Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
J Immunother Cancer ; 11(10)2023 10.
Article in English | MEDLINE | ID: mdl-37852737

ABSTRACT

BACKGROUND: Preclinical studies showed metformin reduces exhaustion of tumor-infiltrating lymphocytes and potentiates programmed cell death protein-1 (PD-1) blockade. We hypothesized that metformin with nivolumab would elicit potent antitumor and immune modulatory activity in metastatic microsatellite stable (MSS) colorectal cancer (CRC). We evaluated this hypothesis in a phase II study. METHODS: Nivolumab (480 mg) was administered intravenously every 4 weeks while metformin (1000 mg) was given orally, two times per day following a 14-day metformin only lead-in phase. Patients ≥18 years of age, with previously treated, stage IV MSS CRC, and Eastern Cooperative Oncology Group 0-1, having received no prior anti-PD-1 agent were eligible. The primary endpoint was overall response rate with secondary endpoints of overall survival (OS) and progression-free survival (PFS). Correlative studies using paired pretreatment/on-treatment biopsies and peripheral blood evaluated a series of immune biomarkers in the tumor microenvironment and systemic circulation using ChipCytometry and flow cytometry. RESULTS: A total of 24 patients were enrolled, 6 patients were replaced per protocol, 18 patients had evaluable disease. Of the 18 evaluable patients, 11/18 (61%) were women and the median age was 58 (IQR 50-67). Two patients had stable disease, but no patients had objective response, hence the study was stopped for futility. Median OS and PFS was 5.2 months (95% CI (3.2 to 11.7)) and 2.3 months (95% CI (1.7 to 2.3)). Most common grade 3/4 toxicities: Anemia (n=2), diarrhea (n=2), and fever (n=2). Metformin alone failed to increase the infiltration of T-cell subsets in the tumor, but combined metformin and nivolumab increased percentages of tumor-infiltrating leukocytes (p=0.031). Dual treatment also increased Tim3+ levels in patient tissues and decreased naïve CD8+T cells (p=0.0475). CONCLUSIONS: Nivolumab and metformin were well tolerated in patients with MSS CRC but had no evidence of efficacy. Correlative studies did not reveal an appreciable degree of immune modulation from metformin alone, but showed trends in tumorous T-cell infiltration as a result of dual metformin and PD-1 blockade despite progression in a majority of patients.


Subject(s)
Colorectal Neoplasms , Metformin , Humans , Female , Middle Aged , Male , Nivolumab/adverse effects , Programmed Cell Death 1 Receptor , Metformin/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Microsatellite Repeats , Tumor Microenvironment
2.
Curr Treat Options Oncol ; 24(7): 892-905, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37191906

ABSTRACT

OPINION STATEMENT: With improvements in treatment and survival from prostate cancer, comorbid cardiac conditions will significantly impact overall morbidity and mortality from prostate cancer. Hypertension is a well-established cardiovascular risk factor that increases the risk of heart failure, myocardial infarction, and stroke. Therapies used in the treatment of prostate cancer, including GnRH agonists, GnRH antagonists, enzalutamide, abiraterone, and others, can directly or indirectly increase the risk of hypertension. In this paper, we review the evidence available on the incidence and mechanism of hypertension in prostate cancer patients. In addition, we provide recommendations on the assessment, treatment, and future directions for hypertension management in the prostate cancer population. We propose an individualized goal for blood pressure in prostate cancer patients, balancing the target goal of 130/80 mmHg with common comorbidities of frailty, orthostatic symptoms, and imbalance in this population. The presence of additional comorbidities (myocardial infarction, heart failure, renal disease, diabetes) can assist in preference of anti-hypertensive drugs.


Subject(s)
Hypertension , Myocardial Infarction , Prostatic Neoplasms , Male , Humans , Hypertension/complications , Hypertension/epidemiology , Myocardial Infarction/drug therapy , Antihypertensive Agents/therapeutic use , Prostatic Neoplasms/complications , Prostatic Neoplasms/epidemiology , Gonadotropin-Releasing Hormone , Androgen Antagonists/adverse effects
3.
Mol Cancer Ther ; 22(6): 751-764, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37014264

ABSTRACT

Non-T-cell-inflamed immunologically "cold" tumor microenvironments (TME) are associated with poor responsiveness to immune checkpoint blockade (ICB) and can be sculpted by tumor cell genomics. Here, we evaluated how retinoblastoma (Rb) tumor-suppressor loss-of-function (LOF), one of the most frequent alterations in human cancer and associated with lineage plasticity, poor prognosis, and therapeutic outcomes, alters the TME, and whether therapeutic strategies targeting the molecular consequences of Rb loss enhance ICB efficacy. We performed bioinformatics analysis to elucidate the impact of endogenous Rb LOF on the immune TME in human primary and metastatic tumors. Next, we used isogenic murine models of Rb-deficient prostate cancer for in vitro and in vivo mechanistic studies to examine how Rb loss and bromodomain and extraterminal (BET) domain inhibition (BETi) reprograms the immune landscape, and evaluated in vivo therapeutic efficacy of BETi, singly and in combination with ICB and androgen deprivation therapy. Rb loss was enriched in non-T-cell-inflamed tumors, and Rb-deficient murine tumors demonstrated decreased immune infiltration in vivo. The BETi JQ1 increased immune infiltration into the TME through enhanced tumor cell STING/NF-κB activation and type I IFN signaling within tumor cells, resulting in differential macrophage and T-cell-mediated tumor growth inhibition and sensitization of Rb-deficient prostate cancer to ICB. BETi can reprogram the immunologically cold Rb-deficient TME via STING/NF-κB/IFN signaling to sensitize Rb-deficient prostate cancer to ICB. These data provide the mechanistic rationale to test combinations of BETi and ICB in clinical trials of Rb-deficient prostate cancer.


Subject(s)
Prostatic Neoplasms , Retinal Neoplasms , Retinoblastoma , Male , Humans , Animals , Mice , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , NF-kappa B , Immune Checkpoint Inhibitors , Androgen Antagonists , Tumor Microenvironment
4.
Clin Cancer Res ; 28(19): 4336-4345, 2022 10 03.
Article in English | MEDLINE | ID: mdl-35833954

ABSTRACT

PURPOSE: Biliary tract cancers (BTC) are aggressive malignancies refractory to chemotherapy and immunotherapy. MEK inhibition (MEKi)-based regimens may have utility in this disease when combined with PD-L1 blockade. We hypothesize that dual MEK/PD-L1 inhibition alters circulating soluble and cellular immune mediators to improve clinical outcomes in patients with advanced BTC. EXPERIMENTAL DESIGN: We examined immune features in peripheral blood from 77 patients with advanced BTC enrolled in a phase II clinical trial investigating atezolizumab with or without cobimetinib. Plasma and peripheral blood mononuclear cells (PBMC) were isolated from whole blood to evaluate soluble factors and immune cell populations. Baseline blood samples were additionally compared with healthy donors to identify immune signatures unique to BTC. RESULTS: At baseline, the soluble factors platelet-derived growth factor B (PDGF)-BB, placental growth factor (PlGF)-1, IL5, and IL17A were elevated in patients with BTC compared with healthy adult donors, and higher baseline frequencies of CD8+BTLA+ T cells correlated with better overall survival (OS) in this trial. There were also significant treatment-related alterations in several factors, including decreased PDGF-BB following combination treatment, that correlated with improved OS and progression-free survival (PFS). Higher baseline levels of IL23 and RANTES corresponded to improved clinical outcomes following combination treatment. Dual MEK/PD-L1 inhibition increased populations of CD4+TIM3+ and decreased CD8+VISTA+ T cells, correlating with worse OS and better PFS, respectively. CONCLUSIONS: This work represents a comprehensive analysis of peripheral immune features in patients with BTC and systemic responses to dual MEK/PD-L1 inhibition. These data support further investigation to understand how MEKi combines with immunotherapeutic approaches to improve clinical outcomes for patients with advanced BTC.


Subject(s)
Bile Duct Neoplasms , Biliary Tract Neoplasms , Adult , B7-H1 Antigen , Becaplermin/therapeutic use , Biliary Tract Neoplasms/drug therapy , Chemokine CCL5 , Cytokines/therapeutic use , Female , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Interleukin-5/therapeutic use , Leukocytes, Mononuclear/metabolism , Lymphocytes/metabolism , Mitogen-Activated Protein Kinase Kinases , Placenta Growth Factor , Proto-Oncogene Proteins c-sis/therapeutic use
5.
Nat Cancer ; 2(4): 444-456, 2021 04.
Article in English | MEDLINE | ID: mdl-33899001

ABSTRACT

Prostate cancers are considered to be immunologically 'cold' tumors given the very few patients who respond to checkpoint inhibitor (CPI) therapy. Recently, enrichment of interferon-stimulated genes (ISGs) predicted a favorable response to CPI across various disease sites. The enhancer of zeste homolog-2 (EZH2) is overexpressed in prostate cancer and known to negatively regulate ISGs. In the present study, we demonstrate that EZH2 inhibition in prostate cancer models activates a double-stranded RNA-STING-ISG stress response upregulating genes involved in antigen presentation, Th1 chemokine signaling and interferon response, including programmed cell death protein 1 (PD-L1) that is dependent on STING activation. EZH2 inhibition substantially increased intratumoral trafficking of activated CD8+ T cells and increased M1 tumor-associated macrophages, overall reversing resistance to PD-1 CPI. Our study identifies EZH2 as a potent inhibitor of antitumor immunity and responsiveness to CPI. These data suggest EZH2 inhibition as a therapeutic direction to enhance prostate cancer response to PD-1 CPI.


Subject(s)
Programmed Cell Death 1 Receptor , Prostatic Neoplasms , CD8-Positive T-Lymphocytes , Enhancer of Zeste Homolog 2 Protein/genetics , Humans , Interferons/pharmacology , Male , Prostatic Neoplasms/drug therapy , RNA, Double-Stranded
6.
Mol Cancer Ther ; 20(1): 150-160, 2021 01.
Article in English | MEDLINE | ID: mdl-33037138

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has a prominent fibrotic stroma, which is a result of interactions between tumor, immune and pancreatic stellate cells (PSC), or cancer-associated fibroblasts (CAF). Targeting inflammatory pathways present within the stroma may improve access of effector immune cells to PDAC and response to immunotherapy. Heat shock protein-90 (Hsp90) is a chaperone protein and a versatile target in pancreatic cancer. Hsp90 regulates a diverse array of cellular processes of relevance to both the tumor and the immune system. However, to date the role of Hsp90 in PSC/CAF has not been explored in detail. We hypothesized that Hsp90 inhibition would limit inflammatory signals, thereby reprogramming the PDAC tumor microenvironment to enhance sensitivity to PD-1 blockade. Treatment of immortalized and primary patient PSC/CAF with the Hsp90 inhibitor XL888 decreased IL6, a key cytokine that orchestrates immune changes in PDAC at the transcript and protein level in vitro XL888 directly limited PSC/CAF growth and reduced Jak/STAT and MAPK signaling intermediates and alpha-SMA expression as determined via immunoblot. Combined therapy with XL888 and anti-PD-1 was efficacious in C57BL/6 mice bearing syngeneic subcutaneous (Panc02) or orthotopic (KPC-Luc) tumors. Tumors from mice treated with both XL888 and anti-PD-1 had a significantly increased CD8+ and CD4+ T-cell infiltrate and a unique transcriptional profile characterized by upregulation of genes associated with immune response and chemotaxis. These data demonstrate that Hsp90 inhibition directly affects PSC/CAF in vitro and enhances the efficacy of anti-PD-1 blockade in vivo.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Azabicyclo Compounds/pharmacology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice, Inbred C57BL , Pancreatic Neoplasms/genetics , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/metabolism , Phenotype , Phthalic Acids/pharmacology , Programmed Cell Death 1 Receptor/metabolism , Treatment Outcome , Tumor Microenvironment , Xenograft Model Antitumor Assays
7.
J Comput Aided Mol Des ; 34(12): 1219-1228, 2020 12.
Article in English | MEDLINE | ID: mdl-32918236

ABSTRACT

SARS-CoV-2 recently jumped species and rapidly spread via human-to-human transmission to cause a global outbreak of COVID-19. The lack of effective vaccine combined with the severity of the disease necessitates attempts to develop small molecule drugs to combat the virus. COVID19_GIST_HSA is a freely available online repository to provide solvation thermodynamic maps of COVID-19-related protein small molecule drug targets. Grid inhomogeneous solvation theory maps were generated using AmberTools cpptraj-GIST, 3D reference interaction site model maps were created with AmberTools rism3d.snglpnt and hydration site analysis maps were created using SSTMap code. The resultant data can be applied to drug design efforts: scoring solvent displacement for docking, rational lead modification, prioritization of ligand- and protein- based pharmacophore elements, and creation of water-based pharmacophores. Herein, we demonstrate the use of the solvation thermodynamic mapping data. It is hoped that this freely provided data will aid in small molecule drug discovery efforts to defeat SARS-CoV-2.


Subject(s)
Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Drug Design , Drug Evaluation, Preclinical , Models, Chemical , Molecular Dynamics Simulation , Molecular Targeted Therapy , Pandemics , Pneumonia, Viral/drug therapy , Thermodynamics , Viral Nonstructural Proteins/drug effects , Antiviral Agents/chemistry , Betacoronavirus/chemistry , Binding Sites , COVID-19 , Catalytic Domain , Humans , Ligands , Models, Molecular , Protein Conformation , SARS-CoV-2 , Small Molecule Libraries , Structure-Activity Relationship , Viral Nonstructural Proteins/chemistry , Water , COVID-19 Drug Treatment
8.
Front Immunol ; 11: 1867, 2020.
Article in English | MEDLINE | ID: mdl-32983104

ABSTRACT

Originally identified as lymphocyte regulation of fellow lymphocytes, our understanding of infectious tolerance has undergone significant evolutions in understanding since being proposed in the early 1970s by Gershon and Kondo and expanded upon by Herman Waldman two decades later. The evolution of our understanding of infectious tolerance has coincided with significant cellular and humoral discoveries. The early studies leading to the isolation and identification of Regulatory T cells (Tregs) and cytokines including TGFß and IL-10 in the control of peripheral tolerance was a paradigm shift in our understanding of infectious tolerance. More recently, another potential, paradigm shift in our understanding of the "infectious" aspect of infectious tolerance was proposed, identifying extracellular vesicles (EVs) as a mechanism for propagating infectious tolerance. In this review, we will outline the history of infectious tolerance, focusing on a potential EV mechanism for infectious tolerance and a novel, EV-associated form for the cytokine IL-35, ideally suited to the task of propagating tolerance by "infecting" other lymphocytes.


Subject(s)
Allergy and Immunology/history , Extracellular Vesicles/immunology , Immune Tolerance/immunology , Interleukins/immunology , Animals , History, 20th Century , History, 21st Century , Humans
9.
ChemRxiv ; 2020 May 13.
Article in English | MEDLINE | ID: mdl-32511289

ABSTRACT

SARS-CoV-2 recently jumped species and rapidly spread via human-to-human transmission to cause a global outbreak of COVID-19. The lack of effective vaccine combined with the severity of the disease necessitates attempts to develop small molecule drugs to combat the virus. COVID19_GIST_HSA is a freely available online repository to provide solvation thermodynamic maps of COVID-19-related protein small molecule drug targets. Grid Inhomogeneous Solvation Theory maps were generated using AmberTools cpptraj-GIST and Hydration Site Analysis maps were created using SSTmap code. The resultant data can be applied to drug design efforts: scoring solvent displacement for docking, rational lead modification, prioritization of ligand- and protein- based pharmacophore elements, and creation of water-based pharmacophores. Herein, we demonstrate the use of the solvation thermodynamic mapping data. It is hoped that this freely provided data will aid in small molecule drug discovery efforts to defeat SARS-CoV-2.

10.
Clin Cancer Res ; 26(19): 5162-5171, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32513836

ABSTRACT

PURPOSE: Preclinical studies demonstrated that a DNA vaccine (pTVG-AR, MVI-118) encoding the androgen receptor ligand-binding domain (AR LBD) augmented antigen-specific CD8+ T cells, delayed prostate cancer progression and emergence of castration-resistant disease, and prolonged survival of tumor-bearing mice. This vaccine was evaluated in a multicenter phase I trial. PATIENTS AND METHODS: Patients with metastatic castration-sensitive prostate cancer (mCSPC) who had recently begun androgen deprivation therapy were randomly assigned to receive pTVG-AR on one of two treatment schedules over one year, and with or without GM-CSF as a vaccine adjuvant. Patients were followed for 18 months. Primary objectives were safety and immune response. Secondary objectives included median time to PSA progression, and 18-month PSA-PFS (PPFS). RESULTS: Forty patients were enrolled at three centers. Twenty-seven patients completed treatment and 18 months of follow-up. Eleven patients (28%) had a PSA progression event before the 18-month time point. No grade 3 or 4 adverse events were observed. Of 30 patients with samples available for immune analysis, 14 (47%) developed Th1-type immunity to the AR LBD, as determined by IFNγ and/or granzyme B ELISPOT. Persistent IFNγ immune responses were observed irrespective of GM-CSF adjuvant. Patients who developed T-cell immunity had a significantly prolonged PPFS compared with patients without immunity (HR = 0.01; 95% CI, 0.0-0.21; P = 0.003). CONCLUSIONS: pTVG-AR was safe and immunologically active in patients with mCSPC. Association between immunity and PPFS suggests that treatment may delay the time to castration resistance, consistent with preclinical findings, and will be prospectively evaluated in future trials.See related commentary by Shenderov and Antonarakis, p. 5056.


Subject(s)
Androgen Antagonists/administration & dosage , Prostatic Neoplasms/drug therapy , Receptors, Androgen/genetics , Vaccines, DNA/administration & dosage , Aged , Androgen Antagonists/adverse effects , CD8-Positive T-Lymphocytes/drug effects , Humans , Interferon-gamma/genetics , Ligands , Male , Middle Aged , Neoplasm Metastasis , Progression-Free Survival , Prostate-Specific Antigen/blood , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Binding/genetics , Receptors, Androgen/drug effects , Vaccines, DNA/adverse effects
11.
Urol Oncol ; 37(8): 535-542, 2019 08.
Article in English | MEDLINE | ID: mdl-30503851

ABSTRACT

The last two decades of cancer research have seen two major advancements in our ability to treat cancer: precision medicine and immunotherapy. While these approaches have shown striking anticancer efficacy in numerous malignancies, they have not shown similar success and applicability in advanced prostate cancer patients. The fields of precision medicine and immunotherapy have come to realize that targeted therapies are capable of not only inhibiting tumor cell growth, but also promoting antitumor immunity by modulating the tumor microenvironment. Here we examine how personalized medicine can be used to target the tumor immune microenvironment in prostate cancer, with the goal of enhancing clinical responses to immunotherapy.


Subject(s)
Immunotherapy/methods , Precision Medicine/methods , Prostatic Neoplasms/therapy , Tumor Microenvironment/immunology , Humans , Male , Prostatic Neoplasms/immunology
12.
Cancer Discov ; 8(11): 1358-1365, 2018 11.
Article in English | MEDLINE | ID: mdl-30309862

ABSTRACT

Immunotherapy has revolutionized cancer therapy, largely attributed to the success of immune-checkpoint blockade. However, there are subsets of patients across multiple cancers who have not shown robust responses to these agents. A major impediment to progress in the field is the availability of faithful mouse models that recapitulate the complexity of human malignancy and immune contexture within the tumor microenvironment. These models are urgently needed across all malignancies to interrogate and predict antitumor immune responses and therapeutic efficacy in clinical trials. Herein, we seek to review pros and cons of different cancer mouse models, and how they can be used as platforms to predict efficacy and resistance to cancer immunotherapies.Significance: Although immunotherapy has shown substantial benefit in the treatment of a variety of malignancies, a key hurdle toward the advancement of these therapies is the availability of immunocompetent preclinical mouse models that recapitulate human disease. Here, we review the evolution of preclinical mouse models and their utility as coclinical platforms for mechanistic interrogation of cancer immunotherapies. Cancer Discov; 8(11); 1358-65. ©2018 AACR.


Subject(s)
Biomedical Research , Disease Models, Animal , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Animals , Humans , Mice
13.
Cancer Immunol Res ; 5(12): 1074-1085, 2017 12.
Article in English | MEDLINE | ID: mdl-29051161

ABSTRACT

Androgen deprivation is the primary therapy for recurrent prostate cancer, and agents targeting the androgen receptor (AR) pathway continue to be developed. Because androgen-deprivation therapy (ADT) has immmunostimulatory effects as well as direct antitumor effects, AR-targeted therapies have been combined with other anticancer therapies, including immunotherapies. Here, we sought to study whether an antigen-specific mechanism of resistance to ADT (overexpression of the AR) may result in enhanced AR-specific T-cell immune recognition, and whether this might be strategically combined with an antitumor vaccine targeting the AR. Androgen deprivation increased AR expression in human and murine prostate tumor cells in vitro and in vivo The increased expression persisted over time. Increased AR expression was associated with recognition and cytolytic activity by AR-specific T cells. Furthermore, ADT combined with vaccination, specifically a DNA vaccine encoding the ligand-binding domain of the AR, led to improved antitumor responses as measured by tumor volumes and delays in the emergence of castrate-resistant prostate tumors in two murine prostate cancer models (Myc-CaP and prostate-specific PTEN-deficient mice). Together, these data suggest that ADT combined with AR-directed immunotherapy targets a major mechanism of resistance, overexpression of the AR. This combination may be more effective than ADT combined with other immunotherapeutic approaches. Cancer Immunol Res; 5(12); 1074-85. ©2017 AACR.


Subject(s)
Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Receptors, Androgen/genetics , T-Cell Antigen Receptor Specificity , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunization , Immunohistochemistry , Male , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/therapy , Receptors, Androgen/immunology , Receptors, Androgen/metabolism , Xenograft Model Antitumor Assays
14.
J Immunother Cancer ; 5(1): 56, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28716080

ABSTRACT

BACKGROUND: Immunotherapies have demonstrated clinical benefit for many types of cancers, however many patients do not respond, and treatment-related adverse effects can be severe. Hence many efforts are underway to identify treatment predictive biomarkers. We have reported the results of two phase I trials using a DNA vaccine encoding prostatic acid phosphatase (PAP) in patients with biochemically recurrent prostate cancer. In both trials, persistent PAP-specific Th1 immunity developed in some patients, and this was associated with favorable changes in serum PSA kinetics. In the current study, we sought to determine if measures of antigen-specific or antigen non-specific immunity were present prior to treatment, and associated with subsequent immune response, to identify possible predictive immune biomarkers. METHODS: Patients who developed persistent PAP-specific, IFNγ-secreting immune responses were defined as immune "responders." The frequency of peripheral T cell and B cell lymphocytes, natural killer cells, monocytes, dendritic cells, myeloid derived suppressor cells, and regulatory T cells were assessed by flow cytometry and clinical laboratory values. PAP-specific immune responses were evaluated by cytokine secretion in vitro, and by antigen-specific suppression of delayed-type hypersensitivity to a recall antigen in an in vivo SCID mouse model. RESULTS: The frequency of peripheral blood cell types did not differ between the immune responder and non-responder groups. Non-responder patients tended to have higher PAP-specific IL-10 production pre-vaccination (p = 0.09). Responder patients had greater preexisting PAP-specific bystander regulatory responses that suppressed DTH to a recall antigen (p = 0.016). CONCLUSIONS: While our study population was small (n = 38), these results suggest that different measures of antigen-specific tolerance or regulation might help predict immunological outcome from DNA vaccination. These will be prospectively evaluated in an ongoing randomized, phase II trial.


Subject(s)
Acid Phosphatase/immunology , Cancer Vaccines/immunology , Immunity, Cellular/immunology , Prostatic Neoplasms/immunology , Vaccines, DNA/immunology , Animals , Antigens, Neoplasm/immunology , Biomarkers, Tumor/immunology , Humans , Immunogenicity, Vaccine , Immunophenotyping , Interferon-gamma/immunology , Interleukin-10/biosynthesis , Leukocyte Count , Leukocytes, Mononuclear/immunology , Male , Mice, SCID , Prostatic Neoplasms/therapy , Vaccines, DNA/therapeutic use
15.
Cancer Discov ; 7(7): 750-765, 2017 07.
Article in English | MEDLINE | ID: mdl-28274958

ABSTRACT

Several kinase inhibitors that target aberrant signaling pathways in tumor cells have been deployed in cancer therapy. However, their impact on the tumor immune microenvironment remains poorly understood. The tyrosine kinase inhibitor cabozantinib showed striking responses in cancer clinical trial patients across several malignancies. Here, we show that cabozantinib rapidly eradicates invasive, poorly differentiated PTEN/p53-deficient murine prostate cancer. This was associated with enhanced release of neutrophil chemotactic factors from tumor cells, including CXCL12 and HMGB1, resulting in robust infiltration of neutrophils into the tumor. Critically, cabozantinib-induced tumor clearance in mice was abolished by antibody-mediated granulocyte depletion or HMGB1 neutralization or blockade of neutrophil chemotaxis with the CXCR4 inhibitor plerixafor. Collectively, these data demonstrate that cabozantinib triggers a neutrophil-mediated anticancer innate immune response, resulting in tumor clearance.Significance: This study is the first to demonstrate that a tyrosine kinase inhibitor can activate neutrophil-mediated antitumor innate immunity, resulting in invasive cancer clearance. Cancer Discov; 7(7); 750-65. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 653.


Subject(s)
Anilides/administration & dosage , Chemokine CXCL12/antagonists & inhibitors , HMGB1 Protein/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/drug therapy , Pyridines/administration & dosage , Tumor Suppressor Protein p53/genetics , Animals , Benzylamines , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL12/genetics , Cyclams , HMGB1 Protein/genetics , Heterocyclic Compounds/administration & dosage , Humans , Immunity, Innate/drug effects , Male , Mice , Neutrophils/drug effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/administration & dosage , Tumor Microenvironment/genetics
16.
Learn Behav ; 45(3): 243-251, 2017 09.
Article in English | MEDLINE | ID: mdl-28181182

ABSTRACT

Two experiments using rats evaluated the susceptibility of CS preexposure to retrograde amnesia induced by the protein synthesis inhibitor cycloheximide and tested whether amnesia for CS preexposure shares similar characteristics with amnesia for other memories. In Experiment 1, rats received cycloheximide either immediately, 60 minutes, or 120 minutes after preexposure. Following preexposure, rats received fear conditioning. When later tested, the subjects that received the amnestic treatment shortly after preexposure showed no CS preexposure effect (i.e., no reduction of fear). The amnesia for CS preexposure was attenuated with longer post-preexposure delays, showing a temporal gradient. In Experiment 2, following the replication of amnesia for CS preexposure, the amnestic treatment was readministered to the rats prior to testing. It was demonstrated that the amnestic-preexposure memory could be recovered (i.e., readministration of the drug alleviated the amnesia for CS preexposure). These two experiments show that memories for CS preexposure are susceptible to retrograde amnesia and share similar characteristics with memories for original acquisition and extinction. The results are explained using a retrieval hypothesis of retrograde amnesia.


Subject(s)
Amnesia, Retrograde/chemically induced , Conditioning, Classical/drug effects , Cycloheximide/pharmacology , Animals , Fear , Female , Male , Rats , Retention, Psychology/drug effects , Time Factors
17.
Prostate ; 77(7): 812-821, 2017 May.
Article in English | MEDLINE | ID: mdl-28181678

ABSTRACT

BACKGROUND: The androgen receptor (AR) is a key oncogenic driver of prostate cancer, and has been the primary focus of prostate cancer treatment for several decades. We have previously demonstrated that the AR is also an immunological target antigen, recognized in patients with prostate cancer, and targetable by means of vaccines in rodent models with delays in prostate tumor growth. The current study was performed to determine the safety and immunological efficacy of a GMP-grade plasmid DNA vaccine encoding the ligand-binding domain (LBD) of the AR, pTVG-AR. METHODS: Groups of male mice (n = 6-10 per group) were evaluated after four or seven immunizations, using different schedules and inclusion of GM-CSF as a vaccine adjuvant. Animals were assessed for toxicity using gross observations, pathological analysis, and analysis of serum chemistries. Animals were analyzed for evidence of vaccine-augmented immunity by tetramer analysis. Survival studies using different immunization schedules and inclusion of GM-CSF were conducted in an autochthonous genetically engineered mouse model. RESULTS: No significant toxicities were observed in terms of animal weights, histopathology, hematological changes, or changes in serum chemistries, although there was a trend to lower serum glucose in animals treated with the vaccine. There was specifically no evidence of toxicity in other tissues that express AR, including liver, muscle, hematopoietic, and brain. Vaccination was found to elicit AR LBD-specific CD8+ T cells. In a subsequent study of tumor-bearing animals, animals treated with vaccine had prolonged survival compared with control-immunized mice. CONCLUSIONS: These studies demonstrate that, in immunocompetent mice expressing the target antigen, immunization with the pTVG-AR vaccine was both safe and effective in eliciting AR-specific cellular immune responses, and prolonged the survival of prostate tumor-bearing mice. These findings support the clinical evaluation of pTVG-AR in patients with recurrent prostate cancer. Prostate 77:812-821, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Prostatic Neoplasms , Receptors, Androgen/immunology , Vaccines, DNA , Adjuvants, Immunologic/administration & dosage , Animals , Male , Mice , Monitoring, Immunologic/methods , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Treatment Outcome , Vaccines, DNA/administration & dosage , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology
18.
Oncoimmunology ; 5(6): e1165377, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27471641

ABSTRACT

We have previously reported that tumor antigen-specific DNA vaccination in mice led to an increase in IFNγ-secreting T cells and an increase in tumor expression of PD-L1. Further, we demonstrated that increasing the encoded antigen's MHC-binding affinity led to increased PD-1 expression on antigen-specific CD8(+) T cells. Together these phenomena provided resistance to antitumor immunization that was abrogated with PD-1/PD-L1 blockade. We consequently sought to determine whether similar regulation occurred in human patients following antitumor immunization. Using clinical samples from prostate cancer patients who were previously immunized with a DNA vaccine, we analyzed changes in checkpoint receptor expression on antigen-specific CD8(+) T cells, the effect of PD-1 blockade on elicited immune responses, and for changes in checkpoint ligand expression on patients' circulating tumor cells (CTCs). We observed no significant changes in T-cell expression of PD-1 or other checkpoint receptors, but antigen-specific immune responses were detected and/or augmented with PD-1 blockade as detected by IFNγ and granzyme B secretion or trans vivo DTH testing. Moreover, PD-L1 expression was increased on CTCs following vaccination, and this PD-L1 upregulation was associated with the development of sustained T-cell immunity and longer progression-free survival. Finally, similar results were observed with patients treated with sipuleucel-T, another vaccine targeting the same prostate antigen. These findings provide in-human rationale for combining anticancer vaccines with PD-1 blocking antibodies, particularly for the treatment of prostate cancer, a disease for which vaccines have demonstrated benefit and yet PD-1 inhibitors have shown little clinical benefit to date as monotherapies.

19.
Prostate ; 76(13): 1192-202, 2016 09.
Article in English | MEDLINE | ID: mdl-27225803

ABSTRACT

INTRODUCTION: Transgenic mouse modeling is a favorable tool to reflect human prostate tumorigenesis and interactions between prostate cancer and the microenvironment. The use of GEMMs and derived cell lines represent powerful tools to study prostate cancer initiation and progression with an associated tumor microenvironment. Notably, such models provide the capacity for rapid preclinical therapy studies including immune therapies for prostate cancer treatment. METHODS: Backcrossing FVB Hi-MYC mice with C57BL/6N mice, we established a Hi-MYC transgenic mouse model on a C57BL/6 background (B6MYC). In addition, using a conditional reprogramming method, a novel C57BL/6 MYC driven prostate adenocarcinoma cell line was generated. RESULTS: Our results demonstrate that disease progression is significantly delayed in B6MYC when compared to their FVB counterparts. Current data also indicates infiltrating immune cells are present in pre-cancer lesions, prostate intraepithelial neoplasia (PIN). Further, immunophenotyping of this immune infiltrate demonstrates the predominant population as myeloid-derived suppressor cells (MDSC). Also, we successfully generated a B6MYC-CaP cell line, and determined that this new PCa cell line express markers of luminal epithelial lineage. DISCUSSION: This novel model of PCa provides a new platform to understand the cross talk between MYC driven prostate cancer and the microenvironment. Importantly, these models will be an ideal tool to support the clinical development of immunotherapy as well as other novel therapeutic strategies for prostate cancer treatment. Prostate 76:1192-1202, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Disease Models, Animal , Disease Progression , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Tumor Microenvironment/physiology , Animals , Cell Line, Tumor , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
20.
Urol Oncol ; 34(4): 193-204, 2016 Apr.
Article in English | MEDLINE | ID: mdl-24332642

ABSTRACT

Prostate cancer is the most commonly diagnosed cancer in the United States. It is also the second leading cause of cancer-related death in men, making it one of the largest public health concerns today. Prostate cancer is an ideal disease for immunotherapies because of the generally slow progression, the dispensability of the target organ in the patient population, and the availability of several tissue-specific antigens. As such, several therapeutic vaccines have entered clinical trials, with one autologous cellular vaccine (sipuleucel-T) recently gaining Food and Drug Administration approval after demonstrating overall survival benefit in randomized phase III clinical trials. DNA-based vaccines are safe, economical, alternative "off-the-shelf" approaches that have undergone extensive evaluation in preclinical models. In fact, the first vaccine approved in the United States for the treatment of cancer was a DNA vaccine for canine melanoma. Several prostate cancer-specific DNA vaccines have been developed in the last decade and have shown promising results in early phase clinical trials. This review summarizes anticancer human DNA vaccine trials, with a focus on those conducted for prostate cancer. We conclude with an outline of special considerations important for the development and successful translation of DNA vaccines from the laboratory to the clinic.


Subject(s)
Cancer Vaccines/immunology , Immunotherapy/methods , Prostatic Neoplasms/therapy , Vaccines, DNA/immunology , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/chemistry , Clinical Trials, Phase III as Topic , Humans , Male , Prostatic Neoplasms/immunology , Randomized Controlled Trials as Topic , Vaccines, DNA/administration & dosage , Vaccines, DNA/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...