1
|
Lunj S, Smith TAD, Reeves KJ, Currell F, Honeychurch J, Hoskin P, Choudhury A. Immune effects of α and β radionuclides in metastatic prostate cancer. Nat Rev Urol 2024; 21:651-661. [PMID: 39192074 DOI: 10.1038/s41585-024-00924-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/29/2024]
Abstract
External beam radiotherapy is used for radical treatment of organ-confined prostate cancer and to treat lesions in metastatic disease whereas molecular radiotherapy with labelled prostate-specific membrane antigen ligands and radium-223 (223Ra) is indicated for metastatic prostate cancer and has demonstrated substantial improvements in symptom control and overall survival compared with standard-of-care treatment. Prostate cancer is considered an immunologically cold tumour, so limited studies investigating the treatment-induced effects on the immune response have been completed. However, emerging data support the idea that radiotherapy induces an immune response in prostate cancer, but whether the response is an antitumour or pro-tumour response is dependent on the radiotherapy regime and is also cell-line dependent. In vitro data demonstrate that single-dose radiotherapy regimes induce a greater immune-suppressive profile than fractionated regimes; less is known about the immune response induced by molecular radiotherapy agents, but evidence suggests that these agents might induce an immune-suppressive systemic immune response, indicated by increased expression of inhibitory checkpoint molecules such as programmed cell death 1 ligand 1 and 2, and that these changes could be associated with clinical response. Different radiotherapy modalities can induce distinct immune profiles, which can either activate or suppress immune-mediated tumour killing and the current preclinical models used for prostate cancer research are not yet optimal for studying the complexity of the radiotherapy-induced immune response.
Collapse
Affiliation(s)
- Sapna Lunj
- Division of Cancer Sciences, Oglesby Cancer Research Building, University of Manchester, Manchester, UK.
| | - Tim Andrew Davies Smith
- Nuclear Futures Institute, School of Computer Science and Engineering, Bangor University, Bangor, UK
| | - Kimberley Jayne Reeves
- Division of Cancer Sciences, Paterson Building, University of Manchester, Manchester, UK
| | - Fred Currell
- The Dalton Cumbria Facility and the Department of Chemistry, University of Manchester, Manchester, UK
| | - Jamie Honeychurch
- Division of Cancer Sciences, Paterson Building, University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Peter Hoskin
- Division of Cancer Sciences, Paterson Building, University of Manchester, Manchester, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, Oglesby Cancer Research Building, University of Manchester, Manchester, UK
- The Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
2
|
Varaprasad GL, Gupta VK, Prasad K, Kim E, Tej MB, Mohanty P, Verma HK, Raju GSR, Bhaskar L, Huh YS. Recent advances and future perspectives in the therapeutics of prostate cancer. Exp Hematol Oncol 2023; 12:80. [PMID: 37740236 PMCID: PMC10517568 DOI: 10.1186/s40164-023-00444-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 09/10/2023] [Indexed: 09/24/2023] Open
Abstract
Prostate cancer (PC) is one of the most common cancers in males and the fifth leading reason of death. Age, ethnicity, family history, and genetic defects are major factors that determine the aggressiveness and lethality of PC. The African population is at the highest risk of developing high-grade PC. It can be challenging to distinguish between low-risk and high-risk patients due to the slow progression of PC. Prostate-specific antigen (PSA) is a revolutionary discovery for the identification of PC. However, it has led to an increase in over diagnosis and over treatment of PC in the past few decades. Even if modifications are made to the standard PSA testing, the specificity has not been found to be significant. Our understanding of PC genetics and proteomics has improved due to advances in different fields. New serum, urine, and tissue biomarkers, such as PC antigen 3 (PCA3), have led to various new diagnostic tests, such as the prostate health index, 4K score, and PCA3. These tests significantly reduce the number of unnecessary and repeat biopsies performed. Chemotherapy, radiotherapy, and prostatectomy are standard treatment options. However, newer novel hormone therapy drugs with a better response have been identified. Androgen deprivation and hormonal therapy are evolving as new and better options for managing hormone-sensitive and castration-resistant PC. This review aimed to highlight and discuss epidemiology, various risk factors, and developments in PC diagnosis and treatment regimens.
Collapse
Affiliation(s)
- Ganji Lakshmi Varaprasad
- Department of Biological Sciences and Bioengineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, 22212, Republic of Korea
| | - Vivek Kumar Gupta
- Department of Biological Sciences and Bioengineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, 22212, Republic of Korea
| | - Kiran Prasad
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Eunsu Kim
- Department of Biological Sciences and Bioengineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, 22212, Republic of Korea
| | - Mandava Bhuvan Tej
- Department of Health Care Informatics, Sacred Heart University, 5151 Park Avenue, Fair Fields, CT, 06825, USA
| | - Pratik Mohanty
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lungs Health and Immunity, Helmholtz Zentrum, 85764, Neuherberg, Munich, Germany
| | - Ganji Seeta Rama Raju
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul, 04620, Republic of Korea.
| | - Lvks Bhaskar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India.
| | - Yun Suk Huh
- Department of Biological Sciences and Bioengineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, 22212, Republic of Korea.
| |
Collapse
|
3
|
Lilleby W, Seierstad T, Inderberg EM, Hole KH. Impact of human telomerase reverse transcriptase peptide vaccine combined with androgen deprivation therapy and radiotherapy in de novo metastatic prostate cancer: Long-term clinical monitoring. Int J Cancer 2023; 152:2166-2173. [PMID: 36715014 DOI: 10.1002/ijc.34448] [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: 11/22/2022] [Revised: 12/31/2022] [Accepted: 01/10/2023] [Indexed: 01/31/2023]
Abstract
Prostate cancer is considered as poorly immunogenic. In a phase I/II study on de novo metastatic prostate cancer we found that a human telomerase reverse transcriptase (hTERT) vaccine induced an early immune response in most of the patients. Here we present the results from the long-term monitoring of the immune responses and clinical outcomes. Twenty-two men with ISUP 4 to 5 and lymph node and/or bone metastases were treated with androgen deprivation therapy (ADT), radiotherapy and the hTERT vaccine UV1 between January 2013 and July 2014. Immune response was monitored before, during and after vaccination and continued every 6 months until PSA progression. All patients had magnetic resonance imaging (MRI) at baseline, and after 6 months, 1 and 2 years, and at progression. The clinical outcome was time to progression, overall survival and prostate cancer-specific survival. The median follow-up was 62 months (range: 19-101). At the last observation, nine of the 22 patients were still alive. Six have no progression, two have castration-resistant disease treated with second-line ADT and one has castration-refractory disease. Median time to PSA progression was 21 months, median overall survival was 62 months and median prostate cancer-specific survival was 84 months. Lack of immune response was an independent marker of prostate cancer death. The long-term monitoring showed that some patients had unanticipated subsequent high immune responses without developing recurrence. This association indicates that there might be a clinical benefit of hTERT vaccination in a subgroup of men with primary metastatic hormone-sensitive prostate cancer treated with ADT and radiotherapy.
Collapse
Affiliation(s)
| | - Therese Seierstad
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | | | - Knut Håkon Hole
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
4
|
Boosting the Immune Response—Combining Local and Immune Therapy for Prostate Cancer Treatment. Cells 2022; 11:cells11182793. [PMID: 36139368 PMCID: PMC9496996 DOI: 10.3390/cells11182793] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 11/29/2022] Open
Abstract
Due to its slow progression and susceptibility to radical forms of treatment, low-grade PC is associated with high overall survival (OS). With the clinical progression of PC, the therapy is becoming more complex. The immunosuppressive tumor microenvironment (TME) makes PC a difficult target for most immunotherapeutics. Its general immune resistance is established by e.g., immune evasion through Treg cells, synthesis of immunosuppressive mediators, and the defective expression of surface neoantigens. The success of sipuleucel-T in clinical trials initiated several other clinical studies that specifically target the immune escape of tumors and eliminate the immunosuppressive properties of the TME. In the settings of PC treatment, this can be commonly achieved with radiation therapy (RT). In addition, focal therapies usually applied for localized PC, such as high-intensity focused ultrasound (HIFU) therapy, cryotherapy, photodynamic therapy (PDT), and irreversible electroporation (IRE) were shown to boost the anti-cancer response. Nevertheless, the present guidelines restrict their application to the context of a clinical trial or a prospective cohort study. This review explains how RT and focal therapies enhance the immune response. We also provide data supporting the combination of RT and focal treatments with immune therapies.
Collapse
|
5
|
Issa M, Tang J, Guo Y, Coss C, Mace TA, Bischof J, Phelps M, Presley CJ, Owen DH. Risk factors and predictors of immune-related adverse events: implications for patients with non-small cell lung cancer. Expert Rev Anticancer Ther 2022; 22:861-874. [PMID: 35786142 DOI: 10.1080/14737140.2022.2094772] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICI) are now utilized as a standard of care treatment for multiple cancers, including in both the metastatic setting as well as in earlier stages of disease. The identification of unique immune-related adverse events (irAE) that occur during ICI treatment has led to intense research to identify potential risk factors and biomarkers that may assist in clinical decision making. Although initial studies in ICI were primarily in advanced stage disease, the use of ICI in earlier stages of disease as adjuvant therapies requires a better understanding of patient risk stratification to mitigate or prevent serious irAE. AREAS COVERED In this review, we set out to describe the current state of research regarding potential risk factors for irAE in patients with non-small cell lung cancer, as well as explore the barriers to understanding irAE. We review data from irAE that occur in large phase 3 trials and prospective studies focusing on irAE, as well as the many retrospective studies that currently form the bulk of our understanding of irAE.
Collapse
Affiliation(s)
- Majd Issa
- Division of Medical Oncology, Department of Internal Medicine, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| | - Joy Tang
- Division of Medical Oncology, Department of Internal Medicine, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| | - Yizhen Guo
- College of Pharmacy, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| | - Chris Coss
- College of Pharmacy, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| | - Thomas A Mace
- Division of Gastroenterology, Hepatology & Nutrition, Department of Internal Medicine, the Ohio State University Wexner Medical Center, Columbus, USA
| | - Jason Bischof
- Department of Emergency Medicine, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| | - Mitch Phelps
- College of Pharmacy, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| | - Carolyn J Presley
- Division of Medical Oncology, Department of Internal Medicine, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| | - Dwight H Owen
- Division of Medical Oncology, Department of Internal Medicine, the Ohio State University Wexner Medical Center - Comprehensive Cancer Center, Columbus, USA
| |
Collapse
|
6
|
López-Campos F, Gajate P, Romero-Laorden N, Zafra-Martín J, Juan M, Hernando Polo S, Conde Moreno A, Couñago F. Immunotherapy in Advanced Prostate Cancer: Current Knowledge and Future Directions. Biomedicines 2022; 10:537. [PMID: 35327339 PMCID: PMC8945350 DOI: 10.3390/biomedicines10030537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 02/22/2022] [Indexed: 02/01/2023] Open
Abstract
The advent of immunotherapy has revolutionized cancer treatment. Unfortunately, this has not been the case for metastatic castration-resistant prostate cancer (mCRPC), likely due to the heterogeneous and immune-suppressive microenvironment present in prostate cancer. The identification of molecular biomarkers that could predict response to immunotherapy represents one of the current challenges in this clinical scenario. The management of advanced castration-resistant prostate cancer is rapidly evolving and immunotherapy treatments, mostly consisting of immune checkpoint inhibitors combinations, BiTE® (bispecific T-cell engager) immune therapies, and chimeric antigen receptors (CAR) are in development with promising results. This review analyses the current evidence of immunotherapy treatments for mCRPC, evaluating past failures and promising approaches and discussing the directions for future research.
Collapse
Affiliation(s)
- Fernando López-Campos
- Radiation Oncology Department, Hospital Universitario Ramón y Cajal, 28024 Madrid, Spain
| | - Pablo Gajate
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, 28024 Madrid, Spain;
| | - Nuria Romero-Laorden
- Medical Oncology Department, Hospital Universitario La Princesa, 28006 Madrid, Spain;
| | - Juan Zafra-Martín
- Department of Radiation Oncology, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain;
| | - Manel Juan
- Servei d’Immunologia, CDB-Hospital Clínic, Plataforma de Inmunoterapia HSJD-Clínic, 08036 Barcelona, Spain;
| | - Susana Hernando Polo
- Medical Oncology Department, Hospital Universitario Fundación Alcorcón, 28922 Alcorcón, Spain;
| | - Antonio Conde Moreno
- Radiation Oncology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain;
| | - Felipe Couñago
- Department of Radiation Oncology, Hospital Universitario Quirónsalud, 28223 Madrid, Spain;
- Department of Radiation Oncology, Hospital La Luz, 28003 Madrid, Spain
- Universidad Europea de Madrid, 28670 Madrid, Spain
| |
Collapse
|
7
|
Vito A, Rathmann S, Mercanti N, El-Sayes N, Mossman K, Valliant J. Combined Radionuclide Therapy and Immunotherapy for Treatment of Triple Negative Breast Cancer. Int J Mol Sci 2021; 22:4843. [PMID: 34063642 PMCID: PMC8124136 DOI: 10.3390/ijms22094843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/28/2021] [Accepted: 05/01/2021] [Indexed: 01/22/2023] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive subtype of the disease with poor clinical outcomes and limited therapeutic options. Immune checkpoint blockade (CP) has surged to the forefront of cancer therapies with widespread clinical success in a variety of cancer types. However, the percentage of TNBC patients that benefit from CP as a monotherapy is low, and clinical trials have shown the need for combined therapeutic modalities. Specifically, there has been interest in combining CP therapy with radiation therapy where clinical studies primarily with external beam have suggested their therapeutic synergy, contributing to the development of anti-tumor immunity. Here, we have developed a therapeutic platform combining radionuclide therapy (RT) and immunotherapy utilizing a radiolabeled biomolecule and CP in an E0771 murine TNBC tumor model. Survival studies show that while neither monotherapy is able to improve therapeutic outcomes, the combination of RT + CP extended overall survival. Histologic analysis showed that RT + CP increased necrotic tissue within the tumor and decreased levels of F4/80+ macrophages. Flow cytometry analysis of the peripheral blood also showed that RT + CP suppressed macrophages and myeloid-derived suppressive cells, both of which actively contribute to immune escape and tumor relapse.
Collapse
Affiliation(s)
- Alyssa Vito
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada; (A.V.); (N.E.-S.)
| | - Stephanie Rathmann
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4K1, Canada; (S.R.); (N.M.)
| | - Natalie Mercanti
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4K1, Canada; (S.R.); (N.M.)
| | - Nader El-Sayes
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada; (A.V.); (N.E.-S.)
| | - Karen Mossman
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada; (A.V.); (N.E.-S.)
| | - John Valliant
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4K1, Canada; (S.R.); (N.M.)
| |
Collapse
|
8
|
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
|
9
|
Philippou Y, Sjoberg HT, Murphy E, Alyacoubi S, Jones KI, Gordon-Weeks AN, Phyu S, Parkes EE, Gillies McKenna W, Lamb AD, Gileadi U, Cerundolo V, Scheiblin DA, Lockett SJ, Wink DA, Mills IG, Hamdy FC, Muschel RJ, Bryant RJ. Impacts of combining anti-PD-L1 immunotherapy and radiotherapy on the tumour immune microenvironment in a murine prostate cancer model. Br J Cancer 2020; 123:1089-1100. [PMID: 32641865 PMCID: PMC7525450 DOI: 10.1038/s41416-020-0956-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/18/2020] [Accepted: 06/04/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Radiotherapy enhances innate and adaptive anti-tumour immunity. It is unclear whether this effect may be harnessed by combining immunotherapy with radiotherapy fractions used to treat prostate cancer. We investigated tumour immune microenvironment responses of pre-clinical prostate cancer models to radiotherapy. Having defined this landscape, we tested whether radiotherapy-induced tumour growth delay could be enhanced with anti-PD-L1. METHODS Hypofractionated radiotherapy was delivered to TRAMP-C1 and MyC-CaP flank allografts. Tumour growth delay, tumour immune microenvironment flow-cytometry, and immune gene expression were analysed. TRAMP-C1 allografts were then treated with 3 × 5 Gy ± anti-PD-L1. RESULTS 3 × 5 Gy caused tumour growth delay in TRAMP-C1 and MyC-CaP. Tumour immune microenvironment changes in TRAMP-C1 at 7 days post-radiotherapy included increased tumour-associated macrophages and dendritic cells and upregulation of PD-1/PD-L1, CD8+ T-cell, dendritic cell, and regulatory T-cell genes. At tumour regrowth post-3 × 5 Gy the tumour immune microenvironment flow-cytometry was similar to control tumours, however CD8+, natural killer and dendritic cell gene transcripts were reduced. PD-L1 inhibition plus 3 × 5 Gy in TRAMP-C1 did not enhance tumour growth delay versus monotherapy. CONCLUSION 3 × 5 Gy hypofractionated radiotherapy can result in tumour growth delay and immune cell changes in allograft prostate cancer models. Adjuncts beyond immunomodulation may be necessary to improve the radiotherapy-induced anti-tumour response.
Collapse
Affiliation(s)
| | - Hanna T Sjoberg
- Department of Oncology, University of Oxford, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Emma Murphy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Said Alyacoubi
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Keaton I Jones
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Su Phyu
- Department of Oncology, University of Oxford, Oxford, UK
| | | | | | - Alastair D Lamb
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Uzi Gileadi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - David A Scheiblin
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, National Institutes of Health, Frederick, 21702, MD, USA
| | - Stephen J Lockett
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, National Institutes of Health, Frederick, 21702, MD, USA
| | - David A Wink
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, 21702, MD, USA
| | - Ian G Mills
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Freddie C Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Ruth J Muschel
- Department of Oncology, University of Oxford, Oxford, UK
| | - Richard J Bryant
- Department of Oncology, University of Oxford, Oxford, UK.
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
10
|
Philippou Y, Sjoberg H, Lamb AD, Camilleri P, Bryant RJ. Harnessing the potential of multimodal radiotherapy in prostate cancer. Nat Rev Urol 2020; 17:321-338. [PMID: 32358562 DOI: 10.1038/s41585-020-0310-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2020] [Indexed: 12/11/2022]
Abstract
Radiotherapy in combination with androgen deprivation therapy (ADT) is a standard treatment option for men with localized and locally advanced prostate cancer. However, emerging clinical evidence suggests that radiotherapy can be incorporated into multimodality therapy regimens beyond ADT, in combinations that include chemotherapy, radiosensitizing agents, immunotherapy and surgery for the treatment of men with localized and locally advanced prostate cancer, and those with oligometastatic disease, in whom the low metastatic burden in particular might be treatable with these combinations. This multimodal approach is increasingly recognized as offering considerable clinical benefit, such as increased antitumour effects and improved survival. Thus, radiotherapy is becoming a key component of multimodal therapy for many stages of prostate cancer, particularly oligometastatic disease.
Collapse
Affiliation(s)
- Yiannis Philippou
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Headington, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Hanna Sjoberg
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Alastair D Lamb
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Philip Camilleri
- Oxford Department of Clinical Oncology, Churchill Hospital Cancer Centre, Oxford University Hospitals NHS Foundation Trust, Headington, Oxford, UK
| | - Richard J Bryant
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Headington, Oxford, UK.
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK.
| |
Collapse
|
11
|
Xu J, Qiu Y. Current opinion and mechanistic interpretation of combination therapy for castration-resistant prostate cancer. Asian J Androl 2020; 21:270-278. [PMID: 30924449 PMCID: PMC6498727 DOI: 10.4103/aja.aja_10_19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Recent advances in genomics technology have led to the massive discovery of new drug targets for prostate cancer; however, none of the currently available therapeutics is curative. One of the greatest challenges is drug resistance. Combinations of therapies with distinct mechanisms of action represent a promising strategy that has received renewed attention in recent years. Combination therapies exert cancer killing functions through either concomitant targeting of multiple pro-cancer factors or more effective inhibition of a single pathway. Theoretically, the combination therapy can improve efficacy and efficiency compared with monotherapy. Although increasing numbers of drug combinations are currently being tested in clinical trials, the mechanisms by which these combinations can overcome drug resistance have yet to be fully understood. The purpose of this review is to summarize recent work on therapeutic combinations in the treatment of castration-resistant prostate cancer and discuss emerging mechanisms underlying drug resistance. In addition, we provide an overview of the current preclinical mechanistic studies on potential therapeutic combinations to overcome drug resistance.
Collapse
Affiliation(s)
- Jin Xu
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yun Qiu
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
12
|
Seifert M, Peitzsch C, Gorodetska I, Börner C, Klink B, Dubrovska A. Network-based analysis of prostate cancer cell lines reveals novel marker gene candidates associated with radioresistance and patient relapse. PLoS Comput Biol 2019; 15:e1007460. [PMID: 31682594 PMCID: PMC6855562 DOI: 10.1371/journal.pcbi.1007460] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 11/14/2019] [Accepted: 10/05/2019] [Indexed: 12/20/2022] Open
Abstract
Radiation therapy is an important and effective treatment option for prostate cancer, but high-risk patients are prone to relapse due to radioresistance of cancer cells. Molecular mechanisms that contribute to radioresistance are not fully understood. Novel computational strategies are needed to identify radioresistance driver genes from hundreds of gene copy number alterations. We developed a network-based approach based on lasso regression in combination with network propagation for the analysis of prostate cancer cell lines with acquired radioresistance to identify clinically relevant marker genes associated with radioresistance in prostate cancer patients. We analyzed established radioresistant cell lines of the prostate cancer cell lines DU145 and LNCaP and compared their gene copy number and expression profiles to their radiosensitive parental cells. We found that radioresistant DU145 showed much more gene copy number alterations than LNCaP and their gene expression profiles were highly cell line specific. We learned a genome-wide prostate cancer-specific gene regulatory network and quantified impacts of differentially expressed genes with directly underlying copy number alterations on known radioresistance marker genes. This revealed several potential driver candidates involved in the regulation of cancer-relevant processes. Importantly, we found that ten driver candidates from DU145 (ADAMTS9, AKR1B10, CXXC5, FST, FOXL1, GRPR, ITGA2, SOX17, STARD4, VGF) and four from LNCaP (FHL5, LYPLAL1, PAK7, TDRD6) were able to distinguish irradiated prostate cancer patients into early and late relapse groups. Moreover, in-depth in vitro validations for VGF (Neurosecretory protein VGF) showed that siRNA-mediated gene silencing increased the radiosensitivity of DU145 and LNCaP cells. Our computational approach enabled to predict novel radioresistance driver gene candidates. Additional preclinical and clinical studies are required to further validate the role of VGF and other candidate genes as potential biomarkers for the prediction of radiotherapy responses and as potential targets for radiosensitization of prostate cancer. Prostate cancer cell lines represent an important model system to characterize molecular alterations that contribute to radioresistance, but irradiation can cause deletions and amplifications of DNA segments that affect hundreds of genes. This in combination with the small number of cell lines that are usually considered does not allow a straight-forward identification of driver genes by standard statistical methods. Therefore, we developed a network-based approach to analyze gene copy number and expression profiles of such cell lines enabling to identify potential driver genes associated with radioresistance of prostate cancer. We used lasso regression in combination with a significance test for lasso to learn a genome-wide prostate cancer-specific gene regulatory network. We used this network for network flow computations to determine impacts of gene copy number alterations on known radioresistance marker genes. Mapping to prostate cancer samples and additional filtering allowed us to identify 14 driver gene candidates that distinguished irradiated prostate cancer patients into early and late relapse groups. In-depth literature analysis and wet-lab validations suggest that our method can predict novel radioresistance driver genes. Additional preclinical and clinical studies are required to further validate these genes for the prediction of radiotherapy responses and as potential targets to radiosensitize prostate cancer.
Collapse
Affiliation(s)
- Michael Seifert
- Institute for Medical Informatics and Biometry (IMB), Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany
- * E-mail:
| | - Claudia Peitzsch
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Ielizaveta Gorodetska
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Caroline Börner
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Barbara Klink
- Institute for Clinical Genetics, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Anna Dubrovska
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiooncology-OncoRay, Dresden, Germany
- German Cancer Consortium (DKTK) Partner Site Dresden, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
13
|
Patel VG, Oh WK. The evolving landscape of immunotherapy in advanced prostate cancer. Immunotherapy 2019; 11:903-912. [PMID: 31161846 DOI: 10.2217/imt-2019-0019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Prostate cancer exists in a clinical continuum of hormone-sensitive to castration-resistant disease. Despite the use of chemotherapy and androgen synthesis inhibitors in the castration-resistant setting, this remains a lethal disease. The advent of immune checkpoint blockade has changed the outlook for cancer treatment and survival for several tumors since its first approval in 2011; however, the clinical benefit in castration-resistant prostate cancer (CRPC) is rather limited. Currently, Sipuleucel-T remains the only immune modality to be approved in CRPC setting. Such immune resistance likely exists due to low immunogenicity of prostate tumor cells and an immunosuppressive tumor microenvironment. In this review, we describe the early experiences of immune checkpoint blockade and therapeutic vaccines in CRPC. We then outline strategies currently being implemented to overcome immune resistance, as well as genomic biomarker investigation to identify patients that may harbor more immunogenic tumors. At last, we preview emerging immunotherapeutic platforms.
Collapse
Affiliation(s)
- Vaibhav G Patel
- Department of Medicine, Division of Hematology & Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10010, USA
| | - William K Oh
- Department of Medicine, Division of Hematology & Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10010, USA
| |
Collapse
|
14
|
Zhang MX, Li XB, Guan BJ, Guan GX, Lin XY, Wu XD, Chi P, Xu BH. Dose escalation of preoperative short-course radiotherapy followed by neoadjuvant chemotherapy in locally advanced rectal cancer: protocol for an open-label, single-centre, phase I clinical trial. BMJ Open 2019; 9:e025944. [PMID: 30904869 PMCID: PMC6475145 DOI: 10.1136/bmjopen-2018-025944] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Preoperative radiotherapy followed by total mesorectal excision with adjuvant chemotherapy has been recommended as the preferred treatment method for locally advanced rectal cancer (LARC). Similar rates of local control, survival and toxicity were observed in preoperative long-course chemoradiotherapy (LCRT) (45-50.4 Gy in 25-28 fractions) and in short-course radiotherapy (SCRT) with 25 Gy over five fractions. Both regimens lower the local recurrence rates compared with that of surgery followed by postoperative radiotherapy. With the simplicity and lower cost of SCRT, a growing number of patients have been receiving SCRT as preoperative radiotherapy. However, the currently established SCRT (25 Gy over five fractions) followed immediately by surgery resulted in poor downstaging and sphincter preservation rate. The pathological complete response (pCR) rate is also markedly lower with SCRT than with LCRT (0.7%vs16%). Several studies recommended SCRT with delayed surgery for more than 4 weeks with expectation of improved pathological outcomes and fewer postoperative complications. While a number of clinical trials demonstrated a persistently better overall local control with SCRT than with LCRT, overall survival advantage has not been observed. Since survival is mainly depended on distant metastases, efforts should be made towards more effective pathological response and systemic treatment. Given the apparent advantages of SCRT, we aimed to establish a dose escalation of SCRT and sequential modified FOLFOX6 (mFOLFOX6) as preoperative therapy for LARC with objectives of achieving an optimal balance of safety, cost effectiveness and clinical outcome, and to support further investigation of this regimen in a phase II/III setting. METHODS In this phase I study, three dose levels (6Gy×5F, 7Gy×5F, 8Gy×5F to gross tumour volume, while keeping the rest of irradiated volume at 5Gy×5) of SCRT followed by four cycles of mFOLFOX6 chemotherapy as neoadjuvant therapy will be tested by using the traditional 3+3 design. The pCR rate, R0 resection rate, sphincter preservation rate and treatment related toxicity will be assessed. ETHICS AND DISSEMINATION The study protocol was approved by the Ethics Committee of Fujian Medical University Union Hospital (No. 2017YF020-02) and all participants provided written informed consent. Results from our study will be disseminated in international peer-reviewed journals. All study procedures were developed in order to assure data protection and confidentiality. TRIAL REGISTRATION NUMBER NCT03466424; Pre-results.
Collapse
Affiliation(s)
- Meng-xia Zhang
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Xiao-bo Li
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Collogy of medical technology and engineering, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Bing-jie Guan
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Guo-xian Guan
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Xiao-yan Lin
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Xiao-dong Wu
- Department of Radiation Oncology, Cancer Hospital of Fudan University, Shanghai, China
- Department of biomedical engineering, Innovative Cancer Institute, Miami, Florida, USA
| | - Pan Chi
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Ben-hua Xu
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
15
|
Sanguineti G, Giannarelli D, Petrongari MG, Arcangeli S, Sangiovanni A, Saracino B, Farneti A, Faiella A, Conte M, Arcangeli G. Leukotoxicity after moderately Hypofractionated radiotherapy versus conventionally fractionated dose escalated radiotherapy for localized prostate Cancer: a secondary analysis from a randomized study. Radiat Oncol 2019; 14:23. [PMID: 30700317 PMCID: PMC6352380 DOI: 10.1186/s13014-019-1223-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 01/20/2019] [Indexed: 11/29/2022] Open
Abstract
Background To compare WBC counts during treatment of localized prostate cancer with either conventionally fractionated (CF) or moderately hypofractionated (HYPO) radiotherapy. Methods Weekly blood test results were extracted from the charts of patients treated within a phase III study comparing HYPO to CF. In order to compare WBC counts at the same nominal dose in both arms and thus to tease out the effect of fractionation, for each recorded WBC value the corresponding cumulative total dose was extracted as well. WBC counts were binned according to percentiles of the delivered dose and three dose levels were identified at median doses of 16, 34.1 and 52 Gy, respectively. A General Linear Model based on mixed design Analysis Of Variance (ANOVA) was used to test variation of WBC counts between the two treatment arms. Results Out of 168 randomized patients, 140 (83.3%) had at least one observation for each one of the selected dose levels and were included in the analysis. Mean counts were lower in the CF than the HYPO arm at all selected dose levels, reaching a statistically significant difference at dose level #3 (5397/mm3 vs 6038/mm3 for CF and HYPO, respectively, p = 0.004). The GLM model confirms that the impact of dose on WBC counts is significantly lower in the HYPO arm over the CF one (Greenhouse-Geisser test, p = 0.04). Interestingly, while WBC counts tend to drop throughout all dose levels in the CF arm, this is the case only in the earlier part of treatment in the HYPO arm. Conclusion This secondary analysis of a phase III study shows that dose fractionation is correlated to WBC drop during treatment of localized prostate cancer, favoring HYPO over CF. Electronic supplementary material The online version of this article (10.1186/s13014-019-1223-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Giuseppe Sanguineti
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.
| | - Diana Giannarelli
- Biostatistical Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Grazia Petrongari
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Stefano Arcangeli
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Angelo Sangiovanni
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Biancamaria Saracino
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Alessia Farneti
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Adriana Faiella
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Mario Conte
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Giorgio Arcangeli
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| |
Collapse
|
16
|
Wang K, Li HL, Xiong YF, Shi Y, Li ZY, Li J, Zhang X, Li HY. Development and validation of nomograms integrating immune-related genomic signatures with clinicopathologic features to improve prognosis and predictive value of triple-negative breast cancer: A gene expression-based retrospective study. Cancer Med 2019; 8:686-700. [PMID: 30677255 PMCID: PMC6382728 DOI: 10.1002/cam4.1880] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/06/2018] [Accepted: 10/27/2018] [Indexed: 02/05/2023] Open
Abstract
Purpose Accumulating evidence indicated that triple‐negative breast cancer (TNBC) can stimulate stronger immune responses than other subtypes of breast cancer. We hypothesized that integrating immune‐related genomic signatures with clinicopathologic factors may yield a predictive accuracy exceeding that of the currently available system. Methods Ten signatures that reflect specific immunogenic or immune microenvironmental features of TNBC were identified and re‐analyzed using bioinformatic methods. Then, clinically annotated TNBC (n = 711) with the corresponding expression profiles, which predicted a patient's probability of disease‐free survival (DFS) and overall survival (OS), was pooled to evaluate their prognostic values and establish a clinicopathologic‐genomic nomogram. Three and two immune features were, respectively, selected out of 10 immune features to construct nomogram for DFS and OS prediction based on multivariate backward stepwise Cox regression analyses. Results By integrating the above immune expression signatures with prognostic clinicopathologic features, clinicopathologic‐genomic nomograms were cautiously constructed, which showed reasonable prediction accuracies (DFS: HR, 1.79; 95% CI, 1.46‐2.18, P < 0.001; AUC, 0.71; OS: HR, 1.96; 95% CI, 1.54‐2.49; P < 0.001; AUC, 0.73). The nomogram showed low‐risk subgroup had higher immune checkpoint molecules (PD‐L1, PD‐1, CTLA‐4, LAG‐3) expression and benefited from radiotherapy (HR, 0.2, 95% CI, 0.05‐0.89; P = 0.034) rather than chemotherapy (HR, 1.26, 95% CI, 0.66‐2.43; P = 0.485). Conclusions These findings offer evidence that immune‐related genomic data provide independent and complementary prognostic information for TNBC, and the nomogram might be a practical predictive tool to identify TNBC patients who would benefit from chemotherapy, radiotherapy, and upcoming popularity of immunotherapy.
Collapse
Affiliation(s)
- Kang Wang
- Department of the Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Hai-Lin Li
- Department of the Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Yong-Fu Xiong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Yang Shi
- Division of Biostatistics and Data Science, Department of Population Health Sciences, Medical College of Georgia, Augusta University, Augusta, Georgia.,West China School of Public Health, Sichuan University, Chengdu, China
| | - Zhu-Yue Li
- Institute of Hospital Management, West China Hospital, Sichuan University, Chengdu, China.,West China Hospital/West China School of Nursing, Sichuan University, Chengdu, China
| | - Jie Li
- Department of the Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Xiang Zhang
- Department of the Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Hong-Yuan Li
- Department of the Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| |
Collapse
|
17
|
Twardowski P, Wong JYC, Pal SK, Maughan BL, Frankel PH, Franklin K, Junqueira M, Prajapati MR, Nachaegari G, Harwood D, Agarwal N. Randomized phase II trial of sipuleucel-T immunotherapy preceded by sensitizing radiation therapy and sipuleucel-T alone in patients with metastatic castrate resistant prostate cancer. Cancer Treat Res Commun 2019; 19:100116. [PMID: 30682445 DOI: 10.1016/j.ctarc.2018.100116] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 12/10/2018] [Indexed: 05/13/2023]
Abstract
BACKGROUND Sipuleucel-T is an autologous cellular immunotherapy indicated for patients with asymptomatic or minimally symptomatic metastatic castration resistant prostate cancer (mCRPC). Since radiation therapy (RT) can suppress bone marrow function and immune responses, previous studies evaluating sipuleucel-T excluded patients who received RT less than or equal to 28 days prior to sipuleucel-T therapy. Recent evidence suggests that RT may act synergistically with immunotherapy to enhance and broaden antitumor immune response. METHODS Patients who met standard criteria for sipuleucel-T were randomized to receive sipuleucel-T alone (Arm A) or sipuleucel-T initiated 1 week after completing sensitizing RT to single metastatic site (Arm B). RT was delivered at 300cGy/day to 3000 cGy total. The primary endpoint was the ability to safely combine sipuleucel-T preceded by RT and generate sipuleucel-T with adequate product immune activation parameters. Secondary endpoints included the measurement of systemic immune responses to prostatic acid phosphatase (PAP), a target for sipuleucel-T immune therapy and PA20204 (recombinant fusion protein utilized in the generation of sipuleucel-T). RESULTS 51 pts were enrolled, 2 did not receive any sipuleucel-T because of vascular access problems and were excluded. 24 were treated on Arm A, 25 on Arm B. 47/49 patients received all 3 sipuleucel-T infusions. Median age was 66 yrs (range 45-90). Sipuleucel-T product parameters including: total nucleated cell (TNC) count, antigen presenting cell (APC) count were similar in both groups. Cumulative APC upregulation was higher in Arm A. 1 patient in Arm A demonstrated PSA response. Median progression free survival (PFS) was 2.46 months on Arm A and 3.65 months on Arm B (p = 0.06). Both arms showed similar increases in humoral responses to PA2024 and PAP. IFN-ƴ ELISPOT T-cell activation responses to PA20204 were observed in both arms, but were more robust in the Arm A (p = 0.028). Both arms were well-tolerated, with fatigue as the most common grade 2 adverse event (1 patient in Arm A and 3 patients in Arm B). CONCLUSIONS Sensitizing RT completed 1 week before generation of sipuleucel-T did not affect the majority of product parameters and the ability to deliver sipuleucel-T therapy. RT did not enhance the humoral and cellular responses associated with sipuleucel-T therapy.
Collapse
Affiliation(s)
| | - Jeffrey Y C Wong
- City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Sumanta K Pal
- City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Benjamin L Maughan
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | | | - Kelly Franklin
- City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | | | | | - Gayatri Nachaegari
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Deborah Harwood
- City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
18
|
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
|
19
|
Lacher MD, Bauer G, Fury B, Graeve S, Fledderman EL, Petrie TD, Coleal-Bergum DP, Hackett T, Perotti NH, Kong YY, Kwok WW, Wagner JP, Wiseman CL, Williams WV. SV-BR-1-GM, a Clinically Effective GM-CSF-Secreting Breast Cancer Cell Line, Expresses an Immune Signature and Directly Activates CD4 + T Lymphocytes. Front Immunol 2018; 9:776. [PMID: 29867922 PMCID: PMC5962696 DOI: 10.3389/fimmu.2018.00776] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 03/28/2018] [Indexed: 12/18/2022] Open
Abstract
Targeted cancer immunotherapy with irradiated, granulocyte–macrophage colony-stimulating factor (GM-CSF)-secreting, allogeneic cancer cell lines has been an effective approach to reduce tumor burden in several patients. It is generally assumed that to be effective, these cell lines need to express immunogenic antigens coexpressed in patient tumor cells, and antigen-presenting cells need to take up such antigens then present them to patient T cells. We have previously reported that, in a phase I pilot study (ClinicalTrials.gov NCT00095862), a subject with stage IV breast cancer experienced substantial regression of breast, lung, and brain lesions following inoculation with clinical formulations of SV-BR-1-GM, a GM-CSF-secreting breast tumor cell line. To identify diagnostic features permitting the prospective identification of patients likely to benefit from SV-BR-1-GM, we conducted a molecular analysis of the SV-BR-1-GM cell line and of patient-derived blood, as well as a tumor specimen. Compared to normal human breast cells, SV-BR-1-GM cells overexpress genes encoding tumor-associated antigens (TAAs) such as PRAME, a cancer/testis antigen. Curiously, despite its presumptive breast epithelial origin, the cell line expresses major histocompatibility complex (MHC) class II genes (HLA-DRA, HLA-DRB3, HLA-DMA, HLA-DMB), in addition to several other factors known to play immunostimulatory roles. These factors include MHC class I components (B2M, HLA-A, HLA-B), ADA (encoding adenosine deaminase), ADGRE5 (CD97), CD58 (LFA3), CD74 (encoding invariant chain and CLIP), CD83, CXCL8 (IL8), CXCL16, HLA-F, IL6, IL18, and KITLG. Moreover, both SV-BR-1-GM cells and the responding study subject carried an HLA-DRB3*02:02 allele, raising the question of whether SV-BR-1-GM cells can directly present endogenous antigens to T cells, thereby inducing a tumor-directed immune response. In support of this, SV-BR-1-GM cells (which also carry the HLA-DRB3*01:01 allele) treated with yellow fever virus (YFV) envelope (Env) 43–59 peptides reactivated YFV-DRB3*01:01-specific CD4+ T cells. Thus, the partial HLA allele match between SV-BR-1-GM and the clinical responder might have enabled patient T lymphocytes to directly recognize SV-BR-1-GM TAAs as presented on SV-BR-1-GM MHCs. Taken together, our findings are consistent with a potentially unique mechanism of action by which SV-BR-1-GM cells can act as APCs for previously primed CD4+ T cells.
Collapse
Affiliation(s)
| | - Gerhard Bauer
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Brian Fury
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Sanne Graeve
- BriaCell Therapeutics Corp., Berkeley, CA, United States
| | - Emily L Fledderman
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Tye D Petrie
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Dane P Coleal-Bergum
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Tia Hackett
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Nicholas H Perotti
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Ying Y Kong
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | | | | | | |
Collapse
|
20
|
Abstract
Harnessing the host immune system to combat genitourinary cancers has key theoretical advantages over other anticancer strategies including specificity and memory which should translate to favorable tolerability and response durability in the clinic. Indeed, key examples of the potential for immunotherapeutic treatment of solid tumors are derived from data in genitourinary cancers including Bacillus Calmette-Guerin for urothelial cancer, sipuleucel-T for prostate cancer, and interleukin-2 for renal cancer. Despite these successes, developing effective immunotherapeutic strategies for the treatment of cancer has largely been hampered by an incomplete understanding of tumor immunobiology and mechanisms of immune resistance. In just a few years since entering the clinic, immune checkpoint blockade has dramatically changed the landscaped of treatment for genitourinary cancer and has secured a place as a standard pillar of treatment. Further iterative bench-bedside-bench research is anticipated to extend the benefits of immunotherapeutic-based approaches to additional patients.
Collapse
Affiliation(s)
- Kyrollis Attalla
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John P Sfakianos
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Galsky
- Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Genitourinary Medical Oncology, Tisch Cancer Institute, New York, NY, USA.
| |
Collapse
|
21
|
Alaia C, Boccellino M, Zappavigna S, Amler E, Quagliuolo L, Rossetti S, Facchini G, Caraglia M. Ipilimumab for the treatment of metastatic prostate cancer. Expert Opin Biol Ther 2017; 18:205-213. [PMID: 29271259 DOI: 10.1080/14712598.2018.1420777] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Immunotherapy with checkpoint inhibitors is beginning to be recognized as a valid weapon for the treatment of metastatic prostate cancer (PCa) when chemotherapy fails. Ipilimumab (ipi) is a fully humanized monoclonal antibody that blocks the activity of CTLA4. It also has a molecular weight of 148 kDa and is water-soluble at physiological pH. Ipi was first approved by the FDA for the treatment of malignant melanoma and is currently being studied in metastatic castration-resistant prostate cancer, with promising early results. Areas covered: The aim of this review is to collate the most significant preclinical and clinical studies available that look at ipi to propose new strategies for the future. Expert opinion: Additional studies are required to reduce toxicity and increase the activity of ipi in PCa. A possible strategy is to combine ipi with standard anti-cancer therapeutics such as vaccines, PDL1 inhibitors, antiandrogen drugs, and chemotherapy agents. Several initial results have suggested that combination strategies are useful to increase the activity in mCRPC, even if the toxicity of the treatment can increase. The activity of combined treatments is still not predictable, but considering the ongoing studies, we believe that they have good potential that will lead to the discovery of an optimal therapeutic strategy.
Collapse
Affiliation(s)
- Concetta Alaia
- a Department of Biochemistry, Biophysics and General Pathology , University of Campania "L. Vanvitelli" , Naples , Italy
| | - Mariarosaria Boccellino
- a Department of Biochemistry, Biophysics and General Pathology , University of Campania "L. Vanvitelli" , Naples , Italy
| | - Silvia Zappavigna
- a Department of Biochemistry, Biophysics and General Pathology , University of Campania "L. Vanvitelli" , Naples , Italy
| | - Evzen Amler
- b Department of Biophysics, 2nd Faculty of Medicine , Charles University Prague , Prague , Czech Republic.,c Laboratory of Tissue Engineering, Institute of Experimental Medicine , Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Lucio Quagliuolo
- a Department of Biochemistry, Biophysics and General Pathology , University of Campania "L. Vanvitelli" , Naples , Italy
| | - Sabrina Rossetti
- d Division of Medical Oncology, Department of Uro-Gynaecological Oncology , Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale" , Napoli , Italy.,e Progetto ONCONET2.0 - Linea progettuale 14 per l'implementazione della prevenzione e diagnosi precoce del tumore alla prostata e testicolo, Uro-Gynaechological Department of the National Institute of Tumours "G. Pascale", Regione Campania , Naples , Italy
| | - Gaetano Facchini
- d Division of Medical Oncology, Department of Uro-Gynaecological Oncology , Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale" , Napoli , Italy
| | - Michele Caraglia
- a Department of Biochemistry, Biophysics and General Pathology , University of Campania "L. Vanvitelli" , Naples , Italy
| |
Collapse
|
22
|
Bilusic M, Madan RA, Gulley JL. Immunotherapy of Prostate Cancer: Facts and Hopes. Clin Cancer Res 2017; 23:6764-6770. [PMID: 28663235 PMCID: PMC5690854 DOI: 10.1158/1078-0432.ccr-17-0019] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/24/2017] [Accepted: 06/23/2017] [Indexed: 11/16/2022]
Abstract
In the last few years, immunotherapy has become an important cancer treatment modality, and although the principles of immunotherapy have evolved over many decades, the FDA approvals of sipuleucel-T and ipilimumab began a new wave in immuno-oncology. Despite the current enthusiasm, it is unlikely that any of the immunotherapeutics alone can dramatically change prostate cancer outcomes, but combination strategies are more promising and provide a reason for optimism. Several completed and ongoing studies have shown that the combination of cancer vaccines or checkpoint inhibitors with different immunotherapeutic agents, hormonal therapy (enzalutamide), radiotherapy (radium 223), DNA-damaging agents (olaparib), or chemotherapy (docetaxel) can enhance immune responses and induce more dramatic, long-lasting clinical responses without significant toxicity. The goal of prostate cancer immunotherapy does not have to be complete eradication of advanced disease but rather the return to an immunologic equilibrium with an indolent disease state. In addition to determining the optimal combination of treatment regimens, efforts are also ongoing to discover biomarkers of immune response. With such concerted efforts, the future of immunotherapy in prostate cancer looks brighter than ever. Clin Cancer Res; 23(22); 6764-70. ©2017 AACR.
Collapse
Affiliation(s)
- Marijo Bilusic
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ravi A Madan
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
23
|
ALBERTI C. Prostate cancer immunotherapy, particularly in combination with androgen deprivation or radiation treatment. Customized pharmacogenomic approaches to overcome immunotherapy cancer resistance. G Chir 2017; 37:225-235. [PMID: 28098061 PMCID: PMC5256907 DOI: 10.11138/gchir/2016.37.5.225] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Conventional therapeutic approaches for advanced prostate cancer - such as androgen deprivation, chemotherapy, radiation - come up often against lack of effectiveness because of possible arising of correlative cancer cell resistance and/or inadequate anti-tumor immune conditions. Whence the timeliness of resorting to immune-based treatment strategies including either therapeutic vaccination-based active immunotherapy or anti-tumor monoclonal antibody-mediated passive immunotherapy. Particularly attractive, as for research studies and clinical applications, results to be the cytotoxic T-lymphocyte check point blockade by the use of anti-CTLA-4 and PD-1 monoclonal antibodies, particularly when combined with androgen deprivation therapy or radiation. Unlike afore said immune check point inhibitors, both cell-based (by the use of prostate specific antigen carriers autologous dendritic cells or even whole cancer cells) and recombinant viral vector vaccines are able to induce immune-mediated focused killing of specific antigen-presenting prostate cancer cells. Such vaccines, either used alone or concurrently/sequentially combined with above-mentioned conventional therapies, led to generally reach, in the field of various clinical trials, reasonable results particularly as regards the patient's overall survival. Adoptive trasferred T-cells, as adoptive T-cell passive immunotherapy, and monoclonal antibodies against specific antigen-endowed prostate cancer cells can improve immune micro-environmental conditions. On the basis of a preliminary survey about various immunotherapy strategies, are here also outlined their effects when combined with androgen deprivation therapy or radiation. What's more, as regard the immune-based treatment effectiveness, it has to be pointed out that suitable personalized epigenetic/gene profile-achieved pharmacogenomic approaches to target identified gene aberrations, may lead to overcome - as well as for conventional therapies - possible prostate cancer resistance to immunotherapy.
Collapse
|
24
|
Schoenhals JE, Skrepnik T, Selek U, Cortez MA, Li A, Welsh JW. Optimizing Radiotherapy with Immunotherapeutic Approaches. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 995:53-71. [PMID: 28321812 DOI: 10.1007/978-3-319-53156-4_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Several factors must be considered to successfully integrate immunotherapy with radiation into clinical practice. One such factor is that concepts arising from preclinical work must be tested in combination with radiation in preclinical models to better understand how combination therapy will work in patients; examples include checkpoint inhibitors, tumor growth factor-beta (TGF-β) inhibitors, and natural killer (NK) cell therapy. Also, many radiation fields and fractionation schedules typically used in radiation therapy had been standardized before the introduction of advanced techniques for radiation planning and delivery that account for changes in tumor size, location, and motion during treatment, as well as uncertainties introduced by variations in patient setup between treatment fractions. As a result, radiation therapy may involve the use of large treatment volumes, often encompassing nodal regions that may not be irradiated with more conformal techniques. Traditional forms of radiation in particular pose challenges for combination trials with immunotherapy. This chapter explores these issues in more detail and provides insights as to how radiation therapy can be optimized to combine with immunotherapy.
Collapse
Affiliation(s)
- Jonathan E Schoenhals
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tijana Skrepnik
- Department of Radiation Oncology, University of Arizona, Tucson, AZ, USA
| | - Ugur Selek
- Department of Radiation Oncology, Koc University, Istanbul, Turkey
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Anderson Central (Y2.5316), 1515 Holcombe Blvd., Unit 0097, Houston, TX, 77030, USA
| | - Maria A Cortez
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ailin Li
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Anderson Central (Y2.5316), 1515 Holcombe Blvd., Unit 0097, Houston, TX, 77030, USA.
| |
Collapse
|
25
|
Orton A, Wright J, Buchmann L, Randall L, Hitchcock YJ. A Case of Complete Abscopal Response in High-Grade Pleiomorphic Sarcoma Treated with Radiotherapy Alone. Cureus 2016; 8:e821. [PMID: 27843739 PMCID: PMC5101400 DOI: 10.7759/cureus.821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background: “Abscopal response” refers to the spontaneous involution of untreated metastatic disease following local primary tumor-directed therapy. We report a case of an abscopal response of untreated lung metastasis in a man with pleomorphic sarcoma of the head and neck treated with hypofractionated radiotherapy. Methods: An inoperable pleomorphic sarcoma of the postauricular soft tissue was treated with 40 Gy of radiation in eight fractions. Untreated disease in the lungs was followed with CT scans. Results: At the two-month post-treatment follow-up, clinical exam and restaging CT demonstrated complete primary tumor involution. Additionally, CT chest images showed a dramatic disease response in the untreated pulmonary disease, which progressed to complete and persistent clinical response at one-year post-treatment follow-up. Conclusions: We report the first described case of a complete abscopal resolution of untreated lung metastases in a patient with a primary pleomorphic sarcoma of the head and neck treated with hypofractionated radiotherapy.
Collapse
Affiliation(s)
- Andrew Orton
- Radiation Oncology, University of Utah School of Medicine, Huntsman Cancer Institute
| | - Jennifer Wright
- Medical Oncology, University of Utah School of Medicine, Huntsman Cancer Institute
| | - Luke Buchmann
- Department of Otolaryngology, University of Utah School of Medicine, Huntsman Cancer Institute
| | - Lor Randall
- Department of Orthopaedic Surgery/Sarcoma, University of Utah School of Medicine, Huntsman Cancer Institute
| | - Ying J Hitchcock
- Radiation Oncology, University of Utah School of Medicine, Huntsman Cancer Institute
| |
Collapse
|
26
|
Silvestri I, Cattarino S, Giantulli S, Nazzari C, Collalti G, Sciarra A. A Perspective of Immunotherapy for Prostate Cancer. Cancers (Basel) 2016; 8:cancers8070064. [PMID: 27399780 PMCID: PMC4963806 DOI: 10.3390/cancers8070064] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/27/2016] [Accepted: 07/01/2016] [Indexed: 12/24/2022] Open
Abstract
In cancer patients, the immune system is often altered with an excess of inhibitory factors, such as immunosuppressive cytokines, produced by regulatory T cells (Treg) or myeloid-derived suppressor cells (MDSC). The manipulation of the immune system has emerged as one of new promising therapies for cancer treatment, and also represents an attractive strategy to control prostate cancer (PCa). Therapeutic cancer vaccines and immune checkpoint inhibitors have been the most investigated in clinical trials. Many trials are ongoing to define the effects of immune therapy with established treatments: androgen deprivation therapy (ADT) and chemotherapy (CT) or radiotherapy (RT). This article discusses some of these approaches in the context of future treatments for PCa.
Collapse
Affiliation(s)
- Ida Silvestri
- Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy.
| | - Susanna Cattarino
- Department of Urology, Sapienza University of Rome, Rome 00161, Italy.
| | - Sabrina Giantulli
- Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy.
| | - Cristina Nazzari
- Department of Public Health hand Infectious Diseases, "Sapienza" University of Rome, Rome 00185, Italy.
| | - Giulia Collalti
- Medicine of Systems, Rheumatology, Allergology and Clinical Immunology, Translational Medicine of the University Tor Vergata, Rome 00133, Italy.
| | | |
Collapse
|
27
|
Schummer V, Flindt S, Hinz T. [Tumor vaccines and peptide-loaded dendritic cells (DCs)]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2016; 58:1254-8. [PMID: 26349562 DOI: 10.1007/s00103-015-2242-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Vaccines are usually intended to prevent the spread of infectious diseases. Due to increasing knowledge about the immune system and its role in malignant disease, the development of therapeutic vaccines, which are intended to treat established tumors, has begun. For the induction of therapeutic immunity towards tumors, either tumor-specific or overexpressed antigens can be used. Tumor-specific antigens are mainly or exclusively expressed in tumors. It is assumed that they can thus be more easily recognized by the immune system than overexpressed antigens. Overexpressed antigens are expressed in both tumors and healthy tissues and therefore bear the risk of autoimmunity. In this review article, we discuss different approaches of therapeutic cancer vaccinations based on cells and on other drug substances. Moreover, we address the possibilities of authorizing cancer vaccines in the EU and in Germany.
Collapse
Affiliation(s)
- Verena Schummer
- Paul-Ehrlich-Institut, Fachgebiet Therapeutische Impfstoffe, Paul-Ehrlich-Straße 51-59, 63225, Langen, Deutschland
| | - Sven Flindt
- Paul-Ehrlich-Institut, Fachgebiet Therapeutische Impfstoffe, Paul-Ehrlich-Straße 51-59, 63225, Langen, Deutschland
| | - Thomas Hinz
- Paul-Ehrlich-Institut, Fachgebiet Therapeutische Impfstoffe, Paul-Ehrlich-Straße 51-59, 63225, Langen, Deutschland.
| |
Collapse
|
28
|
Modena A, Ciccarese C, Iacovelli R, Brunelli M, Montironi R, Fiorentino M, Tortora G, Massari F. Immune Checkpoint Inhibitors and Prostate Cancer: A New Frontier? Oncol Rev 2016; 10:293. [PMID: 27471580 PMCID: PMC4943092 DOI: 10.4081/oncol.2016.293] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 03/23/2016] [Indexed: 12/13/2022] Open
Abstract
Despite recent advances in the treatment of metastatic castration-resistant prostate cancer (mCRPC), agents that provide durable disease control and long-term survival are still needed. It is a fact that a tumor-induced immunosuppressive status (mediated by aberrant activation of inhibitory immune checkpoint pathways as a mechanism to evade host immune surveillance) plays a crucial role in the pathogenesis of cancer, including prostate cancer (PC), making CRPC patients suitable candidates for immunotherapy. Therefore, growing interest of anticancer research aims at blocking immune checkpoints (mainly targeting CTLA-4 and PD1/PD-L1 pathways) to restore and enhance cellular-mediated antitumor immunity and achieve durable tumor regression. In this review, we describe the current knowledge regarding the role of immune checkpoints in mediating PC progression, focusing on CTLA-4 and PD1 pathways. We also provide current clinical data available, an update on ongoing trials of immune checkpoint inhibitors in PC. Finally, we discuss the necessity to identify prognostic and predictive biomarkers of immune activity, and we analyze new immune checkpoints with a role as promising targets for PC therapy.
Collapse
Affiliation(s)
| | | | | | - Matteo Brunelli
- Department of Pathology and Diagnostic, University-Hospital of Verona , Verona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of Marche Region, School of Medicine , Ancona, Italy
| | - Michelangelo Fiorentino
- Pathology Service, Addarii Institute of Oncology, S. Orsola-Malpighi Hospital , Bologna, Italy
| | | | | |
Collapse
|
29
|
Long-term outcome of a phase II trial using immunomodulatory in situ gene therapy in combination with intensity-modulated radiotherapy with or without hormonal therapy in the treatment of prostate cancer. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s13566-015-0239-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
30
|
Cacan E, Greer SF, Garnett-Benson C. Radiation-induced modulation of immunogenic genes in tumor cells is regulated by both histone deacetylases and DNA methyltransferases. Int J Oncol 2015; 47:2264-75. [PMID: 26458736 DOI: 10.3892/ijo.2015.3192] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/16/2015] [Indexed: 11/06/2022] Open
Abstract
Radiation treatment is a pivotal therapy for several cancer types, including colorectal cancer. It has been shown that sublethal doses of radiation modulate gene expression, making tumor cells more susceptible to T-cell-mediated immune attack. We have recently shown that low dose radiation enhances expression of multiple death receptors (Fas, DR4 and DR5) and co-stimulatory molecules (4-1BBL and OX-40L) in colorectal cancer (CRC) cells; however, it is unclear how ionizing radiation (IR) enhances expression of these molecules mechanistically. In the present study, we elucidate the molecular mechanisms by which radiation controls expression of these molecules in CRC. Here we report that, enhanced expression of these genes following radiation treatment of CRC cells is due, in part, to changes in DNA methylation and histone acetylation. We observed that radiation (5 Gy) significantly increased histone acetylation at the promoter regions of 4-1BBL, Fas and DR5 but not OX-40L. However, radiation did not induce changes in the global levels of acetylated histone H3 suggesting specificity of IR-induced changes. Furthermore, evaluation of epigenetic controlling enzymes revealed that IR did not alter overall cellular levels of HDACs (HDAC1, HDAC2 or HDAC3) or DNMTs (DNMT1, DNMT3a, or DNMT3b). Instead, radiation decreased binding of HDAC2 and HDAC3 at the promoter regions of Fas and 4-1BBL, respectively. Radiation also resulted in reduced DNMT1 at both the Fas and 4-1BBL promoter regions but not a control gene. We conclude that single dose radiation can influence the expression of immune response relevant genes in colorectal tumor cells by altering the binding of epigenetic enzymes, and modulating histone acetylation, at specific gene promoters.
Collapse
Affiliation(s)
- Ercan Cacan
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | - Susanna F Greer
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | | |
Collapse
|
31
|
Xu P, Li PJ, Guo K, He Y, Ma C, Jin Z, Zhang Y, Ge Y, Xu A, Zheng S, Li Y, Liu C, Huang P. Remarkable Pathologic Change in Advanced Prostate Cancer Patient Using Dendritic Cell-Cytokine-Induced Killer Combined Therapy: A Case Report. Clin Genitourin Cancer 2015; 13:e379-83. [PMID: 25979641 DOI: 10.1016/j.clgc.2015.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/13/2015] [Accepted: 04/17/2015] [Indexed: 12/30/2022]
Affiliation(s)
- Peng Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Peng Ju Li
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Kai Guo
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yanjie He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Chao Ma
- Department of Urology, Shenzhen Longhua New District Central Hospital, Shenzhen, People's Republic of China
| | - Zhong Jin
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yiming Zhang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yukun Ge
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Abai Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Shaobo Zheng
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Chunxiao Liu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Peng Huang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China.
| |
Collapse
|
32
|
Hojjat-Farsangi M. Novel and emerging targeted-based cancer therapy agents and methods. Tumour Biol 2015; 36:543-56. [PMID: 25663495 DOI: 10.1007/s13277-015-3184-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 01/27/2015] [Indexed: 02/06/2023] Open
Abstract
After several decades of uncovering the cancer features and following the improvement of therapeutic agents, however cancer remains as one of the major reasons of mortality. Chemotherapy is one of the main treatment options and has significantly improved the overall survival of cancer patients, but chemotherapeutic agents are highly toxic for normal cells. Therefore, there is a great unmet medical need to develop new therapeutic principles and agents. Targeted-based cancer therapy (TBCT) agents and methods have revolutionized the cancer treatment efficacy. Monoclonal antibodies (mAbs) and small molecule inhibitors (SMIs) are among the most effective agents of TBCT. These drugs have improved the prognosis and survival of cancer patients; however, the therapeutic resistance has subdued the effects. Several mechanisms lead to drug resistance such as mutations in the drug targets, activation of compensatory pathways, and intrinsic or acquired resistance of cancer stem cells. Therefore, new modalities, improving current generation of inhibitors and mAbs, and optimizing the combinational therapy regimens are necessary to decrease the current obstacles in front of TBCT. Moreover, the success of new TBCT agents such as mAbs, SMIs, and immunomodulatory agents has sparked further therapeutic modalities with novel targets to inhibit. Due to the lack of cumulative information describing different agents and methods of TBCT, this review focuses on the most important agents and methods of TBCT that are currently under investigation.
Collapse
Affiliation(s)
- Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene therapy Lab, Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute, SE-171 76, Stockholm, Sweden,
| |
Collapse
|