1
|
Tseng C, Chen CM, Hsieh YH, Lin CY, Chen JW, Hsiao PH, Fong YC, Wang PH, Chen PN, Lin RC. MTA2 knockdown suppresses human osteosarcoma metastasis by inhibiting uPA expression. Aging (Albany NY) 2024; 16:12239-12251. [PMID: 39248711 PMCID: PMC11424574 DOI: 10.18632/aging.206070] [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: 02/23/2024] [Accepted: 07/17/2024] [Indexed: 09/10/2024]
Abstract
The relationship between metastasis-associated protein 2 (MTA2) overexpression and tumor growth and metastasis has been extensively studied in a variety of tumor cells but not in human osteosarcoma cells. This study aims to elucidate the clinical significance, underlying molecular mechanisms, and biological functions of MTA2 in human osteosarcoma in vitro and in vivo. Our results show that MTA2 was elevated in osteosarcoma cell lines and osteosarcoma tissues and was associated with tumor stage and overall survival of osteosarcoma patients. Knockdown of MTA2 inhibited osteosarcoma cell migration and invasion by reducing the expression of urokinase-type plasminogen activator (uPA). Bioinformatic analysis demonstrated that high levels of uPA in human osteosarcoma tissues correlated positively with MTA2 expression. Furthermore, treatment with recombinant human uPA (Rh-uPA) caused significant restoration of OS cell migration and invasion in MTA2 knockdown osteosarcoma cells. We found that ERK1/2 depletion increased the expression of uPA, facilitating osteosarcoma cell migration and invasion. Finally, MTA2 depletion significantly reduced tumor metastasis and the formation of lung nodules in vivo. Overall, our study suggests that MTA2 knockdown suppresses osteosarcoma cell metastasis by decreasing uPA expression via ERK signaling. This finding provides new insight into potential treatment strategies against osteosarcoma metastasis by targeting MTA2.
Collapse
Affiliation(s)
- Chun Tseng
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Spine Center, China Medical University Hospital, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Chien-Min Chen
- Division of Neurosurgery, Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan
- Department of Leisure Industry Management, National Chin-Yi University of Technology, Taichung, Taiwan
- Department of Biomedical Sciences National Chung Cheng University, Chiayi, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chia-Yu Lin
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Spine Center, China Medical University Hospital, Taichung, Taiwan
| | - Jian-Wen Chen
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Spine Center, China Medical University Hospital, Taichung, Taiwan
| | - Pang-Hsuan Hsiao
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
| | - Yi-Chin Fong
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Spine Center, China Medical University Hospital, Taichung, Taiwan
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
| | - Pei-Han Wang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Pei-Ni Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Renn-Chia Lin
- Department of Orthopedics, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
2
|
Gómez-Aparicio MA, López-Campos F, Buchser D, Lazo A, Willisch P, Ocanto A, Sargos P, Shelan M, Couñago F. Is There an Opportunity to De-Escalate Treatments in Selected Patients with Metastatic Hormone-Sensitive Prostate Cancer? Cancers (Basel) 2024; 16:2331. [PMID: 39001393 PMCID: PMC11240449 DOI: 10.3390/cancers16132331] [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: 05/21/2024] [Revised: 06/16/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
The treatment landscape for metastatic hormone-sensitive prostate cancer continues to evolve, with systemic treatment being the mainstay of current treatment. Prognostic and predictive factors such as tumour volume and disease presentation have been studied to assess responses to different treatments. Intensification and de-escalation strategies arouse great interest, so several trials are being developed to further personalize the therapy in these populations. Is there an optimal sequence and a possible option to de-intensify treatment in selected patients with a favourable profile? This and other goals will be the subject of this review.
Collapse
Affiliation(s)
| | - Fernando López-Campos
- Department of Radiation Oncology, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Department of Radiation Oncology, Hospital Universitario San Francisco de Asis and Hospital Vithas La Milagrosa, GenesisCare, 28002 Madrid, Spain; (A.O.); (F.C.)
| | - David Buchser
- Department of Radiation Oncology, Hospital Universitario Cruces, 48903 Barakaldo, Spain;
| | - Antonio Lazo
- Department of Radiation Oncology, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain;
| | - Patricia Willisch
- Department of Radiation Oncology, Hospital Meixoeiro, 36214 Vigo, Spain;
| | - Abrahams Ocanto
- Department of Radiation Oncology, Hospital Universitario San Francisco de Asis and Hospital Vithas La Milagrosa, GenesisCare, 28002 Madrid, Spain; (A.O.); (F.C.)
| | - Paul Sargos
- Department of Radiation Oncology, Institut Bergonié, 33000 Bordeaux, France;
| | - Mohamed Shelan
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Switzerland;
| | - Felipe Couñago
- Department of Radiation Oncology, Hospital Universitario San Francisco de Asis and Hospital Vithas La Milagrosa, GenesisCare, 28002 Madrid, Spain; (A.O.); (F.C.)
| |
Collapse
|
3
|
Roy S, Sun Y, Morgan SC, Wallis CJD, King K, Zhou YM, D'souza LA, Azem O, Cueto-Marquez AE, Camden NB, Spratt DE, Kishan AU, Saad F, Malone S. Effect of Prior Local Therapy on Response to First-line Androgen Receptor Axis Targeted Therapy in Metastatic Castrate-resistant Prostate Cancer: A Secondary Analysis of the COU-AA-302 Trial. Eur Urol 2023; 83:571-579. [PMID: 36894488 DOI: 10.1016/j.eururo.2023.02.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/09/2023] [Accepted: 02/16/2023] [Indexed: 03/09/2023]
Abstract
BACKGROUND Men with localized prostate cancer are often treated with local therapy (LT). However, a proportion of these patients will eventually develop recurrence and progression requiring systemic therapy. Whether primary LT affects the response to this subsequent systemic treatment is unclear. OBJECTIVE We investigated whether the receipt of prior prostate-directed LT influenced the response to first-line systemic therapy and survival in docetaxel-naïve metastatic castrate-resistant prostate cancer (mCRPC) patients. DESIGN, SETTING, AND PARTICIPANTS This is an exploratory analysis of the COU-AA-302 trial, a multicentric double-blinded phase 3 randomized controlled trial in which mCRPC patients with no to mild symptoms were randomized to receive abiraterone plus prednisone or placebo plus prednisone. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS We compared the time-varying effects of first-line abiraterone in patients with and without prior LT using a Cox proportional hazard model. The cut points were chosen using grid search, and were 6 and 36 mo for radiographic progression-free survival (rPFS) and overall survival (OS), respectively. We also investigated whether there was any difference in treatment effect on score change (relative to baseline) in various patient-reported outcomes (measured by Functional Assessment of Cancer Therapy-Prostate [FACT-P]) over time depending on the receipt of prior LT. The adjusted association of prior LT with survival was determined using weighted Cox regression models. RESULTS AND LIMITATIONS Among 1053 eligible patients, 64% (n = 669) received prior LT. We did not find any statistically significant heterogeneity of time-dependent treatment effect from abiraterone on rPFS in patients with (hazard ratio [HR]: 0.36 [95% confidence interval: 0.27-0.49] at ≤6 mo; 0.64 [0.49-0.83] at >6 mo) or without (HR: 0.37 [0.26-0.55] at ≤6 mo; 0.72 [0.50-1.03] at >6 mo) prior LT. Similarly, there was no significant heterogeneity in time-dependent treatment effect on OS with (HR: 0.88 [0.71-1.10] at ≤36 mo; 0.76 [0.52-1.11] at >36 mo) or without (0.78 [0.60-1.01] at ≤36 mo; 0.55 [0.30-0.99] at >36 mo) prior LT. We did not find sufficient evidence of a difference in treatment effect from abiraterone on score change over time in prostate cancer subscale (interaction p = 0.4), trial outcome index (interaction p = 0.8), and FACT-P total score (interaction p = 0.6) depending on the receipt of prior LT. Receipt of prior LT was associated with a significant improvement in OS (average HR: 0.72 [0.59-0.89]). CONCLUSIONS This study demonstrates that the efficacy of first-line abiraterone and prednisone in docetaxel-naïve mCRPC do not vary significantly based on the receipt of prior prostate-directed LT. Further studies are needed to explore the plausible mechanisms of the association of prior LT with superior OS. PATIENT SUMMARY This secondary analysis of the COU-AA-302 trial suggests that survival benefits and temporal changes in quality of life with first-line abiraterone in docetaxel-naïve mCRPC do not differ significantly among patients who received versus those who did not receive prior prostate-directed local therapy.
Collapse
Affiliation(s)
- Soumyajit Roy
- Department of Radiation Oncology, Rush University Medical Center, Chicago, IL, USA.
| | - Yilun Sun
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Scott C Morgan
- Division of Radiation Oncology, The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa, ON, Canada
| | - Christopher J D Wallis
- Department of Urology, Mount Sinai Hospital and University Health Network, University of Toronto, Toronto, ON, Canada
| | - Kevin King
- Department of Radiation Oncology, Rush University Medical Center, Chicago, IL, USA
| | - Yu M Zhou
- Department of Radiation Oncology, Rush University Medical Center, Chicago, IL, USA
| | - Leah A D'souza
- Department of Radiation Oncology, Rush University Medical Center, Chicago, IL, USA
| | - Omar Azem
- Department of Radiation Oncology, Rush University Medical Center, Chicago, IL, USA
| | | | - Nathaniel B Camden
- Department of Radiation Oncology, Rush University Medical Center, Chicago, IL, USA
| | - Daniel E Spratt
- Department of Radiation Oncology, University Hospital Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Amar U Kishan
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - Fred Saad
- Department of Surgery, Université de Montréal, Montreal, QC, Canada
| | - Shawn Malone
- Division of Radiation Oncology, The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
4
|
Montuori E, Hyde CAC, Crea F, Golding J, Lauritano C. Marine Natural Products with Activities against Prostate Cancer: Recent Discoveries. Int J Mol Sci 2023; 24:1435. [PMID: 36674949 PMCID: PMC9865900 DOI: 10.3390/ijms24021435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
Prostate cancer is the most common cancer in men, with over 52,000 new cases diagnosed every year. Diagnostics and early treatment are potentially hindered by variations in screening protocols, still largely reliant on serum levels of acid phosphatase and prostate-specific antigen, with tumour diagnosis and grading relying on histopathological examination. Current treatment interventions vary in terms of efficacy, cost and severity of side effects, and relapse can be aggressive and resistant to the current standard of care. For these reasons, the scientific community is looking for new chemotherapeutic agents. This review reports compounds and extracts derived from marine organisms as a potential source of new drugs against prostate cancer. Whilst there are several marine-derived compounds against other cancers, such as multiple myeloma, leukemia, breast and lung cancer, already available in the market, the presently collated findings show how the marine environment can be considered to hold potential as a new drug source for prostate cancer, as well. This review presents information on compounds presently in clinical trials, as well as new compounds/extracts that may enter trials in the future. We summarise information regarding mechanisms of action and active concentrations.
Collapse
Affiliation(s)
- Eleonora Montuori
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Acton 55, 80133 Napoli, Italy
| | - Caroline A C Hyde
- Cancer Research Group, School of Life Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK
| | - Francesco Crea
- Cancer Research Group, School of Life Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK
| | - Jon Golding
- Cancer Research Group, School of Life Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK
| | - Chiara Lauritano
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Acton 55, 80133 Napoli, Italy
- Cancer Research Group, School of Life Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK
| |
Collapse
|
5
|
Cailleteau A, Sargos P, Saad F, Latorzeff I, Supiot S. Drug Intensification in Future Postoperative Radiotherapy Practice in Biochemically-Relapsing Prostate Cancer Patients. Front Oncol 2021; 11:780507. [PMID: 35004302 PMCID: PMC8739777 DOI: 10.3389/fonc.2021.780507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/30/2021] [Indexed: 12/26/2022] Open
Abstract
Although salvage prostate bed radiotherapy is highly effective in biochemically-relapsing prostate cancer patients following prostatectomy, relapses remain frequent and improvements are needed. Randomized phase 3 trials have shown the benefit of adding androgen-depriving therapy to irradiation, but not all patients benefit from this combination. Preclinical studies have shown that novel agents targeting the androgen receptor, DNA repair, PI3K/AKT/mTOR pathways, or the hypoxic microenvironment may help increase the response to prostate bed irradiation while minimizing potential side effects. This perspective review focuses on the most relevant molecules that may have an impact when combined with salvage radiotherapy, and underlines the strategies that need to be developed to increase the efficacy of salvage post-prostatectomy radiotherapy in prostate cancer patients.
Collapse
Affiliation(s)
- Axel Cailleteau
- Department of Radiation Oncology, Institut de Cancérologie de l’Ouest, Nantes Saint-Herblain, France
| | - Paul Sargos
- Department of Radiation Oncology, Institut Bergonié, Bordeaux, France
| | - Fred Saad
- Department of Urology, Université de Montréal, Montreal, QC, Canada
| | - Igor Latorzeff
- Department of Radiation Oncology, Oncorad Clinique Pasteur, Toulouse, France
| | - Stéphane Supiot
- Department of Radiation Oncology, Institut de Cancérologie de l’Ouest, Nantes Saint-Herblain, France
- University of Nantes, CRCINA (CNRS, Inserm), Nantes, France
| |
Collapse
|
6
|
Kurganovs N, Wang H, Huang X, Ignatchenko V, Macklin A, Khan S, Downes MR, Boutros PC, Liu SK, Kislinger T. A proteomic investigation of isogenic radiation resistant prostate cancer cell lines. Proteomics Clin Appl 2021; 15:e2100037. [PMID: 34152685 PMCID: PMC8448965 DOI: 10.1002/prca.202100037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 06/19/2021] [Indexed: 11/09/2022]
Abstract
To model the problem of radiation resistance in prostate cancer, cell lines mimicking a clinical course of conventionally fractionated or hypofractionated radiotherapy have been generated. Proteomic analysis of radiation resistant and radiosensitive DU145 prostate cancer cells detected 4410 proteins. Over 400 proteins were differentially expressed across both radiation resistant cell lines and pathway analysis revealed enrichment in epithelial to mesenchymal transition, glycolysis and hypoxia. From the radiation resistant protein candidates, the cell surface protein CD44 was identified in the glycolysis and epithelial to mesenchymal transition pathways and may serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Natalie Kurganovs
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoCanada
| | - Hanzhi Wang
- Sunnybrook Research InstituteSunnybrook Health Sciences CentreTorontoCanada
- Department of Medical BiophysicsUniversity of TorontoTorontoCanada
| | - Xiaoyong Huang
- Sunnybrook Research InstituteSunnybrook Health Sciences CentreTorontoCanada
| | | | - Andrew Macklin
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoCanada
| | - Shahbaz Khan
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoCanada
| | - Michelle R. Downes
- Division of Anatomic PathologyLaboratory Medicine and Molecular DiagnosticsSunnybrook Health Sciences CentreTorontoCanada
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoCanada
| | - Paul C. Boutros
- Departments of Human Genetics & UrologyJonsson Comprehensive Cancer CenterLos AngelesUSA
- Institute for Precision HealthUniversity of CaliforniaLos AngelesUSA
| | - Stanley K. Liu
- Sunnybrook Research InstituteSunnybrook Health Sciences CentreTorontoCanada
| | - Thomas Kislinger
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoCanada
- Department of Medical BiophysicsUniversity of TorontoTorontoCanada
| |
Collapse
|
7
|
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
|
8
|
The Effect of Neoadjuvant Androgen Deprivation Therapy on Tumor Hypoxia in High-Grade Prostate Cancer: An 18F-MISO PET-MRI Study. Int J Radiat Oncol Biol Phys 2018; 102:1210-1218. [DOI: 10.1016/j.ijrobp.2018.02.170] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 02/16/2018] [Accepted: 02/28/2018] [Indexed: 12/16/2022]
|
9
|
Malla B, Aebersold DM, Dal Pra A. Protocol for serum exosomal miRNAs analysis in prostate cancer patients treated with radiotherapy. J Transl Med 2018; 16:223. [PMID: 30103771 PMCID: PMC6090775 DOI: 10.1186/s12967-018-1592-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/30/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Circulating exosomes from prostate cancer (PCa) patients undergoing radiotherapy are attractive candidate biomarkers for monitoring treatment response. Multiple workflows for isolation and content characterization of exosomes in biofluids have been attempted. We report a protocol to isolate and characterize exosomal miRNAs content and assess radiation-induced changes. METHODS In this pilot study, we performed targeted exosomal miRNA profiling of 25 serum samples obtained from PCa patients with intermediate- and high-risk disease treated with curative radiotherapy (RT), and controls. Post-treatment blood samples were collected at least 28 days after radiation therapy as a paired follow-up sample. The complete workflow consisted of two phases: I) filtration and polyethylene glycol salt precipitation phase which enriched particles below 200 nm in size followed by characterization using electron microscopy, and II) flow cytometry. Finally, miRNA expression analysis between untreated and treated patient samples was performed using RNA extraction kit, and qRT-PCR. RESULTS In our preliminary data, 1 ml of serum from PCa patients showed higher exosomal concentration (3.68E+10) compared to controls (6.07E+08). The overall expression of exosomes after RT was found to be higher compared to untreated samples; the median value changed from 3.68E+10 to 5.40E+10; p = 0.52. Using electron microscopy, we were able to visualize cup-shaped vesicles with morphology and size compatible with exosomes. The bead-based flow cytometry showed positivity for exosomal tetraspanins surface markers CD63 and CD9. All five miRNAs (hsa-let-7a-5p, hsa-miR-141-3p, hsa-miR-145-5p, hsa-miR-21-5p, hsa-miR-99b-5p) have been identified in exosomes. Despite overall changes in hsa-let-7a-5p expression after radiation, the difference was significant only in the high-risk group (p = 0.037). In addition, the radiation response to hsa-miR-21-5p was elevated in the high-risk group compared to the intermediate group (p = 0.036). CONCLUSIONS Herewith, we demonstrated a protocol for isolation of serum exosomes and exosomal miRNA amplification. The recovery of exosomal miRNAs and their differential expression after radiation treatment suggests promising biomarker potential that requires further investigation in larger patient cohorts.
Collapse
Affiliation(s)
- Bijaya Malla
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Daniel M. Aebersold
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Alan Dal Pra
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL USA
| |
Collapse
|
10
|
Hompland T, Hole KH, Ragnum HB, Aarnes EK, Vlatkovic L, Lie AK, Patzke S, Brennhovd B, Seierstad T, Lyng H. Combined MR Imaging of Oxygen Consumption and Supply Reveals Tumor Hypoxia and Aggressiveness in Prostate Cancer Patients. Cancer Res 2018; 78:4774-4785. [DOI: 10.1158/0008-5472.can-17-3806] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/14/2018] [Accepted: 06/20/2018] [Indexed: 11/16/2022]
|
11
|
Supiot S, Campion L, Pommier P, Dore M, Palpacuer C, Racadot S, Rio E, Milano GA, Mahier-Ait Oukhatar C, Carrie C. Combined abiraterone acetate plus prednisone, salvage prostate bed radiotherapy and LH-RH agonists (CARLHA-GEP12) in biochemically-relapsing prostate cancer patients following prostatectomy: A phase I study of the GETUG/GEP. Oncotarget 2018; 9:22147-22157. [PMID: 29774129 PMCID: PMC5955159 DOI: 10.18632/oncotarget.25189] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 04/04/2018] [Indexed: 12/20/2022] Open
Abstract
Background To establish the maximum tolerated dose of abiraterone acetate plus prednisone (AA) combined with salvage radiotherapy (SRT) and goserelin in a phase 1 study in men with rising PSA following radical prostatectomy. Methods AA was given during one month before SRT at 1000 mg PO once daily, then 750 mg (Dose Level 1, DL1) or 1000 mg (DL2) during 5 months combined with 6-months goserelin by injection on the first day of irradiation (scheme NEO) or one month before starting SRT (scheme CONCO). Results In scheme NEO at DL1, 2/9 patients did not achieve castration levels of testosterone. 4/9 patients (44%) presented with grade 3 liver enzyme elevation. In scheme CONCO testosterone dropped to undetectable levels. At DL1, 6 patients were recruited, with no dose limiting toxicities. At DL2, 2/3 patients presented with grade 3 liver enzyme elevation occurring during SRT. Conclusions When AA was administered without goserilin, only 78% achieved castration levels. AA combined with SRT and goserilin did not increase pelvic toxicity, but lead to an unsuspected high frequency of grade 3 liver toxicity. The phase II recommended dose of AA combined to goserelin and SRT is 750 mg.
Collapse
Affiliation(s)
- Stéphane Supiot
- Departments of Radiation Oncology and Biostatistics, Institut de Cancérologie de l'Ouest, Nantes, France
| | - Loic Campion
- Departments of Radiation Oncology and Biostatistics, Institut de Cancérologie de l'Ouest, Nantes, France
| | - Pascal Pommier
- Department of Radiation Oncology, Centre Léon Berard, Lyon, France
| | - Mélanie Dore
- Departments of Radiation Oncology and Biostatistics, Institut de Cancérologie de l'Ouest, Nantes, France
| | - Clément Palpacuer
- Departments of Radiation Oncology and Biostatistics, Institut de Cancérologie de l'Ouest, Nantes, France
| | - Séverine Racadot
- Department of Radiation Oncology, Centre Léon Berard, Lyon, France
| | - Emmanuel Rio
- Departments of Radiation Oncology and Biostatistics, Institut de Cancérologie de l'Ouest, Nantes, France
| | - Gérard A Milano
- Laboratoire d'Oncopharmacologie, Centre Antoine-Lacassagne, Nice, France
| | | | - Christian Carrie
- Department of Radiation Oncology, Centre Léon Berard, Lyon, France
| |
Collapse
|
12
|
Feutren T, Herrera FG. Prostate irradiation with focal dose escalation to the intraprostatic dominant nodule: a systematic review. Prostate Int 2018; 6:75-87. [PMID: 30140656 PMCID: PMC6104294 DOI: 10.1016/j.prnil.2018.03.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/10/2018] [Accepted: 03/21/2018] [Indexed: 12/22/2022] Open
Abstract
Radiation therapy (RT) is a curative treatment option for localized prostate cancer. Prostate irradiation with focal dose escalation to the intraprostatic dominant nodule (IDN) is an emerging treatment option that involves the prophylactic irradiation of the whole prostate while increasing RT doses to the visible prostatic tumor. Because of the lack of large multicentre trials, a systematic review was performed in an attempt to get an overview on the feasibility and efficacy of focal dose escalation to the IDN. A bibliographic search for articles in English, which were listed in MEDLINE from 2000 to 2016 to identify publications on RT with focal directed boost to the IDN, was performed. The review was completed following the Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines. Twenty-two articles describing 1,378 patients treated with RT using focal boost were identified and fulfilled the selection criteria. Intensity-modulated radiation therapy (IMRT) was used in 720 patients (52.3%), volumetric modulated arc therapy was used in 45 patients (3.3%), stereotactic body radiation therapy (SBRT) in 113 patients (8.2%), and low–dose rate and high–dose rate brachytherapy (BT) were used in 305 patients (22.1%) and 195 patients (14.1%), respectively. Use of androgen deprivation therapy varied substantially among series. Biochemical disease-free survival at 5 years was reported for a cohort of 812 (58.9%) patients. The combined median biochemical disease-free survival for this group of patients was 85% (range: 78.8–100%; 95% confidence interval: 77.1–82.7%). The average occurrence of grade III or worse gastrointestinal and genitourinary late toxicity was, respectively, 2.5% and 3.1% for intensity-modulated RT boost, 10% and 6% for stereotactic body RT, 6% and 2% for low–dose rate BT, and 4% and 4.3% for high–dose rate BT. This review shows encouraging results for focal dose escalation to the IDN with acceptable short- to medium-term side effects and biochemical disease control rates. However, owing to the heterogeneity of patient population and the short follow-up, the results should be interpreted with caution. Considering that the clinical endpoint in the studies was biochemical recurrence, the use and duration of androgen deprivation therapy administration should be carefully considered before driving definitive conclusions. Randomized trials with long-term follow-up are needed before this technique can be generally recommended.
Collapse
Affiliation(s)
- Thomas Feutren
- Department of Radiotherapy, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Current Position Department of Radiotherapy, Institut de Cancérologie de Lorraine, Nancy, France
| | - Fernanda G. Herrera
- Department of Radiotherapy, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Corresponding author. Rue du Bugnon 46, 1011, Lausanne, Switzerland.
| |
Collapse
|
13
|
Ding M, Van der Kwast TH, Vellanki RN, Foltz WD, McKee TD, Sonenberg N, Pandolfi PP, Koritzinsky M, Wouters BG. The mTOR Targets 4E-BP1/2 Restrain Tumor Growth and Promote Hypoxia Tolerance in PTEN-driven Prostate Cancer. Mol Cancer Res 2018; 16:682-695. [PMID: 29453322 DOI: 10.1158/1541-7786.mcr-17-0696] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 01/11/2018] [Accepted: 01/23/2018] [Indexed: 11/16/2022]
Abstract
The mTOR signaling pathway is a central regulator of protein synthesis and cellular metabolism in response to the availability of energy, nutrients, oxygen, and growth factors. mTOR activation leads to phosphorylation of multiple downstream targets including the eukaryotic initiation factor 4E (eIF4E) binding proteins-1 and -2 (EIF4EBP1/4E-BP1 and EIF4EBP2/4E-BP2). These binding proteins inhibit protein synthesis, but are inactivated by mTOR to stimulate cell growth and metabolism. However, the role of these proteins in the context of aberrant activation of mTOR, which occurs frequently in cancers through loss of PTEN or mutational activation of the PI3K/AKT pathway, is unclear. Here, even under conditions of aberrant mTOR activation, hypoxia causes dephosphorylation of 4E-BP1/4E-BP2 and increases their association with eIF4E to suppress translation. This is essential for hypoxia tolerance as knockdown of 4E-BP1 and 4E-BP2 decreases proliferation under hypoxia and increases hypoxia-induced cell death. In addition, genetic deletion of 4E-BP1 and 4E-BP2 significantly accelerates all phases of cancer development in the context of PTEN loss-driven prostate cancer in mice despite potent PI3K/AKT and mTOR activation. However, even with a more rapid onset, tumors that establish in the absence of 4E-BP1 and 4E-BP2 have reduced levels of tumor hypoxia and show increased cell death within hypoxic tumor regions. Together, these data demonstrate that 4E-BP1 and 4E-BP2 act as essential metabolic breaks even in the context of aberrant mTOR activation and that they are essential for the creation of hypoxia-tolerant cells in prostate cancer. Mol Cancer Res; 16(4); 682-95. ©2018 AACR.
Collapse
Affiliation(s)
- Mei Ding
- Princess Margaret Cancer Centre and Campbell Family Institute for Cancer Research, University Health Network, Toronto, Ontario, Canada
| | | | - Ravi N Vellanki
- Princess Margaret Cancer Centre and Campbell Family Institute for Cancer Research, University Health Network, Toronto, Ontario, Canada
| | - Warren D Foltz
- Radiation Medicine Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Trevor D McKee
- Princess Margaret Cancer Centre and Campbell Family Institute for Cancer Research, University Health Network, Toronto, Ontario, Canada.,Radiation Medicine Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Nahum Sonenberg
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Pier P Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Marianne Koritzinsky
- Princess Margaret Cancer Centre and Campbell Family Institute for Cancer Research, University Health Network, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Bradly G Wouters
- Princess Margaret Cancer Centre and Campbell Family Institute for Cancer Research, University Health Network, Toronto, Ontario, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.,Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Evaluation of tumor hypoxia prior to radiotherapy in intermediate-risk prostate cancer using 18F-fluoromisonidazole PET/CT: a pilot study. Oncotarget 2018. [PMID: 29515786 PMCID: PMC5839367 DOI: 10.18632/oncotarget.24234] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose Hypoxia is a major factor in prostate cancer aggressiveness and radioresistance. Predicting which patients might be bad candidates for radiotherapy may help better personalize treatment decisions in intermediate-risk prostate cancer patients. We assessed spatial distribution of 18F-Misonidazole (FMISO) PET/CT uptake in the prostate prior to radiotherapy treatment. Materials and Methods Intermediate-risk prostate cancer patients about to receive high-dose (>74 Gy) radiotherapy to the prostate without hormonal treatment were prospectively recruited between 9/2012 and 10/2014. Prior to radiotherapy, all patients underwent a FMISO PET/CT as well as a MRI and 18F-choline-PET. 18F-choline and FMISO-positive volumes were semi-automatically determined using the fuzzy locally adaptive Bayesian (FLAB) method. In FMISO-positive patients, a dynamic analysis of early tumor uptake was performed. Group differences were assessed using the Wilcoxon signed rank test. Parameters were correlated using Spearman rank correlation. Results Of 27 patients (median age 76) recruited to the study, 7 and 9 patients were considered positive at 2.5h and 3.5h FMISO PET/CT respectively. Median SUVmax and SUVmax tumor to muscle (T/M) ratio were respectively 3.4 and 3.6 at 2.5h, and 3.2 and 4.4 at 3.5h. The median FMISO-positive volume was 1.1 ml. Conclusions This is the first study regarding hypoxia imaging using FMISO in prostate cancer showing that a small FMISO-positive volume was detected in one third of intermediate-risk prostate cancer patients.
Collapse
|
15
|
Chaiswing L, Weiss HL, Jayswal RD, St. Clair DK, Kyprianou N. Profiles of Radioresistance Mechanisms in Prostate Cancer. Crit Rev Oncog 2018; 23:39-67. [PMID: 29953367 PMCID: PMC6231577 DOI: 10.1615/critrevoncog.2018025946] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Radiation therapy (RT) is commonly used for the treatment of localized prostate cancer (PCa). However, cancer cells often develop resistance to radiation through unknown mechanisms and pose an intractable challenge. Radiation resistance is highly unpredictable, rendering the treatment less effective in many patients and frequently causing metastasis and cancer recurrence. Understanding the molecular events that cause radioresistance in PCa will enable us to develop adjuvant treatments for enhancing the efficacy of RT. Radioresistant PCa depends on the elevated DNA repair system and the intracellular levels of reactive oxygen species (ROS) to proliferate, self-renew, and scavenge anti-cancer regimens, whereas the elevated heat shock protein 90 (HSP90) and the epithelial-mesenchymal transition (EMT) enable radioresistant PCa cells to metastasize after exposure to radiation. The up-regulation of the DNA repairing system, ROS, HSP90, and EMT effectors has been studied extensively, but not targeted by adjuvant therapy of radioresistant PCa. Here, we emphasize the effects of ionizing radiation and the mechanisms driving the emergence of radioresistant PCa. We also address the markers of radioresistance, the gene signatures for the predictive response to radiotherapy, and novel therapeutic platforms for targeting radioresistant PCa. This review provides significant insights into enhancing the current knowledge and the understanding toward optimization of these markers for the treatment of radioresistant PCa.
Collapse
Affiliation(s)
| | - Heidi L. Weiss
- The Markey Biostatistics and Bioinformatics Shared Resource Facility
| | - Rani D. Jayswal
- The Markey Biostatistics and Bioinformatics Shared Resource Facility
| | | | - Natasha Kyprianou
- Department of Toxicology and Cancer Biology
- Department of Urology
- Department of Biochemistry, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
16
|
King AR, Corso CD, Chen EM, Song E, Bongiorni P, Chen Z, Sundaram RK, Bindra RS, Saltzman WM. Local DNA Repair Inhibition for Sustained Radiosensitization of High-Grade Gliomas. Mol Cancer Ther 2017; 16:1456-1469. [PMID: 28566437 DOI: 10.1158/1535-7163.mct-16-0788] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 04/14/2017] [Accepted: 05/16/2017] [Indexed: 11/16/2022]
Abstract
High-grade gliomas, such as glioblastoma (GBM) and diffuse intrinsic pontine glioma (DIPG), are characterized by an aggressive phenotype with nearly universal local disease progression despite multimodal treatment, which typically includes chemotherapy, radiotherapy, and possibly surgery. Radiosensitizers that have improved the effects of radiotherapy for extracranial tumors have been ineffective for the treatment of GBM and DIPG, in part due to poor blood-brain barrier penetration and rapid intracranial clearance of small molecules. Here, we demonstrate that nanoparticles can provide sustained drug release and minimal toxicity. When administered locally, these nanoparticles conferred radiosensitization in vitro and improved survival in rats with intracranial gliomas when delivered concurrently with a 5-day course of fractionated radiotherapy. Compared with previous work using locally delivered radiosensitizers and cranial radiation, our approach, based on the rational selection of agents and a clinically relevant radiation dosing schedule, produces the strongest synergistic effects between chemo- and radiotherapy approaches to the treatment of high-grade gliomas. Mol Cancer Ther; 16(8); 1456-69. ©2017 AACR.
Collapse
Affiliation(s)
- Amanda R King
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Christopher D Corso
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Evan M Chen
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Eric Song
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Paul Bongiorni
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Zhe Chen
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Ranjini K Sundaram
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Ranjit S Bindra
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut. .,Department of Experimental Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut.
| |
Collapse
|
17
|
Malla B, Zaugg K, Vassella E, Aebersold DM, Dal Pra A. Exosomes and Exosomal MicroRNAs in Prostate Cancer Radiation Therapy. Int J Radiat Oncol Biol Phys 2017; 98:982-995. [PMID: 28721912 DOI: 10.1016/j.ijrobp.2017.03.031] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/14/2017] [Accepted: 03/20/2017] [Indexed: 12/11/2022]
Abstract
Despite current risk stratification systems using traditional clinicopathologic factors, many localized and locally advanced prostate cancers fail radical treatment (ie, radical prostatectomy, radiation therapy with or without androgen deprivation therapy). Therefore, a pressing need exists for enhanced methods of disease stratification through novel prognostic and predictive tools that can reliably be applied in clinical practice. Exosomes are 50- to 150-nm small vesicles released by cancer cells that reflect the genetic and nongenetic materials of parent cancer cells. Cancer cells can contain distinct sets of microRNA profiles, the expression of which can change owing to stress such as radiation therapy. These alterations or distinctions in contents allow exosomes to be used as prognostic and/or predictive biomarkers and to monitor the treatment response. Additionally, microRNAs have been shown to influence multiple processes in prostate tumorigenesis, including cell proliferation, induction of apoptosis, migration, oncogene inhibition, and radioresistance. Thus, comparative exosomal microRNA profiling at different levels could help portray tumor aggressiveness and response to radiation therapy. Although technical challenges persist in exosome isolation and characterization, recent improvements in microRNA profiling have evolved toward in-depth analyses of the exosomal cargo and its functions. We have reviewed the role of exosomes and exosomal microRNAs in biologic processes of prostate cancer progression and radiation therapy response, with a particular focus on the development of clinical assays for treatment personalization.
Collapse
Affiliation(s)
- Bijaya Malla
- Department of Radiation Oncology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Kathrin Zaugg
- Department of Radiation Oncology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Erik Vassella
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Daniel M Aebersold
- Department of Radiation Oncology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Alan Dal Pra
- Department of Radiation Oncology, Bern University Hospital, Inselspital, Bern, Switzerland.
| |
Collapse
|
18
|
Gonnissen A, Isebaert S, McKee CM, Dok R, Haustermans K, Muschel RJ. The hedgehog inhibitor GANT61 sensitizes prostate cancer cells to ionizing radiation both in vitro and in vivo. Oncotarget 2016; 7:84286-84298. [PMID: 27713179 PMCID: PMC5356662 DOI: 10.18632/oncotarget.12483] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/28/2016] [Indexed: 02/07/2023] Open
Abstract
Limited data exists regarding the combination of Hedgehog signaling (Hh) inhibition and radiotherapy, even though there are several indications that this might be a promising treatment strategy. In this study, we evaluated the combination of two Hh inhibitors, the SMO inhibitor GDC-0449 and the GLI inhibitor GANT61 with radiotherapy in different prostate cancer (PCa) models. In vitro, GANT61 was able to sensitize 22Rv1 PCa cells but not PC3 and DU145 PCa cells. The lack of radiosensitization in the latter cell lines was shown to be dependent on the presence of mutated p53. Introduction of WT p53 into PC3 cells resulted in radiosensization following GANT61 treatment, suggesting that the p53 transcription factor plays an important role in the GANT61-induced radiosensitization in vitro. Targeting at the level of SMO (GDC-0449) did not show cytotoxicity or synergy with radiation. Furthermore, we confirmed the radiosensitization effect of GANT61 in two in vivo xenograft PCa models. The decrease in tumor growth was associated with decreased proliferation and increased apoptosis. In conclusion, we provide evidence that GANT61 in combination with radiation treatment might represent a promising therapeutic strategy for enhancing the radiation response of PCa patients.
Collapse
Affiliation(s)
- Annelies Gonnissen
- KU Leuven, University of Leuven, Department of Oncology, Experimental Radiotherapy; University Hospitals Leuven, Radiation Oncology, Leuven, Belgium
| | - Sofie Isebaert
- KU Leuven, University of Leuven, Department of Oncology, Experimental Radiotherapy; University Hospitals Leuven, Radiation Oncology, Leuven, Belgium
| | - Chad M McKee
- University of Oxford, Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, Oxford, UK
| | - Rüveyda Dok
- KU Leuven, University of Leuven, Department of Oncology, Experimental Radiotherapy; University Hospitals Leuven, Radiation Oncology, Leuven, Belgium
| | - Karin Haustermans
- KU Leuven, University of Leuven, Department of Oncology, Experimental Radiotherapy; University Hospitals Leuven, Radiation Oncology, Leuven, Belgium
| | - Ruth J Muschel
- University of Oxford, Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, Oxford, UK
| |
Collapse
|
19
|
Herrera FG, Berthold DR. Radiation Therapy after Radical Prostatectomy: Implications for Clinicians. Front Oncol 2016; 6:117. [PMID: 27242957 PMCID: PMC4860423 DOI: 10.3389/fonc.2016.00117] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 04/25/2016] [Indexed: 12/11/2022] Open
Abstract
Depending on the pathological findings, up to 60% of prostate cancer patients who undergo radical prostatectomy (RP) will develop biochemical relapse and require further local treatment. Radiotherapy (RT) immediately after RP may potentially eradicate any residual localized microscopic disease in the prostate bed, and it is associated with improved biochemical, clinical progression-free survival, and overall survival in patients with high-risk pathological features according to published randomized trials. Offering immediate adjuvant RT to all men with high-risk pathological factors we are over-treating around 50% of patients who would anyway be cancer-free, exposing them to unnecessary toxicity and adding costs to the health-care system. The current dilemma is, thus, whether to deliver adjuvant immediate RT solely on the basis of high-risk pathology, but in the absence of measurable prostate-specific antigen, or whether early salvage radiotherapy would yield equivalent outcomes. Randomized trials are ongoing to definitely answer this question. Retrospective analyses suggest that there is a dose–response favoring doses >70 Gy to the prostate bed. The evidence regarding the role of androgen deprivation therapy is emerging, and ongoing randomized trials are underway.
Collapse
Affiliation(s)
- Fernanda G Herrera
- Radiation Oncology Services, Department of Oncology, Lausanne University Hospital , Lausanne , Switzerland
| | - Dominik R Berthold
- Medical Oncology Services, Department of Oncology, Lausanne University Hospital , Lausanne , Switzerland
| |
Collapse
|
20
|
Prostate cancer radiation therapy: A physician’s perspective. Phys Med 2016; 32:438-45. [DOI: 10.1016/j.ejmp.2016.02.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/27/2016] [Accepted: 02/17/2016] [Indexed: 02/07/2023] Open
|