibited T cells through nitric oxide (NO)-related pathways, but expressed different effector inhibitory mechanisms. Specifically, G-MDSC impaired T cells through the production of peroxynitrites (PNT), while Mo-MDSC suppressed by the release of NO. The production of PNT in G-MDSC depended on the expression of gp91(phox) and endothelial NO synthase (eNOS), while inducible NO synthase (iNOS) mediated the generation of NO in Mo-MDSC. Deletion of eNOS and gp91(phox) or scavenging of PNT blocked the suppressive function of G-MDSC and induced anti-tumoral effects, without altering Mo-MDSC inhibitory activity. Furthermore, NO-scavenging or iNOS knockdown prevented Mo-MDSC function, but did not affect PNT production or suppression by G-MDSC. These results suggest that MDSC subpopulations utilize independent effector mechanisms to regulate T cell function. Inhibition of these pathways is expected to specifically block MDSC subsets and overcome immune suppression in cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Granulocytes/immunology , Monocytes/immunology , Nitric Oxide/metabolism , Peroxynitrous Acid/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Lymphocyte Activation/immunology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , NADPH Oxidase 2 , NADPH Oxidases/genetics , Neoplasms/immunology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , Nitrites/metabolism , Peroxynitrous Acid/biosynthesis , Reactive Oxygen Species/metabolism , Signal Transduction/immunology
9.
Immunol Invest ; 41(6-7): 614-34, 2012.
Article in English | MEDLINE | ID: mdl-23017138

ABSTRACT

Patients with cancer have an impaired T cell response that can decrease the potential therapeutic benefit of cancer vaccines and other forms of immunotherapy. The establishment of a chronic inflammatory environment in patients with cancer plays a critical role in the induction of T cell dysfunction. The accumulation of myeloid-derived suppressor cells (MDSC) in tumor bearing hosts is a hallmark of malignancy-associated inflammation and a major mediator of the induction of T cell suppression in cancer. Recent findings in tumor bearing mice and cancer patients indicate that the increased metabolism of L-Arginine (L-Arg) by MDSC producing Arginase I inhibits T cell lymphocyte responses. Here, we discuss some of the most recent concepts of how MDSC expressing Arginase I may regulate T cell function in cancer and suggest possible therapeutic interventions to overcome this inhibitory effect.


Subject(s)
Arginase/immunology , Arginine/metabolism , Gene Expression Regulation, Neoplastic , Inflammation/metabolism , Myeloid Progenitor Cells/metabolism , Neoplasm Proteins/immunology , Neoplasms/metabolism , Animals , Arginase/genetics , Cytokines/genetics , Cytokines/immunology , Humans , Immune Tolerance , Inflammation/immunology , Inflammation/pathology , Mice , Myeloid Progenitor Cells/immunology , Neoplasm Proteins/genetics , Neoplasms/immunology , Neoplasms/pathology , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...