Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 15(719): eadj0740, 2023 10 25.
Article in English | MEDLINE | ID: mdl-37878675

ABSTRACT

Recurrent respiratory papillomatosis (RRP) is a rare, debilitating neoplastic disorder caused by chronic infection with human papillomavirus (HPV) type 6 or 11 and characterized by growth of papillomas in the upper aerodigestive tract. There is no approved medical therapy, and patients require repeated debulking procedures to maintain voice and airway function. PRGN-2012 is a gorilla adenovirus immune-therapeutic capable of enhancing HPV 6/11-specific T cell immunity. This first-in-human, phase 1 study (NCT04724980) of adjuvant PRGN-2012 treatment in adult patients with severe, aggressive RRP demonstrates the overall safety and clinically meaningful benefit observed with PRGN-2012, with a 50% complete response rate in patients treated at the highest dose. Responders demonstrate greater expansion of peripheral HPV-specific T cells compared with nonresponders. Additional correlative studies identify an association between reduced baseline papilloma HPV gene expression, greater interferon responses and expression of CXCL9 and CXCL10, and greater papilloma T cell infiltration in responders. Conversely, nonresponders were characterized by greater HPV and CXCL8 gene expression, increased neutrophilic cell infiltration, and reduced T cell papilloma infiltration. These results suggest that papilloma HPV gene expression may regulate interferon signaling and chemokine expression profiles within the tumor microenvironment that cooperate to govern clinical response to therapeutic HPV vaccination in patients with respiratory papillomatosis.


Subject(s)
Papilloma , Papillomavirus Infections , Respiratory Tract Infections , Adult , Humans , Papillomavirus Infections/therapy , Papillomavirus Infections/pathology , Tumor Microenvironment , Respiratory Tract Infections/therapy , Interferons , Papilloma/therapy , Papilloma/pathology , Vaccination
2.
Int J Mol Sci ; 24(13)2023 Jun 24.
Article in English | MEDLINE | ID: mdl-37445745

ABSTRACT

This study investigated the role of IL-35 in systemic sclerosis (SSc) patients, focusing on CD4+ T cell response and immunomodulatory cytokine production. By comparing the cytokine levels in healthy donors (HD) and SSc patients using ELISAs, we found a significantly lower plasma IL-35 concentration in the SSc patients (52.1 ± 5.6 vs. 143 ± 11.1, p < 0.001). Notably, the IL-35 levels showed a negative correlation with TGF-ß (p < 0.001) and IL-17 (p = 0.04). Assessing the IL-35R expression across cell types in the SSc patients and HDs via flow cytometry, we found higher levels on monocytes (40.7 + 5.7 vs. 20.3 ± 1.9, p < 0.001) and lower levels on CD8+ T cells (61.8 ± 9.2 vs. 83.4 ± 0.8, p < 0.05) in the SSc patients. The addition of recombinant IL-35 to stimulated peripheral blood mononuclear cells reduced the IL-17+CD4+ T cell percentage (9.0 ± 1.5 vs. 4.8 ± 0.7, p < 0.05) and increased the IL-35+CD4+ T percentage (4.1 ± 2.3 vs. 10.2 ± 0.8, p < 0.001). In a Treg:Tresponder cell Sco-culture assay with HD and SSc samples, rIL35 decreased the cell proliferation and levels of IL-17A (178.2 ± 30.5 pg/mL vs. 37.4 ± 6.4 pg/mL, p < 0.001) and TGF-ß (4194 ± 777 pg/mL vs. 2413 ± 608 pg/mL, p < 0.01). Furthermore, we observed a positive correlation between the modified Rodnan skin score (mRSS) and TGF-ß (p < 0.001), while there was a negative correlation between mRSS and IL-35 (p = 0.004). Interestingly, higher levels of plasmatic IL-35 were detected in individuals with limited disease compared to those with diffuse disease (60.1 ± 8.0 vs. 832.3 ± 4.1, p < 0.05). These findings suggest that IL-35 exhibits anti-inflammatory properties in SSc and it may serve as a marker for disease severity and a therapeutic target.


Subject(s)
Interleukin-17 , Scleroderma, Systemic , Humans , Interleukin-17/metabolism , Leukocytes, Mononuclear/metabolism , Scleroderma, Systemic/metabolism , Cytokines/metabolism , Transforming Growth Factor beta
3.
Immunology ; 168(4): 610-621, 2023 04.
Article in English | MEDLINE | ID: mdl-36273280

ABSTRACT

Pulmonary fibrosis involves various types of immune cells and soluble mediators, including TGF-ß and IL-35, a recently identified heterodimeric cytokine that belongs to the IL-12 cytokine family. However, the effect of regulatory IL-35 may play an important role in fibrotic diseases. The aim of this paper is to explore the immunoregulatory role of IL-35 in the development of fibrosis in interstitial lung disease (ILD). To gain a better understanding of this issue, the concentrations of IL-35 and different profibrotic cytokines in fibrotic (F-ILD) and non-fibrotic (NF-ILD) patients by ELISA were compared to that of intracellular IL-35 and IL-17 on CD4+ T cells stimulated in the presence of BAL or with different ratios of recombinant IL-35 (rIL-35) and TGF-ß (rTGF-ß), which were evaluated by flow cytometry. We observed that BAL concentration of IL-35 was lower in F patients (p < 0.001) and was negatively correlated with concentrations of TGF-ß (p < 0.001) and IL-17 (p < 0.001). In supplemented cell cultures, BAL from NF but not F patients enhanced the percentage of IL-35 + CD4+ T (p < 0.001) cells and decreased the percentage of IL-17 + CD4+ T cells (p < 0.001). The percentage of IL-35 + CD4+ T cells correlated positively with BAL concentration of IL-35 (p = 0.02), but correlated negatively with BAL concentrations of IL-17 (p = 0.007) and TGF-ß (p = 0.01). After adjusting the concentrations of recombinant cytokines to establish a TGF-ß: IL-35 ratio of 1:4, an enhanced percentage of IL-35 + CD4+ T cells (p < 0.001) but a decreased percentage of IL-17 + CD4+ T cells (p < 0.001) was observed. After adding recombinant IL-35 to the BAL from F patients until a 1:4 ratio of TGF-ß: IL-35 was reached, a significantly increased percentage of IL-35 + CD4+ T cells (p < 0.001) and a decreased percentage of IL-17 + CD4+ T cells (p = 0.003) was found. These results suggest that IL-35 may induce an anti-fibrotic response, regulating the effect of TGF-ß and the inflammatory response on CD4+ T cells. In addition, the TGF-ß: IL-35 ratio in BAL has been shown to be a potential biomarker to predict the outcome of F patients with ILD.


Subject(s)
Lung Diseases, Interstitial , Pulmonary Fibrosis , Humans , Interleukin-17 , Cytokines/analysis , Transforming Growth Factor beta , Bronchoalveolar Lavage Fluid
4.
NPJ Vaccines ; 6(1): 86, 2021 Jun 18.
Article in English | MEDLINE | ID: mdl-34145272

ABSTRACT

Activation of antigen-specific T-lymphocyte responses may be needed to cure disorders caused by chronic infection with low-risk human papillomavirus (lrHPV). Safe and effective adjuvant therapies for such disorders are needed. The safety and efficacy of a novel gorilla adenovirus vaccine expressing a protein designed to elicit immune responses directed against HPV6 and HPV11, PRGN-2012, was studied using in vitro stimulation of T lymphocytes from patients with recurrent respiratory papillomatosis, in vivo vaccination studies, and therapeutic studies in mice bearing tumors expressing lrHPV antigen. PRGN-2012 treatment induces lrHPV antigen-specific responses in patient T lymphocytes. Vaccination of wild-type mice induces E6-specific T-lymphocyte responses without toxicity. In vivo therapeutic vaccination of mice bearing established HPV6 E6 expressing tumors results in HPV6 E6-specific CD8+ T-lymphocyte immunity of sufficient magnitude to induce tumor growth delay. The clinical study of PRGN-2012 in patients with disorders caused by chronic infection with lrHPV is warranted.

5.
JCI Insight ; 6(7)2021 04 08.
Article in English | MEDLINE | ID: mdl-33651712

ABSTRACT

There are approximately 44,000 cases of human papillomavirus-associated (HPV-associated) cancer each year in the United States, most commonly caused by HPV types 16 and 18. Prophylactic vaccines successfully prevent healthy people from acquiring HPV infections via HPV-specific antibodies. In order to treat established HPV-associated malignancies, however, new therapies are necessary. Multiple recombinant gorilla adenovirus HPV vaccine constructs were evaluated in NSG-ß2m-/- peripheral blood mononuclear cell-humanized mice bearing SiHa, a human HPV16+ cervical tumor, and/or in the syngeneic HPV16+ TC-1 model. PRGN-2009 is a therapeutic gorilla adenovirus HPV vaccine containing multiple cytotoxic T cell epitopes of the viral oncoproteins HPV 16/18 E6 and E7, including T cell enhancer agonist epitopes. PRGN-2009 treatment reduced tumor volume and increased CD8+ and CD4+ T cells in the tumor microenvironment of humanized mice bearing the human cervical tumor SiHa. PRGN-2009 monotherapy in the syngeneic TC-1 model also reduced tumor volumes and weights, generated high levels of HPV16 E6-specific T cells, and increased multifunctional CD8+ and CD4+ T cells in the tumor microenvironment. These studies provide the first evaluation to our knowledge of a therapeutic gorilla adenovirus HPV vaccine, PRGN-2009, showing promising preclinical antitumor efficacy and induction of HPV-specific T cells, along with the rationale for its evaluation in clinical trials.


Subject(s)
Adenoviruses, Simian/genetics , Cancer Vaccines/pharmacology , Papillomavirus Vaccines/pharmacology , Uterine Cervical Neoplasms/drug therapy , Vaccines, Synthetic/pharmacology , Animals , CD8-Positive T-Lymphocytes , Cancer Vaccines/genetics , Cell Line, Tumor , Epitopes , Female , Human papillomavirus 16/immunology , Humans , Mice, Inbred C57BL , Neutrophils , Oncogene Proteins, Viral/immunology , Papillomavirus Vaccines/genetics , Repressor Proteins/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Microenvironment/drug effects , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology , Vaccines, Synthetic/genetics , Xenograft Model Antitumor Assays
6.
PLoS Negl Trop Dis ; 12(4): e0006404, 2018 04.
Article in English | MEDLINE | ID: mdl-29668679

ABSTRACT

A number of features at the host-parasite interface are reminiscent of those that are also observed at the host-tumor interface. Both cancer cells and parasites establish a tissue microenvironment that allows for immune evasion and may reflect functional alterations of various innate cells. Here, we investigated how the phenotype and function of human monocytes is altered by exposure to cancer cell lines and if these functional and phenotypic alterations parallel those induced by exposure to helminth parasites. Thus, human monocytes were exposed to three different cancer cell lines (breast, ovarian, or glioblastoma) or to live microfilariae (mf) of Brugia malayi-a causative agent of lymphatic filariasis. After 2 days of co-culture, monocytes exposed to cancer cell lines showed markedly upregulated expression of M1-associated (TNF-α, IL-1ß), M2-associated (CCL13, CD206), Mreg-associated (IL-10, TGF-ß), and angiogenesis associated (MMP9, VEGF) genes. Similar to cancer cell lines, but less dramatically, mf altered the mRNA expression of IL-1ß, CCL13, TGM2 and MMP9. When surface expression of the inhibitory ligands PDL1 and PDL2 was assessed, monocytes exposed to both cancer cell lines and to live mf significantly upregulated PDL1 and PDL2 expression. In contrast to exposure to mf, exposure to cancer cell lines increased the phagocytic ability of monocytes and reduced their ability to induce T cell proliferation and to expand Granzyme A+ CD8+ T cells. Our data suggest that despite the fact that helminth parasites and cancer cell lines are extraordinarily disparate, they share the ability to alter the phenotype of human monocytes.


Subject(s)
Brugia malayi/immunology , Filariasis/immunology , Immune Evasion , Monocytes/immunology , Monocytes/parasitology , Neoplasms/immunology , Animals , Brugia malayi/genetics , Brugia malayi/physiology , Cell Line, Tumor , Filariasis/parasitology , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lymphocyte Activation , Phagocytosis , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
7.
Sci Transl Med ; 10(424)2018 01 17.
Article in English | MEDLINE | ID: mdl-29343622

ABSTRACT

Antibodies targeting immune checkpoints are emerging as potent and viable cancer therapies, but not all patients respond to these as single agents. Concurrently targeting additional immunosuppressive pathways is a promising approach to enhance immune checkpoint blockade, and bifunctional molecules designed to target two pathways simultaneously may provide a strategic advantage over the combination of two single agents. M7824 (MSB0011359C) is a bifunctional fusion protein composed of a monoclonal antibody against programmed death ligand 1 (PD-L1) fused to the extracellular domain of human transforming growth factor-ß (TGF-ß) receptor II, which functions as a "trap" for all three TGF-ß isoforms. We demonstrate that M7824 efficiently, specifically, and simultaneously binds PD-L1 and TGF-ß. In syngeneic mouse models, M7824 suppressed tumor growth and metastasis more effectively than treatment with either an anti-PD-L1 antibody or TGF-ß trap alone; furthermore, M7824 extended survival and conferred long-term protective antitumor immunity. Mechanistically, the dual anti-immunosuppressive function of M7824 resulted in activation of both the innate and adaptive immune systems, which contributed to M7824's antitumor activity. Finally, M7824 was an effective combination partner for radiotherapy or chemotherapy in mouse models. Collectively, our preclinical data demonstrate that simultaneous blockade of the PD-L1 and TGF-ß pathways by M7824 elicits potent and superior antitumor activity relative to monotherapies.


Subject(s)
Antibodies, Monoclonal/immunology , Programmed Cell Death 1 Receptor/immunology , Transforming Growth Factor beta/immunology , Animals , Antibodies, Monoclonal/chemistry , Immunotherapy/methods , Mice , Programmed Cell Death 1 Receptor/chemistry , Transforming Growth Factor beta/chemistry
8.
AIDS ; 30(16): 2487-2493, 2016 10 23.
Article in English | MEDLINE | ID: mdl-27490642

ABSTRACT

OBJECTIVE: The programed death-1 (PD1)/programed death-ligand 1 (PD-L1) pathway plays a critical role in balancing immunity and host immunopathology. During chronic HIV/SIV infection, there is persistent immune activation accompanied by accumulation of virus-specific cells with terminally differentiated phenotypes and expression of regulatory receptors such as PD1. These observations led us to hypothesize that the PD1/PD-L1 pathway contributes to the functional dysregulation and ineffective viral control, and its blockade may be a potential immunotherapeutic target. METHODS: Lymph node biopsies from HIV-infected patients (n = 23) were studied for expression of PD1 and PD-L1. In addition, we assessed the safety and biological activity of a human anti-PD-L1 antibody (Avelumab) in chronically SIV-infected rhesus macaques. RESULTS: PD-L1 expression was observed in cells with myloid/macrophage morphology in HIV-infected lymph nodes. Administration of anti-PD-L1 was well tolerated, and no changes in body weights, hematologic, or chemistry parameters were observed during the study. Blockade of PD-L1 led to a trend of transient viral control after discontinuation of treatment. CONCLUSION: Administration of anti-PD-L1 in chronic SIV-infected rhesus macaques was well tolerated. Overall, these data warrant further investigation to assess the efficacy of anti-PD-L1 treatment on viral control in chronic SIV infection as a prelude to such therapy in humans.


Subject(s)
Antibodies, Monoclonal/adverse effects , B7-H1 Antigen/analysis , HIV Infections/pathology , Immunologic Factors/adverse effects , Lymph Nodes/pathology , Programmed Cell Death 1 Receptor/analysis , Simian Acquired Immunodeficiency Syndrome/pathology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Humans , Immunologic Factors/administration & dosage , Macaca mulatta , Pilot Projects , Treatment Outcome
9.
Cancer Immunol Res ; 4(5): 452-62, 2016 05.
Article in English | MEDLINE | ID: mdl-26921031

ABSTRACT

Bacillus Calmette-Guerin (BCG) is the standard of care for intravesical therapy for carcinoma in situ and non-muscle invasive, nonmetastatic human urothelial carcinoma. Although the responsiveness to this immunotherapeutic is believed to be linked with (i) a high number of somatic mutations and (ii) a large number of tumor-infiltrating lymphocytes, recent findings of the roles that inhibitory immune receptors and their ligands play in tumor evasion may provide insights into the limitations of the effectiveness of BCG and offer new targets for immune-based therapy. In this study, an aggressive, bioluminescent orthotopic bladder cancer model, MB49 tumor cells transfected with luciferase (MB49(luc)), was used to study the antitumor effects of avelumab, an antibody to PD-L1. MB49(luc) murine tumor cells form multifocal tumors on the mucosal wall of the bladder reminiscent of non-muscle invasive, nonmetastatic urothelial carcinomas. MB49(luc) bladder tumors are highly positive for the expression of PD-L1, and avelumab administration induced significant (P < 0.05) antitumor effects. These antitumor effects were more dependent on the presence of CD4 than CD8 T cells, as determined by in vivo immune cell depletions. The findings suggest that in this bladder tumor model, interruption of the immune-suppressive PD-1/PD-L1 complex releases a local adaptive immune response that, in turn, reduces tumor growth. This bladder tumor model can be used to further identify host antitumor immune mechanisms and evaluate combinations of immune-based therapies for carcinoma in situ and non-muscle invasive, nonmetastatic urothelial carcinoma, to provide the rationale for subsequent clinical studies. Cancer Immunol Res; 4(5); 452-62. ©2016 AACR.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Transitional Cell/drug therapy , Urinary Bladder Neoplasms/drug therapy , Adaptive Immunity , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Transitional Cell/immunology , Carcinoma, Transitional Cell/pathology , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Female , Immune Tolerance/immunology , Immunotherapy/methods , Mice, Inbred C57BL , Mycobacterium bovis/immunology , Neoplasm Transplantation , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
10.
J Immunol ; 194(3): 878-82, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25548231

ABSTRACT

CD4(+) regulatory T cells (Tregs) are critical for maintaining self-tolerance and function to prevent autoimmune disease. High densities of intratumoral Tregs are generally associated with poor patient prognosis, a correlation attributed to their broad immune-suppressive features. Two major populations of Tregs have been defined, thymically derived natural Tregs (nTregs) and peripherally induced Tregs (iTregs). However, the relative contribution of nTregs versus iTregs to the intratumoral Treg compartment remains controversial. Demarcating the proportion of nTregs versus iTregs has important implications in the design of therapeutic strategies to overcome their antagonistic effects on antitumor immune responses. We used epigenetic, phenotypic, and functional parameters to evaluate the composition of nTregs versus iTregs isolated from mouse tumor models and primary human tumors. Our findings failed to find evidence for extensive intratumoral iTreg induction. Rather, we identified a population of Foxp3-stable nTregs in tumors from mice and humans.


Subject(s)
Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Antigens, Surface/metabolism , Cell Line, Tumor , CpG Islands , DNA Methylation , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Humans , Immunophenotyping , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Transgenic , Neoplasms/metabolism , Transforming Growth Factor beta/metabolism
11.
Oncotarget ; 5(7): 1869-84, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24681847

ABSTRACT

Targeted delivery of IL-12 might turn this cytokine into a safer, more effective cancer therapeutic. Here we describe a novel immunocytokine, NHS-IL12, consisting of two molecules of IL-12 fused to a tumor necrosis-targeting human IgG1 (NHS76). The addition of the human IgG1 moiety resulted in a longer plasma half-life of NHS-IL12 than recombinant IL-12, and a selective targeting to murine tumors in vivo. Data from both in vitro assays using human PBMCs and in vivo primate studies showed that IFN-gamma production by immune cells is attenuated following treatment with the immunocytokine, suggesting an improved toxicity profile than seen with recombinant IL-12 alone. NHS-IL12 was superior to recombinant IL-12 when evaluated as an anti-tumor agent in three murine tumor models. Mechanistic studies utilizing immune cell subset-depleting antibodies, flow cytometric methods, and in vitro cytotoxicity and ELISA assays all indicated that the anti-tumor effects of NHS-IL12 were primarily CD8+ T cell-dependent and likely IL-12-mediated. Combining NHS-IL12 treatment with a cancer vaccine, radiation, or chemotherapy resulted in greater anti-tumor effects than each individual therapy alone. These preclinical findings provide a rationale for the clinical testing of this immunocytokine, both as a single agent and in combination with vaccines, radiation and chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Immunoglobulin G/pharmacology , Interferon-gamma/metabolism , Interleukin-12/pharmacology , Neoplasms, Experimental/therapy , Recombinant Fusion Proteins/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD8-Positive T-Lymphocytes , Cancer Vaccines/administration & dosage , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Docetaxel , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin G/therapeutic use , Immunomodulation , Indoles/administration & dosage , Interleukin-12/immunology , Interleukin-12/therapeutic use , Lymphocyte Activation/drug effects , Macaca fascicularis , Membrane Glycoproteins/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Engineering , Pyrroles/administration & dosage , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Sunitinib , Survival Rate , Taxoids/administration & dosage , Tumor Burden
12.
J Immunol ; 188(9): 4323-33, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22450814

ABSTRACT

Immunologic memory involving CD8(+) T cells is a hallmark of an adaptive Ag-specific immune response and constitutes a critical component of protective immunity. Designing approaches that enhance long-term T cell memory would, for the most part, fortify vaccines and enhance host protection against infectious diseases and, perhaps, cancer immunotherapy. A better understanding of the cellular programs involved in the Ag-specific T cell response has led to new approaches that target the magnitude and quality of the memory T cell response. In this article, we show that T cells from TCR transgenic mice for the nucleoprotein of influenza virus NP68 exhibit the distinct phases--priming, expansion, contraction, and memory--of an Ag-specific T cell response when exposed in vitro to the cognate peptide. Saracatinib, a specific inhibitor of Src family kinases, administered at low doses during the expansion or contraction phases, increased CD62L(high)/CD44(high) central memory CD8(+) T cells and IFN-γ production but suppressed immunity when added during the priming phase. These effects by saracatinib were not accompanied by the expected decline of Src family kinases but were accompanied by Akt-mammalian target of rapamycin suppression and/or mediated via another pathway. Increased central memory cells by saracatinib were recapitulated in mice using a poxvirus-based influenza vaccine, thus underscoring the importance of dose and timing of the inhibitor in the context of memory T cell differentiation. Finally, vaccine plus saracatinib treatment showed better protection against tumor challenge. The immune-potentiating effects on CD8(+) T cells by a low dose of saracatinib might afford better protection from pathogens or cancer when combined with vaccine.


Subject(s)
Benzodioxoles/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Immunologic Memory/drug effects , Quinazolines/pharmacology , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , Antigens, Viral/metabolism , CD8-Positive T-Lymphocytes/enzymology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cancer Vaccines/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Hyaluronan Receptors/metabolism , Immunologic Memory/genetics , Influenza A virus/genetics , Influenza A virus/immunology , Influenza A virus/metabolism , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , L-Selectin/genetics , L-Selectin/immunology , L-Selectin/metabolism , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics , src-Family Kinases/immunology , src-Family Kinases/metabolism
13.
Blood ; 117(15): 4032-40, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21304101

ABSTRACT

IL-15 has growth-promoting effects on select lymphoid subsets, including natural killer (NK) cells, NK T cells, intraepithelial lymphocytes (IELs), CD8 T cells, and γδ-T cells. Constitutive expression of murine IL-15 in IL-15-transgenic mice was reported to cause T-NK leukemia. We investigated whether IL-15 expression is sufficient for leukemic transformation using a human IL-15-transgenic (IL-15Tg) mouse model. We noted that 100% of the mice observed over a 2-year period (n > 150) developed fatal expansions of CD8 T cells with NK markers, and determined that these cells expressed IL-15 receptor alpha (IL-15Rα). The expression of IL-15Rα on CD8 T cells appears to be required for uncontrolled aggressive lymphoproliferation, because none of the IL-15Rα(-/-)-IL-15Tg mice that we followed for more than 2 years developed the fatal disease despite controlled expansion of CD8 T cells. In addition, in contrast to IL-15Tg mice, in which leukemia-like CD8 T cells expressed IL-15Rα persistently, acutely activated normal CD8 T cells only transiently expressed IL-15Rα. Inhibition of DNA methylation enabled sustained IL-15Rα expression induced by activation. We present a scenario for IL-15Tg mice in which CD8 T cells that acquire constitutive persistent IL-15Rα expression are at a selective advantage and become founder cells, outgrow other lymphocytes, and lead to the establishment of a leukemia-like condition.


Subject(s)
Autocrine Communication/immunology , CD8-Positive T-Lymphocytes/physiology , Interleukin-15 Receptor alpha Subunit/genetics , Interleukin-15/genetics , Leukemia, T-Cell , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Epigenesis, Genetic/physiology , Gene Silencing/physiology , Genetic Complementation Test , Homeostasis/immunology , Humans , Interleukin-15/metabolism , Interleukin-15 Receptor alpha Subunit/metabolism , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation
14.
Int J Cancer ; 127(7): 1603-13, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20091862

ABSTRACT

Small molecule BCL-2 inhibitors are being examined as monotherapy in phase I/II clinical trials for several types of tumors. However, few data are available about the effect of BCL-2 inhibitors on immune function. The aims of our study were to investigate the effect of a small molecule BCL-2 inhibitor on immune function and determine the most effective way of combining this inhibitor with a recombinant vaccine to treat tumors. The in vitro effect of the pan-BCL-2 inhibitor GX15-070 was assessed in mouse CD8 T lymphocytes at 2 different stages of activation as well as regulatory T lymphocytes (Treg). The in vivo effect of GX15-070 after recombinant vaccinia/fowlpox CEA-TRICOM vaccination was analyzed in tumor-infiltrating lymphocytes, and in splenocytes of mice bearing subcutaneous tumors. The therapeutic efficacy of such sequential therapy was measured as a reduction of pulmonary tumor nodules. Activated mature CD8 T lymphocytes were more resistant to GX15-070 as compared to early-activated cells. Treg function was significantly decreased after treatment with the BCL-2 inhibitor. In vivo, GX15-070 was given after vaccination so as to not negatively impact the induction of vaccine-mediated immunity, resulting in increased intratumoral activated CD8:Treg ratio and significant reduction of pulmonary tumor nodules. Our study is the first to show the effect of a small molecule BCL-2 inhibitor on the immune system and following a vaccine. It is also the first to demonstrate the efficacy of this sequence in reducing tumors in mouse models, providing a rationale for the design of combinational clinical studies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Vaccines, Synthetic/immunology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell Cycle , Flow Cytometry , Hyaluronan Receptors/immunology , Immunoblotting , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , Vaccines, Synthetic/therapeutic use
15.
Cancer Immunol Immunother ; 58(11): 1809-18, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19319531

ABSTRACT

This study demonstrates that CD8+ T cells in the tumor microenvironment display reduced functionality and hyporesponsiveness. TGF-beta contributed markedly to the tumor-infiltrating CD8+ T cells' (TILs) reduced functionality, which could be reversed using a small molecule TGF-beta inhibitor. Upon T-cell receptor (TCR) activation, the activation of ITK and ERK kinases were reduced in CD8+ TILs, as compared to splenic CD8+ T cells: TGF-beta inhibitor could reverse this phenomenon. This study demonstrates for the first time the association of the Spred-1 gene, an inhibitor of the Ras/MAPK pathway, with CD8+ TILs and TGF-beta activity. Spred-1 was upregulated in CD8+ TILs and TGF-beta enhanced the expression of Spred-1 in effector/memory CD8+ T cells and not in rested/memory CD8+ T cells. Based on these findings, this study supports the hypothesis that TGF-beta mediates an inhibitory mechanism on CD8+ TILs involving TCR-signaling blockade and the upregulation of Spred-1, thus implicating Spred-1 as a potential new target for future anti-tumor immune studies.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Lymphocytes, Tumor-Infiltrating/physiology , Neoplasms, Experimental/immunology , Receptors, Antigen, T-Cell/physiology , Repressor Proteins/physiology , Transforming Growth Factor beta/physiology , Adaptor Proteins, Signal Transducing , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic , Immune Tolerance , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Phosphorylation , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors
16.
Cancer Immunol Immunother ; 58(6): 955-65, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19009294

ABSTRACT

In chronic lymphocytic leukemia (CLL), malignant B cells and nonmalignant T cells exhibit dysfunction. We previously demonstrated that infection of CLL cells with modified vaccinia Ankara (MVA) expressing the costimulatory molecules B7-1, ICAM-1, and LFA-3 (designated TRICOM) increased expression of these costimulatory molecules on the surface of CLL cells and thus augmented their antigen-presenting capability. Here, we evaluate the effect of MVA-TRICOM-modified CLL cells on T cells. Following incubation with irradiated MVA-TRICOM-modified CLL cells, allogeneic and autologous CD4(+) and CD8(+) T cells expressed significantly higher levels of B7-1, ICAM-1, and LFA-3. We show that this increase was the result of physical acquisition from the antigen-presenting cells (APCs), and that purified T cells that acquired costimulatory molecules from MVA-TRICOM-modified CLL cells were able to stimulate the proliferation of untreated T cells. These results demonstrate for the first time that T cells from CLL patients can acquire multiple costimulatory molecules from autologous CLL cells and can then act as APCs themselves. Given the immunodeficiencies characteristic of CLL, enhancing the antigen-presenting function of CLL cells and T cells simultaneously could be a distinct advantage in the effort to elicit antitumor immune responses.


Subject(s)
Antigen-Presenting Cells/immunology , B7-1 Antigen/metabolism , CD58 Antigens/metabolism , Intercellular Adhesion Molecule-1/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , T-Lymphocytes/immunology , B7-1 Antigen/genetics , CD58 Antigens/genetics , Humans , Intercellular Adhesion Molecule-1/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Vaccinia virus/genetics
17.
Vaccine ; 27(7): 987-94, 2009 Feb 11.
Article in English | MEDLINE | ID: mdl-19110021

ABSTRACT

Tumor-associated antigens are weakly immunogenic. Human carcinoembryonic antigen (CEA) is overexpressed on a wide range of human carcinomas and represents an attractive target for cancer immunotherapy. This study analyzes the ability of a Saccharomyces cerevisiae vector containing the transgene encoding CEA (yeast-CEA) to activate human dendritic cells (DCs) and stimulate CEA-specific T-cell responses. We demonstrate for the first time that treatment with yeast-CEA can activate human DCs, resulting in increases in surface expression of CD80, CD83, CD54, CD58, and MHC class II, and increased production by DCs of IL-12p70, TNF-alpha, IFN-gamma, IL-8, IL-2, IL-13, IL-10, and IL-1beta. We also show that human DCs treated with yeast-CEA can activate CEA-specific T-cell lines and can act as antigen-presenting cells (APCs) to generate CEA-specific T-cell lines capable of lysing CEA(+) human tumor cells. Gene expression profiles of human DCs treated with yeast-CEA show increased expression of numerous genes involved in the production of chemokines and cytokines and their receptors, and genes related to antigen uptake, antigen presentation, and signal transduction.


Subject(s)
Carcinoembryonic Antigen/biosynthesis , Carcinoembryonic Antigen/immunology , Dendritic Cells/immunology , Saccharomyces cerevisiae/genetics , Antigens, CD/analysis , Carcinoembryonic Antigen/genetics , Cell Line, Tumor , Cytokines/biosynthesis , Cytotoxicity Tests, Immunologic , Dendritic Cells/chemistry , Gene Expression Profiling , Humans , Lymphocyte Activation , Saccharomyces cerevisiae/immunology , T-Lymphocytes, Cytotoxic/immunology
18.
Blood ; 112(4): 1290-8, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18541719

ABSTRACT

Patent lymphatic filariasis is characterized by antigen-specific T-cell unresponsiveness with diminished IFN-gamma and IL-2 production and defects in dendritic cell (DC) function. Because Toll-like receptors (TLRs) play an important role in pathogen recognition and TLR expression is diminished on B and T cells of filaria-infected individuals, we examined the effect of live microfilariae (mf) on expression and function of TLRs in human DCs. We show that mf-exposed monocyte-derived human DCs (mhDCs) demonstrate marked diminution of TLR3 and TLR4 mRNA expression compared with mf-unexposed mhDCs that translated into loss of function in response to appropriate TLR ligands. Exposure to mf significantly down-regulated production of IFN-alpha, MIP-1alpha, IL-12p70, and IL-1alpha following activation with poly I:C, and of IL-12p40 following activation with poly I:C or LPS. mRNA expression of MyD88, the adaptor molecule involved in TLR4 signaling, was significantly diminished in mhDCs after exposure to mf. Moreover, mf interfered with NF-kappaB activation (particularly p65 and p50) following stimulation with poly I:C or LPS. These data suggest that mf interfere with mhDC function by altering TLR expression and interfering with both MyD88-dependent signaling and a pathway that ultimately diminishes NF-kappaB activity. This down-regulated NF-kappaB activity impairs mhDC-produced cytokines needed for full T-cell activation.


Subject(s)
Dendritic Cells/parasitology , Helminths/immunology , Toll-Like Receptor 3/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Cells, Cultured , Cytokines/biosynthesis , Dendritic Cells/immunology , Down-Regulation , Humans , Lymphocyte Activation , Myeloid Differentiation Factor 88/genetics , NF-kappa B/metabolism , RNA, Messenger/analysis , Toll-Like Receptor 3/genetics , Toll-Like Receptor 4/genetics
19.
Cancer Res ; 68(10): 3835-43, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18483268

ABSTRACT

Overexpression of transforming growth factor beta (TGF-beta) is frequently associated with metastasis and poor prognosis, and TGF-beta antagonism has been shown to prevent metastasis in preclinical models with surprisingly little toxicity. Here, we have used the transplantable 4T1 model of metastatic breast cancer to address underlying mechanisms. We showed that efficacy of the anti-TGF-beta antibody 1D11 in suppressing metastasis was dependent on a synergistic combination of effects on both the tumor parenchyma and microenvironment. The main outcome was a highly significant enhancement of the CD8+ T-cell-mediated antitumor immune response, but effects on the innate immune response and on angiogenesis also contributed to efficacy. Treatment with 1D11 increased infiltration of natural killer cells and T cells at the metastatic site, and enhanced expression of coactivators (NKG2D) and cytotoxic effectors (perforin and granzyme B) on CD8+ T cells. On the tumor cells, increased expression of an NKG2D ligand (Rae1gamma) and of a death receptor (TNFRSF1A) contributed to enhanced immune cell-mediated recognition and lysis. The data suggest that elevated TGF-beta expression in the tumor microenvironment modulates a complex web of intercellular interactions that aggregately promote metastasis and progression. TGF-beta antibodies reverse this effect, and the absence of a major effect of TGF-beta antagonism on any one cell compartment may be critical for a good therapeutic window and the avoidance of autoimmune complications.


Subject(s)
Immunotherapy/methods , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/therapy , Transforming Growth Factor beta/chemistry , Animals , Apoptosis , CD8-Positive T-Lymphocytes/metabolism , Culture Media, Conditioned/pharmacology , Killer Cells, Natural/metabolism , Ligands , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Prognosis , Transfection
20.
J Immunol ; 180(9): 5871-81, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18424706

ABSTRACT

This study demonstrates, for the first time, that murine regulatory T (Treg) cells in the tumor microenvironment display both enhanced proliferation and reduced functionality. This enhanced proliferation, combined with decreased apoptosis, leads to an intratumoral accumulation of Treg cells with a unique phenotype: CD4(+)CD25(+)FoxP3(+)GITR(high)CD27(low)CD62L(-). The loss of functionality is associated with down-regulation of the TCR signaling complex, including IL-2-inducible T cell kinase. It is also demonstrated that tumor-infiltrating Treg cells have impaired TCR-mediated signaling and calcium influx. Based on these findings, this study supports the hypothesis that 1) tumor-infiltrating Treg cells lose functionality due to their diminished ability to become effectively activated and 2) intratumoral accumulation of Treg cells may compensate for the impaired functionality, thus maintaining immune tolerance to the tumor.


Subject(s)
Apoptosis/immunology , Calcium Signaling/immunology , Cell Proliferation , Neoplasms, Experimental/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Down-Regulation/immunology , Female , Immune Tolerance , Lymphocyte Activation , Mice , Mice, Knockout , Neoplasms, Experimental/enzymology , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...