1
|
Rastogi I, Mannone JA, Gibadullin R, Moseman JE, Sidney J, Sette A, McNeel DG, Gellman SH. β-amino acid substitution in the SIINFEKL antigen alters immunological recognition. Cancer Biol Ther 2025; 26:2486141. [PMID: 40200635 PMCID: PMC11988276 DOI: 10.1080/15384047.2025.2486141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Peptide vaccines offer a direct way to initiate an immunogenic response to a defined antigen epitope. However, peptide vaccines are unstable in vivo, subject to rapid enzymatic proteolysis. Replacement of an α-amino acid residue with a homologous β-amino acid residue (native side chain, but backbone extended by a single CH2 unit) impairs proteolysis at nearby amide bonds. Therefore, antigen analogues containing α-to-β replacements have been examined for functional mimicry of native all-α antigens. Another group previously took this approach in the ovalbumin (OVA) antigen model by evaluating single α-to-β analogues of the murine major histocompatibility complex (MHC) I-restricted peptide SIINFEKL. METHODS We re-examined this set of α/β SIINFEKL antigens. We tested the susceptibility to proteolysis in mouse serum and their ability to activate OVA-antigen-specific CD8 T cells in vitro. Additionally, we tested the α/β antigens in vivo for their ability to induce an antigen-specific immunogenic response in naïve mice and in OVA-expressing tumor-bearing mice. RESULTS The α/β antigens were comparable to the native antigen in their susceptibility to proteolysis in serum. Each α/β antigen was capable of activating antigen-specific CD8 T cells in vitro. However, antigen-specific CD8 T cells induced against α/β antigens in vivo were not cross-reactive to the native antigen. Moreover, immunization with α/β analogues did not elicit anti-tumor effects in tumor-bearing mice. CONCLUSIONS We conclude that even though α/β analogues of the SIINFEKL antigen can elicit a T cell-based response, this class of backbone-modified peptides is not promising from the perspective of antitumor vaccine development.
Collapse
Affiliation(s)
- Ichwaku Rastogi
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - John A. Mannone
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Ruslan Gibadullin
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jena E. Moseman
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California, San Diego, CA, USA
| | - Douglas G. McNeel
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Samuel H. Gellman
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
2
|
Gomari MM, Ghantabpour T, Pourgholam N, Rostami N, Hatfield SM, Namazifar F, Abkhiz S, Eslami SS, Ramezanpour M, Darestanifarahani M, Astsaturov I, Bencherif SA. Breaking barriers: Smart vaccine platforms for cancer immunomodulation. Cancer Commun (Lond) 2025; 45:529-571. [PMID: 39901621 PMCID: PMC12067400 DOI: 10.1002/cac2.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Despite significant advancements in cancer treatment, current therapies often fail to completely eradicate malignant cells. This shortfall underscores the urgent need to explore alternative approaches such as cancer vaccines. Leveraging the immune system's natural ability to target and kill cancer cells holds great therapeutic potential. However, the development of cancer vaccines is hindered by several challenges, including low stability, inadequate immune response activation, and the immunosuppressive tumor microenvironment, which limit their efficacy. Recent progress in various fields, such as click chemistry, nanotechnology, exosome engineering, and neoantigen design, offer innovative solutions to these challenges. These achievements have led to the emergence of smart vaccine platforms (SVPs), which integrate protective carriers for messenger ribonucleic acid (mRNA) with functionalization strategies to optimize targeted delivery. Click chemistry further enhances SVP performance by improving the encapsulation of mRNA antigens and facilitating their precise delivery to target cells. This review highlights the latest developments in SVP technologies for cancer therapy, exploring both their opportunities and challenges in advancing these transformative approaches.
Collapse
Affiliation(s)
- Mohammad Mahmoudi Gomari
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
| | - Taha Ghantabpour
- Department of AnatomySchool of MedicineQazvin University of Medical SciencesQazvinIran
| | - Nima Pourgholam
- School of Nursing and MidwiferyIran University of Medical ScienceTehranIran
| | - Neda Rostami
- Department of Chemical EngineeringArak UniversityArakIran
| | - Stephen M. Hatfield
- New England Inflammation and Tissue Protection InstituteDepartment of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
- Department of Chemical EngineeringNortheastern UniversityBostonMassachusettsUSA
| | | | - Shadi Abkhiz
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
| | - Seyed Sadegh Eslami
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
- Molecular Proteomics LaboratoryBaker Heart and Diabetes InstituteMelbourneVictoriaAustralia
| | - Mahsa Ramezanpour
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
| | | | - Igor Astsaturov
- Marvin and Concetta Greenberg Pancreatic Cancer InstituteFox Chase Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Sidi A. Bencherif
- Department of Chemical EngineeringNortheastern UniversityBostonMassachusettsUSA
- Harvard John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Polymers, Biopolymers Surfaces (PBS) LaboratoryNational Center for Scientific Research (CNRS) Mixed Research Unit (UMR) 6270University Rouen NormandieRouenFrance
| |
Collapse
|
3
|
Yaraghi P, Kheyri A, Mikaeili N, Boroumand A, Abbasifard M, Farhangnia P, Rezagholizadeh F, Khorramdelazad H. Nanoparticle-mediated enhancement of DNA Vaccines: Revolutionizing immunization strategies. Int J Biol Macromol 2025; 302:140558. [PMID: 39900152 DOI: 10.1016/j.ijbiomac.2025.140558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/25/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025]
Abstract
DNA vaccines are a novel form of vaccination that aims to harness genetic material to produce targeted immune responses. Nevertheless, their therapeutic application is hampered by low transfection efficacy, immunogenicity, and instability. Nanoparticle (NP) - based delivery systems are beneficial in enhancing DNA stability, increasing DNA uptake by antigen-presenting cells (APCs), and controlling antigen release. Some key progress includes the polymeric, lipid-based, and hybrid NPs and biocompatible carriers with inherent adjuvant effects. These systems have helped to enhance the antigen cross-presentation and T-cell activation significantly. In addition, biocompatible hybrid nanocarriers, antigen cross-presentation strategies, and next-generation sequencing (NGS) technologies are speeding up the identification of new antigens, while AI and machine learning are facilitating the development of efficient delivery systems. This review aims to assess how NPs have contributed to improving the effectiveness of DNA vaccines for treating diseases, cancer, and emerging diseases, as well as advancing the next generation of DNA vaccines.
Collapse
Affiliation(s)
- Pegah Yaraghi
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Abbas Kheyri
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Narges Mikaeili
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Armin Boroumand
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mitra Abbasifard
- Department of Internal Medicine, School of Medicine, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Pooya Farhangnia
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Rezagholizadeh
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
4
|
Tonelli TP, Eickhoff JC, Johnson LE, Liu G, McNeel DG. Long-term follow up of patients treated with a DNA vaccine (pTVG-hp) for PSA-recurrent prostate cancer. Hum Vaccin Immunother 2024; 20:2395680. [PMID: 39208856 PMCID: PMC11364063 DOI: 10.1080/21645515.2024.2395680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/25/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
We have previously reported two single-agent phase I trials, evaluating the dose or schedule, of a DNA vaccine (pTVG-HP) encoding prostatic acid phosphatase (PAP) administered with GM-CSF as the adjuvant. These were in patients with PSA-recurrent, radiographically non-metastatic, prostate cancer (PCa). We report here the long-term safety and overall survival of these patients. Specifically, 22 patients with non-metastatic, castration-sensitive PCa (nmCSPC) were treated with pTVG-HP, 100-1500 µg, administered over 12 weeks and followed for 15 y. 17 patients with non-metastatic castration-resistant PCa (nmCRPC) were treated with 100 µg pTVG-HP with different schedules of administration over 1 y and followed for 5 y. No adverse events were detected in long-term follow-up from either trial that were deemed possibly related to vaccination. Patients with nmCSPC had a median overall survival of 12.3 y, with 5/22 (23%) alive at 15 y. 8/22 (36%) died due to prostate cancer with a median survival of 11.0 y, and 9/22 (41%) died of other causes. Patients with nmCRPC had a median overall survival of 4.5 y, with 8/17 (47%) alive at 5 y. The presence of T-cells specific for the PAP target antigen was detectable in 6/10 (60%) individuals with nmCSPC, and 3/5 (60%) individuals with nmCRPC, many years after immunization. The detection of immune responses to the vaccine target years after immunization suggests durable immunity can be elicited in patients using a DNA vaccine encoding a tumor-associated antigen.Trial Registration: NCT00582140 and NCT00849121.
Collapse
Affiliation(s)
- Tommaso P. Tonelli
- University of Wisconsin Carbone Comprehensive Cancer Center, Madison, WI, USA
| | - Jens C. Eickhoff
- University of Wisconsin Carbone Comprehensive Cancer Center, Madison, WI, USA
- Department of Biostatistics, University of Wisconsin, Madison, WI, USA
| | - Laura E. Johnson
- University of Wisconsin Carbone Comprehensive Cancer Center, Madison, WI, USA
| | - Glenn Liu
- University of Wisconsin Carbone Comprehensive Cancer Center, Madison, WI, USA
- Department of Medicine, University of Wisconsin, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin, Madison, WI, USA
| | - Douglas G. McNeel
- University of Wisconsin Carbone Comprehensive Cancer Center, Madison, WI, USA
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
5
|
Liu Q, Ma H. Cancer biotherapy: review and prospect. Clin Exp Med 2024; 24:114. [PMID: 38801637 PMCID: PMC11130057 DOI: 10.1007/s10238-024-01376-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
Malignant tumors pose a grave threat to the quality of human life. The prevalence of malignant tumors in China is steadily rising. Presently, clinical interventions encompass surgery, radiotherapy, and pharmaceutical therapy in isolation or combination. Nonetheless, these modalities fail to completely eradicate malignant tumor cells, frequently leading to metastasis and recurrence. Conversely, tumor biotherapy has emerged as an encouraging fourth approach in preventing and managing malignant tumors owing to its safety, efficacy, and minimal adverse effects. Currently, a range of tumor biotherapy techniques are employed, including gene therapy, tumor vaccines, monoclonal antibody therapy, cancer stem cell therapy, cytokine therapy, and adoptive cellular immunotherapy. This study aims to comprehensively review the latest developments in biological treatments for malignant tumors.
Collapse
Affiliation(s)
- Qi Liu
- Zunyi Medical University, Zunyi, Guizhou, 563000, China
- Department of Thoracic Oncology, The Second Affiliated Hospital of Zunyi Medical University, Guizhou, 56300, Zunyi, China
| | - Hu Ma
- Zunyi Medical University, Zunyi, Guizhou, 563000, China.
- Department of Thoracic Oncology, The Second Affiliated Hospital of Zunyi Medical University, Guizhou, 56300, Zunyi, China.
| |
Collapse
|
6
|
Abstract
Prostate cancer is a leading cause of death in men worldwide. For over 30 years, growing interest has focused on the development of vaccines as treatments for prostate cancer, with the goal of using vaccines to activate immune cells capable of targeting prostate cancer to either eradicate recurrent disease or at least delay disease progression. This interest has been prompted by the prevalence and long natural history of the disease and by the fact that the prostate is an expendable organ. Thus, an immune response elicited by vaccination might not need to target the tumour uniquely but could theoretically target any prostate tissue. To date, different vaccine approaches and targets for prostate cancer have been evaluated in clinical trials. Overall, five approaches have been assessed in randomized phase III trials and sipuleucel-T was approved as a treatment for metastatic castration-resistant prostate cancer, being the only vaccine approved to date by the FDA as a treatment for cancer. Most vaccine approaches showed safety and some evidence of immunological activity but had poor clinical activity when used as monotherapies. However, increased activity has been observed when these vaccines were used in combination with other immune-modulating therapies. This evidence suggests that, in the future, prostate cancer vaccines might be used to activate and expand tumour-specific T cells as part of combination approaches with agents that target tumour-associated immune mechanisms of resistance.
Collapse
Affiliation(s)
- Ichwaku Rastogi
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Anusha Muralidhar
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Douglas G McNeel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
7
|
Hawlina S, Zorec R, Chowdhury HH. Potential of Personalized Dendritic Cell-Based Immunohybridoma Vaccines to Treat Prostate Cancer. Life (Basel) 2023; 13:1498. [PMID: 37511873 PMCID: PMC10382052 DOI: 10.3390/life13071498] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Prostate cancer (PCa) is the most commonly diagnosed cancer and the second most common cause of death due to cancer. About 30% of patients with PCa who have been castrated develop a castration-resistant form of the disease (CRPC), which is incurable. In the last decade, new treatments that control the disease have emerged, slowing progression and spread and prolonging survival while maintaining the quality of life. These include immunotherapies; however, we do not yet know the optimal combination and sequence of these therapies with the standard ones. All therapies are not always suitable for every patient due to co-morbidities or adverse effects of therapies or both, so there is an urgent need for further work on new therapeutic options. Advances in cancer immunotherapy with an immune checkpoint inhibition mechanism (e.g., ipilimumab, an anti-CTLA-4 inhibitor) have not shown a survival benefit in patients with CRPC. Other immunological approaches have also not given clear results, which has indirectly prevented breakthrough for this type of therapeutic strategy into clinical use. Currently, the only approved form of immunotherapy for patients with CRPC is a cell-based medicine, but it is only available to patients in some parts of the world. Based on what was gained from recently completed clinical research on immunotherapy with dendritic cell-based immunohybridomas, the aHyC dendritic cell vaccine for patients with CRPC, we highlight the current status and possible alternatives that should be considered in the future.
Collapse
Affiliation(s)
- Simon Hawlina
- Clinical Department of Urology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Surgery, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Helena H Chowdhury
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
8
|
Physical and in silico immunopeptidomic profiling of a cancer antigen prostatic acid phosphatase reveals targets enabling TCR isolation. Proc Natl Acad Sci U S A 2022; 119:e2203410119. [PMID: 35878026 PMCID: PMC9351518 DOI: 10.1073/pnas.2203410119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tissue-specific antigens can serve as targets for adoptive T cell transfer-based cancer immunotherapy. Recognition of tumor by T cells is mediated by interaction between peptide-major histocompatibility complexes (pMHCs) and T cell receptors (TCRs). Revealing the identity of peptides bound to MHC is critical in discovering cognate TCRs and predicting potential toxicity. We performed multimodal immunopeptidomic analyses for human prostatic acid phosphatase (PAP), a well-recognized tissue antigen. Three physical methods, including mild acid elution, coimmunoprecipitation, and secreted MHC precipitation, were used to capture a thorough signature of PAP on HLA-A*02:01. Eleven PAP peptides that are potentially A*02:01-restricted were identified, including five predicted strong binders by NetMHCpan 4.0. Peripheral blood mononuclear cells (PBMCs) from more than 20 healthy donors were screened with the PAP peptides. Seven cognate TCRs were isolated which can recognize three distinct epitopes when expressed in PBMCs. One TCR shows reactivity toward cell lines expressing both full-length PAP and HLA-A*02:01. Our results show that a combined multimodal immunopeptidomic approach is productive in revealing target peptides and defining the cloned TCR sequences reactive with prostatic acid phosphatase epitopes.
Collapse
|
9
|
Liu C, Cong X, Wang Y, Guo Q, Xie Y, Geng F, Guo J, Dong L, Zhou Y, Wu H, Yu B, Wu J, Zhang H, Yu X, Kong W. Fast DNA Vaccination Strategy Elicits a Stronger Immune Response Dependent on CD8 +CD11c + Cell Accumulation. Front Oncol 2021; 11:752444. [PMID: 34950581 PMCID: PMC8691261 DOI: 10.3389/fonc.2021.752444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
Conventional DNA vaccine strategies usually employ a regimen of immunizations at 2-week or longer intervals to induce effective memory cell-dependent immune responses. Clinical cancer treatment requires a faster immunization strategy to contend with tumor progression. In this study, a novel fast immunization strategy was established, wherein a DNA vaccine was intramuscularly administered on days 0, 2, and 5 in a murine lung cancer model. Effector cells peaked 7 to 10 days after the last vaccination. Compared with traditional 2-week-interval immunization strategies, antigen-specific cytolysis and INF-γ secretion were significantly enhanced under the fast vaccination approach. As a result, the rapidly administered DNA vaccine elicited stronger and more prompt antitumor effects. The probable underlying mechanism of fast immunization was the accumulation of CD8+CD11c+ antigen-presenting cells at the injection site, which enhanced subsequent antigen presentation. In conclusion, the fast DNA vaccination strategy shortened vaccination time to 5 days and elicited a stronger antitumor immune response.
Collapse
Affiliation(s)
- Chenlu Liu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Biobank, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Xianling Cong
- Biobank, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Yuqian Wang
- Biobank, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Qianqian Guo
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Yu Xie
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Fei Geng
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Jie Guo
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Ling Dong
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Yi Zhou
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Hui Wu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Jiaxin Wu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Haihong Zhang
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, College of Life Science, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, College of Life Science, Jilin University, Changchun, China
| |
Collapse
|
10
|
Movassaghi M, Chung R, Anderson CB, Stein M, Saenger Y, Faiena I. Overcoming Immune Resistance in Prostate Cancer: Challenges and Advances. Cancers (Basel) 2021; 13:cancers13194757. [PMID: 34638243 PMCID: PMC8507531 DOI: 10.3390/cancers13194757] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Immunotherapy has changed the landscape of treatment modalities available for many different types of malignancies. However, the factors that influence the success of immunotherapeutics have not been as clearly seen in advanced prostate cancer, likely due to immunosuppressive factors that exist within the prostate cancer tumor microenvironment. While there have been many immunotherapeutics used for prostate cancer, the majority have targeted a single immunosuppressive mechanism resulting in limited clinical efficacy. More recent research centered on elucidating the key mechanisms of immune resistance in the prostate tumor microenvironment has led to the discovery of a range of new treatment targets. With that in mind, many clinical trials have now set out to evaluate combination immunotherapeutic strategies in patients with advanced prostate cancer, in the hopes of circumventing the immunosuppressive mechanisms. Abstract The use of immunotherapy has become a critical treatment modality in many advanced cancers. However, immunotherapy in prostate cancer has not been met with similar success. Multiple interrelated mechanisms, such as low tumor mutational burden, immunosuppressive cells, and impaired cellular immunity, appear to subvert the immune system, creating an immunosuppressive tumor microenvironment and leading to lower treatment efficacy in advanced prostate cancer. The lethality of metastatic castrate-resistant prostate cancer is driven by the lack of therapeutic regimens capable of generating durable responses. Multiple strategies are currently being tested to overcome immune resistance including combining various classes of treatment modalities. Several completed and ongoing trials have shown that combining vaccines or checkpoint inhibitors with hormonal therapy, radiotherapy, antibody–drug conjugates, chimeric antigen receptor T cell therapy, or chemotherapy may enhance immune responses and induce long-lasting clinical responses without significant toxicity. Here, we review the current state of immunotherapy for prostate cancer, as well as tumor-specific mechanisms underlying therapeutic resistance, with a comprehensive look at the current preclinical and clinical immunotherapeutic strategies aimed at overcoming the immunosuppressive tumor microenvironment and impaired cellular immunity that have largely limited the utility of immunotherapy in advanced prostate cancer.
Collapse
Affiliation(s)
- Miyad Movassaghi
- Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA; (R.C.); (C.B.A.)
- Correspondence: (M.M.); (I.F.)
| | - Rainjade Chung
- Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA; (R.C.); (C.B.A.)
| | - Christopher B. Anderson
- Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA; (R.C.); (C.B.A.)
| | - Mark Stein
- Department of Medicine, Division of Medical Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (Y.S.)
| | - Yvonne Saenger
- Department of Medicine, Division of Medical Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA; (M.S.); (Y.S.)
| | - Izak Faiena
- Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA; (R.C.); (C.B.A.)
- Correspondence: (M.M.); (I.F.)
| |
Collapse
|
11
|
Barqawi AB, Rodrigues Pessoa R, Crawford ED, Al-Musawi M, MacDermott T, O'Donell C, Kendl RM. Boosting immune response with GM-CSF optimizes primary cryotherapy outcomes in the treatment of prostate cancer: a prospective randomized clinical trial. Prostate Cancer Prostatic Dis 2021; 24:750-757. [PMID: 33558662 DOI: 10.1038/s41391-021-00321-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/22/2020] [Accepted: 01/14/2021] [Indexed: 02/01/2023]
Abstract
OBJECTIVE We explored the association of prostate cryotherapy and immunomodulation with granulocyte-macrophage colony-stimulating factor (GMCSF) in the generation of detectable tumor-specific T- and B-cell responses in men with prostate cancer. MATERIALS AND METHODS A randomized pilot study of patients assigned to either cryotherapy alone (Control group) or in combination with GMCSF (Treatment group). The impact of therapy on the development of T- and B-cell responses against tumor-related antigens was studied using enzyme-linked immune absorbent spot (ELISpot) and protein microarray panels (Sematrix) assays, respectively. Fold changes in response to treatment were calculated by normalization of post-treatment ELISpot values against the mean pre-cryoablation response. Student t tests between treatment and control groups at 4 weeks and 12 weeks across all the antigens were performed. RESULTS A total of 20 patients were randomized to either control or treatment arm. At 4 weeks after cryotherapy, the treatment group demonstrated an average fold change in cancer antigen-related antibodies of 2.8% above their mean baseline values, whereas controls averaged an 18% change below mean baseline (p < 0.05). At 12 weeks, antibody response in treatment group increased to 25% above baseline, while the average of control group patients remained 9% below baseline (p < 0.05). Patients in treatment group displayed, on average, higher ELISPOT readings for the 4- and 12-week times points (527 vs 481 for PSA and 748 vs 562 for PAP). CONCLUSIONS GMCSF appeared to broadly elevate antibodies against prostate-specific and nonspecific antigens. Prostate antigen-specific T-cell responses were more enhanced over non-prostate-specific responses, preferentially in the treatment group. Our findings suggest a possible therapeutic effect of adjuvant immunotherapy in association with cryotherapy for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Al Baha Barqawi
- Division of Urology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Rodrigo Rodrigues Pessoa
- Division of Urology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - E David Crawford
- Division of Urology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mohammed Al-Musawi
- Clinical Research and Trials Unit, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Tracey MacDermott
- Clinical Research and Trials Unit, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Colin O'Donell
- Clinical Research and Trials Unit, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ross M Kendl
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
12
|
Gregg JR, Thompson TC. Considering the potential for gene-based therapy in prostate cancer. Nat Rev Urol 2021; 18:170-184. [PMID: 33637962 DOI: 10.1038/s41585-021-00431-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2021] [Indexed: 01/31/2023]
Abstract
Therapeutic gene manipulation has been at the forefront of popular scientific discussion and basic and clinical research for decades. Basic and clinical research applications of CRISPR-Cas9-based technologies and ongoing clinical trials in this area have demonstrated the potential of genome editing to cure human disease. Evaluation of research and clinical trials in gene therapy reveals a concentration of activity in prostate cancer research and practice. Multiple aspects of prostate cancer care - including anatomical considerations that enable direct tumour injections and sampling, the availability of preclinical immune-competent models and the delineation of tumour-related antigens that might provide targets for an induced immune system - make gene therapy an appealing treatment option for this common malignancy. Vaccine-based therapies that induce an immune response and new technologies exploiting CRISPR-Cas9-assisted approaches, including chimeric antigen receptor (CAR) T cell therapies, are very promising and are currently under investigation both in the laboratory and in the clinic. Although laboratory and preclinical advances have, thus far, not led to oncologically relevant outcomes in the clinic, future studies offer great promise for gene therapy to become established in prostate cancer care.
Collapse
Affiliation(s)
- Justin R Gregg
- Department of Urology and Health Disparities Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Timothy C Thompson
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
13
|
Johnson LE, Frye TP, McNeel DG. Immunization with a prostate cancer xenoantigen elicits a xenoantigen epitope-specific T-cell response. Oncoimmunology 2021; 1:1546-1556. [PMID: 23264901 PMCID: PMC3525610 DOI: 10.4161/onci.22564] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Vaccines encoding xenoantigens, “non-self” proteins that are highly homologous to their autologous counterparts, have been investigated as a means to increase immunogenicity and overcome tolerance to “self” antigens. We have previously shown that DNA vaccines encoding native prostatic acid phosphatase (PAP) were able to elicit PAP-specific T cells in both rats and humans, but required multiple immunization courses. In this study, we investigated in a preclinical model whether immunizations with a DNA vaccine encoding a xenoantigen could elicit a cross-reactive immune response to the native protein, potentially requiring fewer immunizations. Lewis rats were immunized with a DNA vaccine encoding human PAP and splenocytes from immunized rats were screened with a human peptide library containing overlapping, 15-mer PAP-derived peptides using T-cell proliferation and interferon γ (IFNγ) release as measures of the immune response. One dominant PAP-specific, RT1.Al-restricted, epitope was identified. Direct immunization with the immunodominant peptide (HP201–215) containing this epitope demonstrated that it included a naturally presented MHC Class I epitope recognized by CD8+ T cells in Lewis rats. However, no cross-reactive immune response was elicited to the corresponding rat peptide despite a difference of only three amino acids. Immunization with DNA vaccines encoding rat PAP (rPAP) in which this foreign dominant epitope was included as well as with DNA vaccines coding for a variant of the xenoantigen from which this epitope was deleted, did not elicit responses to the native antigen. Overall, these results indicate that the immunization with a xenoantigen-coding DNA vaccine can lead to an immune response which potentially favors foreign epitopes and hence limits any cross-reactive response to the native antigen.
Collapse
Affiliation(s)
- Laura E Johnson
- Department of Medicine; University of Wisconsin; Madison, WI USA
| | | | | |
Collapse
|
14
|
Droplet-based mRNA sequencing of fixed and permeabilized cells by CLInt-seq allows for antigen-specific TCR cloning. Proc Natl Acad Sci U S A 2021; 118:2021190118. [PMID: 33431692 PMCID: PMC7826406 DOI: 10.1073/pnas.2021190118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
T cell receptors (TCRs) surveil cellular environment by recognizing peptides presented by the major histocompatibility complex. TCR sequencing allows for understanding the scope of T cell reactivity in health and disease. Specific TCR clones can be used as therapeutics in cancer and autoimmune disease. We present a technique that allows for TCR sequencing based on intracellular signaling molecules, such as cytokines and transcription factors. The core concept is highly generalizable and should be applicable to global gene expression analysis where intracellular marker-based cell isolation is required. T cell receptors (TCRs) are generated by somatic recombination of V/D/J segments to produce up to 1015 unique sequences. Highly sensitive and specific techniques are required to isolate and identify the rare TCR sequences that respond to antigens of interest. Here, we describe the use of mRNA sequencing via cross-linker regulated intracellular phenotype (CLInt-Seq) for efficient recovery of antigen-specific TCRs in cells stained for combinations of intracellular proteins such as cytokines or transcription factors. This method enables high-throughput identification and isolation of low-frequency TCRs specific for any antigen. As a proof of principle, intracellular staining for TNFα and IFNγ identified cytomegalovirus (CMV)- and Epstein-Barr virus (EBV)-reactive TCRs with efficiencies similar to state-of-the-art peptide-MHC multimer methodology. In a separate experiment, regulatory T cells were profiled based on intracellular FOXP3 staining, demonstrating the ability to examine phenotypes based on transcription factors. We further optimized the intracellular staining conditions to use a chemically cleavable primary amine cross-linker compatible with current single-cell sequencing technology. CLInt-Seq for TNFα and IFNγ performed similarly to isolation with multimer staining for EBV-reactive TCRs. We anticipate CLInt-Seq will enable droplet-based single-cell mRNA analysis from any tissue where minor populations need to be isolated by intracellular markers.
Collapse
|
15
|
Handa S, Hans B, Goel S, Bashorun HO, Dovey Z, Tewari A. Immunotherapy in prostate cancer: current state and future perspectives. Ther Adv Urol 2020; 12:1756287220951404. [PMID: 32952615 PMCID: PMC7476347 DOI: 10.1177/1756287220951404] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/25/2020] [Indexed: 02/06/2023] Open
Abstract
Metastatic castrate resistant prostate cancer (PCa) remains an incurable entity. In the era of immunotherapy, the complex PCa microenvironment poses a unique challenge to the successful application of this class of agents. However, in the last decade, a tremendous effort has been made to explore this field of therapeutics. In this review, the physiology of the cancer immunity cycle is highlighted in the context of the prostate tumor microenvironment, and the current evidence for use of various classes of immunotherapy agents including vaccines (dendritic cell based, viral vector based and DNA/mRNA based), immune checkpoint inhibitors, Chimeric antigen receptor T cell therapy, antibody-mediated radioimmunotherapy, antibody drug conjugates, and bispecific antibodies, is consolidated. Finally, the future directions for combinatorial approaches to combat PCa are discussed.
Collapse
Affiliation(s)
- Shivani Handa
- Department of Internal Medicine, Icahn School of Medicine, Mount Sinai Morningside and West Hospital, New York, NY, 10019, USA
| | - Bandhul Hans
- Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, PA, USA
| | - Shokhi Goel
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Hafis O Bashorun
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Zach Dovey
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Ashutosh Tewari
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| |
Collapse
|
16
|
Cappuccini F, Bryant R, Pollock E, Carter L, Verrill C, Hollidge J, Poulton I, Baker M, Mitton C, Baines A, Meier A, Schmidt G, Harrop R, Protheroe A, MacPherson R, Kennish S, Morgan S, Vigano S, Romero PJ, Evans T, Catto J, Hamdy F, Hill AVS, Redchenko I. Safety and immunogenicity of novel 5T4 viral vectored vaccination regimens in early stage prostate cancer: a phase I clinical trial. J Immunother Cancer 2020; 8:e000928. [PMID: 32591433 PMCID: PMC7319775 DOI: 10.1136/jitc-2020-000928] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Prostate cancer (PCa) has been under investigation as a target for antigen-specific immunotherapies in metastatic disease settings for the last two decades leading to a licensure of the first therapeutic cancer vaccine, Sipuleucel-T, in 2010. However, neither Sipuleucel-T nor other experimental PCa vaccines that emerged later induce strong T-cell immunity. METHODS In this first-in-man study, VANCE, we evaluated a novel vaccination platform based on two replication-deficient viruses, chimpanzee adenovirus (ChAd) and MVA (Modified Vaccinia Ankara), targeting the oncofetal self-antigen 5T4 in early stage PCa. Forty patients, either newly diagnosed with early-stage PCa and scheduled for radical prostatectomy or patients with stable disease on an active surveillance protocol, were recruited to the study to assess the vaccine safety and T-cell immunogenicity. Secondary and exploratory endpoints included immune infiltration into the prostate, prostate-specific antigen (PSA) change, and assessment of phenotype and functionality of antigen-specific T cells. RESULTS The vaccine had an excellent safety profile. Vaccination-induced 5T4-specific T-cell responses were measured in blood by ex vivo IFN-γ ELISpot and were detected in the majority of patients with a mean level in responders of 198 spot-forming cells per million peripheral blood mononuclear cells. Flow cytometry analysis demonstrated the presence of both CD8+ and CD4+ polyfunctional 5T4-specific T cells in the circulation. 5T4-reactive tumor-infiltrating lymphocytes were isolated from post-treatment prostate tissue. Some of the patients had a transient PSA rise 2-8 weeks following vaccination, possibly indicating an inflammatory response in the target organ. CONCLUSIONS An excellent safety profile and T-cell responses elicited in the circulation and also detected in the prostate gland support the evaluation of the ChAdOx1-MVA 5T4 vaccine in efficacy trials. It remains to be seen if this vaccination strategy generates immune responses of sufficient magnitude to mediate clinical efficacy and whether it can be effective in late-stage PCa settings, as a monotherapy in advanced disease or as part of multi-modality PCa therapy. To address these questions, the phase I/II trial, ADVANCE, is currently recruiting patients with intermediate-risk PCa, and patients with advanced metastatic castration-resistant PCa, to receive this vaccine in combination with nivolumab. TRIAL REGISTRATION The trial was registered with the U.S. National Institutes of Health (NIH) Clinical Trials Registry (ClinicalTrials.gov identifier NCT02390063).
Collapse
Affiliation(s)
- Federica Cappuccini
- Nuffield Department of Medicine, The Jenner Institute, Oxford University, Oxford, UK
| | - Richard Bryant
- Nuffield Department of Surgical Sciences, Oxford University, Oxford, UK
- Department of Urology, Churchill Hospital, Oxford, UK
| | - Emily Pollock
- Nuffield Department of Medicine, The Jenner Institute, Oxford University, Oxford, UK
| | - Lucy Carter
- Nuffield Department of Medicine, The Jenner Institute, Oxford University, Oxford, UK
| | - Clare Verrill
- Nuffield Department of Surgical Sciences, Oxford University, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University, Oxford, UK
| | - Julianne Hollidge
- Nuffield Department of Surgical Sciences, Oxford University, Oxford, UK
| | - Ian Poulton
- Nuffield Department of Medicine, The Jenner Institute, Oxford University, Oxford, UK
| | - Megan Baker
- Nuffield Department of Medicine, The Jenner Institute, Oxford University, Oxford, UK
| | - Celia Mitton
- Nuffield Department of Medicine, The Jenner Institute, Oxford University, Oxford, UK
| | - Andrea Baines
- Nuffield Department of Medicine, The Jenner Institute, Oxford University, Oxford, UK
| | | | | | | | - Andrew Protheroe
- Department of Oncology, Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford, UK
| | | | - Steven Kennish
- Department of Radiology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Susan Morgan
- Department of Pathology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Selena Vigano
- Oncology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Pedro J Romero
- Oncology, University Hospital of Lausanne, Lausanne, Switzerland
| | | | - James Catto
- Academic Urology Unit, The University of Sheffield, Sheffield, UK
| | - Freddie Hamdy
- Nuffield Department of Surgical Sciences, Oxford University, Oxford, UK
- Department of Urology, Churchill Hospital, Oxford, UK
| | - Adrian V S Hill
- Nuffield Department of Medicine, The Jenner Institute, Oxford University, Oxford, UK
| | - Irina Redchenko
- Nuffield Department of Medicine, The Jenner Institute, Oxford University, Oxford, UK
| |
Collapse
|
17
|
Bouzid R, Peppelenbosch M, Buschow SI. Opportunities for Conventional and in Situ Cancer Vaccine Strategies and Combination with Immunotherapy for Gastrointestinal Cancers, A Review. Cancers (Basel) 2020; 12:cancers12051121. [PMID: 32365838 PMCID: PMC7281593 DOI: 10.3390/cancers12051121] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/28/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022] Open
Abstract
Survival of gastrointestinal cancer remains dismal, especially for metastasized disease. For various cancers, especially melanoma and lung cancer, immunotherapy has been proven to confer survival benefits, but results for gastrointestinal cancer have been disappointing. Hence, there is substantial interest in exploring the usefulness of adaptive immune system education with respect to anti-cancer responses though vaccination. Encouragingly, even fairly non-specific approaches to vaccination and immune system stimulation, involving for instance influenza vaccines, have shown promising results, eliciting hopes that selection of specific antigens for vaccination may prove useful for at least a subset of gastrointestinal cancers. It is widely recognized that immune recognition and initiation of responses are hampered by a lack of T cell help, or by suppressive cancer-associated factors. In this review we will discuss the hurdles that limit efficacy of conventional cancer therapeutic vaccination methods (e.g., peptide vaccines, dendritic cell vaccination). In addition, we will outline other forms of treatment (e.g., radiotherapy, chemotherapy, oncolytic viruses) that also cause the release of antigens through immunogenic tumor cell death and can thus be considered unconventional vaccination methods (i.e., in situ vaccination). Finally, we focus on the potential additive value that vaccination strategies may have for improving the effect immunotherapy. Overall, a picture will emerge that although the field has made substantial progress, successful immunotherapy through the combination with cancer antigen vaccination, including that for gastrointestinal cancers, is still in its infancy, prompting further intensification of the research effort in this respect.
Collapse
|
18
|
Ihle CL, Owens P. Integrating the immune microenvironment of prostate cancer induced bone disease. Mol Carcinog 2020; 59:822-829. [PMID: 32233011 DOI: 10.1002/mc.23192] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/03/2020] [Accepted: 03/17/2020] [Indexed: 12/11/2022]
Abstract
Prostate cancer (PCa) is the most frequently diagnosed cancer for men in the U.S. but does not impede patient survival until the disease is metastatic. Metastatic lesions most frequently occur in the bone, which exhibits a distinct microenvironment of immune and bone cell populations. Advances in the diagnosis and treatment of primary PCa allow for the use of tailored therapeutic approaches based on biomarkers, protein expression, and histopathology. Understanding the molecular and cellular characteristics of primary tumors has advanced therapeutic development and survival for patients with PCa. Personalized medicine has only recently emerged for the treatment of metastatic bone lesions. Tumor induced bone disease (TIBD) in patients with PCa can be classified into lytic, blastic, or mixed pathologies, with most patients exhibiting the blastic phenotype. Progress has been made in treating TIBD, but metastatic PCa has yet to be cured. Immune checkpoint inhibitors have exhibited limited responses in immunosuppressive PCa tumors, but have yet to be assessed in metastatic sites which may be susceptible to an increased inflammatory response. Recent discoveries have uncovered distinct tumor microenvironments (TMEs) of blastic and lytic bone metastases from patients with PCa, identifying actionable targets for therapeutic applications, including immune checkpoint inhibitors and targeted therapeutics. Enrichment for macrophages and T cells in patient samples suggests metastatic sites may be reappraised as immunologically targetable, despite their immunologically "cold" primary tumors. The practice of performing bone biopsies will help identify unique cellular and protein targets in the bone TME that can guide therapy decisions.
Collapse
Affiliation(s)
- Claire L Ihle
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Philip Owens
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Department of Veterans Affairs, Research Service, Eastern Colorado Health Care System, Aurora, Colorado
| |
Collapse
|
19
|
McNeel DG, Eickhoff JC, Johnson LE, Roth AR, Perk TG, Fong L, Antonarakis ES, Wargowski E, Jeraj R, Liu G. Phase II Trial of a DNA Vaccine Encoding Prostatic Acid Phosphatase (pTVG-HP [MVI-816]) in Patients With Progressive, Nonmetastatic, Castration-Sensitive Prostate Cancer. J Clin Oncol 2019; 37:3507-3517. [PMID: 31644357 DOI: 10.1200/jco.19.01701] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
PURPOSE We previously reported the safety and immunologic effects of a DNA vaccine (pTVG-HP [MVI-816]) encoding prostatic acid phosphatase (PAP) in patients with recurrent, nonmetastatic prostate cancer. The current trial evaluated the effects of this vaccine on metastatic progression. PATIENTS AND METHODS Ninety-nine patients with castration-sensitive prostate cancer and prostate-specific antigen (PSA) doubling time (DT) of less than 12 months were randomly assigned to treatment with either pTVG-HP co-administered intradermally with 200 μg granulocyte-macrophage colony-stimulating factor (GM-CSF) adjuvant or 200 μg GM-CSF alone six times at 14-day intervals and then quarterly for 2 years. The primary end point was 2-year metastasis-free survival (MFS). Secondary and exploratory end points were median MFS, changes in PSA DT, immunologic effects, and changes in quantitative 18F-sodium fluoride (NaF) positron emission tomography/computed tomography (PET/CT) imaging. RESULTS Two-year MFS was not different between study arms (41.8% vaccine v 42.3%; P = .97). Changes in PSA DT and median MFS were not different between study arms (18.9 v 18.3 months; hazard ratio [HR], 1.6; P = .13). Preplanned subset analysis identified longer MFS in vaccine-treated patients with rapid (< 3 months) pretreatment PSA DT (12.0 v 6.1 months; n = 21; HR, 4.4; P = .03). PAP-specific T cells were detected in both cohorts, including multifunctional PAP-specific T-helper 1-biased T cells. Changes in total activity (total standardized uptake value) on 18F-NaF PET/CT from months 3 to 6 increased 50% in patients treated with GM-CSF alone and decreased 23% in patients treated with pTVG-HP (n = 31; P = .07). CONCLUSION pTVG-HP treatment did not demonstrate an overall increase in 2-year MFS in patients with castration-sensitive prostate cancer, with the possible exception of a subgroup with rapidly progressive disease. Prespecified 18F-NaF PET/CT imaging conducted in a subset of patients suggests that vaccination had detectable effects on micrometastatic bone disease. Additional trials using pTVG-HP in combination with PD-1 blockade are under way.
Collapse
Affiliation(s)
| | | | | | | | | | - Lawrence Fong
- University of California, San Francisco, San Francisco, CA
| | | | | | | | - Glenn Liu
- University of Wisconsin, Madison, WI
| |
Collapse
|
20
|
Boettcher AN, Usman A, Morgans A, VanderWeele DJ, Sosman J, Wu JD. Past, Current, and Future of Immunotherapies for Prostate Cancer. Front Oncol 2019; 9:884. [PMID: 31572678 PMCID: PMC6749031 DOI: 10.3389/fonc.2019.00884] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022] Open
Abstract
Prostate cancer (PCa) is the most common cancer in men, and the second leading cause of cancer related death in men in Western countries. The standard therapy for metastatic PCa is androgen suppression therapy (AST). Men undergoing AST eventually develop metastatic castration-resistant prostate cancer (mCRPC), of which there are limited treatment options available. Immunotherapy has presented substantial benefits for many types of cancer, but only a marginal benefit for mCRPC, at least in part, due to the immunosuppressive tumor microenvironment (TME). Current clinical trials are investigating monotherapies or combination therapies involving adoptive cellular therapy, viral, DNA vaccines, oncolytic viruses, and immune checkpoint inhibitors (ICI). Immunotherapies are also being combined with chemotherapy, radiation, and AST. Additionally, preclinical investigations show promise with the recent description of alternative ways to circumvent the immunosuppressive nature of the prostate tumor microenvironment, including harnessing the immune stimulatory NKG2D pathway, inhibiting myeloid derived suppressor cells, and utilizing immunomodulatory oncolytic viruses. Herein we provide an overview of recent preclinical and clinical developments in cancer immunotherapies and discuss the perspectives for future immunotherapies in PCa.
Collapse
Affiliation(s)
- Adeline N. Boettcher
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Ahmed Usman
- Department of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Alicia Morgans
- Department of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - David J. VanderWeele
- Department of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jeffrey Sosman
- Department of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jennifer D. Wu
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
21
|
Olson B, Patnaik A. Utilizing precision medicine to modulate the prostate tumor microenvironment and enhance immunotherapy. Urol Oncol 2019; 37:535-542. [PMID: 30503851 PMCID: PMC8725604 DOI: 10.1016/j.urolonc.2018.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/31/2018] [Accepted: 11/06/2018] [Indexed: 12/25/2022]
Abstract
The last two decades of cancer research have seen two major advancements in our ability to treat cancer: precision medicine and immunotherapy. While these approaches have shown striking anticancer efficacy in numerous malignancies, they have not shown similar success and applicability in advanced prostate cancer patients. The fields of precision medicine and immunotherapy have come to realize that targeted therapies are capable of not only inhibiting tumor cell growth, but also promoting antitumor immunity by modulating the tumor microenvironment. Here we examine how personalized medicine can be used to target the tumor immune microenvironment in prostate cancer, with the goal of enhancing clinical responses to immunotherapy.
Collapse
Affiliation(s)
- Brian Olson
- Department of Hematology and Medical Oncology and Department of Urology, Emory University, Atlanta, GA.
| | - Akash Patnaik
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL
| |
Collapse
|
22
|
Hossain MK, Nahar K, Donkor O, Apostolopoulos V. Immune-based therapies for metastatic prostate cancer: an update. Immunotherapy 2019; 10:283-298. [PMID: 29421982 DOI: 10.2217/imt-2017-0123] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Prostate cancer (PC) is a common malignancy among elderly males and is noncurable once it becomes metastatic. In recent years, a number of antigen-delivery systems have emerged as viable and promising immunotherapeutic agents against PC. The approval of sipuleucel-T by the US FDA for the treatment of males with asymptomatic or minimally symptomatic castrate resistant PC was a landmark in cancer immunotherapy, making this the first approved immunotherapeutic. A number of vaccines are under clinical investigation, each having its own set of advantages and disadvantages. Here, we discuss the basic technologies underlying these different delivery modes, we discuss the completed and current human clinical trials, as well as the use of vaccines in combination with immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Kamrun Nahar
- Vetafarm Pty Ltd, Wagga Wagga, NSW, 2650, Australia
| | - Osaana Donkor
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Australia
| | - Vasso Apostolopoulos
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Australia
| |
Collapse
|
23
|
Itraconazole as a Noncastrating Treatment for Biochemically Recurrent Prostate Cancer: A Phase 2 Study. Clin Genitourin Cancer 2019; 17:e92-e96. [DOI: 10.1016/j.clgc.2018.09.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/19/2018] [Accepted: 09/19/2018] [Indexed: 11/18/2022]
|
24
|
Scarpelli M, Zahm C, Perlman S, McNeel DG, Jeraj R, Liu G. FLT PET/CT imaging of metastatic prostate cancer patients treated with pTVG-HP DNA vaccine and pembrolizumab. J Immunother Cancer 2019; 7:23. [PMID: 30700328 PMCID: PMC6354338 DOI: 10.1186/s40425-019-0516-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 01/20/2019] [Indexed: 12/17/2022] Open
Abstract
Background Immunotherapy has demonstrated remarkable success in treating different cancers. Nonetheless, a large number of patients do not respond, many respond without immediate changes detectable with conventional imaging, and many have unusual immune-related adverse events that cannot be predicted in advance. In this exploratory study, we investigate how 3′-Deoxy-3’-18F-fluorothymidine (FLT) positron emission tomography (PET) measurements of tumor and immune cell proliferation might be utilized as biomarkers in immunotherapy. Methods Seventeen patients with metastatic castrate resistant prostate cancer were treated with combination pTVG-HP DNA vaccine and pembrolizumab. Patients underwent baseline and 12-week FLT PET/CT scans. FLT PET standardized uptake values (SUVs) were extracted from tumors, non-metastatic lymph nodes, spleen, bone marrow, pancreas, and thyroid to quantify cell proliferation in these tissues. Regional immune cell responses to pTVG-HP DNA vaccine were assessed by comparing FLT uptake changes in vaccine draining and non-draining lymph nodes. Cox proportional hazards regression was utilized to relate FLT uptake and other clinical markers (PSA and tumor size) to progression-free survival. Area under receiver operating characteristic (AUC) curves and concordance indices were used to assess the predictive capabilities of FLT uptake. Results Changes in FLT uptake in vaccine draining lymph nodes were significantly greater than changes in non-draining lymph nodes (P = 0.02), suggesting a regional immune response to vaccination. However, the changes in FLT uptake in lymph nodes were not significantly predictive of progression-free survival. Increases in tumor FLT uptake were significantly predictive of shorter progression-free survival (concordance index = 0.83, P < 0.01). Baseline FLT uptake in the thyroid was significantly predictive of whether or not a patient would subsequently experience a thyroid-related adverse event (AUC = 0.97, P < 0.01). Conclusions FLT PET uptake was significantly predictive of progression-free survival and the occurrence of adverse events relating to thyroid function. The results suggest FLT PET imaging has potential as a biomarker in immunotherapy, providing a marker of tumor and immune responses, and as a possible means of anticipating specific immune-related adverse events. Trial registration NCT02499835. Electronic supplementary material The online version of this article (10.1186/s40425-019-0516-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Matthew Scarpelli
- Department of Medical Physics, University of Wisconsin, 1111 Highland Ave, Madison, WI, 53792, USA
| | - Christopher Zahm
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave, Madison, WI, 53792, USA
| | - Scott Perlman
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave, Madison, WI, 53792, USA.,Department of Radiology, Section of Nuclear Medicine, University of Wisconsin, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Douglas G McNeel
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave, Madison, WI, 53792, USA.,Department of Medicine, Division of Hematology/Oncology, University of Wisconsin, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Robert Jeraj
- Department of Medical Physics, University of Wisconsin, 1111 Highland Ave, Madison, WI, 53792, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Ave, Madison, WI, 53792, USA.,Department of Radiology, Section of Nuclear Medicine, University of Wisconsin, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Glenn Liu
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave, Madison, WI, 53792, USA. .,Department of Medicine, Division of Hematology/Oncology, University of Wisconsin, 600 Highland Avenue, Madison, WI, 53792, USA.
| |
Collapse
|
25
|
Abstract
There have been a number of recent developments in the treatment of castration-resistant prostate cancer which seek to exploit the hormonal axis. Still, the castration-resistant prostate cancer remains a major challenge since this is the lethal and incurable phenotype which results in tens of thousands of deaths every year. There has been emerging interest in utilizing anticancer immunotherapy in prostate cancer, especially since the development of sipuleucel-T. Several other prostate cancer therapeutic vaccines including autologous and allogeneic vaccines, as well as viral vector-based vaccines, have demonstrated promising results in early trials. The checkpoint inhibitors which have shown some dramatic results in other cancers are now being studied in advanced prostate cancer setting. Studies are examining the therapeutic effects for both CTLA-4 inhibitors and PD-1/PD-L1 inhibitors. It appears that definitions and measurements of response used in cytotoxic therapies may not be valid in determining response to immunotherapy. Early reports suggest that combination therapies, either concurrent or sequential, may be needed to achieve the desired response against advanced prostate cancer.
Collapse
Affiliation(s)
- Adam Schatz
- Division of Urology, Albany Medical College, Albany, NY 12054, USA
| | - Badar M Mian
- Division of Urology, Albany Medical College, Albany, NY 12054, USA
| |
Collapse
|
26
|
Tian JY, Guo FJ, Zheng GY, Ahmad A. Prostate cancer: updates on current strategies for screening, diagnosis and clinical implications of treatment modalities. Carcinogenesis 2018; 39:307-317. [PMID: 29216344 DOI: 10.1093/carcin/bgx141] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/29/2017] [Indexed: 01/23/2023] Open
Abstract
Prostate cancer is the most common cancer in men by way of diagnosis and a leading cause of cancer-related deaths. Early detection and intervention remains key to its optimum clinical management. This review provides the most updated information on the recent methods of prostate cancer screening, imaging and treatment modalities. Wherever possible, clinical trial data has been supplemented to provide a comprehensive overview of current prostate cancer research and development. Considering the recent success of immunotherapy in prostate cancer, we discuss cell, DNA and viruses based, as well as combinatorial immunotherapeutic strategies in detail. Furthermore, the potential of nanotechnology is increasingly being realized, especially in prostate cancer research, and we provide an overview of nanotechnology-based strategies, with special emphasis on nanotheranostics and multifunctional nanoconstructs. Understanding these recent developments is critical to the design of future therapeutic strategies to counter prostate cancer.
Collapse
Affiliation(s)
- Jing-Yan Tian
- Department of Urology, Second Division of the First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Feng-Jun Guo
- Department of Gynaecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Guo-You Zheng
- Department of Urology, Second Division of the First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Aamir Ahmad
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| |
Collapse
|
27
|
Wang X, Fan S, Pan H, Chen W, Wang H. Cancer immunotherapy for metastasis: past, present and future. Brief Funct Genomics 2018; 18:140-146. [PMID: 29992233 DOI: 10.1093/bfgp/ely022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Xiaobo Wang
- Department of Orthopaedics, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, P.R. China
| | - Shaoyi Fan
- Department of Traditional Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Hehai Pan
- Department of Orthopaedics, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, P.R. China
| | - Wenli Chen
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, P.R. China
| | - Hua Wang
- Department of Orthopaedics, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, P.R. China
| |
Collapse
|
28
|
Pilla L, Ferrone S, Maccalli C. Methods for improving the immunogenicity and efficacy of cancer vaccines. Expert Opin Biol Ther 2018; 18:765-784. [PMID: 29874943 PMCID: PMC8670419 DOI: 10.1080/14712598.2018.1485649] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/04/2018] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Cancer vaccines represent one of the oldest immunotherapy strategies. A variety of tumor-associated antigens have been exploited to investigate their immunogenicity as well as multiple strategies for vaccine administration. These efforts have led to the development of several clinical trials in tumors with different histological origins to test the clinical efficacy of cancer vaccines. However, suboptimal clinical results have been reported mainly due to the lack of optimized strategies to induce strong and sustained systemic tumor antigen-specific immune responses. AREAS COVERED We provide an overview of different types of cancer vaccines that have been developed and used in the context of clinical studies. Moreover, we review different preclinical and clinical strategies pursued to enhance the immunogenicity, stability, and targeting at tumor site of cancer vaccines. EXPERT OPINION Additional and appropriate preclinical studies are warranted to optimize the immunogenicity and delivery of cancer vaccines. The appropriate choice of target antigens is challenging; however, the exploitation of neoantigens generated from somatic mutations of tumor cells represents a promising approach to target highly immunogenic tumor-specific antigens. Remarkably, the investigation of the combination of cancer vaccines with immunomodulating agents able to skew the tumor microenvironment from immunosuppressive to immunostimulating will dramatically improve their clinical efficacy.
Collapse
Affiliation(s)
- Lorenzo Pilla
- Medical Oncology Unit, San Gerardo Hospital, Monza, Italy
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina Maccalli
- Clinical Research Center, Division of Translational Medicine, Sidra Medicine, Doha, Qatar
| |
Collapse
|
29
|
Wang K, Ruan H, Xu T, Liu L, Liu D, Yang H, Zhang X, Chen K. Recent advances on the progressive mechanism and therapy in castration-resistant prostate cancer. Onco Targets Ther 2018; 11:3167-3178. [PMID: 29881290 PMCID: PMC5983013 DOI: 10.2147/ott.s159777] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Background Although there have been great advances in mechanisms and therapeutic methods of prostate cancer, the mortality rate of prostate cancer remains high. The castration-resistant prostate cancer (CRPC), which develops from hormone-sensitive prostate cancer, foreshadows a more dismal outcome. Concomitant with the researches in the mechanism of CRPC and therapy for CRPC, more and more landmark progress has been made in recent years. Methods A number of clinical and experimental studies were reviewed to indicate the novel advancement in the progressive mechanism and therapy of CRPC. Results The androgen receptor (AR) is still a vital driver in the progression of CRPC, while other multiple mechanisms also contribute to this progression, such as tumor immunity, cancer stem cells, epithelial–mesenchymal transition and DNA repair disorder. In terms of the therapeutic methods of CRPC, chemotherapy with drugs, such as docetaxel, has been the first-line therapy for CRPC for many years. Besides, newer agents, which target some of the above mechanisms, show additional overall survival benefits for CRPC patients. These therapies include drugs targeting the androgen axis pathway (androgen synthesis, androgen receptor splice variants, coactivators of AR and so on), PI3K-AKT pathway, WNT pathway, DNA repair, rearrangement of ETS gene, novel chemotherapy and immunotherapy, bone metastasis therapy and so on. Understanding these novel findings on the mechanisms of CRPC and the latest potential CRPC therapies will direct us for further exploration of CRPC. Conclusion Through comprehensive consideration, the predominant mechanism of CRPC might be the AR signal axis concomitant with tumor microenvironment, stress, immunity, tumor microenvironment and so on. For CRPC therapy, targeting the AR axis pathway and chemotherapy are the first-line treatments at present. However, with the advancements in CRPC therapy made by the researchers, other novel potential methods will occupy more and more important position in the treatment of CRPC, especially the therapies targeting the tumor microenviroment, tumor immunity and DNA repair and so on.
Collapse
Affiliation(s)
- Keshan Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hailong Ruan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tianbo Xu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Di Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ke Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
30
|
Concurrent, but not sequential, PD-1 blockade with a DNA vaccine elicits anti-tumor responses in patients with metastatic, castration-resistant prostate cancer. Oncotarget 2018; 9:25586-25596. [PMID: 29876010 PMCID: PMC5986652 DOI: 10.18632/oncotarget.25387] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/26/2018] [Indexed: 01/03/2023] Open
Abstract
T-cell checkpoint inhibitors have demonstrated dramatic clinical activity against multiple cancer types, however little activity in patients with prostate cancer. Conversely, an anti-tumor vaccine was approved for the treatment of prostate cancer, having demonstrated an improvement in overall survival, despite few objective disease responses. In murine studies, we found that PD-1 expression on CD8+ T cells increased following anti-tumor vaccination, and that PD-1/PD-L1 blockade at the time of immunization elicited greater anti-tumor responses. Based on these data we initiated a pilot trial evaluating the immunological and clinical efficacy of a DNA encoding prostatic acid phosphatase (PAP) when delivered in combination with pembrolizumab. 26 patients were treated for 12 weeks with vaccine and received pembrolizumab either during this time or during the subsequent 12 weeks. Adverse events included grade 2 and 3 fatigue, diarrhea, thyroid dysfunction, and hepatitis. Median time to radiographic progression was not different between study arms. 8/13 (62%) of patients treated concurrently, and 1/12 (8%, p=0.01) of patients treated sequentially, experienced PSA declines from baseline. Of these, two were over 50% and one was a complete PSA response. No confirmed CR or PR were observed, however 4/5 patients treated concurrently had measurable decreases in tumor volume at 12 weeks. PSA declines were associated with the development of PAP-specific Th1-biased T cell immunity and CD8+ T cell infiltration in metastatic tumor biopsy specimens. These data are the first report of a clinical trial demonstrating that the efficacy of an anti-tumor vaccine can be augmented by concurrent PD-1 blockade.
Collapse
|
31
|
Johnson LE, Brockstedt D, Leong M, Lauer P, Theisen E, Sauer JD, McNeel DG. Heterologous vaccination targeting prostatic acid phosphatase (PAP) using DNA and Listeria vaccines elicits superior anti-tumor immunity dependent on CD4+ T cells elicited by DNA priming. Oncoimmunology 2018; 7:e1456603. [PMID: 30221049 DOI: 10.1080/2162402x.2018.1456603] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/14/2018] [Accepted: 03/15/2018] [Indexed: 12/18/2022] Open
Abstract
Background. Sipuleucel T, an autologous cell-based vaccine targeting prostatic acid phosphatase (PAP), has demonstrated efficacy for the treatment of advanced prostate cancer. DNA vaccines encoding PAP and live attenuated Listeria vaccines have entered clinical trials for patients with prostate cancer, and have advantages in terms of eliciting predominantly Th1-biased immunity. In this study, we investigated whether the immunogenicity and anti-tumor efficacy of a DNA and Listeria vaccine, each encoding PAP, could be enhanced by using them in a heterologous prime/boost approach. Methods. Transgenic mice expressing HLA-A2.01 and HLA-DRB1*0101 were immunized alone or with a heterologous prime/boost strategy. Splenocytes were evaluated for MHC class I and II-restricted, PAP-specific immune responses by IFNγ ELISPOTs. Anti-tumor activity to a syngeneic, PAP-expressing tumor line was evaluated. Results. PAP-specific cellular immunity and anti-tumor activity were elicited in mice after immunization with DNA- or listeria-based vaccines. Greater CD4+ and CD8+ responses, and anti-tumor responses, were elicited when mice were immunized first with DNA and boosted with Listeria, but not when administered in the opposite order. This was found to be dependent on CD4+ T cells elicited with DNA priming, and was not due to inflammatory signals by Listeria itself or due to B cells serving as antigen-presenting cells for DNA during priming. Conclusions. Heterologous prime/boost vaccination using DNA priming with Listeria boosting may provide better anti-tumor immunity, similar to many reports evaluating DNA priming with vaccines targeting foreign microbial antigens. These findings have implications for the design of future clinical trials.
Collapse
Affiliation(s)
- Laura E Johnson
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI
| | | | | | | | - Erin Theisen
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI
| | - Douglas G McNeel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI
| |
Collapse
|
32
|
Wargowski E, Johnson LE, Eickhoff JC, Delmastro L, Staab MJ, Liu G, McNeel DG. Prime-boost vaccination targeting prostatic acid phosphatase (PAP) in patients with metastatic castration-resistant prostate cancer (mCRPC) using Sipuleucel-T and a DNA vaccine. J Immunother Cancer 2018. [PMID: 29534736 PMCID: PMC5850960 DOI: 10.1186/s40425-018-0333-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Prostatic acid phosphatase (PAP) is a prostate tumor antigen, and the target of the only FDA-approved anti-tumor vaccine, sipuleucel-T. We have previously reported in two clinical trials that a DNA vaccine encoding PAP (pTVG-HP) could elicit PAP-specific, Th1-biased T cells in patients with PSA-recurrent prostate cancer. In the current pilot trial we sought to evaluate whether this vaccine could augment PAP-specific immunity when used as a booster to immunization with sipuleucel-T in patients with metastatic, castration-resistant prostate cancer (mCRPC). Methods Eigthteen patients with mCRPC were randomized to receive sipuleucel-T alone or followed by intradermal immunization with pTVG-HP DNA vaccine. Patients were followed for time to progression, and immune monitoring was conducted at defined intervals. Results Overall, patients were followed for a median of 24 months. 11/18 patients completed treatments as per protocol. No treatment-associated events > grade 2 were observed. Th1-biased PAP-specific T-cell responses were detected in 11/18 individuals, and were not statistically different between study arms. Higher titer antibody responses to PAP were detectable in patients who received pTVG-HP booster immunizations. Median time to progression was less than 6 months and not statistically different between study arms. The median overall survival for all patients was 28 months. Conclusions These findings suggest that prime-boost vaccination can augment and diversify the type of immunity elicited with anti-tumor vaccination in terms of T-cell and humoral immunity. Future studies will explore DNA as priming immunization rather than a booster immunization. Trial registration NCT01706458.
Collapse
Affiliation(s)
- Ellen Wargowski
- University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Laura E Johnson
- University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Jens C Eickhoff
- University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, Madison, WI, 53705, USA.,Department of Biostatistics, University of Wisconsin, Madison, WI, 53792, USA
| | - Lauren Delmastro
- University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Mary Jane Staab
- University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Glenn Liu
- University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Douglas G McNeel
- University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
33
|
Affiliation(s)
- John C. Henegan
- Division of Hematology and Oncology, Department of Medicine, University of Mississippi Cancer Center, Jackson, MS, USA
| | - Guru Sonpavde
- Department of Medical Oncology, GU section, Dana Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
34
|
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
|
35
|
Yan WL, Shen KY, Tien CY, Chen YA, Liu SJ. Recent progress in GM-CSF-based cancer immunotherapy. Immunotherapy 2017; 9:347-360. [PMID: 28303764 DOI: 10.2217/imt-2016-0141] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cancer immunotherapy is a growing field. GM-CSF, a potent cytokine promoting the differentiation of myeloid cells, can also be used as an immunostimulatory adjuvant to elicit antitumor immunity. Additionally, GM-CSF is essential for the differentiation of dendritic cells, which are responsible for processing and presenting tumor antigens for the priming of antitumor cytotoxic T lymphocytes. Some strategies have been developed for GM-CSF-based cancer immunotherapy in clinical practice: GM-CSF monotherapy, GM-CSF-secreting cancer cell vaccines, GM-CSF-fused tumor-associated antigen protein-based vaccines, GM-CSF-based DNA vaccines and GM-CSF combination therapy. GM-CSF also contributes to the regulation of immunosuppression in the tumor microenvironment. This review provides recommendations regarding GM-CSF-based cancer immunotherapy.
Collapse
Affiliation(s)
- Wan-Lun Yan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,National Institute of Infectious Diseases & Vaccinology, National Health Research Institutes, No. 35 Keyan Road, Zhunan Town, Miaoli County 350, Taiwan
| | - Kuan-Yin Shen
- National Institute of Infectious Diseases & Vaccinology, National Health Research Institutes, No. 35 Keyan Road, Zhunan Town, Miaoli County 350, Taiwan.,Graduate Instituteof Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Chun-Yuan Tien
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,National Institute of Infectious Diseases & Vaccinology, National Health Research Institutes, No. 35 Keyan Road, Zhunan Town, Miaoli County 350, Taiwan
| | - Yu-An Chen
- Graduate Instituteof Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Shih-Jen Liu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,National Institute of Infectious Diseases & Vaccinology, National Health Research Institutes, No. 35 Keyan Road, Zhunan Town, Miaoli County 350, Taiwan
| |
Collapse
|
36
|
Kates M, Drake CG. Immunotherapy for Prostate Cancer-Why Now? UROLOGY PRACTICE 2017; 4:329-334. [PMID: 37592679 DOI: 10.1016/j.urpr.2016.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Immunotherapy is transforming cancer care with the potential for an important role in prostate cancer. METHODS In this review we discuss the rationale for immunotherapy for prostate cancer and the treatment strategies currently under way. RESULTS Cell and viral based vaccines have shown some promise, as has immune checkpoint inhibition using CTLA-4 blocking agents. CONCLUSIONS The future will likely involve combination immunotherapy to maximize treatment effects and improve survival for patients with prostate cancer.
Collapse
Affiliation(s)
- Max Kates
- James Buchanan Brady Urological Institute, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Charles G Drake
- Herbert Irving Comprehensive Cancer Center, NewYork-Presbyterian/Columbia University Medical Center, New York, New York
| |
Collapse
|
37
|
Maia MC, Hansen AR. A comprehensive review of immunotherapies in prostate cancer. Crit Rev Oncol Hematol 2017; 113:292-303. [PMID: 28427519 DOI: 10.1016/j.critrevonc.2017.02.026] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/28/2017] [Accepted: 02/28/2017] [Indexed: 01/21/2023] Open
Abstract
Prostate cancer is the second most common malignant neoplasm in men worldwide and the fifth cause of cancer-related death. Although multiple new agents have been approved for metastatic castration resistant prostate cancer over the last decade, it is still an incurable disease. New strategies to improve cancer control are needed and agents targeting the immune system have shown encouraging results in many tumor types. Despite being attractive for immunotherapies due to the expression of various tumor associated antigens, the microenvironment in prostate cancer is relatively immunosuppressive and may be responsible for the failures of various agents targeting the immune system in this disease. To date, sipuleucel-T is the only immunotherapy that has shown significant clinical efficacy in this setting, although the high cost and potential trial flaws have precluded its widespread incorporation into clinical practice. Issues with patient selection and trial design may have contributed to the multiple failures of immunotherapy in prostate cancer and provides an opportunity to tailor future studies to evaluate these agents more accurately. We have reviewed all the completed immune therapy trials in prostate cancer and highlight important considerations for the next generation of clinical trials.
Collapse
Affiliation(s)
- Manuel Caitano Maia
- Department of Medical Oncology, Instituto do Câncer do Estado de São Paulo (ICESP), Av. Dr Arnaldo, 251, Cerqueira César, CEP 01246-000, São Paulo, Brazil.
| | - Aaron R Hansen
- Department of Medical Oncology and Hematology, Princess Margaret Hospital, 610 University Ave, Toronto, ON, Canada; Department of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Cir#3172, Toronto, ON, Canada
| |
Collapse
|
38
|
Abstract
DNA vaccines offer many advantages over other anti-tumor vaccine approaches due to their simplicity, ease of manufacturing, and safety. Results from several clinical trials in patients with cancer have demonstrated that DNA vaccines are safe and can elicit immune responses. However, to date few DNA vaccines have progressed beyond phase I clinical trial evaluation. Studies into the mechanism of action of DNA vaccines in terms of antigen-presenting cell types able to directly present or cross-present DNA-encoded antigens, and the activation of innate immune responses due to DNA itself, have suggested opportunities to increase the immunogenicity of these vaccines. In addition, studies into the mechanisms of tumor resistance to anti-tumor vaccination have suggested combination approaches that can increase the anti-tumor effect of DNA vaccines. This review focuses on these mechanisms of action and mechanisms of resistance using DNA vaccines, and how this information is being used to improve the anti-tumor effect of DNA vaccines. These approaches are then specifically discussed in the context of human prostate cancer, a disease for which DNA vaccines have been and continue to be explored as treatments.
Collapse
Affiliation(s)
- Christopher D Zahm
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Viswa Teja Colluru
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Douglas G McNeel
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, United States.
| |
Collapse
|
39
|
Affiliation(s)
- Mingming Zhang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, 236 Baidi Road, Nankai District, Tianjin 300192, China
| | - Yanhang Hong
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, 236 Baidi Road, Nankai District, Tianjin 300192, China
| | - Wenjuan Chen
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, 236 Baidi Road, Nankai District, Tianjin 300192, China
| | - Chun Wang
- Department
of Biomedical Engineering, University of Minnesota, 7-105 Hasselmo
Hall, 312 Church Street S. E., Minneapolis, Minnesota 55455, United States
| |
Collapse
|
40
|
Tiptiri-Kourpeti A, Spyridopoulou K, Pappa A, Chlichlia K. DNA vaccines to attack cancer: Strategies for improving immunogenicity and efficacy. Pharmacol Ther 2016; 165:32-49. [DOI: 10.1016/j.pharmthera.2016.05.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
41
|
Lee SH, Danishmalik SN, Sin JI. DNA vaccines, electroporation and their applications in cancer treatment. Hum Vaccin Immunother 2016; 11:1889-900. [PMID: 25984993 DOI: 10.1080/21645515.2015.1035502] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Numerous animal studies and recent clinical studies have shown that electroporation-delivered DNA vaccines can elicit robust Ag-specific CTL responses and reduce disease severity. However, cancer antigens are generally poorly immunogenic, requiring special conditions for immune response induction. To date, many different approaches have been used to elicit Ag-specific CTL and anti-neoplastic responses to DNA vaccines against cancer. In vivo electroporation is one example, whereas others include DNA manipulation, xenogeneic antigen use, immune stimulatory molecule and immune response regulator application, DNA prime-boost immunization strategy use and different DNA delivery methods. These strategies likely increase the immunogenicity of cancer DNA vaccines, thereby contributing to cancer eradication. However, cancer cells are heterogeneous and might become CTL-resistant. Thus, understanding the CTL resistance mechanism(s) employed by cancer cells is critical to develop counter-measures for this immune escape. In this review, the use of electroporation as a DNA delivery method, the strategies used to enhance the immune responses, the cancer antigens that have been tested, and the escape mechanism(s) used by tumor cells are discussed, with a focus on the progress of clinical trials using cancer DNA vaccines.
Collapse
Key Words
- AFP, α-fetoprotein
- APCs, antigen presenting cells
- CEA, carcinoembryonic antigen
- CTLA-4, cytotoxic T lymphocyte-associated antigen-4
- DCs, dendritic cells
- DNA vaccine
- EP, electroporation
- GITR, glucocorticoid-induced tumor necrosis factor receptor family-related gene
- HPV, human papillomavirus
- HSP, heat shock protein
- HSV, herpes simplex virus
- ID, intradermal
- IM, intramuscular
- MAGE, melanoma-associated antigen
- MART, melanoma antigen recognized by T cells
- PAP, prostatic acid phosphatase
- PD, programmed death
- PRAME, preferentially expressed antigen in melanoma
- PSA, prostate-specific antigen
- PSMA, prostate-specific membrane antigen
- WT1, Wilm's tumor
- anti-tumor immunity
- cancer
- hTERT, human telomerase reverse transcriptase
- tumor immune evasion
Collapse
Affiliation(s)
- Si-Hyeong Lee
- a BK21 Plus Graduate Program; Department of Microbiology ; School of Medicine; Kangwon National University ; Chuncheon , Gangwon-do , Korea
| | | | | |
Collapse
|
42
|
Graham L, Schweizer MT. Targeting persistent androgen receptor signaling in castration-resistant prostate cancer. Med Oncol 2016; 33:44. [DOI: 10.1007/s12032-016-0759-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 03/24/2016] [Indexed: 12/19/2022]
|
43
|
Rekoske BT, Olson BM, McNeel DG. Antitumor vaccination of prostate cancer patients elicits PD-1/PD-L1 regulated antigen-specific immune responses. Oncoimmunology 2016; 5:e1165377. [PMID: 27471641 DOI: 10.1080/2162402x.2016.1165377] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/05/2016] [Accepted: 03/09/2016] [Indexed: 12/30/2022] Open
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.
Collapse
Affiliation(s)
- Brian T Rekoske
- Department of Medicine, University of Wisconsin-Madison , Madison, WI, USA
| | - Brian M Olson
- Carbone Cancer Center, University of Wisconsin-Madison , Madison, WI, USA
| | - Douglas G McNeel
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
44
|
Noguchi M, Koga N, Moriya F, Itoh K. Immunotherapy in prostate cancer: challenges and opportunities. Immunotherapy 2016; 8:69-77. [DOI: 10.2217/imt.15.101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Although treatment options for castration-resistant prostate cancer (CRPC) have increased over the last decade, there remains a need for strategies that can provide durable disease control and long-term benefit. Recently, immunotherapy has emerged as a viable and attractive strategy for the treatment of CRPC. To date, there are multiple strategies to target the immune system, and several approaches including therapeutic cancer vaccines and immune checkpoint inhibitors have been most successful in clinical trials. With regard to this, we report the results of the most recent clinical trials investigating immunotherapy in CRPC and discuss the future development of immunotherapy for CRPC, as well as the potential importance of biomarkers in the future progress of this field.
Collapse
Affiliation(s)
- Masanori Noguchi
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Noriko Koga
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
| | - Fukuko Moriya
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
45
|
Abstract
DNA vaccination has emerged as an attractive immunotherapeutic approach against cancer
due to its simplicity, stability, and safety. Results from numerous clinical trials have
demonstrated that DNA vaccines are well tolerated by patients and do not trigger major
adverse effects. DNA vaccines are also very cost effective and can be administered
repeatedly for long-term protection. Despite all the practical advantages, DNA vaccines
face challenges in inducing potent antigen specific cellular immune responses as a result
of immune tolerance against endogenous self-antigens in tumors. Strategies to enhance
immunogenicity of DNA vaccines against self-antigens have been investigated including
encoding of xenogeneic versions of antigens, fusion of antigens to molecules that activate
T cells or trigger associative recognition, priming with DNA vectors followed by boosting
with viral vector, and utilization of immunomodulatory molecules. This review will focus
on discussing strategies that circumvent immune tolerance and provide updates on findings
from recent clinical trials.
Collapse
Key Words
- APCs, antigen presenting cells
- CEA, carcinoembryonic antigen
- CIN, cervical intraepithelial neoplasia
- CT antigens, cancer-testis antigens
- CTLs, cytotoxic lymphocytes
- DNA vaccines
- DOM, fragment c domain
- EP, electroporation
- GITR, glucocorticoid-induced tumor necrosis factor receptor family-related genes
- HER2, Her2/neu
- HSP70, heat shock protein 70
- IFNs, interferons
- IRF, interferon regulatory factor
- Id, idiotype
- MHC, major histocompatibility complex
- Mam-A, Mammaglobin-A
- NHP, non-human primate
- PAP, Prostatic acid phosphatase
- PMED, particle mediated epidermal delivery
- PSMA, prostate-specific membrane antigen
- SCT, single-chain trimer
- STING, stimulator of interferon genes
- TAAs, tumor-associated antigens
- TBK1, Tank-binding kinase 1
- TLRs, Toll-like receptors
- TT, tetanus toxin
- Trp2, tyrosinase related protein 2
- cellular immune response
- hTERT, human telomerase reverse transcriptase
- humoral immune response
- immune tolerance
- phTERT, optimized full-length hTERT
- tumor antigens
- vaccine delivery
Collapse
Affiliation(s)
- Benjamin Yang
- a Department of Pathology ; Johns Hopkins University ; Baltimore , MD USA
| | | | | | | | | |
Collapse
|
46
|
Abstract
Metastatic prostate cancer is the second leading cause of death from cancer in the United States. It is the first prevalent cancer in which overall survival in advanced disease is modestly, but objectively, improved with outpatient delivered dendritic cell-based immunotherapy. More prostate cancer patients have enrolled through Facebook and trusted-site Internet searches in clinical trials for prostate cancer vaccine-based immunotherapy than in immunotherapy trials for lung, breast, colon, pancreas, ovarian, and bladder cancer combined in the past 7 years. Exceptional responses to anti-CTLA-4 treatment have been documented in clinics, and prostate cancer neoantigen characterization and T-cell clonotyping are in their research ascendancy. The prostate is an accessory organ; it is not required for fertility, erectile function, or urinary continence. The true evolutionary advantage of having a prostate for male mammalian physiology is a topic of speculation in seminar rooms and on bar stools, but it remains unknown. Hundreds of prostate lineage-unique proteins (PLUP) exist among the >37,000 normal human prostate lineage-unique open reading frames that can be targeted for immunologic ablation of PLUP(+) prostate cancer cells by prostate-specific autoimmunity. This bioengineered graft-versus-prostate disease is a powerful strategy that can eliminate deaths from prostate cancer. Immunologic tolerance to prostate cancer can be overcome at every clinical stage of presentation. This Cancer Immunology at the Crossroads article aims to present advances in the past two decades of basic, translational, and clinical research in prostate cancer, including bioengineering B-cell and T-cell responses, and ongoing prostate cancer immunotherapy trials.
Collapse
|
47
|
Cole G, McCaffrey J, Ali AA, McCarthy HO. DNA vaccination for prostate cancer: key concepts and considerations. Cancer Nanotechnol 2015; 6:2. [PMID: 26161151 PMCID: PMC4488504 DOI: 10.1186/s12645-015-0010-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 05/16/2015] [Indexed: 11/10/2022] Open
Abstract
While locally confined prostate cancer is associated with a low five year mortality rate, advanced or metastatic disease remains a major challenge for healthcare professionals to treat and is usually terminal. As such, there is a need for the development of new, efficacious therapies for prostate cancer. Immunotherapy represents a promising approach where the host's immune system is harnessed to mount an anti-tumour effect, and the licensing of the first prostate cancer specific immunotherapy in 2010 has opened the door for other immunotherapies to gain regulatory approval. Among these strategies DNA vaccines are an attractive option in terms of their ability to elicit a highly specific, potent and wide-sweeping immune response. Several DNA vaccines have been tested for prostate cancer and while they have demonstrated a good safety profile they have faced problems with low efficacy and immunogenicity compared to other immunotherapeutic approaches. This review focuses on the positive aspects of DNA vaccines for prostate cancer that have been assessed in preclinical and clinical trials thus far and examines the key considerations that must be employed to improve the efficacy and immunogenicity of these vaccines.
Collapse
Affiliation(s)
- Grace Cole
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL Northern Ireland UK
| | - Joanne McCaffrey
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL Northern Ireland UK
| | - Ahlam A Ali
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL Northern Ireland UK
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL Northern Ireland UK
| |
Collapse
|
48
|
Bloy N, Buqué A, Aranda F, Castoldi F, Eggermont A, Cremer I, Sautès-Fridman C, Fucikova J, Galon J, Spisek R, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Naked and vectored DNA-based anticancer vaccines. Oncoimmunology 2015; 4:e1026531. [PMID: 26155408 PMCID: PMC4485755 DOI: 10.1080/2162402x.2015.1026531] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 02/27/2015] [Indexed: 12/28/2022] Open
Abstract
One type of anticancer vaccine relies on the administration of DNA constructs encoding one or multiple tumor-associated antigens (TAAs). The ultimate objective of these preparations, which can be naked or vectored by non-pathogenic viruses, bacteria or yeast cells, is to drive the synthesis of TAAs in the context of an immunostimulatory milieu, resulting in the (re-)elicitation of a tumor-targeting immune response. In spite of encouraging preclinical results, the clinical efficacy of DNA-based vaccines employed as standalone immunotherapeutic interventions in cancer patients appears to be limited. Thus, efforts are currently being devoted to the development of combinatorial regimens that allow DNA-based anticancer vaccines to elicit clinically relevant immune responses. Here, we discuss recent advances in the preclinical and clinical development of this therapeutic paradigm.
Collapse
Key Words
- AFP, α-fetoprotein
- APC, antigen-presenting cell
- CDR, complementarity-determining region
- CEA, carcinoembryonic antigen
- CIN, cervical intraepithelial neoplasia
- CTLA4, cytotoxic T lymphocyte protein 4
- DAMP, damage-associated molecular pattern
- DC, dendritic cell
- FDA, Food and Drug Administration
- GM-CSF, granulocyte macrophage colony-stimulating factor
- GX-188E
- HCC, hepatocellular carcinoma
- HNSCC, head and neck squamous cell carcinoma
- HPV, human papillomavirus
- IL, interleukin
- OS, overall survival
- OVA, ovalbumin
- PAP, prostate acid phosphatase
- SCGB2A2, secretoglobin, family 2A, member 2
- SOX2, SRY (sex determining region Y)-box 2
- T, brachyury homolog
- TAA, tumor-associated antigen
- TLR, Toll-like receptor
- TRA, tumor rejection antigen
- Treg, regulatory T cell
- VGX-3100
- WT1, Wilms tumor 1
- adjuvants
- dendritic cell
- electroporation
- mucosal immunity
Collapse
Affiliation(s)
- Norma Bloy
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
| | - Aitziber Buqué
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
| | - Fernando Aranda
- Group of Immune receptors of the Innate and Adaptive System; Institut d’Investigacions Biomédiques August Pi i Sunyer (IDIBAPS); Barcelona, Spain
| | - Francesca Castoldi
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- Faculté de Medicine; Université Paris Sud/Paris XI; Le Kremlin-Bicêtre, France
- Sotio a.c; Prague, Czech Republic
| | | | - Isabelle Cremer
- INSERM, U1138; Paris, France
- Equipe 13; Center de Recherche des Cordeliers; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
| | - Catherine Sautès-Fridman
- INSERM, U1138; Paris, France
- Equipe 13; Center de Recherche des Cordeliers; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
| | - Jitka Fucikova
- Sotio a.c; Prague, Czech Republic
- Dept. of Immunology; 2 Faculty of Medicine and University Hospital Motol; Charles University; Prague, Czech Republic
| | - Jérôme Galon
- INSERM, U1138; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Laboratory of Integrative Cancer Immunology; Center de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| | - Radek Spisek
- Sotio a.c; Prague, Czech Republic
- Dept. of Immunology; 2 Faculty of Medicine and University Hospital Motol; Charles University; Prague, Czech Republic
| | - Eric Tartour
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- INSERM, U970; Paris, France
- Paris-Cardiovascular Research Center (PARCC); Paris, France
- Service d'Immunologie Biologique; Hôpital Européen Georges Pompidou (HEGP); AP-HP; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1015, CICBT507; Villejuif, France
| | - Guido Kroemer
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
49
|
Schweizer MT, Drake CG. Immunotherapy for prostate cancer: recent developments and future challenges. Cancer Metastasis Rev 2015; 33:641-55. [PMID: 24477411 DOI: 10.1007/s10555-013-9479-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Since the approval of sipuleucel-T for men with metastatic castrate resistant prostate cancer in 2010, great strides in the development of anti-cancer immunotherapies have been made. Current drug development in this area has focused primarily on antigen-specific (i.e. cancer vaccines and antibody based therapies) or checkpoint inhibitor therapies, with the checkpoint inhibitors perhaps gaining the most attention as of late. Indeed, drugs blocking the inhibitory signal generated by the engagement of cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death-1 (PD-1) found on T-cells has emerged as potent means to combat the immunosuppressive milieu. The anti-CTLA-4 monoclonal antibody ipilimumab has already been approved in advanced melanoma and two phase III trials evaluating ipilimumab in men with metastatic castrate-resistant prostate cancer are underway. A phase III trial evaluating ProstVac-VF, a poxvirus-based therapeutic prostate cancer vaccine, is also underway. While there has been reason for encouragement over the past few years, many questions regarding the use of immunotherapies remain. Namely, it is unclear what stage of disease is most likely to benefit from these approaches, how best to incorporate said treatments with each other and into our current treatment regimens and which therapy is most appropriate for which disease. Herein we review some of the recent advances in immunotherapy as related to the treatment of prostate cancer and outline some of the challenges that lie ahead.
Collapse
Affiliation(s)
- Michael T Schweizer
- Medical Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA,
| | | |
Collapse
|
50
|
Uhlman MA, Bing MT, Lubaroff DM. Prostate cancer vaccines in combination with additional treatment modalities. Immunol Res 2015; 59:236-42. [PMID: 24838261 DOI: 10.1007/s12026-014-8532-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Immunotherapy has been investigated in both preclinical studies and clinical trials as a new therapy for prostate cancer. Vaccines, including those that utilize dendritic cells, viruses, or DNA, immunize against prostate-specific antigen and prostatic acid phosphatase. The vaccines have long been studied as monotherapy for the cancer, but increasingly more trials have been initiated in combination with other modalities. These include radiation, chemotherapy, and androgen deprivation therapy. This review describes and discusses the various combinations of vaccine immunotherapies.
Collapse
Affiliation(s)
- Matthew A Uhlman
- Department of Urology, University of Iowa, 375 Newton Road, Iowa City, IA, 52242, USA
| | | | | |
Collapse
|