1
|
Sun BL. Immunotherapy in treatment of metastatic prostate cancer: An approach to circumvent immunosuppressive tumor microenvironment. Prostate 2021; 81:1125-1134. [PMID: 34435699 DOI: 10.1002/pros.24213] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 01/21/2023]
Abstract
Prostate cancer is the second most common cause of cancer-related death in men in the United States and the fifth worldwide. Most prostate cancer arises as an androgen-dependent tumor but eventually progresses into castration-resistance prostate cancer, incurable by the current androgen deprivation therapy and chemotherapy. The development of immunotherapy in cancer treatment has brought an exciting era of antiprostate cancer therapy through antitumor immune responses. Prostate cancer is recognized as a poorly immunogenic tissue with immunological ignorance showing low levels of antigen-presenting process and cytotoxic T-cell activation, high levels of immune checkpoint molecules and immunosuppressive cytokines/chemokines, and recruitment of immunosuppressive cells. Immunotherapies for prostate cancer have been developed to activate the innate and adaptive immune responses, such as vaccines and adoptive CAR-T cells, or to inhibit immunosuppressive molecules, such as immune checkpoint inhibitors or antibodies. The U.S Food and Drug Administration has approved Sipuleucel-T for the treatment of asymptomatic or minimally symptomatic metastatic castrate-resistant prostate cancer (mCRPC) and immune checkpoint inhibitor pembrolizumab for the treatment of all solid tumors, including prostate cancer, with impaired mismatch repair genes/microsatellite instability; however, the current clinical outcomes still need to be improved. As various immunosuppressive mechanisms coexist and cross-interact within the tumor microenvironment, different immunotherapy approaches may have to be combined and selected in a highly personalized way. It is hoped that this rapidly evolving field of immunotherapy will achieve successful treatment for mCRPC and will be applied to a wider range of prostate cancer patients.
Collapse
Affiliation(s)
- Belinda L Sun
- Department of Pathology, Banner-University Medical Center, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
2
|
Potluri HK, Ng TL, Newton MA, Zhang J, Maher CA, Nelson PS, McNeel DG. Antibody profiling of patients with prostate cancer reveals differences in antibody signatures among disease stages. J Immunother Cancer 2020; 8:e001510. [PMID: 33335027 PMCID: PMC7745697 DOI: 10.1136/jitc-2020-001510] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Previous studies of prostate cancer autoantibodies have largely focused on diagnostic applications. So far, there have been no reports attempting to more comprehensively profile the landscape of prostate cancer-associated antibodies. Specifically, it is unknown whether the quantity of antibodies or the types of proteins recognized change with disease progression. METHODS A peptide microarray spanning the amino acid sequences of the gene products of 1611 prostate cancer-associated genes was synthesized. Serum samples from healthy male volunteers (n=15) and patients with prostate cancer (n=85) were used to probe the array. These samples included patients with various clinical stages of disease: newly diagnosed localized prostate cancer (n=15), castration-sensitive non-metastatic prostate cancer (nmCSPC, n=40), castration-resistant non-metastatic prostate cancer (n=15) and castration-resistant metastatic disease (n=15). The patients with nmCSPC received treatment with either standard androgen deprivation therapy (ADT) or an antitumor DNA vaccine encoding prostatic acid phosphatase. Serial sera samples from these individuals were also used to probe the array, to secondarily determine whether this approach could be used to detect treatment-related changes. RESULTS We demonstrated that this peptide array yielded highly reproducible measurements of serum IgG levels. We found that the overall number of antibody responses did not increase with disease burden. However, the composition of recognized proteins shifted with clinical stage of disease. Our analysis revealed that the largest difference was between patients with castration-sensitive and castration-resistant disease. Patients with castration-resistant disease recognized more proteins associated with nucleic acid binding and gene regulation compared with men in other groups. Our longitudinal data showed that treatments can elicit antibodies detectable by this array, and notably vaccine-treated patients developed increased responses to more proteins over the course of treatment than did ADT-treated patients. CONCLUSIONS This study represents the largest survey of prostate cancer-associated antibodies to date. We have been able to characterize the classes of proteins recognized by patients and determine how they change with disease burden. Our findings further demonstrate the potential of this platform for measuring antigen spread and studying responses to immunomodulatory therapies.
Collapse
Affiliation(s)
| | - Tun Lee Ng
- Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Michael A Newton
- Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jin Zhang
- Medicine, Washington University in Saint Louis, Saint Louis, Missouri, USA
| | | | - Peter S Nelson
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Douglas G McNeel
- Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
3
|
Kyriakopoulos CE, Eickhoff JC, Ferrari AC, Schweizer MT, Wargowski E, Olson BM, McNeel DG. Multicenter Phase I Trial of a DNA Vaccine Encoding the Androgen Receptor Ligand-binding Domain (pTVG-AR, MVI-118) in Patients with Metastatic Prostate Cancer. Clin Cancer Res 2020; 26:5162-5171. [PMID: 32513836 DOI: 10.1158/1078-0432.ccr-20-0945] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/29/2020] [Accepted: 06/01/2020] [Indexed: 12/18/2022]
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.
Collapse
Affiliation(s)
- Christos E Kyriakopoulos
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jens C Eickhoff
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Biostatistics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Anna C Ferrari
- Department of Oncology, Albert Einstein College of Medicine, Montefiore Medical Center for Cancer Care, New York, New York
| | - Michael T Schweizer
- University of Washington/Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ellen Wargowski
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | | | - Douglas G McNeel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin.
| |
Collapse
|
4
|
Krueger TE, Thorek DLJ, Meeker AK, Isaacs JT, Brennen WN. Tumor-infiltrating mesenchymal stem cells: Drivers of the immunosuppressive tumor microenvironment in prostate cancer? Prostate 2019; 79:320-330. [PMID: 30488530 PMCID: PMC6549513 DOI: 10.1002/pros.23738] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/17/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Prostate cancer is characterized by T-cell exclusion, which is consistent with their poor responses to immunotherapy. In addition, T-cells restricted to the adjacent stroma and benign areas are characterized by anergic and immunosuppressive phenotypes. In order for immunotherapies to produce robust anti-tumor responses in prostate cancer, this exclusion barrier and immunosuppressive microenvironment must first be overcome. We have previously identified mesenchymal stem cells (MSCs) in primary and metastatic human prostate cancer tissue. METHODS An Opal Multiplex immunofluorescence assay based on CD73, CD90, and CD105 staining was used to identify triple-labeled MSCs in human prostate cancer tissue. T-cell suppression assays and flow cytometry were used to demonstrate the immunosuppressive potential of primary MSCs expanded from human bone marrow and prostate cancer tissue from independent donors. RESULTS Endogenous MSCs were confirmed to be present at sites of human prostate cancer. These prostate cancer-infiltrating MSCs suppress T-cell proliferation in a dose-dependent manner similar to their bone marrow-derived counterparts. Also similar to bone marrow-derived MSCs, prostate cancer-infiltrating MSCs upregulate expression of PD-L1 and PD-L2 on their cell surface in the presence of IFNγ and TNFα. CONCLUSION Prostate cancer-infiltrating MSCs suppress T-cell proliferation similar to canonical bone marrow-derived MSCs, which have well-documented immunosuppressive properties with numerous effects on both innate and adaptive immune system function. Thus, we hypothesize that selective depletion of MSCs infiltrating sites of prostate cancer should restore immunologic recognition and elimination of malignant cells via broad re-activation of cytotoxic pro-inflammatory pathways.
Collapse
Affiliation(s)
- Timothy E. Krueger
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Daniel L. J. Thorek
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, Missouri
- Department of Biomedical Engineering, Washington University School of Medicine, Saint Louis, Missouri
| | - Alan K. Meeker
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland
| | - John T. Isaacs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - W. Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
5
|
Heninger E, Krueger TEG, Thiede SM, Sperger JM, Byers BL, Kircher MR, Kosoff D, Yang B, Jarrard DF, McNeel DG, Lang JM. Inducible expression of cancer-testis antigens in human prostate cancer. Oncotarget 2018; 7:84359-84374. [PMID: 27769045 PMCID: PMC5341296 DOI: 10.18632/oncotarget.12711] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 10/11/2016] [Indexed: 12/23/2022] Open
Abstract
Immune tolerance to self-antigens can limit robust anti-tumor immune responses in the use of tumor vaccines. Expression of novel tumor associated antigens can improve immune recognition and lysis of tumor cells. The cancer-testis antigen (CTA) family of proteins has been hypothesized to be an ideal class of antigens due to tumor-restricted expression, a subset of which have been found to induce antibody responses in patients with prostate disease. We demonstrate that CTA expression is highly inducible in five different Prostate Cancer (PC) cell lines using a hypomethylating agent 5-Aza-2′-deoxycytidine (5AZA) and/or a histone deacetylase inhibitor LBH589. These CTAs include NY-ESO1, multiple members of the MAGE and SSX families and NY-SAR35. A subset of CTAs is synergistically induced by the combination of 5AZA and LBH589. We developed an ex vivo organ culture using human PC biopsies for ex vivo drug treatments to evaluate these agents in clinical samples. These assays found significant induction of SSX2 in 9/9 distinct patient samples and NY-SAR35 in 7/9 samples. Further, we identify expression of SSX2 in circulating tumor cells (CTC) from patients with advanced PC. These results indicate that epigenetic modifying agents can induce expression of a broad range of neoantigens in human PC and may serve as a useful adjunctive therapy with novel tumor vaccines and checkpoint inhibitors.
Collapse
Affiliation(s)
- Erika Heninger
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Timothy E G Krueger
- University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Stephanie M Thiede
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Jamie M Sperger
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Brianna L Byers
- University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Madison R Kircher
- University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - David Kosoff
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Bing Yang
- Department of Urology, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - David F Jarrard
- Department of Urology, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Douglas G McNeel
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Joshua M Lang
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| |
Collapse
|
6
|
Johnson LE, Olson BM, McNeel DG. Pretreatment antigen-specific immunity and regulation - association with subsequent immune response to anti-tumor DNA vaccination. J Immunother Cancer 2017; 5:56. [PMID: 28716080 PMCID: PMC5514519 DOI: 10.1186/s40425-017-0260-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 06/23/2017] [Indexed: 12/26/2022] Open
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.
Collapse
Affiliation(s)
- Laura E Johnson
- University of Wisconsin Carbone Cancer Center, 7007 Wisconsin Institutes for Medical Research, University of Wisconsin, Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Brian M Olson
- University of Wisconsin Carbone Cancer Center, 7007 Wisconsin Institutes for Medical Research, University of Wisconsin, Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Douglas G McNeel
- University of Wisconsin Carbone Cancer Center, 7007 Wisconsin Institutes for Medical Research, University of Wisconsin, Madison, 1111 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
7
|
Olson BM, Bradley ES, Sawicki T, Zhong W, Ranheim EA, Bloom JE, Colluru VT, Johnson LE, Rekoske BT, Eickhoff JC, McNeel DG. Safety and Immunological Efficacy of a DNA Vaccine Encoding the Androgen Receptor Ligand-Binding Domain (AR-LBD). Prostate 2017; 77:812-821. [PMID: 28181678 PMCID: PMC5382038 DOI: 10.1002/pros.23321] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/24/2017] [Indexed: 11/11/2022]
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.
Collapse
Affiliation(s)
- Brian M Olson
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| | - Eric S Bradley
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| | - Thomas Sawicki
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| | - Weixiong Zhong
- Department of Pathology, University of Wisconsin, Madison, Wisconsin
| | - Erik A Ranheim
- Department of Pathology, University of Wisconsin, Madison, Wisconsin
| | - Jordan E Bloom
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| | - Viswa T Colluru
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| | - Laura E Johnson
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| | - Brian T Rekoske
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| | - Jens C Eickhoff
- Department of Biostatistics, University of Wisconsin, Madison, Wisconsin
| | - Douglas G McNeel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
8
|
McNeel DG, Bander NH, Beer TM, Drake CG, Fong L, Harrelson S, Kantoff PW, Madan RA, Oh WK, Peace DJ, Petrylak DP, Porterfield H, Sartor O, Shore ND, Slovin SF, Stein MN, Vieweg J, Gulley JL. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of prostate carcinoma. J Immunother Cancer 2016; 4:92. [PMID: 28031820 PMCID: PMC5170901 DOI: 10.1186/s40425-016-0198-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/23/2016] [Indexed: 12/22/2022] Open
Abstract
Prostate cancer is the most commonly diagnosed malignancy and second leading cause of cancer death among men in the United States. In recent years, several new agents, including cancer immunotherapies, have been approved or are currently being investigated in late-stage clinical trials for the management of advanced prostate cancer. Therefore, the Society for Immunotherapy of Cancer (SITC) convened a multidisciplinary panel, including physicians, nurses, and patient advocates, to develop consensus recommendations for the clinical application of immunotherapy for prostate cancer patients. To do so, a systematic literature search was performed to identify high-impact papers from 2006 until 2014 and was further supplemented with literature provided by the panel. Results from the consensus panel voting and discussion as well as the literature review were used to rate supporting evidence and generate recommendations for the use of immunotherapy in prostate cancer patients. Sipuleucel-T, an autologous dendritic cell vaccine, is the first and currently only immunotherapeutic agent approved for the clinical management of metastatic castrate resistant prostate cancer (mCRPC). The consensus panel utilized this model to discuss immunotherapy in the treatment of prostate cancer, issues related to patient selection, monitoring of patients during and post treatment, and sequence/combination with other anti-cancer treatments. Potential immunotherapies emerging from late-stage clinical trials are also discussed. As immunotherapy evolves as a therapeutic option for the treatment of prostate cancer, these recommendations will be updated accordingly.
Collapse
Affiliation(s)
- Douglas G McNeel
- University of Wisconsin Carbone Cancer Center, 7007 WIMR, 1111 Highland Avenue, Madison, WI 53705 USA
| | - Neil H Bander
- Weill Medical College of Cornell University, Laboratory of Urological Oncology E-300, 525 East 68th Street, New York, NY 10021 USA
| | - Tomasz M Beer
- Oregon Health and Science University Knight Cancer Institute, 3181 SW Sam Jackson Park Road, Portland, OR 97239 USA
| | - Charles G Drake
- Johns Hopkins University, 1650 Orleans Street Room 410, Baltimore, MD 21287 USA
| | - Lawrence Fong
- University of California, San Francisco, 513 Parnassus Ave, Room HSF 301, Box 1270, San Francisco, CA 94143 USA
| | - Stacey Harrelson
- Carolina Urologic Research Center, 823 82nd Parkway, Suite B, Myrtle Beach, SC 29572 USA
| | - Philip W Kantoff
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10021 USA
| | - Ravi A Madan
- National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892 USA
| | - William K Oh
- Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1079, New York, NY 10029 USA
| | - David J Peace
- University of Illinois, 840 S Wood Street, Suite 820, Chicago, IL 60612 USA
| | | | - Hank Porterfield
- Alliance for Prostate Cancer Prevention, 17660 Tamiami Trail, Suite 106, Fort Myers, FL 33908 USA
| | - Oliver Sartor
- Tulane University School of Medicine, 1430 Tulane Ave, SL-42, New Orleans, LA 70112 USA
| | - Neal D Shore
- Carolina Urologic Research Center, 823 82nd Parkway, Suite B, Myrtle Beach, SC 29572 USA
| | - Susan F Slovin
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10021 USA
| | - Mark N Stein
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903 USA
| | - Johannes Vieweg
- Nova Southeastern University, 3200 South University Drive, Fort Lauderdale, FL 33328 USA
| | - James L Gulley
- Genitourinary Malignancies Branch, 10 Center Drive, 13N240, Bethesda, MD 20892 USA
| |
Collapse
|
9
|
Abstract
Overexpressed tumor-self antigens represent the largest group of candidate vaccine targets. Those exhibiting a role in oncogenesis may be some of the least studied but perhaps most promising. This review considers this subset of self antigens by highlighting vaccine efforts for some of the better known members and focusing on TPD52, a new promising vaccine target. We shed light on the importance of both preclinical and clinical vaccine studies demonstrating that tolerance and autoimmunity (presumed to preclude this class of antigens from vaccine development) can be overcome and do not present the obstacle that might have been expected. The potential of this class of antigens for broad application is considered, possibly in the context of low tumor burden or adjuvant therapy, as is the need to understand mechanisms of tolerance that are relatively understudied.
Collapse
Key Words
- ALK, Anaplastic lymphoma kinase
- AR, androgen receptor
- CTL, cytotoxic T lymphocyte
- CTLA-4, cytotoxic T lymphocyte-associated antigen 4
- HLA, human leukocyte antigen
- Her-2/neu, human epithelial growth factor receptor 2
- ODN, oligodeoxynucleotide
- Overexpressed tumor-self antigen
- TAA, tumor associated antigen
- TPD52
- TRAMP, Transgenic adenocarcinoma of the mouse prostate
- Treg, T regulatory cell
- VEGFR2, vascular endothelial growth factor receptor 2
- WT-1, Wilms tumor-1
- hD52
- hD52, human TPD52
- mD52
- mD52, murine TPD52
- oncogenic
- shared
- tumor protein D52
- universal
- vaccine
Collapse
Affiliation(s)
- Robert K Bright
- a Department of Immunology and Molecular Microbiology and the TTUHSC Cancer Center ; Texas Tech University Health Sciences Center ; Lubbock , TX USA
| | | | | |
Collapse
|
10
|
Abstract
Cancer immunotherapy was selected as the Breakthrough of the Year 2013 by the editors of Science, in part because of the successful treatment of refractory hematological malignancies with adoptive transfer of chimeric antigen receptor (CAR)-engineered T cells. Effective treatment of B cell leukemia may pave the road to future treatment of solid tumors, using similar approaches. The prostate expresses many unique proteins and, since the prostate gland is a dispensable organ, CAR T cells can potentially be used to target these tissue-specific antigens. However, the location and composition of prostate cancer metastases complicate the task of treating these tumors. It is therefore likely that more sophisticated CAR T cell approaches are going to be required for prostate metastasis than for B cell malignancies. Two main challenges that need to be resolved are how to increase the migration and infiltration of CAR T cells into prostate cancer bone metastases and how to counteract the immunosuppressive microenvironment found in bone lesions. Inclusion of homing (chemokine) receptors in CAR T cells may improve their recruitment to bone metastases, as may antibody-based combination therapies to normalize the tumor vasculature. Optimal activation of CAR T cells through the introduction of multiple costimulatory domains would help to overcome inhibitory signals from the tumor microenvironment. Likewise, combination therapy with checkpoint inhibitors that can reduce tumor immunosuppression may help improve efficacy. Other elegant approaches such as induced expression of immune stimulatory cytokines upon target recognition may also help to recruit other effector immune cells to metastatic sites. Although toxicities are difficult to predict in prostate cancer, severe on-target/off-tumor toxicities have been observed in clinical trials with use of CAR T cells against hematological malignancies; therefore, the choice of the target antigen is going to be crucial. This review focuses on different means of accomplishing maximal effectiveness of CAR T cell therapy for prostate cancer bone metastases while minimizing side effects and CAR T cell-associated toxicities. CAR T cell-based therapies for prostate cancer have the potential to be a therapy model for other solid tumors.
Collapse
|
11
|
Arlen PM. Prostate cancer vaccines: an old yet novel target, the androgen receptor. Expert Rev Vaccines 2013; 12:249-51. [PMID: 23496664 DOI: 10.1586/erv.13.17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cancer vaccines have been utilized as a therapeutic modality to treat prostate cancer in clinical studies for several decades. Recently with the approval of sipuleucel-T, vaccines have now been accepted as standard therapy for this disease. The androgen receptorhas long been recognized as a therapeutic target for the treatment of patients with locally advanced as well as metastatic disease. Recent preclinical studies described by Olson et al. have focused on the androgen receptor as a target for prostate cancer immunotherapy. They have developed and tested a DNA vaccine targeting the ligand-binding domain of the androgen receptor and have demonstrated in animal studies the ability to elicit T-cell responses towards the vaccine that have resulted in both antitumor activity as well as increased survival in the animal models described.
Collapse
Affiliation(s)
- Philip M Arlen
- Precision Biologics, Inc., 9700 Great Seneca Hwy, Suite 321, Rockville, MD 20850, USA.
| |
Collapse
|
12
|
Ingrosso G, Fantini M, Nardi A, Benvenuto M, Sacchetti P, Masuelli L, Ponti E, Frajese GV, Lista F, Schillaci O, Santoni R, Modesti A, Bei R. Local radiotherapy increases the level of autoantibodies to ribosomal P0 protein but not to heat shock proteins, extracellular matrix molecules and EGFR/ErbB2 receptors in prostate cancer patients. Oncol Rep 2012; 29:1167-74. [PMID: 23254686 DOI: 10.3892/or.2012.2197] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 10/22/2012] [Indexed: 11/06/2022] Open
Abstract
Prostate cancer is a common cancer among men in developed countries. Although hormonotherapy and radiotherapy (RT) represent valid therapies for prostate cancer treatment, novel immunological approaches have been explored. The development of clinical trials employing cancer vaccines has indicated that immune response to tumor antigens can be boosted and that vaccine administration can improve patient survival. Immune response to tumor antigens could also be enhanced after standard therapies. In the present study, we determined the occurrence of antibodies to extracellular matrix (ECM) molecules, heat shock protein (HSP), ribosomal P0 protein, EGFR, ErbB2 and prostate-specific antigen (PSA) in 35 prostate cancer patients prior to and following local RT and hormonotherapy. We demonstrated that immunity to P0, ECM molecules [collagens (C) CI, CIII, CV, fibronectin (FN) and laminin (LM)] and to HSP90 was associated with malignancy in untreated patients. None of the patient sera showed antibodies to EGFR, while 2 and 1 patients showed reactivity to ErbB2 and PSA, respectively. We also demonstrated that 8 months after therapy the IgG serum levels to CI, CIII, FN and HSP90 significantly decreased. Conversely, the level of P0 autoantibodies increased after therapy in 10 patients. Five of the 10 patients with increased levels of P0 autoantibodies were treated with RT plus hormonotherapy. Treatment of patients did not change the levels of antibodies against EGFR, ErbB2 and PSA. Our results indicated that the modification of antibody level to self molecules after standard treatment of prostate cancer patients is influenced by the type of antigen. Ribosomal P0 protein appears to be a high immunogenic antigen and its immunogenicity increases following RT. In addition, 10 patients with increased levels of autoantibodies to P0 showed PSA mean levels lower than the remaining 25 patients at 18 months. This study may contribute to a better understanding of the immunobiological behavior of prostate cancer patients following standard treatment.
Collapse
Affiliation(s)
- Gianluca Ingrosso
- Department of Diagnostic Imaging, Molecular Imaging, Interventional Radiology and Radiotherapy, University of Rome Tor Vergata, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Olson BM, Johnson LE, McNeel DG. The androgen receptor: a biologically relevant vaccine target for the treatment of prostate cancer. Cancer Immunol Immunother 2012; 62:585-96. [PMID: 23108626 DOI: 10.1007/s00262-012-1363-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 10/12/2012] [Indexed: 10/27/2022]
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.
Collapse
Affiliation(s)
- Brian M Olson
- University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, Madison, WI 53705, USA.
| | | | | |
Collapse
|
14
|
Johnson LE, McNeel DG. Identification of prostatic acid phosphatase (PAP) specific HLA-DR1-restricted T-cell epitopes. Prostate 2012; 72:730-40. [PMID: 22529020 DOI: 10.1002/pros.21477] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 07/18/2011] [Indexed: 11/08/2022]
Abstract
BACKGROUND Prostatic acid phosphatase (PAP) is a prostate cancer tumor antigen and is an immunological target in several active immunotherapy clinical trials for the treatment of prostate cancer. We and others have demonstrated that PAP-specific T-cell responses can be elicited and augmented following antigen-specific immunization in both humans and animal models. We have previously reported that prostate cancer patients immunized with a DNA vaccine encoding PAP (pTVG-HP) developed both CD4+ and CD8+ T-cell responses. PAP-specific, CD4+ T-cell proliferative responses were generated in three out of four HLA-DRB1*0101 patients suggesting the possibility that DR1-restricted epitopes exist. METHODS To identify PAP-specific HLA-DRB1*0101 restricted epitopes, we immunized HLA-A2.01/HLA-DRB1*0101 (A2/DR1) transgenic mice with the pTVG-HP DNA vaccine. To map DRB1*0101-restricted epitopes, splenocytes from immunized mice were screened against a library of overlapping 15-residue, PAP-derived peptides using an IFNγ ELISPOT assay. RESULTS We identified four HLA-DRB1*0101 epitopes for PAP in A2/DR1 mice (PAP(161-175) , PAP(181-195) , PAP(191-205) , and PAP (351-365) ). T cells specific for one epitope (PAP(181-195) ) were found to be augmented after immunization in a HLA-DRB1*0101+ prostate cancer patient. CONCLUSIONS The identification of MHC class II epitopes may provide tools to directly monitor immune responses after vaccination and may be important for the design of future prostate cancer vaccines.
Collapse
Affiliation(s)
- Laura E Johnson
- Department of Medicine, University of Wisconsin, Madison, Wisconsin 53705, USA
| | | |
Collapse
|
15
|
Zabransky DJ, Smith HA, Thoburn CJ, Zahurak M, Keizman D, Carducci M, Eisenberger MA, McNeel DG, Drake CG, Antonarakis ES. Lenalidomide modulates IL-8 and anti-prostate antibody levels in men with biochemically recurrent prostate cancer. Prostate 2012; 72:487-98. [PMID: 21748755 PMCID: PMC3248613 DOI: 10.1002/pros.21449] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 06/10/2011] [Indexed: 12/25/2022]
Abstract
BACKGROUND We retrospectively explored changes in immunological parameters in men with biochemically recurrent prostate cancer treated with either 5 or 25 mg of lenalidomide in a randomized phase 2 trial, and determined whether those changes correlated with disease progression. METHODS Cytokine levels were compared for each patient at baseline and after 6 months of treatment with lenalidomide. Regression models for correlated data were used to assess associations of cytokine levels with lenalidomide treatment effect. Estimates were obtained using generalized estimating equations. Changes in circulating anti-prostate antibodies were evaluated using a high-throughput immunoblot technique. RESULTS Treatment with lenalidomide was associated with global changes in immunoreactivity to a number of prostate-associated antigens, as well as with changes in circulating levels of the T(H) 2 cytokines IL-4, IL-5, IL-10, and IL-13. Disease progression in treated patients was associated with an increase in circulating IL-8 levels, while IL-8 levels decreased significantly in non-progressors. CONCLUSIONS Lenalidomide demonstrates immunomodulatory properties in patients with biochemically recurrent prostate cancer. The induction of novel anti-prostate antibodies is a potential mechanism for lenalidomide response. Changes in serum IL-8 levels may serve as a potential biomarker in treated patients. These hypotheses require formal testing in future prospective trials.
Collapse
Affiliation(s)
- Daniel J. Zabransky
- Johns Hopkins Kimmel Cancer Center, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Heath A. Smith
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI, 53705, USA
| | | | - Marianna Zahurak
- Johns Hopkins Kimmel Cancer Center, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Daniel Keizman
- Johns Hopkins Kimmel Cancer Center, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Michael Carducci
- Johns Hopkins Kimmel Cancer Center, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Mario A. Eisenberger
- Johns Hopkins Kimmel Cancer Center, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Douglas G. McNeel
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI, 53705, USA
| | - Charles G. Drake
- Johns Hopkins Kimmel Cancer Center, Johns Hopkins University, Baltimore, MD, 21231, USA
| | | |
Collapse
|
16
|
Abstract
Prostate cancer continues to be one of the most serious afflictions of men of advanced age, remaining the most commonly diagnosed and second leading cause of cancer-related deaths in American men. The treatment options for patients with incurable metastatic, castrate-resistant disease have long focused on various chemotherapeutic approaches, which provide a slight survival benefit while being associated with potentially significant side effects. However, the recent approval of sipuleucel-T has given patients with advanced disease an additional treatment option that has demonstrated benefit without the side effects associated with chemotherapy. Sipuleucel-T is an antigen-presenting cell-based active immunotherapy that utilizes a patient’s own immune cells, presumably to activate an antigen-specific immune response against tumor cells. This review focuses on the development and implementation of sipuleucel-T as a therapy for prostate cancer. Specifically, we present some of the issues associated with the management of advanced prostate cancer, the research and development that led to the approval of sipuleucel-T, how the approval of sipuleucel-T could change the clinical management of prostate cancer, and current and future areas of investigation that are being pursued with regard to sipuleucel-T and other treatments for advanced prostate cancer.
Collapse
Affiliation(s)
- Brian M Olson
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | | |
Collapse
|
17
|
CD8+ T cells specific for the androgen receptor are common in patients with prostate cancer and are able to lyse prostate tumor cells. Cancer Immunol Immunother 2011; 60:781-92. [PMID: 21350948 DOI: 10.1007/s00262-011-0987-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Accepted: 02/06/2011] [Indexed: 12/29/2022]
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.
Collapse
|
18
|
Maricque BB, Eickhoff JC, McNeel DG. Antibody responses to prostate-associated antigens in patients with prostatitis and prostate cancer. Prostate 2011; 71:134-46. [PMID: 20632317 PMCID: PMC2978272 DOI: 10.1002/pros.21229] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND An important focus of tumor immunotherapy has been the identification of appropriate antigenic targets. Serum-based screening approaches have led to the discovery of hundreds of tumor-associated antigens recognized by IgG. Our efforts to identify immunologically recognized proteins in prostate cancer have yielded a multitude of antigens; however, prioritizing these antigens as targets for evaluation in immunotherapies has been challenging. In this report, we set out to determine whether the evaluation of multiple antigenic targets would allow the identification of a subset of antigens that are common immunologic targets in patients with prostate cancer. METHODS Using a phage immunoblot approach, we evaluated IgG responses in patients with prostate cancer (n = 126), patients with chronic prostatitis (n = 45), and men without prostate disease (n = 53). RESULTS We found that patients with prostate cancer or prostatitis have IgG specific for multiple common antigens. A subset of 23 proteins was identified to which IgG were detected in 38% of patients with prostate cancer and 33% patients with prostatitis versus 6% of controls (P < 0.001 and P = 0.003, respectively). Responses to multiple members were not higher in patients with advanced disease, suggesting antibody immune responses occur early in the natural history of cancer progression. CONCLUSIONS These findings suggest an association between inflammatory conditions of the prostate and prostate cancer, and suggest that IgG responses to a panel of commonly recognized prostate antigens could be potentially used in the identification of patients at risk for prostate cancer or as a tool to identify immune responses elicited to prostate tissue.
Collapse
Affiliation(s)
- Brett B. Maricque
- University of Wisconsin Carbone Comprehensive Cancer Center, 1111 Highland Avenue, Madison, WI 53705
| | - Jens C. Eickhoff
- Colorado State University, Department of Statistics, Fort Collins, CO 80523
| | - Douglas G. McNeel
- University of Wisconsin Carbone Comprehensive Cancer Center, 1111 Highland Avenue, Madison, WI 53705
- To whom correspondence should be addressed: 7007 Wisconsin Institutes for Medical Research, 1111 Highland Avenue, Madison, WI 53705. Tel: (608) 265-8131 Fax: (608) 265-0614
| |
Collapse
|
19
|
Abstract
Prostate cancer is a significant public health problem, and the most commonly diagnosed cancer in the USA. The long natural history of prostate cancer, the presence of a serum biomarker that can be used to detect very early recurrences, and the previous identification of multiple potential tissue-specific target antigens are all features that make this disease suitable for the development of anti-tumor vaccines. To date, many anti-tumor vaccines have entered clinical testing for patients with prostate cancer, and some have demonstrated clinical benefit. DNA vaccines represent one vaccine approach that has been evaluated in multiple preclinical models and clinical trials. The safety, specificity for the target antigen, ease of manufacturing and ease of incorporating other immune-modulating approaches make DNA vaccines particularly relevant for future development. This article focuses on DNA vaccines specifically in the context of prostate cancer treatment, focusing on antigens targeted in preclinical models, recent clinical trials and efforts to improve the potency of these vaccines.
Collapse
Affiliation(s)
- Sheeba Alam
- Department of Medicine, University of Wisconsin Carbone Comprehensive Cancer Center, Madison, WI, USA
| | | |
Collapse
|
20
|
Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, Mellman I, Prindiville SA, Viner JL, Weiner LM, Matrisian LM. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res 2009; 15:5323-37. [PMID: 19723653 DOI: 10.1158/1078-0432.ccr-09-0737] [Citation(s) in RCA: 1017] [Impact Index Per Article: 63.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The purpose of the National Cancer Institute pilot project to prioritize cancer antigens was to develop a well-vetted, priority-ranked list of cancer vaccine target antigens based on predefined and preweighted objective criteria. An additional aim was for the National Cancer Institute to test a new approach for prioritizing translational research opportunities based on an analytic hierarchy process for dealing with complex decisions. Antigen prioritization involved developing a list of "ideal" cancer antigen criteria/characteristics, assigning relative weights to those criteria using pairwise comparisons, selecting 75 representative antigens for comparison and ranking, assembling information on the predefined criteria for the selected antigens, and ranking the antigens based on the predefined, preweighted criteria. Using the pairwise approach, the result of criteria weighting, in descending order, was as follows: (a) therapeutic function, (b) immunogenicity, (c) role of the antigen in oncogenicity, (d) specificity, (e) expression level and percent of antigen-positive cells, (f) stem cell expression, (g) number of patients with antigen-positive cancers, (h) number of antigenic epitopes, and (i) cellular location of antigen expression. None of the 75 antigens had all of the characteristics of the ideal cancer antigen. However, 46 were immunogenic in clinical trials and 20 of them had suggestive clinical efficacy in the "therapeutic function" category. These findings reflect the current status of the cancer vaccine field, highlight the possibility that additional organized efforts and funding would accelerate the development of therapeutically effective cancer vaccines, and accentuate the need for prioritization.
Collapse
Affiliation(s)
- Martin A Cheever
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Sfanos KS, Bruno TC, Meeker AK, De Marzo AM, Isaacs WB, Drake CG. Human prostate-infiltrating CD8+ T lymphocytes are oligoclonal and PD-1+. Prostate 2009; 69:1694-703. [PMID: 19670224 PMCID: PMC2782577 DOI: 10.1002/pros.21020] [Citation(s) in RCA: 183] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Prostate-infiltrating CD8(+) T lymphocytes (CD8(+) PIL) are prevalent in men with prostate cancer (PCa), however, it is unclear whether the presence of such cells reflects a non-specific immune infiltrate or an oligoclonal, antigen-driven adaptive immune response. METHODS We investigated the complexity of the T-cell receptor (TCR) repertoire in the prostate gland by examining the diversity of CD8(+) TCR beta chain variable region (Vbeta) gene sequences in both the peripheral blood and prostates of cancer patients. Vbeta repertoire analysis was performed by family-specific Vbeta spectratyping and flow cytometry, as well as direct sequence analysis (5' RACE and cloning). Programmed cell death 1 (PD-1 or PDCD1) expression on peripheral blood CD8(+) T cells and CD8(+) PIL was analyzed by flow cytometry. RESULTS CD8(+) PIL isolated from cancer patients exhibited restricted TCR Vbeta gene usage, and identical clones were identified in multiple sites within the prostate. Furthermore, CD8(+) PIL express high levels of the inhibitory receptor PD-1, a cell surface protein associated with an "exhausted" CD8(+) T-cell phenotype. CONCLUSIONS CD8(+) PIL appear to have undergone clonal expansion in response to an as yet unidentified antigen; however, due to the high expression of PD-1, these cells are likely incapable of mounting an effective immune response. The results provide an important basis for further efforts aimed at the identification of specific antigens involved in prostatic inflammation, and suggest that PD-1 blockade may be useful in immunotherapy for PCa.
Collapse
MESH Headings
- Adenocarcinoma/immunology
- Adult
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Antigens, CD/immunology
- Apoptosis Regulatory Proteins/biosynthesis
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/immunology
- Biopsy
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Clone Cells/immunology
- Clone Cells/pathology
- Flow Cytometry
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Male
- Middle Aged
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/pathology
- Programmed Cell Death 1 Receptor
- Prostatic Neoplasms/immunology
- Random Amplified Polymorphic DNA Technique
- Receptors, Antigen, T-Cell/biosynthesis
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Karen S. Sfanos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Tullia C. Bruno
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Alan K. Meeker
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Angelo M. De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - William B. Isaacs
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Charles G. Drake
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Correspondence to: Charles G. Drake, Johns Hopkins SKCCC, 1650 Orleans Street, CRBI 410, Baltimore, MD 21231. Phone: 410-502-7523; Fax: 443-287-4653;
| |
Collapse
|
22
|
Arredouani MS, Lu B, Bhasin M, Eljanne M, Yue W, Mosquera JM, Bubley GJ, Li V, Rubin MA, Libermann TA, Sanda MG. Identification of the transcription factor single-minded homologue 2 as a potential biomarker and immunotherapy target in prostate cancer. Clin Cancer Res 2009; 15:5794-802. [PMID: 19737960 DOI: 10.1158/1078-0432.ccr-09-0911] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Identification of novel biomarkers and immunotherapy targets for prostate cancer (PCa) is crucial to better diagnosis and therapy. We sought to identify novel PCa tumor-associated antigens (TAA) that are expressed in PCa, absent in nonprostate human tissue, and immunogenic for immune responses restricted by human HLA. EXPERIMENTAL DESIGN AND RESULTS Using microarray analysis of normal and cancerous human prostate tissues, we identified 1,063 genes overexpressed in PCa. After validating 195 transcripts in publicly available array data sets, we interrogated expression of these TAAs in normal human tissues to identify genes that are not expressed at detectable levels in normal, nonprostate adult human tissue. We identified 23 PCa TAA candidates. Real-time PCR confirmed that 15 of these genes were overexpressed in PCa (P< 0.05 for each). The most frequently overexpressed gene, single-minded homologue 2 (SIM2), was selected for further evaluation as a potential target for immunotherapy. ELISA assay revealed that a fraction of PCa patients exhibited immune responsiveness to SIM2 as evidenced by the presence of autoantibodies to SIM2 in their sera. We next showed binding of putative HLA-A2.1-restricted SIM2 epitopes to human A2.1, and immunization of transgenic HLA-A2.1 mice showed induction of SIM2-specific CTL responses in vivo. CONCLUSIONS Our findings that SIM2 is selectively expressed in PCa, that human HLA-A2.1-restricted SIM2 epitopes induce specific T cells in vivo, and that anti-SIM2 antibodies are detectable in PCa patients' sera implicate SIM2 as a PCa-associated antigen that is a suitable potential target for PCa immunotherapy.
Collapse
Affiliation(s)
- Mohamed S Arredouani
- Division of Urology, Department of Surgery, Genomics Center, and Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|