1
|
Bhojnagarwala PS, Jose J, Zhao S, Weiner DB. DNA-based immunotherapy for cancer: In vivo approaches for recalcitrant targets. Mol Ther 2025:S1525-0016(25)00282-5. [PMID: 40211538 DOI: 10.1016/j.ymthe.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 05/10/2025] Open
Abstract
Immunotherapy has revolutionized cancer treatment and complements traditional therapies, including surgery, chemotherapy, radiation therapy, and targeted therapies. Immunotherapy redirects the patient's immune system against tumors via several immune-mediated approaches. Over the past few years, therapeutic immunization, which enable the patient's T cells to better recognize and kill tumors, have been increasingly tested in the clinic, with several approaches demonstrating treatment improvements. There has been a renewed interest in cancer vaccines due to advances in tumor antigen identification, immune response optimization, novel adjuvants, next-generation vaccine delivery platforms, and antigen designs. The COVID-19 pandemic accelerated progress in nucleic acid-based vaccine manufacturing, which spurred broader interest in mRNA or plasmid platforms. Enhanced DNA vaccine designs, including optimized leader sequences and RNA and codon optimizations, improved formulations, and delivery via adaptive electroporation using stereotactic intramuscular/intradermal methods have improved T cell responses to plasmid-delivered tumor antigens. Additionally, advancements for direct in vivo delivery of DNA-encoded monospecific/bispecific antibodies offer novel tumor-targeting strategies. This review summarizes the recent clinical data for therapeutic cancer vaccines utilizing the DNA platform, including vaccines targeting common tumor-associated and viral antigens and neoantigen vaccines using nucleic acid technologies. We also summarize preclinical data using DNA-launched monoclonal/bispecific antibodies, underscoring their potential as a novel cancer therapy tool.
Collapse
Affiliation(s)
- Pratik S Bhojnagarwala
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA
| | - Joshua Jose
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA
| | - Shushu Zhao
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Zhang L, Ren X, An R, Song H, Tian Y, Wei X, Shi M, Wang Z. The Role of STEAP1 in Prostate Cancer: Implications for Diagnosis and Therapeutic Strategies. Biomedicines 2025; 13:794. [PMID: 40299363 PMCID: PMC12024770 DOI: 10.3390/biomedicines13040794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/21/2025] [Accepted: 03/23/2025] [Indexed: 04/30/2025] Open
Abstract
Prostate cancer (PCa) is one of the most common malignancies and the second leading cause of cancer-related death in men worldwide. The six-transmembrane epithelial antigen of the prostate 1 (STEAP1) is exceptionally overexpressed in PCa, maintaining high expression even in the castration-resistant prostate cancer (CRPC) stage, making it a promising target for diagnosis and treatment. STEAP1-positive extracellular vesicles and STEAP1-PET imaging are optimistic approaches for the non-invasive detection of different stages of PCa. STEAP1-targeted therapy includes an antibody-drug conjugate (ADC), chimeric antigen receptor T cell (CAR-T), T-cell engager (TCE), and vaccines, which demonstrate valuable therapeutic prospects. This review presents the structure and pathophysiological function of STEAP1, synthesizes cutting-edge advances in STEAP1-targeted molecular imaging and clinical applications, and critically analyzes their translational potential to overcome the limitations of current PCa diagnosis and treatment.
Collapse
Affiliation(s)
- Lingling Zhang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (L.Z.); (X.R.); (R.A.); (Y.T.); (X.W.)
| | - Xinyi Ren
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (L.Z.); (X.R.); (R.A.); (Y.T.); (X.W.)
| | - Ran An
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (L.Z.); (X.R.); (R.A.); (Y.T.); (X.W.)
| | - Hongchen Song
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China;
| | - Yaqi Tian
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (L.Z.); (X.R.); (R.A.); (Y.T.); (X.W.)
| | - Xuan Wei
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (L.Z.); (X.R.); (R.A.); (Y.T.); (X.W.)
| | - Mingjun Shi
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China;
- Institute of Urology, Beijing Municipal Health Commission, Beijing 100054, China
| | - Zhenchang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (L.Z.); (X.R.); (R.A.); (Y.T.); (X.W.)
| |
Collapse
|
3
|
Gu Q, Qi A, Wang N, Zhou Z, Zhou X. Unlocking Immunity: Innovative prostate cancer vaccine strategies. Int Immunopharmacol 2024; 142:113137. [PMID: 39276448 DOI: 10.1016/j.intimp.2024.113137] [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: 06/24/2024] [Revised: 09/02/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
OBJECTIVE Prostate Cancer (PCa) is a leading cause of cancer-related mortality in men, especially in Western societies. The objective of this research is to address the unmet need for effective treatments in advanced or recurrent PCa, where current strategies fall short of offering a cure. The focus is on leveraging immunotherapy and cancer vaccines to target the tumor's unique immunological microenvironment. MAIN RESULTS Despite immunotherapy's success in other cancers, its effectiveness in PCa has been limited by the tumor's immunosuppressive characteristics. However, cancer vaccines that engage Tumor-Specific Antigens (TSA) and Tumor-Associated Antigens (TAA) have emerged as a promising approach. Preclinical and clinical investigations of Dendritic Cell (DC) vaccines, DNA vaccines, mRNA vaccines, peptide vaccines, and viral vectors have shown their potential to elicit anti-tumor immune responses. The exploration of combination therapies with immune checkpoint inhibitors and the advent of novel adjuvants and oral microparticle vaccines present innovative strategies to improve efficacy and compliance. CONCLUSION The development of cancer vaccines for PCa holds significant potential. Future directions include optimizing vaccine design, refining combination therapy strategies, and creating patient-friendly administration methods. The integration of interdisciplinary knowledge and innovative clinical trial designs is essential for advancing personalized and precision immunotherapy for PCa.
Collapse
Affiliation(s)
- Qiannan Gu
- China Pharmaceutical University, School of Basic Medicine and Clinical Pharmacy, Nanjing, Jiangsu 210009, China
| | - Anning Qi
- Medical Laboratory, Liuhe People's Hospital of Jiangsu Province, Nanjing 211500, Jiangsu, China
| | - Ne Wang
- Jiangning Hospital Tiandi New City Branch, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211198, Jiangsu Province, China
| | - Zhenxian Zhou
- Nanjing Second People's Hospital, 211103, Jiangsu Province, China
| | - Xiaohui Zhou
- China Pharmaceutical University, School of Basic Medicine and Clinical Pharmacy, Nanjing, Jiangsu 210009, China; Jiangning Outpatient Department of China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
4
|
Novysedlak R, Guney M, Al Khouri M, Bartolini R, Koumbas Foley L, Benesova I, Ozaniak A, Novak V, Vesely S, Pacas P, Buchler T, Ozaniak Strizova Z. The Immune Microenvironment in Prostate Cancer: A Comprehensive Review. Oncology 2024:1-25. [PMID: 39380471 DOI: 10.1159/000541881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND Prostate cancer (PCa) is a malignancy with significant immunosuppressive properties and limited immune activation. This immunosuppression is linked to reduced cytotoxic T cell activity, impaired antigen presentation, and elevated levels of immunosuppressive cytokines and immune checkpoint molecules. Studies demonstrate that cytotoxic CD8+ T cell infiltration correlates with improved survival, while increased regulatory T cells (Tregs) and tumor-associated macrophages (TAMs) are associated with worse outcomes and therapeutic resistance. Th1 cells are beneficial, whereas Th17 cells, producing interleukin-17 (IL-17), contribute to tumor progression. Tumor-associated neutrophils (TANs) and immune checkpoint molecules, such as PD-1/PD-L1 and T cell immunoglobulin-3 (TIM-3) are also linked to advanced stages of PCa. Chemotherapy holds promise in converting the "cold" tumor microenvironment (TME) to a "hot" one by depleting immunosuppressive cells and enhancing tumor immunogenicity. SUMMARY This comprehensive review examines the immune microenvironment in PCa, focusing on the intricate interactions between immune and tumor cells in the TME. It highlights how TAMs, Tregs, cytotoxic T cells, and other immune cell types contribute to tumor progression or suppression and how PCa's low immunogenicity complicates immunotherapy. KEY MESSAGES The infiltration of cytotoxic CD8+ T cells and Th1 cells correlates with better outcomes, while elevated T regs and TAMs promote tumor growth, metastasis, and resistance. TANs and natural killer (NK) cells exhibit dual roles, with higher NK cell levels linked to better prognoses. Immune checkpoint molecules like PD-1, PD-L1, and TIM-3 are associated with advanced disease. Chemotherapy can improve tumor immunogenicity by depleting T regs and myeloid-derived suppressor cells, offering therapeutic promise.
Collapse
Affiliation(s)
- Rene Novysedlak
- Third Department of Surgery, 1st Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Miray Guney
- Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Majd Al Khouri
- Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Robin Bartolini
- Lausanne Center for Immuno-oncology Toxicities (LCIT), Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Lily Koumbas Foley
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Iva Benesova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Andrej Ozaniak
- Third Department of Surgery, 1st Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Vojtech Novak
- Department of Urology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Stepan Vesely
- Department of Urology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Pavel Pacas
- Department of Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Tomas Buchler
- Department of Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Zuzana Ozaniak Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| |
Collapse
|
5
|
He J, Wu J, Li Z, Zhao Z, Qiu L, Zhu X, Liu Z, Xia H, Hong P, Yang J, Ni L, Lu J. Immunotherapy Vaccines for Prostate Cancer Treatment. Cancer Med 2024; 13:e70294. [PMID: 39463159 PMCID: PMC11513549 DOI: 10.1002/cam4.70294] [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: 07/24/2024] [Revised: 09/09/2024] [Accepted: 09/21/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Therapeutic tumor vaccines have emerged as a compelling avenue for treating patients afflicted with advanced prostate cancer (PCa), particularly those experiencing biochemical relapse or ineligible for surgical intervention. This study serves to consolidate recent research findings on therapeutic vaccines targeting prostate tumors while delineating prevalent challenges within vaccine research and development. METHODS We searched electronic databases, including PubMed, Web of Science, Embase, and Scopus, up to August 31, 2024, using keywords such as 'vaccine', 'prostate cancer', 'immunotherapy', and others. We reviewed studies on various therapeutic vaccines, including dendritic cell-based, antigen, nucleic acid, and tumor cell vaccines. RESULTS Studies consistently showed that therapeutic vaccines, notably DC vaccines, had favorable safety profiles with few adverse effects. These vaccines, with varied antigenic formulations, demonstrated strong clinical outcomes, as indicated by metrics such as PSA response rates (9.5%-58%), extended PSA doubling times (52.9%-89.7%), overall survival durations (17.7-33.8 months), two-year mortality rates (0%-12.5%), biochemical relapse rates (42%-73%), and antigen-specific immune responses (33.3%-71.4% in responsive groups). CONCLUSION While clinical data for tumor vaccines have illuminated robust evidence of tumoricidal activity, the processes of their formulation and deployment are riddled with complexities. Combining vaccines with other therapies may enhance outcomes, and future research should focus on early interventions and deciphering the immune system's role in oncogenesis.
Collapse
Affiliation(s)
- Jide He
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Jialong Wu
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Ziang Li
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Zhenkun Zhao
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Lei Qiu
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Xuehua Zhu
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Zenan Liu
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Haizhui Xia
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Peng Hong
- Department of UrologyPeking University Third HospitalBeijingChina
| | - Jianling Yang
- Institute of Medical Innovation and ResearchPeking University Third HospitalBeijingChina
| | - Ling Ni
- Institute for Immunology and School of MedicineTsinghua University, Medical Research BuildingBeijingChina
| | - Jian Lu
- Department of UrologyPeking University Third HospitalBeijingChina
- State Key Laboratory of Natural and Biomimetic DrugsPeking University
| |
Collapse
|
6
|
Bordoloi D, Kulkarni AJ, Adeniji OS, Pampena MB, Bhojnagarwala PS, Zhao S, Ionescu C, Perales-Puchalt A, Parzych EM, Zhu X, Ali AR, Cassel J, Zhang R, Betts MR, Abdel-Mohsen M, Weiner DB. Siglec-7 glyco-immune binding mAbs or NK cell engager biologics induce potent antitumor immunity against ovarian cancers. SCIENCE ADVANCES 2023; 9:eadh4379. [PMID: 37910620 PMCID: PMC10619929 DOI: 10.1126/sciadv.adh4379] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/27/2023] [Indexed: 11/03/2023]
Abstract
Ovarian cancer (OC) is a lethal gynecologic malignancy, with modest responses to CPI. Engagement of additional immune arms, such as NK cells, may be of value. We focused on Siglec-7 as a surface antigen for engaging this population. Human antibodies against Siglec-7 were developed and characterized. Coculture of OC cells with PBMCs/NKs and Siglec-7 binding antibodies showed NK-mediated killing of OC lines. Anti-Siglec-7 mAb (DB7.2) enhanced survival in OC-challenged mice. In addition, the combination of DB7.2 and anti-PD-1 demonstrated further improved OC killing in vitro. To use Siglec-7 engagement as an OC-specific strategy, we engineered an NK cell engager (NKCE) to simultaneously engage NK cells through Siglec-7, and OC targets through FSHR. The NKCE demonstrated robust in vitro killing of FSHR+ OC, controlled tumors, and improved survival in OC-challenged mice. These studies support additional investigation of the Siglec-7 targeting approaches as important tools for OC and other recalcitrant cancers.
Collapse
Affiliation(s)
- Devivasha Bordoloi
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | | | - Opeyemi S. Adeniji
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - M. Betina Pampena
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Shushu Zhao
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Candice Ionescu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | | | | | - Xizhou Zhu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Ali R. Ali
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Joel Cassel
- Molecular Screening and Protein Expression facility, The Wistar Institute, Philadelphia, PA, USA
| | - Rugang Zhang
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Michael R. Betts
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - David B. Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| |
Collapse
|
7
|
Wang J, Zhou K, Zhu H, Wei F, Ma S, Kan Y, Li B, Mao L. Current status and progress of the development of prostate cancer vaccines. J Cancer 2023; 14:835-842. [PMID: 37056394 PMCID: PMC10088880 DOI: 10.7150/jca.80803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 03/08/2023] [Indexed: 04/15/2023] Open
Abstract
At present, common treatments of prostate cancer mainly include surgery, radiotherapy, chemotherapy and hormone therapy. However, patients have high recurrence rate after treatment, and are prone to castration-resistant prostate cancer. Tumor vaccine is based on tumor specific antigen (TSA) and tumor associated antigen (TAA) to activate specific immune response of the body to cancer cells. With continuous maturity of tumor vaccine technology, different forms of prostate cancer vaccines have been developed, such as cellular vaccines, extracellular-based anti-tumor vaccines, polypeptide vaccines, and nucleic acid vaccines. In this review, we summarize current status and progress in the development of prostate cancer vaccines.
Collapse
Affiliation(s)
- Jie Wang
- Department of Urology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Kaichen Zhou
- Department of Urology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Huihuang Zhu
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical University, Xuzhou 221002, China
| | - Fukun Wei
- Department of Urology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Sai Ma
- Department of Urology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Yi Kan
- Department of Urology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Bingheng Li
- Department of Urology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Lijun Mao
- Department of Urology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical University, Xuzhou 221002, China
| |
Collapse
|
8
|
Bordoloi D, Bhojnagarwala PS, Perales-Puchalt A, Kulkarni AJ, Zhu X, Liaw K, O’Connell RP, Park DH, Kulp DW, Zhang R, Weiner DB. A mAb against surface-expressed FSHR engineered to engage adaptive immunity for ovarian cancer immunotherapy. JCI Insight 2022; 7:e162553. [PMID: 36509287 PMCID: PMC9746812 DOI: 10.1172/jci.insight.162553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/20/2022] [Indexed: 11/22/2022] Open
Abstract
Despite advances in ovarian cancer (OC) therapy, recurrent OC remains a poor-prognosis disease. Because of the close interaction between OC cells and the tumor microenvironment (TME), it is important to develop strategies that target tumor cells and engage components of the TME. A major obstacle in the development of OC therapies is the identification of targets with expression limited to tumor surface to avoid off-target interactions. The follicle-stimulating hormone receptor (FSHR) has selective expression on ovarian granulosa cells and is expressed on 50%-70% of serous OCs. We generated mAbs targeting the external domain of FSHR using in vivo-expressed FSHR vector. By high-throughput flow analysis, we identified multiple clones and downselected D2AP11, a potent FSHR surface-targeted mAb. D2AP11 identifies important OC cell lines derived from tumors with different mutations, including BRCA1/2, and lines resistant to a wide range of therapies. We used D2AP11 to develop a bispecific T cell engager. In vitro addition of PBMCs and T cells to D2AP11-TCE induced specific and potent killing of different genetic and immune escape OC lines, with EC50s in the ng/ml range, and attenuated tumor burden in OC-challenged mouse models. These studies demonstrate the potential utility of biologics targeting FSHR for OC and perhaps other FSHR-positive cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Rugang Zhang
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
9
|
STEAP1-4 (Six-Transmembrane Epithelial Antigen of the Prostate 1-4) and Their Clinical Implications for Prostate Cancer. Cancers (Basel) 2022; 14:cancers14164034. [PMID: 36011027 PMCID: PMC9406800 DOI: 10.3390/cancers14164034] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Despite recent therapeutic advances in the treatment of prostate cancer, metastatic castration-resistant prostate cancer continues to cause significant morbidity and mortality. New research into highly expressed proteins in metastatic castration-resistant prostate cancer shows that Six-Transmembrane Epithelial Antigen of the Prostate 1–4 (STEAP1–4) are significant drivers of prostate cancer aggressiveness and metastasis. STEAP1, in particular, is highly expressed on the plasma membrane of prostate cancer cells and has received significant attention as a potential therapeutic target. This review highlights what is known about STEAP1–4 and identifies knowledge gaps that require further research. Abstract Six-Transmembrane Epithelial Antigen of the Prostate 1–4 (STEAP1–4) compose a family of metalloproteinases involved in iron and copper homeostasis and other cellular processes. Thus far, five homologs are known: STEAP1, STEAP1B, STEAP2, STEAP3, and STEAP4. In prostate cancer, STEAP1, STEAP2, and STEAP4 are overexpressed, while STEAP3 expression is downregulated. Although the metalloreductase activities of STEAP1–4 are well documented, their other biological functions are not. Furthermore, the properties and expression levels of STEAP heterotrimers, homotrimers, heterodimers, and homodimers are not well understood. Nevertheless, studies over the last few decades have provided sufficient impetus to investigate STEAP1–4 as potential biomarkers and therapeutic targets for prostate cancer. In particular, STEAP1 is the target of many emerging immunotherapies. Herein, we give an overview of the structure, physiology, and pathophysiology of STEAP1–4 to provide context for past and current efforts to translate STEAP1–4 into the clinic.
Collapse
|
10
|
Tretyakova I, Tomai M, Vasilakos J, Pushko P. Live-Attenuated VEEV Vaccine Delivered by iDNA Using Microneedles Is Immunogenic in Rabbits. FRONTIERS IN TROPICAL DISEASES 2022; 3:813671. [PMID: 37854093 PMCID: PMC10583749 DOI: 10.3389/fitd.2022.813671] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023] Open
Abstract
Effective and simple delivery of DNA vaccines remains a key to successful clinical applications. Previously, we developed a novel class of DNA vaccines, sometimes called iDNA, which encodes the whole live-attenuated vaccine viruses. Compared to a standard DNA vaccine, an iDNA vaccine required a low dose to launch a live-attenuated vaccine in vitro or in vivo. The goal of this pilot study was to investigate if iDNA vaccine encoding live-attenuated Venezuelan equine encephalitis virus (VEEV) can be efficiently delivered in vivo by a microneedle device using a single-dose vaccination with naked iDNA plasmid. For this purpose, we used pMG4020 plasmid encoding live-attenuated V4020 vaccine of VEE virus. The V4020 virus contains structural gene rearrangement, as well as attenuating mutations genetically engineered to prevent reversion mutations. The pMG4020 was administered to experimental rabbits by using a hollow microstructured transdermal system (hMTS) microneedle device. No adverse events to vaccination were noted. Animals that received pMG4020 plasmid have successfully seroconverted, with high plaque reduction neutralization test (PRNT) antibody titers, similar to those observed in animals that received V4020 virus in place of the pMG4020 iDNA plasmid. We conclude that naked iDNA vaccine can be successfully delivered in vivo by using a single-dose vaccination with a microneedle device.
Collapse
|