Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
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
2.
J Immunol ; 189(12): 5590-601, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23152566

ABSTRACT

Regulatory T cells play important roles in cancer development and progression by limiting the generation of innate and adaptive anti-tumor immunity. We hypothesized that in addition to natural CD4(+)CD25(+) regulatory T cells (Tregs) and myeloid-derived suppressor cells, tumor Ag-specific Tregs interfere with the detection of anti-tumor immunity after immunotherapy. Using samples from prostate cancer patients immunized with a DNA vaccine encoding prostatic acid phosphatase (PAP) and a trans-vivo delayed-type hypersensitivity (tvDTH) assay, we found that the detection of PAP-specific effector responses after immunization was prevented by the activity of PAP-specific regulatory cells. These regulatory cells were CD8(+)CTLA-4(+), and their suppression was relieved by blockade of CTLA-4, but not IL-10 or TGF-ß. Moreover, Ag-specific CD8(+) Tregs were detected prior to immunization in the absence of PAP-specific effector responses. These PAP-specific CD8(+)CTLA-4(+) suppressor T cells expressed IL-35, which was decreased after blockade of CTLA-4, and inhibition of either CTLA-4 or IL-35 reversed PAP-specific suppression of tvDTH response. PAP-specific CD8(+)CTLA-4(+) T cells also suppressed T cell proliferation in an IL-35-dependent, contact-independent fashion. Taken together, these findings suggest a novel population of CD8(+)CTLA-4(+) IL-35-secreting tumor Ag-specific Tregs arise spontaneously in some prostate cancer patients, persist during immunization, and can prevent the detection of Ag-specific effector responses by an IL-35-dependent mechanism.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/antagonists & inhibitors , Epitopes, T-Lymphocyte/immunology , Growth Inhibitors/antagonists & inhibitors , Interleukins/antagonists & inhibitors , Prostatic Neoplasms/immunology , Protein Tyrosine Phosphatases/physiology , T-Lymphocytes, Regulatory/immunology , Acid Phosphatase , Animals , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/pathology , CTLA-4 Antigen/biosynthesis , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cells, Cultured , Clinical Trials as Topic/methods , Coculture Techniques , Growth Inhibitors/biosynthesis , Humans , Interleukins/biosynthesis , Interleukins/metabolism , Male , Mice , Mice, SCID , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/prevention & control , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/pathology , Tumor Escape/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
3.
Cancer Immunol Immunother ; 62(3): 585-96, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23108626

ABSTRACT

The androgen receptor (AR) plays an essential role in the development and progression of prostate cancer. However, while it has long been the primary molecular target of metastatic prostate cancer therapies, it has not been explored as an immunotherapeutic target. In particular, the AR ligand-binding domain (LBD) is a potentially attractive target, as it has an identical sequence among humans as well as among multiple species, providing a logical candidate for preclinical evaluation. In this report, we evaluated the immune and anti-tumor efficacy of a DNA vaccine targeting the AR LBD (pTVG-AR) in relevant rodent preclinical models. We found immunization of HHDII-DR1 mice, which express human HLA-A2 and HLA-DR1, with pTVG-AR augmented AR LBD HLA-A2-restricted peptide-specific, cytotoxic immune responses in vivo that could lyse human prostate cancer cells. Using an HLA-A2-expressing autochthonous model of prostate cancer, immunization with pTVG-AR augmented HLA-A2-restricted immune responses that could lyse syngeneic prostate tumor cells and led to a decrease in tumor burden and an increase in overall survival of tumor-bearing animals. Finally, immunization decreased prostate tumor growth in Copenhagen rats that was associated with a Th1-type immune response. These data show that the AR is as a prostate cancer immunological target antigen and that a DNA vaccine targeting the AR LBD is an attractive candidate for clinical evaluation.


Subject(s)
Prostatic Neoplasms/therapy , Receptors, Androgen/immunology , Adenocarcinoma/prevention & control , Animals , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Cytotoxicity, Immunologic , HLA-A2 Antigen/genetics , HLA-DR1 Antigen/genetics , Humans , Immunization , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/immunology , Prostatic Neoplasms/mortality , Prostatic Neoplasms/prevention & control , Protein Structure, Tertiary , Rats , Vaccines, DNA/therapeutic use
4.
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
5.
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
6.
Cancer Immunol Immunother ; 60(6): 781-92, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21350948

ABSTRACT

The androgen receptor (AR) is a hormone receptor that plays a critical role in prostate cancer, and depletion of its ligand has long been the cornerstone of treatment for metastatic disease. Here, we evaluate the AR ligand-binding domain (LBD) as an immunological target, seeking to identify HLA-A2-restricted epitopes recognized by T cells in prostate cancer patients. Ten AR LBD-derived, HLA-A2-binding peptides were identified and ranked with respect to HLA-A2 affinity and were used to culture peptide-specific T cells from HLA-A2+ prostate cancer patients. These T-cell cultures identified peptide-specific T cells specific for all ten peptides in at least one patient, and T cells specific for peptides AR805 and AR811 were detected in over half of patients. Peptide-specific CD8+ T-cell clones were then isolated and characterized for prostate cancer cytotoxicity and cytokine expression, identifying that AR805 and AR811 CD8+ T-cell clones could lyse prostate cancer cells in an HLA-A2-restricted fashion, but only AR811 CTL had polyfunctional cytokine expression. Epitopes were confirmed using immunization studies in HLA-A2 transgenic mice, in which the AR LBD is an autologous antigen with an identical protein sequence, which showed that mice immunized with AR811 developed peptide-specific CTL that lyse HLA-A2+ prostate cancer cells. These data show that AR805 and AR811 are HLA-A2-restricted epitopes for which CTL can be commonly detected in prostate cancer patients. Moreover, CTL responses specific for AR811 can be elicited by direct immunization of A2/DR1 mice. These findings suggest that it may be possible to elicit an anti-prostate tumor immune response by augmenting CTL populations using AR LBD-based vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Prostatic Neoplasms/immunology , Receptors, Androgen/immunology , Amino Acid Sequence , Animals , Animals, Genetically Modified , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cytokines/biosynthesis , Cytokines/immunology , HLA-A2 Antigen/biosynthesis , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , HLA-DR1 Antigen/genetics , HLA-DR1 Antigen/immunology , Humans , Ligands , Male , Mice , Mice, Knockout , Mice, Transgenic , Oligopeptides/immunology , Oligopeptides/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Protein Structure, Tertiary , Receptors, Androgen/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
7.
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
8.
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
9.
Cancer Immunol Immunother ; 59(6): 943-53, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20140431

ABSTRACT

Prostatic acid phosphatase (PAP) has been investigated as the target of several antigen-specific anti-prostate tumor vaccines. The goal of antigen-specific active immunotherapies targeting PAP would ideally be to elicit PAP-specific CD8+ effector T cells. The identification of PAP-specific CD8+ T-cell epitopes should provide a means of monitoring the immunological efficacy of vaccines targeting PAP, and these epitopes might themselves be developed as vaccine antigens. In the current report, we hypothesized that PAP-specific epitopes might be identified by direct identification of pre-existing CD8+ T cells specific for HLA-A2-restricted peptides derived from PAP in the blood of HLA-A2-expressing individuals. 11 nonamer peptides derived from the amino acid sequence of PAP were used as stimulator antigens in functional ELISPOT assays with peripheral blood mononuclear cells from 20 HLA-A2+ patients with prostate cancer or ten healthy blood donors. Peptide-specific T cells were frequently identified in both groups for three of the peptides, p18-26, p112-120, and p135-143. CD8+ T-cell clones specific for three peptides, p18-26, p112-120, and p299-307, confirmed that these are HLA-A2-restricted T-cell epitopes. Moreover, HLA-A2 transgenic mice immunized with a DNA vaccine encoding PAP developed epitope-specific responses for one or more of these three peptide epitopes. We propose that this method to first identify epitopes for which there are pre-existing epitope-specific T cells could be used to prioritize MHC class I-specific epitopes for other antigens. In addition, we propose that the epitopes identified here could be used to monitor immune responses in HLA-A2+ patients receiving vaccines targeting PAP to identify potentially therapeutic immune responses.


Subject(s)
Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines , Epitopes, T-Lymphocyte/metabolism , Peptide Fragments/metabolism , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Protein Tyrosine Phosphatases/metabolism , Acid Phosphatase , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Clone Cells , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Humans , Immunization , Interferon-gamma/metabolism , Lymphocyte Activation , Male , Mice , Mice, Transgenic , Peptide Fragments/genetics , Peptide Fragments/immunology , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Protein Binding , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/immunology
10.
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
11.
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
12.
Methods Mol Biol ; 542: 211-43, 2009.
Article in English | MEDLINE | ID: mdl-19565905

ABSTRACT

An antitumor DNA vaccine is a bacterial DNA plasmid that encodes the complementary DNA (cDNA) of a tumor antigen. When injected into recipients, antitumor DNA vaccines have been shown to elicit both humoral and cellular immunity against the encoded tumor antigen. These vaccines represent a relatively new immunotherapeutic technique being investigated as a means to deliver a target antigen and elicit or augment antitumor antigen-specific immune responses. One of the primary advantages of DNA vaccines as opposed to some other methods of antigen delivery is that they can be easily constructed, purified, and delivered to recipients. In this review we describe this process, detailing the procedures used to construct, purify, deliver, and evaluate the efficacy of DNA vaccines. We begin by describing the process of molecularly constructing the vaccine, from selecting a bacterial plasmid to form the backbone of the vaccine, cloning the antigen cDNA into this plasmid, and confirming the sequence and orientation of the completed vaccine. This is then followed by a series of experiments that can be used to ensure that the antigen encoded by the vaccine is transcribed and translated after being taken up by eukaryotic cells. We then describe large-scale purification procedures that can be used to obtain sufficient quantities of plasmid DNA to conduct in vivo immunization experiments. Finally, we provide an immunization protocol that can be used to evaluate the immunological efficacy of the constructed DNA vaccine. By following these protocols, it is possible to construct, purify, deliver, and evaluate the efficacy of antitumor DNA vaccines.


Subject(s)
Cancer Vaccines/biosynthesis , Cancer Vaccines/immunology , Molecular Biology/methods , Vaccines, DNA/biosynthesis , Vaccines, DNA/immunology , Animals , Antigens, Neoplasm/metabolism , CHO Cells , Cancer Vaccines/isolation & purification , Cricetinae , Cricetulus , Mice , Rats , Reproducibility of Results , Vaccines, DNA/isolation & purification
13.
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
14.
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
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.
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.

17.
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
18.
Cancer Immunol Res ; 3(8): 946-55, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26041735

ABSTRACT

DNA vaccines have demonstrated antitumor efficacy in multiple preclinical models, but low immunogenicity has been observed in several human clinical trials. This has led to many approaches seeking to improve the immunogenicity of DNA vaccines. We previously reported that a DNA vaccine encoding the cancer-testis antigen SSX2, modified to encode altered epitopes with increased MHC class I affinity, elicited a greater frequency of cytolytic, multifunctional CD8(+) T cells in non-tumor-bearing mice. We sought to test whether this optimized vaccine resulted in increased antitumor activity in mice bearing an HLA-A2-expressing tumor engineered to express SSX2. We found that immunization of tumor-bearing mice with the optimized vaccine elicited a surprisingly inferior antitumor effect relative to the native vaccine. Both native and optimized vaccines led to increased expression of PD-L1 on tumor cells, but antigen-specific CD8(+) T cells from mice immunized with the optimized construct expressed higher PD-1. Splenocytes from immunized animals induced PD-L1 expression on tumor cells in vitro. Antitumor activity of the optimized vaccine could be increased when combined with antibodies blocking PD-1 or PD-L1, or by targeting a tumor line not expressing PD-L1. These findings suggest that vaccines aimed at eliciting effector CD8(+) T cells, and DNA vaccines in particular, might best be combined with PD-1 pathway inhibitors in clinical trials. This strategy may be particularly advantageous for vaccines targeting prostate cancer, a disease for which antitumor vaccines have demonstrated clinical benefit and yet PD-1 pathway inhibitors alone have shown little efficacy to date.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Cancer Vaccines/immunology , Epitopes/immunology , Neoplasms/immunology , Peptide Fragments/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Vaccines, DNA/immunology , Animals , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Disease Models, Animal , Epitopes/genetics , Gene Expression , Humans , Immunization , Mice , Mice, Knockout , Neoplasms/metabolism , Neoplasms/therapy , Peptide Fragments/genetics , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Vaccines, DNA/administration & dosage
19.
Clin Cancer Res ; 20(14): 3692-704, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24850844

ABSTRACT

PURPOSE: We have previously reported that a DNA vaccine encoding prostatic acid phosphatase (PAP) could elicit PAP-specific T cells in patients with early recurrent prostate cancer. In the current pilot trial, we sought to evaluate whether prolonged immunization with regular booster immunizations, or "personalized" schedules of immunization determined using real-time immune monitoring, could elicit persistent, antigen-specific T cells, and whether treatment was associated with changes in PSA doubling time (PSA DT). EXPERIMENTAL DESIGN: Sixteen patients with castration-resistant, nonmetastatic prostate cancer received six immunizations at 2-week intervals and then either quarterly (arm 1) or as determined by multiparameter immune monitoring (arm 2). RESULTS: Patients were on study a median of 16 months; four received 24 vaccinations. Only one event associated with treatment >grade 2 was observed. Six of 16 (38%) remained metastasis-free at 2 years. PAP-specific T cells were elicited in 12 of 16 (75%), predominantly of a Th1 phenotype, which persisted in frequency and phenotype for at least 1 year. IFNγ-secreting T-cell responses measured by ELISPOT were detectable in 5 of 13 individuals at 1 year, and this was not statistically different between study arms. The overall median fold change in PSA DT from pretreatment to posttreatment was 1.6 (range, 0.6-7.0; P = 0.036). CONCLUSIONS: Repetitive immunization with a plasmid DNA vaccine was safe and elicited Th1-biased antigen-specific T cells that persisted over time. Modifications in the immunization schedule based on real-time immune monitoring did not increase the frequency of patients developing effector and memory T-cell responses with this DNA vaccine.


Subject(s)
Acid Phosphatase/immunology , Adenocarcinoma/therapy , Cancer Vaccines/administration & dosage , Plasmids/administration & dosage , Prostatic Neoplasms, Castration-Resistant/therapy , Vaccines, DNA/administration & dosage , Adenocarcinoma/blood , Adenocarcinoma/immunology , Aged , Aged, 80 and over , Humans , Immunization , Immunization Schedule , Male , Middle Aged , Prostate-Specific Antigen/blood , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/immunology , Treatment Outcome
20.
Front Oncol ; 3: 109, 2013.
Article in English | MEDLINE | ID: mdl-23653893

ABSTRACT

While immune monitoring of tumor immunotherapy often focuses on the generation of productive Th1-type inflammatory immune responses, the importance of regulatory immune responses is often overlooked, despite the well-documented effects of regulatory immune responses in suppressing anti-tumor immunity. In a variety of malignancies, the frequency of regulatory cell populations has been shown to correlate with disease progression and a poor prognosis, further emphasizing the importance of characterizing the effects of immunotherapy on these populations. This review focuses on the role of suppressive immune populations (regulatory T cells, myeloid-derived suppressor cells, and tumor-associated macrophages) in inhibiting anti-tumor immunity, how these populations have been used in the immune monitoring of clinical trials, the prognostic value of these responses, and how the monitoring of these regulatory responses can be improved in the future.

SELECTION OF CITATIONS
SEARCH DETAIL