1
|
Febronio EM, Secaf ADF, Chahud F, Elias J, Reis RB, Muglia VF. Pilot Study Examining the Use of DCE MRI With Pharmacokinetic Analysis to Evaluate Hypoxia in Prostate Cancer. J Comput Assist Tomogr 2025:00004728-990000000-00409. [PMID: 40263980 DOI: 10.1097/rct.0000000000001707] [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: 06/25/2024] [Accepted: 10/23/2024] [Indexed: 04/24/2025]
Abstract
PURPOSE This study aimed to investigate the association between tumor hypoxia, assessed through anti-HIF (hypoxia-inducible factor) staining, and aggressiveness in prostate cancer using a pharmacokinetic model, particularly those derived from the Tofts model, in predicting tumor aggressiveness. MATERIAL AND METHODS From January 2019 to April 2021, we conducted a retrospective search of patients with confirmed prostate cancer and a previous magnetic resonance imaging (MRI) examination. After exclusions, a total of 57 consecutive patients were enrolled. Patient data, including demographic, laboratory, and pathologic variables, were collected. MRI acquisition followed PI-RADS guidelines, encompassing T2-weighted, diffusion-weighted imaging, and dynamic contrast-enhanced imaging. An experienced abdominal radiologist conducted both morphologic and quantitative MRI analyses, evaluating parameters such as lesion size, apparent diffusion coefficient values, and the Tofts pharmacokinetics (TF) model. The histopathologic analysis included the International Society of Uropathology (ISUP) score and hypoxia marker immunohistochemistry. RESULTS There were no significant demographic and imaging differences between hypoxic and nonhypoxic tumors, except for elevated prostate-specific antigen levels in the latter and decreased normalized apparent diffusion coefficient in the former. Morphologic assessments revealed larger lesions in the hypoxia group. While Ktrans showed a positive association with hypoxia, it did not independently predict high-risk lesions. CONCLUSIONS Our results suggest that pharmacokinetic analysis by the Tofts model was associated with tumors with hypoxia. However, this parameter was not an independent predictor of more aggressive tumors. Further studies, with a larger number of patients, multi-institutional and prospective, are needed to verify this possible association.
Collapse
Affiliation(s)
| | | | | | - Jorge Elias
- Department of Imaging, Oncology and Hematology
| | - Rodolfo B Reis
- Department of Surgery and Anatomy, Urology Division, Ribeirao Preto School of Medicin, University of Sao Paulo, Sao Paulo, Brazil
| | | |
Collapse
|
2
|
Elming PB, Busk M, Wittenborn TR, Bussink J, Horsman MR, Lønbro S. The effect of single bout and prolonged aerobic exercise on tumor hypoxia in mice. J Appl Physiol (1985) 2023; 134:692-702. [PMID: 36727633 DOI: 10.1152/japplphysiol.00561.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 02/03/2023] Open
Abstract
The objectives of this study were to investigate 1) the effect of acute aerobic exercise on tumor hypoxia and blood perfusion, 2) the impact of exercise intensity, 3) the duration of the effect, and 4) the effect of prolonged training on tumor hypoxia and tumor growth. Female CDF1 mice were inoculated with the C3H mammary carcinoma either in the mammary fat pad or subcutaneously in the back. For experiments on the effect of different intensities in a single exercise bout, mice were randomized to 30-min treadmill running at low-, moderate-, or high-intensity speeds or no exercise. To investigate the prolonged effect on hypoxia and tumor growth, tumor-bearing mice were randomized to no exercise (CON) or daily 30-min high-intensity exercise averaging 2 wk (EX). Tumor hypoxic fraction was quantified using the hypoxia marker Pimonidazole. Initially, high-intensity exercise reduced tumor hypoxic fraction by 37% compared with CON [P = 0.046; 95% confidence interval (CI): 0.1; 10.3] in fat pad tumors. Low- and moderate-intensity exercises did not. Following experiments investigating the duration of the effect-as well as experiments in mice with back tumors-failed to show any exercise-induced changes in hypoxia. Interestingly, prolonged daily training significantly reduced hypoxic fraction by 60% (P = 0.002; 95% CI: 2.5; 10.1) compared with CON. Despite diverging findings on the acute effect of exercise on hypoxia, our data indicate that if exercise has a diminishing effect, high-intensity exercise is needed. Prolonged training reduced tumor hypoxic fraction-cautiously suggesting a potential clinical potential.NEW & NOTEWORTHY This study provides novel information on the effects of acute and chronic exercise on tumor hypoxia in mice. In contrast to the few related existing studies, diverging findings on tumor hypoxia after acute exercise were observed, suggesting that tumor model and location should be considered in future studies. Highly significant reductions in tumor hypoxia following chronic high-intensity exercise propose a future clinical potential but this should be investigated in patients.
Collapse
Affiliation(s)
| | - Morten Busk
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas Rea Wittenborn
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Michael R Horsman
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Simon Lønbro
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Section for Sports Science, Department of Public Health, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
3
|
Miller KJ, Henry I, Maylin Z, Smith C, Arunachalam E, Pandha H, Asim M. A compendium of Androgen Receptor Variant 7 target genes and their role in Castration Resistant Prostate Cancer. Front Oncol 2023; 13:1129140. [PMID: 36937454 PMCID: PMC10014620 DOI: 10.3389/fonc.2023.1129140] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/13/2023] [Indexed: 03/05/2023] Open
Abstract
Persistent androgen receptor (AR) signalling is the main driver of prostate cancer (PCa). Truncated isoforms of the AR called androgen receptor variants (AR-Vs) lacking the ligand binding domain often emerge during treatment resistance against AR pathway inhibitors such as Enzalutamide. This review discusses how AR-Vs drive a more aggressive form of PCa through the regulation of some of their target genes involved in oncogenic pathways, enabling disease progression. There is a pressing need for the development of a new generation of AR inhibitors which can repress the activity of both the full-length AR and AR-Vs, for which the knowledge of differentially expressed target genes will allow evaluation of inhibition efficacy. This review provides a detailed account of the most common variant, AR-V7, the AR-V7 regulated genes which have been experimentally validated, endeavours to understand their relevance in aggressive AR-V driven PCa and discusses the utility of the downstream protein products as potential drug targets for PCa treatment.
Collapse
Affiliation(s)
| | | | - Zoe Maylin
- *Correspondence: Zoe Maylin, ; Mohammad Asim,
| | | | | | | | | |
Collapse
|
4
|
Djurhuus SS, Schauer T, Simonsen C, Toft BG, Jensen ARD, Erler JT, Røder MA, Hojman P, Brasso K, Christensen JF. Effects of acute exercise training on tumor outcomes in men with localized prostate cancer: A randomized controlled trial. Physiol Rep 2022; 10:e15408. [PMID: 36199257 PMCID: PMC9535256 DOI: 10.14814/phy2.15408] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 05/28/2022] [Accepted: 07/14/2022] [Indexed: 06/16/2023] Open
Abstract
Postdiagnosis physical activity is associated with improved cancer outcomes, but biological mechanisms mediating anticancer effects remain unclear. Recent findings suggest that physiological adaptations to acute exercise comprise potential anticancer effects, but these remain poorly explored in clinical settings. The objective of this study was to explore the effects of a single exercise bout on tumor oxygenation and immune cell infiltration in patients with prostate cancer. Thirty patients with localized prostate cancer were randomized (2:1) to either one high-intensity interval training bout or no exercise on the day before radical prostatectomy. Immunohistochemical analyses were performed on prostatic tissue from surgery and assessed for tumor hypoxia, natural killer (NK) cell infiltration, and microvessel density (MVD). Acute systemic response in blood lymphocytes, epinephrine, norepinephrine, IL-6, tumor necrosis factor, cortisol, lactate, and glucose was also evaluated. We did not find between-group differences in tumor hypoxia (Mann-Whitney U test, U = 83.5, p = 0.604) or NK cell infiltration (U = 77.0, p = 0.328). Also, no significant correlation was found between MVD and tumor hypoxia or NK cell infiltration. One exercise bout is likely insufficient to modulate tumor hypoxia or NK cell infiltration. Future studies may elucidate if an accumulation of several exercise bouts can impact these outcomes (NCT03675529, www.clinicaltrials.gov).
Collapse
Affiliation(s)
| | - Tim Schauer
- Centre for Physical Activity ResearchCopenhagen University Hospital – RigshospitaletCopenhagenDenmark
| | - Casper Simonsen
- Centre for Physical Activity ResearchCopenhagen University Hospital – RigshospitaletCopenhagenDenmark
| | - Birgitte Grønkær Toft
- Department of PathologyCopenhagen University Hospital – RigshospitaletCopenhagenDenmark
| | | | - Janine Terra Erler
- Biotech Research and Innovation Centre (BRIC)University of Copenhagen (UCPH)CopenhagenDenmark
| | - Martin Andreas Røder
- Department of UrologyCopenhagen Prostate Cancer Center, Copenhagen University Hospital –RigshospitaletCopenhagenDenmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Pernille Hojman
- Centre for Physical Activity ResearchCopenhagen University Hospital – RigshospitaletCopenhagenDenmark
| | - Klaus Brasso
- Department of UrologyCopenhagen Prostate Cancer Center, Copenhagen University Hospital –RigshospitaletCopenhagenDenmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Jesper Frank Christensen
- Centre for Physical Activity ResearchCopenhagen University Hospital – RigshospitaletCopenhagenDenmark
- The Department of Sports Science and Clinical BiomechanicsFaculty of Health Sciences at the University of Southern DenmarkDenmark
- Digestive Disease CenterBispebjerg HospitalCopenhagenDenmark
| |
Collapse
|
5
|
Xie B, Meng Q, Yu H, Shen K, Cheng Y, Dong C, Zhou HB. Estrogen receptor β-targeted hypoxia-responsive near-infrared fluorescence probes for prostate cancer study. Eur J Med Chem 2022; 238:114506. [DOI: 10.1016/j.ejmech.2022.114506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 12/01/2022]
|
6
|
Abu el Maaty MA, Terzic J, Keime C, Rovito D, Lutzing R, Yanushko D, Parisotto M, Grelet E, Namer IJ, Lindner V, Laverny G, Metzger D. Hypoxia-mediated stabilization of HIF1A in prostatic intraepithelial neoplasia promotes cell plasticity and malignant progression. SCIENCE ADVANCES 2022; 8:eabo2295. [PMID: 35867798 PMCID: PMC9307253 DOI: 10.1126/sciadv.abo2295] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Prostate cancer (PCa) is a leading cause of cancer-related deaths. The slow evolution of precancerous lesions to malignant tumors provides a broad time frame for preventing PCa. To characterize prostatic intraepithelial neoplasia (PIN) progression, we conducted longitudinal studies on Pten(i)pe-/- mice that recapitulate prostate carcinogenesis in humans. We found that early PINs are hypoxic and that hypoxia-inducible factor 1 alpha (HIF1A) signaling is activated in luminal cells, thus enhancing malignant progression. Luminal HIF1A dampens immune surveillance and drives luminal plasticity, leading to the emergence of cells that overexpress Transglutaminase 2 (TGM2) and have impaired androgen signaling. Elevated TGM2 levels in patients with PCa are associated with shortened progression-free survival after prostatectomy. Last, we show that pharmacologically inhibiting HIF1A impairs cell proliferation and induces apoptosis in PINs. Therefore, our study demonstrates that HIF1A is a target for PCa prevention and that TGM2 is a promising prognostic biomarker of early relapse after prostatectomy.
Collapse
Affiliation(s)
- Mohamed A. Abu el Maaty
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Julie Terzic
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Céline Keime
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Daniela Rovito
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Régis Lutzing
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Darya Yanushko
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Maxime Parisotto
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Elise Grelet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Izzie Jacques Namer
- Université de Strasbourg, Strasbourg, France
- ICube, CNRS, UMR 7357, Strasbourg, France
| | - Véronique Lindner
- Département de Pathologie, Les Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Gilles Laverny
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
- Corresponding author. (D.M.); (G.L.)
| | - Daniel Metzger
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
- Corresponding author. (D.M.); (G.L.)
| |
Collapse
|
7
|
A prognostic hypoxia gene signature with low heterogeneity within the dominant tumour lesion in prostate cancer patients. Br J Cancer 2022; 127:321-328. [PMID: 35332267 PMCID: PMC9296675 DOI: 10.1038/s41416-022-01782-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/10/2022] [Accepted: 03/08/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Gene signatures measured in a biopsy have been proposed as hypoxia biomarkers in prostate cancer. We assessed a previously developed signature, and aimed to determine its relationship to hypoxia and its heterogeneity within the dominant (index) lesion of prostate cancer. METHODS The 32-gene signature was assessed from gene expression data of 141 biopsies from the index lesion of 94 patients treated with prostatectomy. A gene score calculated from the expression levels was applied in the analyses. Hypoxic fraction from pimonidazole immunostained whole-mount and biopsy sections was used as reference standard for hypoxia. RESULTS The gene score was correlated with pimonidazole-defined hypoxic fraction in whole-mount sections, and the two parameters showed almost equal association with clinical markers of tumour aggressiveness. Based on the gene score, incorrect classification according to hypoxic fraction in whole-mount sections was seen in one third of the patients. The incorrect classifications were apparently not due to intra-tumour heterogeneity, since the score had low heterogeneity compared to pimonidazole-defined hypoxic fraction in biopsies. The score showed prognostic significance in uni-and multivariate analysis in independent cohorts. CONCLUSIONS Our signature from the index lesion reflects tumour hypoxia and predicts prognosis in prostate cancer, independent of intra-tumour heterogeneity in pimonidazole-defined hypoxia.
Collapse
|
8
|
Immunofluorescence-Based Method to Assess Cancer Biomarker in the Hypoxic Region of the Tumor. Methods Mol Biol 2022; 2413:37-43. [PMID: 35044652 PMCID: PMC9116888 DOI: 10.1007/978-1-0716-1896-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The development of novel imaging technologies allows the analysis of the expression and spatial distribution of multiple markers simultaneously, providing necessary information about a cellular identity and the surrounding microenvironment. This chapter describes the utilization of immunofluorescence to identify such biomarkers in fixed tissue from prostate cancer (PCa) xenografts. One such marker detectable by immunofluorescence is pimonidazole, which has been utilized to locate areas of low oxygen (hypoxia). Pimonidazole, in combination with other biomarkers, could be utilized to identify "niches" in the microenvironment harboring more aggressive cells both within and outside hypoxic areas. Specifically, we describe the method to use pimonidazole for the identification of hypoxic regions in PCa xenograft tumors along with CPT1A (carnitine palmitoyltransferase 1A) expression, an indicator of β-oxidation. This approach could be useful to characterize various biomarkers in the complex hypoxic tumor microenvironment.
Collapse
|
9
|
Rios-Colon L, Kumar P, Kim S, Sharma M, Su Y, Kumar A, Singh S, Stocks N, Liu L, Joshi M, Schlaepfer IR, Kumar D, Deep G. Carnitine Palmitoyltransferase 1 Regulates Prostate Cancer Growth under Hypoxia. Cancers (Basel) 2021; 13:cancers13246302. [PMID: 34944922 PMCID: PMC8699124 DOI: 10.3390/cancers13246302] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/07/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Cancer cell survival in hypoxia areas, with low oxygen and food supply as well as abundant waste material, is critical to their aggressiveness and associated with disease relapse and mortality. Therefore, it is vital to understand the molecular regulators of cancer cell survival under these harsh physiological conditions. In the present study, we assessed the role of a mitochondrial protein carnitine palmitoyltransferase (CPT1A) in regulating prostate cancer (PCa) cell survival and proliferation under hypoxic conditions in both cell culture and animal models. The results showed that CPT1A expression in PCa cells is key to their survival and proliferation in the hypoxic tumor microenvironment. These results have high translational significance in improving cancer prognosis and therapy. Abstract Hypoxia and hypoxia-related biomarkers are the major determinants of prostate cancer (PCa) aggressiveness. Therefore, a better understanding of molecular players involved in PCa cell survival under hypoxia could offer novel therapeutic targets. We previously reported a central role of mitochondrial protein carnitine palmitoyltransferase (CPT1A) in PCa progression, but its role in regulating PCa survival under hypoxia remains unknown. Here, we employed PCa cells (22Rv1 and MDA-PCa-2b) with knockdown or overexpression of CPT1A and assessed their survival under hypoxia, both in cell culture and in vivo models. The results showed that CPT1A knockdown in PCa cells significantly reduced their viability, clonogenicity, and sphere formation under hypoxia, while its overexpression increased their proliferation, clonogenicity, and sphere formation. In nude mice, 22Rv1 xenografts with CPT1A knockdown grew significantly slower compared to vector control cells (~59% reduction in tumor volume at day 29). On the contrary, CPT1A-overexpressing 22Rv1 xenografts showed higher tumor growth compared to vector control cells (~58% higher tumor volume at day 40). Pathological analyses revealed lesser necrotic areas in CPT1A knockdown tumors and higher necrotic areas in CPT1A overexpressing tumors. Immunofluorescence analysis of tumors showed that CPT1A knockdown strongly compromised the hypoxic areas (pimonidazole+), while CPT1A overexpression resulted in more hypoxia areas with strong expression of proliferation biomarkers (Ki67 and cyclin D1). Finally, IHC analysis of tumors revealed a significant decrease in VEGF or VEGF-D expression but without significant changes in biomarkers associated with microvessel density. These results suggest that CPT1A regulates PCa survival in hypoxic conditions and might contribute to their aggressiveness.
Collapse
Affiliation(s)
- Leslimar Rios-Colon
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA;
| | - Pawan Kumar
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
- Division of Pathology, ICAR—Indian Veterinary Research Institute, Izatnagar 243122, India
| | - Susy Kim
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
| | - Mitu Sharma
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
| | - Yixin Su
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
| | - Ashish Kumar
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
| | - Sangeeta Singh
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
| | - Nalexus Stocks
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
| | - Liang Liu
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
- Center for Cancer Genomics and Precision Oncology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
| | - Molishree Joshi
- Functional Genomics Facility, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Isabel R. Schlaepfer
- Division of Medical Oncology, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA;
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA;
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
- Correspondence: ; Tel.: +336-716-9363
| |
Collapse
|
10
|
Wadsworth BJ, Decotret LR, Villamil C, Yapp D, Wilson D, Benard F, McKenzie M, Bennewith KL. Evaluation of 18F-EF5 for detection of hypoxia in localized adenocarcinoma of the prostate. Acta Oncol 2021; 60:1489-1498. [PMID: 34379579 DOI: 10.1080/0284186x.2021.1959636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND A common feature of solid tumours that are resistant to therapy is the presence of regions with low oxygen content (i.e., hypoxia). Oxygen electrode studies suggest that localized prostate adenocarcinoma is commonly hypoxic, although conflicting data have been reported between immunohistochemical detection of hypoxia-induced proteins in biopsy specimens and positron emission tomography (PET) imaging of 18F-labeled hypoxia reporters. Although the 2-nitroimidazole 18F-EF5 is well-established to label hypoxic tumour cells in pre-clinical tumour models and clinical trials of multiple primary tumour sites, it has yet to be tested in prostate cancer. The purpose of this study was to evaluate the feasibility of using 18F-EF5 to detect hypoxia in clinical prostate tumours. MATERIAL AND METHODS Patients with localized adenocarcinoma of the prostate were recruited for pre-treatment 18F-EF5 PET scans. Immunohistochemistry was conducted on diagnostic biopsies to assess the expression of glucose transporter 1 (GLUT1), osteopontin (OPN), and carbonic anhydrase IX (CAIX). Immunoreactivity scores of staining intensity and frequency were used to indicate the presence of tumour hypoxia. RESULTS We found low tumour-to-muscle ratios of 18F-EF5 uptake that were not consistent with tumour hypoxia, causing early termination of the study. However, we observed GLUT1 and OPN expression in all prostate tumour biopsies, indicating the presence of hypoxia in all tumours. CONCLUSION Our data do not support the use of 18F-EF5 PET to detect hypoxia in prostate adenocarcinoma, and suggest the use of immunohistochemistry to quantify expression of the hypoxia-inducible proteins GLUT1 and OPN as indications of prostate tumour hypoxia.
Collapse
Affiliation(s)
- Brennan J. Wadsworth
- Integrative Oncology, BC Cancer, Vancouver, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Lisa R. Decotret
- Integrative Oncology, BC Cancer, Vancouver, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | | | - Donald Yapp
- Experimental Therapeutics, BC Cancer, Vancouver, Canada
| | - Don Wilson
- Functional Imaging, BC Cancer, Vancouver, Canada
| | - Francois Benard
- Functional Imaging, BC Cancer, Vancouver, Canada
- Molecular Oncology, BC Cancer, Vancouver, Canada
| | | | - Kevin L. Bennewith
- Integrative Oncology, BC Cancer, Vancouver, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
11
|
Elming PB, Wittenborn TR, Busk M, Sørensen BS, Thomsen MBH, Strandgaard T, Dyrskjøt L, Nielsen S, Horsman MR. Refinement of an Established Procedure and Its Application for Identification of Hypoxia in Prostate Cancer Xenografts. Cancers (Basel) 2021; 13:2602. [PMID: 34073301 PMCID: PMC8198481 DOI: 10.3390/cancers13112602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND This pre-clinical study was designed to refine a dissection method for validating the use of a 15-gene hypoxia classifier, which was previously established for head and neck squamous cell carcinoma (HNSCC) patients, to identify hypoxia in prostate cancer. METHODS PC3 and DU-145 adenocarcinoma cells, in vitro, were gassed with various oxygen concentrations (0-21%) for 24 h, followed by real-time PCR. Xenografts were established in vivo, and the mice were injected with the hypoxic markers [18F]-FAZA and pimonidazole. Subsequently, tumors were excised, frozen, cryo-sectioned, and analyzed using autoradiography ([18F]-FAZA) and immunohistochemistry (pimonidazole); the autoradiograms used as templates for laser capture microdissection of hypoxic and non-hypoxic areas, which were lysed, and real-time PCR was performed. RESULTS In vitro, all 15 genes were increasingly up-regulated as oxygen concentrations decreased. With the xenografts, all 15 genes were up-regulated in the hypoxic compared to non-hypoxic areas for both cell lines, although this effect was greater in the DU-145. CONCLUSIONS We have developed a combined autoradiographic/laser-guided microdissection method with broad applicability. Using this approach on fresh frozen tumor material, thereby minimizing the degree of RNA degradation, we showed that the 15-gene hypoxia gene classifier developed in HNSCC may be applicable for adenocarcinomas such as prostate cancer.
Collapse
Affiliation(s)
- Pernille B. Elming
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| | - Thomas R. Wittenborn
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| | - Morten Busk
- Danish Center for Particle Therapy, Aarhus University Hospital, 8200 Aarhus, Denmark;
| | - Brita S. Sørensen
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
- Danish Center for Particle Therapy, Aarhus University Hospital, 8200 Aarhus, Denmark;
| | - Mathilde Borg Houlberg Thomsen
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.B.H.T.); (T.S.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Trine Strandgaard
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.B.H.T.); (T.S.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Lars Dyrskjøt
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.B.H.T.); (T.S.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Steffen Nielsen
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| | - Michael R. Horsman
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| |
Collapse
|
12
|
Thiruthaneeswaran N, Bibby BAS, Yang L, Hoskin PJ, Bristow RG, Choudhury A, West C. Lost in application: Measuring hypoxia for radiotherapy optimisation. Eur J Cancer 2021; 148:260-276. [PMID: 33756422 DOI: 10.1016/j.ejca.2021.01.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/15/2022]
Abstract
The history of radiotherapy is intertwined with research on hypoxia. There is level 1a evidence that giving hypoxia-targeting treatments with radiotherapy improves locoregional control and survival without compromising late side-effects. Despite coming in and out of vogue over decades, there is now an established role for hypoxia in driving molecular alterations promoting tumour progression and metastases. While tumour genomic complexity and immune profiling offer promise, there is a stronger evidence base for personalising radiotherapy based on hypoxia status. Despite this, there is only one phase III trial targeting hypoxia modification with full transcriptomic data available. There are no biomarkers in routine use for patients undergoing radiotherapy to aid management decisions, and a roadmap is needed to ensure consistency and provide a benchmark for progression to application. Gene expression signatures address past limitations of hypoxia biomarkers and could progress biologically optimised radiotherapy. Here, we review recent developments in generating hypoxia gene expression signatures and highlight progress addressing the challenges that must be overcome to pave the way for their clinical application.
Collapse
Affiliation(s)
- Niluja Thiruthaneeswaran
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.
| | - Becky A S Bibby
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Lingjang Yang
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Peter J Hoskin
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; Mount Vernon Cancer Centre, Northwood, UK
| | - Robert G Bristow
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; CRUK Manchester Institute and Manchester Cancer Research Centre, Manchester, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, The University of Manchester, Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Catharine West
- Division of Cancer Sciences, The University of Manchester, Christie Hospital NHS Foundation Trust, Manchester, UK
| |
Collapse
|
13
|
Abstract
Over the last few years, cancer immunotherapy experienced tremendous developments and it is nowadays considered a promising strategy against many types of cancer. However, the exclusion of lymphocytes from the tumor nest is a common phenomenon that limits the efficiency of immunotherapy in solid tumors. Despite several mechanisms proposed during the years to explain the immune excluded phenotype, at present, there is no integrated understanding about the role played by different models of immune exclusion in human cancers. Hypoxia is a hallmark of most solid tumors and, being a multifaceted and complex condition, shapes in a unique way the tumor microenvironment, affecting gene transcription and chromatin remodeling. In this review, we speculate about an upstream role for hypoxia as a common biological determinant of immune exclusion in solid tumors. We also discuss the current state of ex vivo and in vivo imaging of hypoxic determinants in relation to T cell distribution that could mechanisms of immune exclusion and discover functional-morphological tumor features that could support clinical monitoring.
Collapse
|
14
|
Chen Z, Xue Y, Zhang Z, Li W, Wen M, Zhao Y, Li J, Weng Z, Ye Q. The performance of intravoxel-incoherent motion diffusion-weighted imaging derived hypoxia for the risk stratification of prostate cancer in peripheral zone. Eur J Radiol 2020; 125:108865. [PMID: 32058895 DOI: 10.1016/j.ejrad.2020.108865] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE To assess the association between intravoxel-incoherent motion diffusion-weighted imaging (IVIM) derived hypoxia and the aggressiveness of prostate cancer (PCa) and to explore its contribution to the risk stratification of PCa. METHODS Seventy-five peripheral zone (PZ) PCa patients, who underwent multiparametric MRI (mpMRI), were included in this study. Systematic ultrasound guided biopsy was used as reference. IVIM was acquired with 5 b values (b = 0∼750 s/mm2). Apparent diffusion coefficient (ADC), pure tissue diffusion (Ds), volume fraction of pseudo-diffusion (fp), hypoxic fraction (HFDWI), hypoxia score (HSDWI) and relative oxygen saturation(rOSDWI), were calculated and histogram analysis was applied. Groups comparison was performed between low-intermediate-grade group (LG, the International Society of Urological Pathology (ISUP) Gleason Grade (GG) ≤2) and high-grade (HG, ISUP GG ≥ 3) group. The correlation between diffusion parameters and ISUP GG was assessed. Cross-validated Support Vector Machine (SVM) Classification was performed and compared with univariate ROC analysis to explore the risk stratification of PZ PCa. RESULTS Mean, median, and the 10th percentile of Ds differed significantly between groups (p < 0.05). Several parameters significantly correlated with ISUP grade, and the 10th percentile of Ds showed the strongest correlation (ρ= - 0.284). The prediction model containing IVIM derived hypoxia yielded an area under the receiver operating characteristics curve (AUC) ranging 0.749-0.786 for cross-validation. The AUCs of the SVM modeling were higher than that of any single parameter. CONCLUSION IVIM derived hypoxia demonstrated significant correlation with the aggressiveness of PCa. It's supplemental to the MRI assessment of PCa with a promising stratification of risk stratification of PZ PCa.
Collapse
Affiliation(s)
- Zhongwei Chen
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Yingnan Xue
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Zhao Zhang
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Weikang Li
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Min Wen
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Youfan Zhao
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Jiance Li
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Zhiliang Weng
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
| | - Qiong Ye
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China; High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, PR China.
| |
Collapse
|
15
|
O'Reilly D, Johnson P, Buchanan PJ. Hypoxia induced cancer stem cell enrichment promotes resistance to androgen deprivation therapy in prostate cancer. Steroids 2019; 152:108497. [PMID: 31521707 DOI: 10.1016/j.steroids.2019.108497] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 09/03/2019] [Accepted: 09/09/2019] [Indexed: 02/06/2023]
Abstract
Androgen deprivation therapy (ADT) is the main treatment to prolong survival in advance stage prostate cancer (PCa) but associated resistance leads to the development of terminal castrate resistant PCa (CRPC). Current research demonstrates that prostate cancer stem cells (PCSC) play a critical role in the development of treatment resistance and subsequent disease progression. Despite uncertainty surrounding the origin of these cells, studies clearly show they are associated with poorer outcomes and that ADT significantly enhances their numbers. Here in we highlight how activation of HIF signalling, in response to hypoxic conditions within the tumour microenvironment, results in the expression of genes associated with stemness and EMT promoting PCSC emergence which ultimately drives tumour relapse to CRPC. Hypoxic conditions are not only enhanced by ADT but the associated decrease in AR activation also promotes PI3K/AKT signalling which actively enhances HIF and its effects on PCSC's. Furthermore, emerging evidence now indicates that HIF-2α, rather than the commonly considered HIF-1α, is the main family member that drives PCSC emergence. Taken together this clearly identifies HIF and associated pathways as key targets for new therapeutic strategies that could potentially prevent or slow PCSC promoted resistance to ADT, thus holding potential to prolong patient survival.
Collapse
Affiliation(s)
- Debbie O'Reilly
- School of Nursing & Human Sciences, Dublin City University, Dublin, Ireland; National Institute of Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Patricia Johnson
- School of Nursing & Human Sciences, Dublin City University, Dublin, Ireland
| | - Paul J Buchanan
- School of Nursing & Human Sciences, Dublin City University, Dublin, Ireland; National Institute of Cellular Biotechnology, Dublin City University, Dublin, Ireland.
| |
Collapse
|
16
|
Hypoxia and angiogenic biomarkers in prostate cancer after external beam radiotherapy (EBRT) alone or combined with high-dose-rate brachytherapy boost (HDR-BTb). Radiother Oncol 2019; 137:38-44. [PMID: 31059955 DOI: 10.1016/j.radonc.2019.04.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 04/06/2019] [Accepted: 04/17/2019] [Indexed: 01/18/2023]
Abstract
PURPOSE To investigate angiogenic and hypoxia biomarkers to predict outcome in patients receiving external beam radiotherapy (EBRT) alone or combined with high-dose-rate brachytherapy boost (HDR-BTb) for localised prostate cancer. METHODS Prostate biopsy samples were collected prospectively in patients entered into a phase 3 randomised controlled trial of patients receiving EBRT or EBRT + HDR-BTb. Univariate and multivariate analyses using Cox proportional hazards model were performed to identify associations between immunohistochemical staining of hypoxia inducible factor 1 alpha (HIF1α), glucose transporter 1 (GLUT1), osteopontin (OPN) and microvessel density (MVD) using CD-34 antibody with clinical outcome. The primary endpoint was biochemical relapse free survival (BRFS) and secondary endpoint was distant metastasis free survival (DMFS). RESULTS Immunohistochemistry was available for 204 patients. Increased OPN (Hazard ratio [HR] 2.38, 95% Confidence Interval [CI] 1.06-5.34, p < 0.036) and GLUT1 (HR 2.36, 95%CI 1.39-4.01, p < 0.001) expression were predictive of worse BRFS. Increased GLUT1 expression (HR 2.22, 1.02-4.84, p = 0.045) was predictive of worse DMFS. Increased MVD (CD-34) (HR 1.82, 95%CI 1.06-3.14, p = 0.03) and OPN (HR 1.82, 95%CI 1.06-3.14, p = 0.03) but reduced GLUT1 expression (HR 0.40, 95%CI 0.20-0.79, p = 0.009) were predictive of improved BRFS in patients receiving EBRT + HDR-BTb. CONCLUSION Our data suggest angiogenic and hypoxia biomarkers may predict outcome and benefit of dose escalation, however further validation in prospective studies including hypoxia modification is needed. Trial registration number ISRCTN98241100, registered with ISRCTN at http://www.controlled-trials.com/isrctn/.
Collapse
|
17
|
Macharia LW, Wanjiru CM, Mureithi MW, Pereira CM, Ferrer VP, Moura-Neto V. MicroRNAs, Hypoxia and the Stem-Like State as Contributors to Cancer Aggressiveness. Front Genet 2019; 10:125. [PMID: 30842790 PMCID: PMC6391339 DOI: 10.3389/fgene.2019.00125] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 02/04/2019] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules that play key regulatory roles in cancer acting as both oncogenes and tumor suppressors. Due to their potential roles in improving cancer prognostic, predictive, diagnostic and therapeutic approaches, they have become an area of intense research focus in recent years. Several studies have demonstrated an altered expression of several miRNAs under hypoxic condition and even shown that the hypoxic microenvironment drives the selection of a more aggressive cancer cell population through cellular adaptations referred as the cancer stem-like cell. These minor fractions of cells are characterized by their self-renewal abilities and their ability to maintain the tumor mass, suggesting their crucial roles in cancer development. This review aims to highlight the interconnected role between miRNAs, hypoxia and the stem-like state in contributing to the cancer aggressiveness as opposed to their independent contributions, and it is based in four aggressive tumors, namely glioblastoma, cervical, prostate, and breast cancers.
Collapse
Affiliation(s)
- Lucy Wanjiku Macharia
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina da Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Caroline Muriithi Wanjiru
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Instituto de Ciências Biomédicas da Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Valéria Pereira Ferrer
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina da Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivaldo Moura-Neto
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina da Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
18
|
Harryman WL, Warfel NA, Nagle RB, Cress AE. The Tumor Microenvironments of Lethal Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:149-170. [PMID: 31900909 DOI: 10.1007/978-3-030-32656-2_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Localized prostate cancer (confined to the gland) generally is considered curable, with nearly a 100% 5-year-survival rate. When the tumor escapes the prostate capsule, leading to metastasis, there is a poorer prognosis and higher mortality rate, with 5-year survival dropping to less than 30%. A major research question has been to understand the transition from indolent (low risk) disease to aggressive (high risk) disease. In this chapter, we provide details of the changing tumor microenvironments during prostate cancer invasion and their role in the progression and metastasis of lethal prostate cancer. Four microenvironments covered here include the muscle stroma, perineural invasion, hypoxia, and the role of microvesicles in altering the extracellular matrix environment. The adaptability of prostate cancer to these varied microenvironments and the cues for phenotypic changes are currently understudied areas. Model systems for understanding smooth muscle invasion both in vitro and in vivo are highlighted. Invasive human needle biopsy tissue and mouse xenograft tumors both contain smooth muscle invasion. In combination, the models can be used in an iterative process to validate molecular events for smooth muscle invasion in human tissue. Understanding the complex and interacting microenvironments in the prostate holds the key to early detection of high-risk disease and preventing tumor invasion through escape from the prostate capsule.
Collapse
Affiliation(s)
| | - Noel A Warfel
- University of Arizona Cancer Center, Tucson, AZ, USA
| | - Raymond B Nagle
- Department of Pathology, University of Arizona Cancer Center, Tucson, AZ, USA
| | - Anne E Cress
- University of Arizona Cancer Center, Tucson, AZ, USA.
| |
Collapse
|
19
|
Analysis of regulator of G-protein signalling 2 (RGS2) expression and function during prostate cancer progression. Sci Rep 2018; 8:17259. [PMID: 30467386 PMCID: PMC6250724 DOI: 10.1038/s41598-018-35332-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/30/2018] [Indexed: 01/19/2023] Open
Abstract
Prostate cancer (PC) represents the second highest cancer-related mortality among men and the call for biomarkers for early discrimination between aggressive and indolent forms is essential. Downregulation of Regulator of G-protein signaling 2 (RGS2) has been shown in PC, however the underlying mechanism has not been described. Aberrant RGS2 expression has also been reported for other carcinomas in association to both positive and negative prognosis. In this study, we assessed RGS2 expression during PC progression in terms of regulation and impact on tumour phenotype and evaluated its prognostic value. Our experimental data suggest that the RGS2 downregulation seen in early PC is caused by hypoxia. In line with the common indolent phenotype of a primary PC, knockdown of RGS2 induced epithelial features and impaired metastatic properties. However, increased STAT3, TWIST1 and decreased E-cadherin expression suggest priming for EMT. Additionally, improved tumour cell survival and increased BCL-2 expression linked decreased RGS2 levels to fundamental tumour advantages. In contrast, high RGS2 levels in advanced PC were correlated to poor patient survival and a positive metastatic status. This study describes novel roles for RGS2 during PC progression and suggests a prognostic potential discriminating between indolent and metastatic forms of PC.
Collapse
|
20
|
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]
|
21
|
Levesque C, Nelson PS. Cellular Constituents of the Prostate Stroma: Key Contributors to Prostate Cancer Progression and Therapy Resistance. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a030510. [PMID: 28490538 DOI: 10.1101/cshperspect.a030510] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Reciprocal signaling between prostate stroma and its epithelium are fundamental to organ development and homeostasis. Similarly, interactions between tumor cells and stromal constituents are central to key aspects of carcinogenesis and malignancy growth involving tumor cell invasion, dissemination, and growth in distant sites. The prostate stroma is complex with several distinct resident cell types, infiltrating nonresident cell types and an amalgam of structural matrix factors, matricellular proteins, metabolites, growth factors, and cytokines. Of importance, the stroma is dynamic with changes in composition as a cause or consequence of intrinsic and extrinsic factors. In the context of epithelial neoplasia, the prostate stroma undergoes phenotypic changes with a loss of well-differentiated smooth muscle cell population and the expansion of cancer-associated fibroblast populations. This reactive stroma further coevolves with tumor progression. Recent studies show the role of tumor microenvironment components in therapy resistance and highlight the importance of a thorough knowledge of cross talk between tumor cells and microenvironment niches to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Christine Levesque
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Peter S Nelson
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| |
Collapse
|
22
|
Abstract
The concept of tumour hypoxia as a cause of radiation resistance has been prevalent for over 100 years. During this time, our understanding of tumour hypoxia has matured with the recognition that oxygen tension within a tumour is influenced by both diffusion and perfusion mechanisms. In parallel, clinical strategies to modify tumour hypoxia with the expectation that this will improve response to radiation have been developed and tested in clinical trials. Despite many disappointments, meta-analysis of the data on hypoxia modification confirms a significant impact on both tumour control and survival. Early trials evaluated hyperbaric oxygen followed by a generation of studies testing oxygen mimetics such as misonidazole, pimonidazole and etanidazole. One highly significant result stands out from the use of nimorazole in advanced laryngeal cancer with a significant advantage seen for locoregional control using this radiosensitiser. More recent studies have evaluated carbogen and nicotinamide targeting both diffusion related and perfusion related hypoxia. A significant survival advantage is seen in muscle invasive bladder cancer and also for locoregional control in hypopharygeal cancer associated with a low haemoglobin. New developments include the recognition that mitochondrial complex inhibitors reducing tumour oxygen consumption are potential radiosensitising agents and atovaquone is currently in clinical trials. One shortcoming of past hypoxia modifying trials is the failure to identify oxygenation status and select those patient with significant hypoxia. A range of biomarkers are now available including histological necrosis, immunohistochemical intrinsic markers such as CAIX and Glut 1 and hypoxia gene signatures which have been shown to predict outcome and will inform the next generation of hypoxia modifying clinical trials.
Collapse
Affiliation(s)
- Hannah Tharmalingham
- Mount Vernon Cancer Centre, Northwood, UK.,University of Manchester, Manchester, UK.,Christie Hospital, Manchester, UK
| | - Peter Hoskin
- Mount Vernon Cancer Centre, Northwood, UK.,University of Manchester, Manchester, UK.,Christie Hospital, Manchester, UK.,Manchester Cancer Research Centre, Manchester, UK
| |
Collapse
|
23
|
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]
|
24
|
Thomann B, Sachpazidis I, Koubar K, Zamboglou C, Mavroidis P, Wiehle R, Grosu AL, Baltas D. Influence of inhomogeneous radiosensitivity distributions and intrafractional organ movement on the tumour control probability of focused IMRT in prostate cancer. Radiother Oncol 2018; 127:62-67. [PMID: 29548559 DOI: 10.1016/j.radonc.2018.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/29/2018] [Accepted: 02/05/2018] [Indexed: 12/25/2022]
Abstract
PURPOSE To evaluate the influence of radioresistance and intrafractional movement on the tumour control probability (TCP) in IMRT prostate treatments using simultaneous integrated boosts to PSMA-PET/CT-delineated GTVs. MATERIALS AND METHODS 13 patients had PSMA-PET/CT prior to prostatectomy and histopathological examination. Two GTVs were available: GTV-PET and GTV-histo, which is the true cancer volume. Focused IMRT plans delivering 77 Gy in 35 fractions to the prostate and 95 Gy to PTV-PET were produced. For random portions of the true cancer volume, α and α/β were uniformly changed to represent different radiosensitivity reductions. TCP was calculated (linear quadratic model) for the true cancer volume with and without simulated intrafractional movement. RESULTS Intrafractional movement increased the TCP by up to 10.2% in individual cases and 1.2% averaged over all cases for medium radiosensitivity levels. At lower levels of radiosensitivity, movement decreased the TCP. Radiosensitivity reductions of 10-20% led to TCP reductions of 1-24% and 10-68% for 1% and 5% affected cancer volume, respectively. There is no linear correlation but a sudden breakdown of TCPs within a small range of radiosensitivity levels. CONCLUSION TCP drops significantly within a narrow range of radiosensitivity levels. Intrafractional movement can increase TCP when the boost volume is surrounded by a sufficiently high dose plateau.
Collapse
Affiliation(s)
- Benedikt Thomann
- Division of Medical Physics, Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Ilias Sachpazidis
- Division of Medical Physics, Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Khodor Koubar
- Division of Medical Physics, Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Panayiotis Mavroidis
- University of North Carolina at Chapel Hill, Department of Radiation Oncology, USA
| | - Rolf Wiehle
- Division of Medical Physics, Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dimos Baltas
- Division of Medical Physics, Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
25
|
Panigrahi GK, Praharaj PP, Peak TC, Long J, Singh R, Rhim JS, Abd Elmageed ZY, Deep G. Hypoxia-induced exosome secretion promotes survival of African-American and Caucasian prostate cancer cells. Sci Rep 2018; 8:3853. [PMID: 29497081 PMCID: PMC5832762 DOI: 10.1038/s41598-018-22068-4] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/15/2018] [Indexed: 12/14/2022] Open
Abstract
African American men in the United States have higher mortality due to prostate cancer (PCa) compared to other races. One reason for this disparity is the lack of in-depth understanding of the PCa biology in African Americans. For example, hypoxia in prostate tumor microenvironment is associated with adverse prognosis; still, no hypoxia-related studies have been reported in African Americans. Here, we compared African-American and Caucasian PCa cells for exosome secretion under normoxic (21% O2) and hypoxic (1% O2) conditions. All cell lines showed higher exosome secretion under hypoxia but it was clearly more prominent in African-American PCa cells. Further, under hypoxia, Rab5 (a biomarker for early endosome) was clustered in perinuclear region; and CD63 (a biomarker for exosomes and multivesicular endosomes) showed greater co-localization with actin cytoskeleton especially in African American PCa cells. Importantly, exosome biogenesis inhibitors GW4869 (10-20 µM) or DMA (10-20 µg/ml) significantly decreased cell viability and clonogenicity in PCa cells. Interestingly, we also observed higher level of lactic acid loaded in exosomes secreted under hypoxia. Overall, under chronic hypoxia, PCa cells secrete more exosomes as a survival mechanism to remove metabolic waste.
Collapse
Affiliation(s)
- Gati K Panigrahi
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Prakash P Praharaj
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Taylor C Peak
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Jessica Long
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Ravi Singh
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Johng S Rhim
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of Health Sciences, Bethesda, MD, USA
| | - Zakaria Y Abd Elmageed
- Department of Pharmaceutical Sciences, Texas A&M Rangel College of Pharmacy, College Station, Texas, USA
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.
| |
Collapse
|
26
|
Deep G, Panigrahi GK. Hypoxia-Induced Signaling Promotes Prostate Cancer Progression: Exosomes Role as Messenger of Hypoxic Response in Tumor Microenvironment. Crit Rev Oncog 2018; 20:419-34. [PMID: 27279239 DOI: 10.1615/critrevoncog.v20.i5-6.130] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prostate cancer (PCA) is the leading malignancy in men and the second leading cause of cancer-related deaths. Hypoxia (low O2 condition) is considered an early event in prostate carcinogenesis associated with an aggressive phenotype. In fact, clinically, hypoxia and hypoxia-related biomarkers are associated with treatment failure and disease progression. Hypoxia-inducible factor 1 (HIF-1) is the key factor that is activated under hypoxia, and mediates adaptation of cells to hypoxic conditions through regulating the expression of genes associated with angiogenesis, epithelial-to-mesenchymal transition (EMT), metastasis, survival, proliferation, metabolism, sternness, hormone-refractory progression, and therapeutic resistance. Besides HIF-1, several other signaling pathways including PI3K/Akt/mTOR, NADPH oxidase (NOX), Wnt/b-catenin, and Hedgehog are activated in cancer cells under hypoxic conditions, and also contribute in hypoxia-induced biological effects in HIF-1-dependent and -independent manners. Hypoxic cancer cells cause extensive changes in the tumor microenvironment both local and distant, and recent studies have provided ample evidence supporting the crucial role of nanosized vesicles "exosomes" in mediating hypoxia-induced tumor microenvironment remodeling. Exosomes' role has been reported in hypoxia-induced angiogenesis, sternness, activation of cancer-associated fibroblasts (CAFs), and EMT. Together, existing literature suggests that hypoxia plays a predominant role in PCA growth and progression, and PCA could be effectively prevented and treated via targeting hypoxia/hypoxia-related signaling pathways.
Collapse
Affiliation(s)
- Gagan Deep
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO; University of Colorado Cancer Center, University of Colorado Denver, Aurora, CO
| | - Gati K Panigrahi
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO
| |
Collapse
|
27
|
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
|
28
|
Low-intensity pulsed ultrasound enhances angiogenesis and ameliorates contractile dysfunction of pressure-overloaded heart in mice. PLoS One 2017; 12:e0185555. [PMID: 28957396 PMCID: PMC5619801 DOI: 10.1371/journal.pone.0185555] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 09/14/2017] [Indexed: 11/19/2022] Open
Abstract
Introduction Chronic left ventricular (LV) pressure overload causes relative ischemia with resultant LV dysfunction. We have recently demonstrated that low-intensity pulsed ultrasound (LIPUS) improves myocardial ischemia in a pig model of chronic myocardial ischemia through enhanced myocardial angiogenesis. In the present study, we thus examined whether LIPUS also ameliorates contractile dysfunction in LV pressure-overloaded hearts. Methods and results Chronic LV pressure overload was induced with transverse aortic constriction (TAC) in mice. LIPUS was applied to the whole heart three times in the first week after TAC and was repeated once a week for 7 weeks thereafter (n = 22). Animals in the control groups received the sham treatment without LIPUS (n = 23). At 8 weeks after TAC, LV fractional shortening was depressed in the TAC-Control group, which was significantly ameliorated in the TAC-LIPUS group (30.4±0.5 vs. 36.2±3.8%, P<0.05). Capillary density was higher and perivascular fibrosis was less in the LV in the TAC-LIPUS group than in the TAC-Control group. Myocardial relative ischemia evaluated with hypoxyprobe was noted in the TAC-Control group, which was significantly attenuated in the TAC-LIPUS group. In the TAC-LIPUS group, as compared with the control group, mRNA expressions of BNP and collagen III were significantly lower (both P<0.05) and protein expressions of VEGF and eNOS were significantly up-regulated associated with Akt activation (all P<0.05). No adverse effect related to the LIPUS therapy was noted. Conclusions These results indicate that the LIPUS therapy ameliorates contractile dysfunction in chronically pressure-overloaded hearts through enhanced myocardial angiogenesis and attenuated perivascular fibrosis. Thus, the LIPUS therapy may be a promising, non-invasive treatment for cardiac dysfunction due to chronic pressure overload.
Collapse
|
29
|
Jonsson M, Ragnum HB, Julin CH, Yeramian A, Clancy T, Frikstad KAM, Seierstad T, Stokke T, Matias-Guiu X, Ree AH, Flatmark K, Lyng H. Hypoxia-independent gene expression signature associated with radiosensitisation of prostate cancer cell lines by histone deacetylase inhibition. Br J Cancer 2016; 115:929-939. [PMID: 27599042 PMCID: PMC5061908 DOI: 10.1038/bjc.2016.278] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/22/2016] [Accepted: 08/11/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Histone deacetylase inhibitors (HDACis) like vorinostat are promising radiosensitisers in prostate cancer, but their effect under hypoxia is not known. We investigated gene expression associated with radiosensitisation of normoxic and hypoxic prostate cancer cells by vorinostat. METHODS Cells were exposed to vorinostat under normoxia or hypoxia and subjected to gene expression profiling before irradiation and clonogenic survival analysis. RESULTS Pretreatment with vorinostat led to radiosensitisation of the intrinsically radioresistant DU 145 cells, but not the radiosensitive PC-3 and 22Rv1 cells, and was independent of hypoxia status. Knockdown experiments showed that the sensitisation was not caused by repression of hypoxia-inducible factor HIF1 or tumour protein TP53. Global deregulation of DNA repair and chromatin organisation genes was associated with radiosensitisation under both normoxia and hypoxia. A radiosensitisation signature with expression changes of 56 genes was generated and valid for both conditions. For eight signature genes, baseline expression also correlated with sensitisation, showing potential as pretreatment biomarker. The hypoxia independence of the signature was confirmed in a clinical data set. CONCLUSIONS Pretreatment with HDACi may overcome radioresistance of hypoxic prostate tumours by similar mechanisms as under normoxia. We propose a gene signature to predict radiosensitising effects independent of hypoxia status.
Collapse
Affiliation(s)
- Marte Jonsson
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Pb 4950, Nydalen, 0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Harald Bull Ragnum
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Pb 4950, Nydalen, 0424 Oslo, Norway
| | - Cathinka Halle Julin
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Pb 4950, Nydalen, 0424 Oslo, Norway
| | - Andree Yeramian
- Department of Pathology and Molecular Genetics HUAV, University of Lleida, Lleida, Spain
| | - Trevor Clancy
- Department of Tumor Biology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Kari-Anne Myrum Frikstad
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Pb 4950, Nydalen, 0424 Oslo, Norway
| | - Therese Seierstad
- Department of Radiology and Nuclear Medicine, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Trond Stokke
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Pb 4950, Nydalen, 0424 Oslo, Norway
| | - Xavier Matias-Guiu
- Department of Pathology and Molecular Genetics HUAV, University of Lleida, Lleida, Spain
| | - Anne Hansen Ree
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Tumor Biology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| | - Kjersti Flatmark
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Tumor Biology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Gastroenterological Surgery, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Heidi Lyng
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Pb 4950, Nydalen, 0424 Oslo, Norway
| |
Collapse
|
30
|
Schlaepfer IR, Nambiar DK, Ramteke A, Kumar R, Dhar D, Agarwal C, Bergman B, Graner M, Maroni P, Singh RP, Agarwal R, Deep G. Hypoxia induces triglycerides accumulation in prostate cancer cells and extracellular vesicles supporting growth and invasiveness following reoxygenation. Oncotarget 2016; 6:22836-56. [PMID: 26087400 PMCID: PMC4673203 DOI: 10.18632/oncotarget.4479] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 05/22/2015] [Indexed: 12/21/2022] Open
Abstract
Hypoxia is an independent prognostic indicator of poor outcome in several malignancies. However, precise mechanism through which hypoxia promotes disease aggressiveness is still unclear. Here, we report that under hypoxia (1% O2), human prostate cancer (PCA) cells, and extracellular vesicles (EVs) released by these cells, are significantly enriched in triglycerides due to the activation of lipogenesis-related enzymes and signaling molecules. This is likely a survival response to hypoxic stress as accumulated lipids could support growth following reoxygenation. Consistent with this, significantly higher proliferation was observed in hypoxic PCA cells following reoxygenation associated with rapid use of accumulated lipids. Importantly, lipid utilization inhibition by CPT1 inhibitor etomoxir and shRNA-mediated CPT1-knockdown significantly compromised hypoxic PCA cell proliferation following reoxygenation. Furthermore, COX2 inhibitor celecoxib strongly reduced growth and invasiveness following hypoxic PCA cells reoxygenation, and inhibited invasiveness induced by hypoxic PCA EVs. This establishes a role for COX2 enzymatic products in the enhanced PCA growth and invasiveness. Importantly, concentration and loading of EVs secreted by PCA cells were significantly compromised under delipidized serum condition and by lipogenesis inhibitors (fatostatin and silibinin). Overall, present study highlights the biological significance of lipid accumulation in hypoxic PCA cells and its therapeutic relevance in PCA.
Collapse
Affiliation(s)
- Isabel R Schlaepfer
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Dhanya K Nambiar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA.,Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Anand Ramteke
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA.,Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, India
| | - Rahul Kumar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA
| | - Deepanshi Dhar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA.,University of Colorado Cancer Center, University of Colorado Denver, Aurora, Colorado, USA
| | - Bryan Bergman
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Denver, Aurora, Colorado, USA
| | - Michael Graner
- Department of Neurosurgery, University of Colorado Denver, Aurora, Colorado, USA
| | - Paul Maroni
- Department of Surgery, University of Colorado Denver, Aurora, Colorado, USA
| | - Rana P Singh
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA.,University of Colorado Cancer Center, University of Colorado Denver, Aurora, Colorado, USA
| | - Gagan Deep
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA.,University of Colorado Cancer Center, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
31
|
Rupp NJ, Schüffler PJ, Zhong Q, Falkner F, Rechsteiner M, Rüschoff JH, Fankhauser C, Drach M, Largo R, Tremp M, Poyet C, Sulser T, Kristiansen G, Moch H, Buhmann J, Müntener M, Wild PJ. Oxygen supply maps for hypoxic microenvironment visualization in prostate cancer. J Pathol Inform 2016; 7:3. [PMID: 26955501 PMCID: PMC4763504 DOI: 10.4103/2153-3539.175376] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/26/2015] [Indexed: 12/04/2022] Open
Abstract
Background: Intratumoral hypoxia plays an important role with regard to tumor biology and susceptibility to radio- and chemotherapy. For further investigation of hypoxia-related changes, areas of certain hypoxia must be reliably detected within cancer tissues. Pimonidazole, a 2-nitroimindazole, accumulates in hypoxic tissue and can be easily visualized using immunohistochemistry. Materials and Methods: To improve detection of highly hypoxic versus normoxic areas in prostate cancer, immunoreactivity of pimonidazole and a combination of known hypoxia-related proteins was used to create computational oxygen supply maps of prostate cancer. Pimonidazole was intravenously administered before radical prostatectomy in n = 15 patients, using the da Vinci robot-assisted surgical system. Prostatectomy specimens were immediately transferred into buffered formaldehyde, fixed overnight, and completely embedded in paraffin. Pimonidazole accumulation and hypoxia-related protein expression were visualized by immunohistochemistry. Oxygen supply maps were created using the normalized information from pimonidazole and hypoxia-related proteins. Results: Based on pimonidazole staining and other hypoxia.related proteins (osteopontin, hypoxia-inducible factor 1-alpha, and glucose transporter member 1) oxygen supply maps in prostate cancer were created. Overall, oxygen supply maps consisting of information from all hypoxia-related proteins showed high correlation and mutual information to the golden standard of pimonidazole. Here, we describe an improved computer-based ex vivo model for an accurate detection of oxygen supply in human prostate cancer tissue. Conclusions: This platform can be used for precise colocalization of novel candidate hypoxia-related proteins in a representative number of prostate cancer cases, and improve issues of single marker correlations. Furthermore, this study provides a source for further in situ tests and biochemical investigations
Collapse
Affiliation(s)
- Niels J Rupp
- Institute of Surgical Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Peter J Schüffler
- Department of Computer Science, ETH Zurich, Universitaetstr 6, 8092 Zurich, Switzerland
| | - Qing Zhong
- Institute of Surgical Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Florian Falkner
- Institute of Pathology, Cantonal Hospital Winterthur, Brauerstrasse 15, 8401 Winterthur, Switzerland
| | - Markus Rechsteiner
- Institute of Surgical Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Jan H Rüschoff
- Institute of Surgical Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Christian Fankhauser
- Institute of Surgical Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Matthias Drach
- Institute of Surgical Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Remo Largo
- Department of Urology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Mathias Tremp
- Department of Plastic, Reconstructive and Aesthetic Surgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Cedric Poyet
- Department of Urology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Tullio Sulser
- Department of Urology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Glen Kristiansen
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Strasse 25, 53127 Bonn, Germany
| | - Holger Moch
- Institute of Surgical Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Joachim Buhmann
- Department of Computer Science, ETH Zurich, Universitaetstr 6, 8092 Zurich, Switzerland
| | - Michael Müntener
- Department of Urology, City Hospital Triemli, Birmensdorferstrasse 497, 8063 Zurich, Switzerland
| | - Peter J Wild
- Institute of Surgical Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| |
Collapse
|
32
|
Dhani N, Fyles A, Hedley D, Milosevic M. The clinical significance of hypoxia in human cancers. Semin Nucl Med 2015; 45:110-21. [PMID: 25704384 DOI: 10.1053/j.semnuclmed.2014.11.002] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hypoxia is present to some extent in most solid malignant human cancers because of an imbalance between the limited oxygen delivery capacity of the abnormal vasculature and the high oxygen consumption of tumor cells. This drives a complex and dynamic compensatory response to enable continued cell survival, including genomic changes leading to selection of hypoxia-adapted cells with a propensity to invade locally, metastasize, and recur following surgery or radiotherapy. There is indisputable clinical evidence from numerous observational and therapeutic studies across a range of tumor types to implicate hypoxia as a key determinant of cancer behavior and treatment outcome. Despite this, hypoxia-targeted treatment has failed to influence clinical practice. This is explained, in part, by emerging findings to indicate that hypoxia is not equally important in all patients even when present to the same extent. The impact of hypoxia on patient outcome and the benefit of hypoxia-targeted treatments are greatest in situations where hypoxia is a primary biological driver of disease behavior-patients with tumors having a "hypoxic driver" phenotype. The challenge for the clinical and scientific communities moving forward is to develop robust precision cancer medicine strategies for identifying these patients in the setting of other etiologic, genomic, and host-tumor factors, considering not only the state of the tumor at diagnosis but also changing patient and tumor characteristics over time.
Collapse
Affiliation(s)
- Neesha Dhani
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Canada
| | - Anthony Fyles
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - David Hedley
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Canada
| | - Michael Milosevic
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Canada.
| |
Collapse
|
33
|
Dhani NC, Serra S, Pintilie M, Schwock J, Xu J, Gallinger S, Hill RP, Hedley DW. Analysis of the intra- and intertumoral heterogeneity of hypoxia in pancreatic cancer patients receiving the nitroimidazole tracer pimonidazole. Br J Cancer 2015; 113:864-71. [PMID: 26325106 PMCID: PMC4578083 DOI: 10.1038/bjc.2015.284] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 06/29/2015] [Accepted: 07/11/2015] [Indexed: 12/18/2022] Open
Abstract
Background: Hypoxia is thought to be an adverse feature of pancreatic cancer, but direct measurement in patients is technically challenging. To address this, we characterised the intra/interpatient heterogeneity of hypoxia in surgical specimens from patients who received the 2-nitroimidazole tracer pimonidazole pre-operatively. Methods: Pimondazole was given intravenously 16–20 h before pancreatectomy, and the extent and intratumoral heterogeneity of hypoxia determined by image analysis applied to multiple tissue blocks stained by immunohistochemistry. Intra/interpatient heterogeneity was estimated by variance component analysis. Results: Pimonidazole staining was analysed in 10 tumours. The extent of labelling varied amongst patients (0–26%), with a broader range of hypoxia in the epithelial (1–39%) compared with the stromal (1–13%) compartments. Variance component analysis demonstrated greater inter- than intrapatient variability of hypoxia, and that multiple (4–5) tumour sections are required to provide a consistent evaluation of its extent in individual tumours. Conclusions: There is significant intra- and intertumoral heterogeneity of hypoxia in pancreatic cancers, and these do not appear to be generally more hypoxic than other cancer types. This study establishes the feasibility to assess hypoxia in pancreatic cancer patients using pimonidazole, but questions the reliability of measurements made using a single tissue section.
Collapse
Affiliation(s)
- N C Dhani
- Division of Medical Oncology and Hematology, University Health Network, Princess Margaret Cancer Centre/Ontario Cancer Institute, 610 University Avenue, Toronto, ON, Canada M5G 2M9
| | - S Serra
- Department of Laboratory Medicine and Pathobiology, University Health Network, Princess Margaret Cancer Centre/Ontario Cancer Institute, 610 University Avenue, Toronto, ON, Canada M5G 2M9
| | - M Pintilie
- Department of Biostatistics, University Health Network, Princess Margaret Cancer Centre/Ontario Cancer Institute, 610 University Avenue, Toronto, ON, M5G 2M9
| | - J Schwock
- Department of Laboratory Medicine and Pathobiology, University Health Network, Princess Margaret Cancer Centre/Ontario Cancer Institute, 610 University Avenue, Toronto, ON, Canada M5G 2M9
| | - J Xu
- Applied Molecular Profiling Laboratory, University Health Network, Princess Margaret Cancer Centre/Ontario Cancer Institute, 610 University Avenue, Toronto, ON, Canada M5G 2M9
| | - S Gallinger
- Division of Hepato-biliary Pancreatic Surgical Oncology, University Health Network and Mount Sinai Hospital, University Health Network, Princess Margaret Cancer Centre/Ontario Cancer Institute, 610 University Avenue, Toronto, ON, Canada M5G 2M9
| | - R P Hill
- Radiation Medicine Program, University Health Network, Princess Margaret Cancer Centre/Ontario Cancer Institute, 610 University Avenue, Toronto, ON, Canada M5G 2M9
| | - D W Hedley
- Division of Medical Oncology and Hematology, University Health Network, Princess Margaret Cancer Centre/Ontario Cancer Institute, 610 University Avenue, Toronto, ON, Canada M5G 2M9
| |
Collapse
|
34
|
Ranasinghe WK, Baldwin GS, Bolton D, Shulkes A, Ischia J, Patel O. HIF1α Expression under Normoxia in Prostate Cancer— Which Pathways to Target? J Urol 2015; 193:763-70. [DOI: 10.1016/j.juro.2014.10.085] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2014] [Indexed: 10/24/2022]
Affiliation(s)
| | - Graham S. Baldwin
- Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Damien Bolton
- Department of Urology, Austin Health, Heidelberg, Victoria, Australia
| | - Arthur Shulkes
- Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Joseph Ischia
- Department of Urology, Austin Health, Heidelberg, Victoria, Australia
| | - Oneel Patel
- Department of Surgery, University of Melbourne, Melbourne, Australia
| |
Collapse
|
35
|
Ragnum HB, Vlatkovic L, Lie AK, Axcrona K, Julin CH, Frikstad KM, Hole KH, Seierstad T, Lyng H. The tumour hypoxia marker pimonidazole reflects a transcriptional programme associated with aggressive prostate cancer. Br J Cancer 2014; 112:382-90. [PMID: 25461803 PMCID: PMC4453458 DOI: 10.1038/bjc.2014.604] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/15/2014] [Accepted: 11/04/2014] [Indexed: 12/20/2022] Open
Abstract
Background: The hypoxia marker pimonidazole is a candidate biomarker of cancer aggressiveness. We investigated the transcriptional programme associated with pimonidazole staining in prostate cancer. Methods: Index tumour biopsies were taken by image guidance from an investigation cohort of 52 patients, where 43 patients received pimonidazole before prostatectomy. Biopsy location within the index tumour was verified for 46 (88%) patients, who were included for gene expression profiling and immunohistochemistry. Two independent cohorts of 59 and 281 patients were used for validation. Results: Expression of genes in proliferation, DNA repair and hypoxia response was a major part of the transcriptional programme associated with pimonidazole staining. A signature of 32 essential genes was constructed and showed positive correlation to Ki67 staining, confirming the increased proliferation in hypoxic tumours as suggested from the gene data. Positive correlations were also found to tumour stage and lymph node status, but not to blood prostate-specific antigen level, consistent with the findings for pimonidazole staining. The association with aggressiveness was confirmed in validation cohorts, where the signature correlated with Gleason score and had independent prognostic impact, respectively. Conclusions: Pimonidazole staining reflects an aggressive hypoxic phenotype of prostate cancer characterised by upregulation of proliferation, DNA repair and hypoxia response genes.
Collapse
Affiliation(s)
- H B Ragnum
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Pb 4950, Nydalen, Oslo 0424, Norway
| | - L Vlatkovic
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - A K Lie
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - K Axcrona
- Department of Urology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - C H Julin
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Pb 4950, Nydalen, Oslo 0424, Norway
| | - K M Frikstad
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Pb 4950, Nydalen, Oslo 0424, Norway
| | - K H Hole
- Department of Radiology and Nuclear Medicine, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - T Seierstad
- Department of Radiology and Nuclear Medicine, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - H Lyng
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Pb 4950, Nydalen, Oslo 0424, Norway
| |
Collapse
|
36
|
|
37
|
Bristow RG, Berlin A, Dal Pra A. An arranged marriage for precision medicine: hypoxia and genomic assays in localized prostate cancer radiotherapy. Br J Radiol 2014; 87:20130753. [PMID: 24588670 DOI: 10.1259/bjr.20130753] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Prostate cancer (CaP) is the most commonly diagnosed malignancy in males in the Western world with one in six males diagnosed in their lifetime. Current clinical prognostication groupings use pathologic Gleason score, pre-treatment prostatic-specific antigen and Union for International Cancer Control-TNM staging to place patients with localized CaP into low-, intermediate- and high-risk categories. These categories represent an increasing risk of biochemical failure and CaP-specific mortality rates, they also reflect the need for increasing treatment intensity and justification for increased side effects. In this article, we point out that 30-50% of patients will still fail image-guided radiotherapy or surgery despite the judicious use of clinical risk categories owing to interpatient heterogeneity in treatment response. To improve treatment individualization, better predictors of prognosis and radiotherapy treatment response are needed to triage patients to bespoke and intensified CaP treatment protocols. These should include the use of pre-treatment genomic tests based on DNA or RNA indices and/or assays that reflect cancer metabolism, such as hypoxia assays, to define patient-specific CaP progression and aggression. More importantly, it is argued that these novel prognostic assays could be even more useful if combined together to drive forward precision cancer medicine for localized CaP.
Collapse
Affiliation(s)
- R G Bristow
- Princess Margaret Cancer Center (University Health Network), Toronto, ON, Canada
| | | | | |
Collapse
|
38
|
Intratumoral Hypoxia as the Genesis of Genetic Instability and Clinical Prognosis in Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 772:189-204. [DOI: 10.1007/978-1-4614-5915-6_9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
39
|
Gomez CR, Kosari F, Munz JM, Schreiber CA, Knutson GJ, Ida CM, El Khattouti A, Karnes RJ, Cheville JC, Vasmatzis G, Vuk-Pavlović S. Prognostic value of discs large homolog 7 transcript levels in prostate cancer. PLoS One 2013; 8:e82833. [PMID: 24349376 PMCID: PMC3857287 DOI: 10.1371/journal.pone.0082833] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 10/29/2013] [Indexed: 01/25/2023] Open
Abstract
Hypoxia has been associated with malignant progression, metastasis and resistance to therapy. Hence, we studied expression of hypoxia–regulated genes in 100 prostate cancer (CaP) bulk tissues and 71 adjacent benign tissues. We found 24 transcripts significantly overexpressed (p≤0.02). Importantly, higher transcript levels of disc large (drosophila) homolog-associated protein 5 (DLGAP5)/discs large homolog 7 (DLG7)/hepatoma up-regulated protein (HURP), hyaluronan-mediated motility receptor (HMMR) and cyclin B1 (CCNB1) were associated with higher Gleason score and more advanced systemic progression. Since the products of HMMR and CCNB1 have been identified recently as molecular markers of CaP progression, we postulated that DLG7 has prognostic value too. To test this hypothesis, we measured transcript levels for DLG7 in a 150-pair case-control cohort. The cases (progression to systemic disease within six years of surgery) and controls (no progression within eight years) were matched for clinical and pathologic prognostic variables, including grade, stage, and preoperative serum levels of PSA. The overall prognostic ability of DLG7, as tested in receiver operating characteristic analysis was of 0.74 (95% CI, 0.68 to 0.8). Overall, our data indicate that expression of DLG7, a hypoxia-controlled gene, holds prognostic potential in high-risk CaP; this also demonstrates that variation of oxygen tension may constitute a tool for identification of novel biomarkers for CaP.
Collapse
Affiliation(s)
- Christian R. Gomez
- Stem Cell Laboratory, Mayo Clinic Cancer Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- Division of Preventive and Occupational Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| | - Farhad Kosari
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jan-Marie Munz
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Claire A. Schreiber
- Stem Cell Laboratory, Mayo Clinic Cancer Center, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Gaylord J. Knutson
- Stem Cell Laboratory, Mayo Clinic Cancer Center, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Cristiane M. Ida
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | | | - R. Jeffrey Karnes
- Department of Urology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - John C. Cheville
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - George Vasmatzis
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Stanimir Vuk-Pavlović
- Stem Cell Laboratory, Mayo Clinic Cancer Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- Division of Preventive and Occupational Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
40
|
McCullough DJ, Nguyen LMD, Siemann DW, Behnke BJ. Effects of exercise training on tumor hypoxia and vascular function in the rodent preclinical orthotopic prostate cancer model. J Appl Physiol (1985) 2013; 115:1846-54. [PMID: 24177690 DOI: 10.1152/japplphysiol.00949.2013] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Regular physical exercise is considered to be an integral component of cancer care strategies. However, the effect of exercise training on tumor microvascular oxygenation, hypoxia, and vascular function, all of which can affect the tumor microenvironment, remains unknown. Using an orthotopic preclinical model of prostate cancer, we tested the hypotheses that, after exercise training, in the tumor, there would be an enhanced microvascular Po2, increased number of patent vessels, and reduced hypoxia. We also investigated tumor resistance artery contractile properties. Dunning R-3327 AT-1 tumor cells (10(4)) were injected into the ventral prostate of 4-5-mo-old male Copenhagen or Nude rats, which were randomly assigned to tumor-bearing exercise trained (TB-Ex trained; n = 15; treadmill exercise for 5-7 wk) or sedentary groups (TB-Sedentary; n = 12). Phosphorescence quenching was used to measure tumor microvascular Po2, and Hoechst-33342 and EF-5 were used to measure patent vessels and tumor hypoxia, respectively. Tumor resistance artery function was assessed in vitro using the isolated microvessel technique. Compared with sedentary counterparts, tumor microvascular Po2 increased ∼100% after exercise training (TB-Sedentary, 6.0 ± 0.3 vs. TB-Ex Trained, 12.2 ± 1.0 mmHg, P < 0.05). Exercise training did not affect the number of patent vessels but did significantly reduce tumor hypoxia in the conscious, resting condition from 39 ± 12% of the tumor area in TB-Sedentary to 4 ± 1% in TB-Ex Trained. Exercise training did not affect vessel contractile function. These results demonstrate that after exercise training, there is a large increase in the driving force of O2 from the tumor microcirculation, which likely contributes to the considerable reduction in tumor hypoxia. These results suggest that exercise training can modulate the microenvironment of the tumor, such that a sustained reduction in tumor hypoxia occurs, which may lead to a less aggressive phenotype and improve patient prognosis.
Collapse
Affiliation(s)
- Danielle J McCullough
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, Florida
| | | | | | | |
Collapse
|
41
|
Abstract
Prostate cancer hypoxia is associated with inferior prognosis and resistance to treatment. The use of androgen deprivation therapy, both prior to and during radiotherapy, may exacerbate underlying hypoxia. Whilst larger radiation doses per fraction may achieve therapeutic gain, this is balanced by the reduced opportunity for re-oxygenation to take place during the course of treatment. Improving the underlying hypoxic tumour environment may therefore improve the treatment outcomes. Strategies to combat tumour hypoxia, with particular focus on the use of carbogen gas breathing concurrently with radiotherapy, is the subject of this review.
Collapse
Affiliation(s)
- Kent Yip
- Department of Oncology, Mount Vernon Cancer Centre, Northwood, UK
| | | |
Collapse
|
42
|
Ranasinghe WKB, Xiao L, Kovac S, Chang M, Michiels C, Bolton D, Shulkes A, Baldwin GS, Patel O. The role of hypoxia-inducible factor 1α in determining the properties of castrate-resistant prostate cancers. PLoS One 2013; 8:e54251. [PMID: 23342109 PMCID: PMC3546972 DOI: 10.1371/journal.pone.0054251] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 12/10/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Castrate-resistant prostate cancer (CRPC) is a lethal condition in patients receiving androgen deprivation therapy for prostate cancer (PC). Despite numerous studies showing the expression of HIF1α protein under normoxia in PC cell lines, the role of this normoxic HIF1α expression in chemo-resistance and migration has not been investigated previously. As no method is currently available to determine which tumors will progress to CRPC, the role of HIF1α in PC and its potential for predicting the development of CRPC was also investigated. METHODS The effect of HIF1α protein knockdown on chemo-resistance and migration of PC3 cells was assessed by cell counting and Transwell assays, respectively. Translation efficiency of HIF1α mRNA was determined in PC cells using a HIF1α 5'UTR-luciferase construct. Clinical outcomes were correlated following the staining of 100 prostate tumors for HIF1α expression. RESULTS The CRPC-like cell lines (PC3 and DU145) expressed more HIF1α protein than an androgen sensitive cell line (LNCaP). Migration rate and chemo-resistance were higher in the PC3 cells and both were decreased when HIF1α expression was reduced. Increased translation of HIF1α mRNA may be responsible for HIF1α overexpression in PC3 cells. Patients whose tumors expressed HIF1α had significantly decreased metastasis-free survival and the patients who were on androgen-deprivation therapy had decreased CRPC-free survival on Kaplan-Meier analysis. On multivariate analysis HIF1α was an independent risk factor for progression to metastatic PC (Hazard ratio (HR) 9.8, p = 0.017) and development of CRPC (HR 10.0, p = 0.021) in patients on androgen-deprivation therapy. Notably the tumors which did not express HIF1α did not metastasize or develop CRPC. CONCLUSIONS HIF1α is likely to contribute to metastasis and chemo-resistance of CRPC and targeted reduction of HIF1α may increase the responsiveness of CRPCs to chemotherapy. Expression of HIF1α may be a useful screening tool for development of CRPC.
Collapse
Affiliation(s)
- Weranja K B Ranasinghe
- Department of Surgery, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Milosevic M, Warde P, Ménard C, Chung P, Toi A, Ishkanian A, McLean M, Pintilie M, Sykes J, Gospodarowicz M, Catton C, Hill RP, Bristow R. Tumor hypoxia predicts biochemical failure following radiotherapy for clinically localized prostate cancer. Clin Cancer Res 2012; 18:2108-14. [PMID: 22465832 DOI: 10.1158/1078-0432.ccr-11-2711] [Citation(s) in RCA: 214] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Tumor hypoxia is an important determinant of outcome in many human malignancies and is associated with treatment resistance and metastases. The aim of this study was to determine the effect of hypoxia in patients with prostate cancer treated with radiotherapy. EXPERIMENTAL DESIGN Tumor hypoxia was measured in 247 patients with clinically localized prostate cancer before radiotherapy, with or without hormonal therapy. The median pO(2) was 6.8 mm Hg and the median hypoxic percentage less than 10 mm Hg (HP(10)) was 63%. The median follow-up was 6.6 years. RESULTS The 5-year biochemical relapse-free rate (bRFR) was 78%. Prostrate-specific antigen and Gleason score were both associated with biochemical relapse and formed a baseline clinical model. The effect of hypoxia was found to vary with the duration of patient follow-up. HP(10), when added to the clinical model, was an independent predictor of early bRFR (P = 0.019). The relationship between hypoxia and early bRFR was more pronounced when the analysis was restricted to 142 patients with bulk tumor at the site of the oxygen measurements (P = 0.004). Hypoxia was the only factor predictive of local recurrence in 70 patients who had biopsies conducted during follow-up (P = 0.043), again with the effect being greatest early after completing treatment. CONCLUSIONS This is the largest clinical study of prostate cancer hypoxia with direct measurement of tumor oxygen levels. It shows that hypoxia is associated with early biochemical relapse after radiotherapy and also with local recurrence in the prostate gland.
Collapse
Affiliation(s)
- Michael Milosevic
- Radiation Medicine Program, Princess Margaret Hospital and Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, ON, Canada M5G 2M9.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Wagh NK, Zhou Z, Ogbomo SM, Shi W, Brusnahan SK, Garrison JC. Development of hypoxia enhanced 111In-labeled Bombesin conjugates: design, synthesis, and in vitro evaluation in PC-3 human prostate cancer. Bioconjug Chem 2012; 23:527-37. [PMID: 22296619 DOI: 10.1021/bc200600w] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The gastrin-releasing peptide receptor (BB2r) has shown great promise for tumor targeting due to the increase of the receptor expression in a variety of human cancers including prostate, breast, small-cell lung, and pancreatic cancer. From clinical investigations, prostate cancer has been shown to be among the most hypoxic of the cancers investigated. Many solid tumors contain regions of hypoxia due to poor organization and efficiency of the vasculature. However, hypoxia is typically not present in normal tissue. Nitroimidazoles, a thoroughly investigated class of hypoxia selective drugs, have been shown to be highly retained in hypoxic tissues. The purpose of this study is to determine if the incorporation of hypoxia trapping moieties into the structural paradigm of BB2r-targeted peptides will increase the retention time of the agents in prostate cancer tumors. The present work involves the design, syntheses, purification, and in vitro investigation of hypoxia enhanced (111)In-BB2r-targeted radioconjugates. A total of four BB2r-targeted conjugates (1-4) were synthesized and coupled with increasing numbers of 2-nitroimidazoles, a hypoxia trapping moiety. Conjugates were radiolabeled with (111)In and purified by HPLC prior to in vitro studies. Receptor saturation assays under both normoxic and hypoxic conditions showed that the BB2r receptor expression on the PC-3 human prostate cancer cell line was not significantly affected by oxygen levels. Competitive binding assays revealed that incorporation of 2-nitroimidazoles had a detrimental effect to BB2r binding when adequate spacer groups, between the hypoxia trapping agent and the pharmacophore, were not employed. All of the 2-nitroimidazole containing BB2r-targeted agents exhibited significantly higher longitudinal retention in PC-3 cells under hypoxic conditions compared to the analogous normoxic studies. Protein association analysis revealed a 3-fold increase in binding of a 2-nitroimidazole containing BB2r-targeted agent under hypoxic relative to normoxic conditions. The positive nature of these results indicate that further exploration into the potential of hypoxia selective trapping agents for BB2r-targeted agents, as well as other targeted compounds, is warranted.
Collapse
Affiliation(s)
- Nilesh K Wagh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | | | | | | | |
Collapse
|
45
|
Liu SK, Bham SAS, Fokas E, Beech J, Im J, Cho S, Harris AL, Muschel RJ. Delta-like ligand 4-notch blockade and tumor radiation response. J Natl Cancer Inst 2011; 103:1778-98. [PMID: 22010178 DOI: 10.1093/jnci/djr419] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The microenvironment plays an important role in regulating tumor response to radiotherapy. Ionizing radiation can disrupt tumor vasculature, and Notch pathway inhibition can interfere with functional angiogenesis. We explored the potential cooperativity between Notch inhibition and ionizing radiation in delaying tumor growth. METHODS Human colorectal carcinoma LS174T cells, which express the Notch ligand delta-like ligand 4 (DLL4), and human head and neck cancer FaDu cells, which do not, were grown as subcutaneous xenografts in nude mice. The mice were treated with dibenzazepine (DBZ), a γ-secretase inhibitor that blocks all Notch signaling, or a DLL4-specific blocking monoclonal antibody, alone or in combination with ionizing radiation (n = 5-10 mice per group), and response was assessed by tumor growth delay. Microbubble contrast Doppler ultrasound was used to measure tumor blood flow. Tumor Notch activity was monitored by in vivo bioluminescence from a Notch luciferase reporter. Vessel density was assessed using Chalkley vessel counting. All statistical tests were two-sided. RESULTS In LS174T xenografts, the average time for tumor volumes to reach four times the starting volume was longer for mice treated with the DLL4 monoclonal antibody than for mice treated with DBZ (16.4 vs 9.5 days, difference = 6.9 days, 95% confidence interval [CI] = 3.7 to 10.1 days, P < .001). Both Notch inhibitors suppressed tumor Notch activity within 24 hours of administration compared with vehicle (change in luciferase activity, vehicle vs DBZ: 103% vs 28%, difference = 75%, 95% CI = 39% to 109%, P = .002; vehicle vs DLL4 antibody: 172% vs 26%, difference = 146%, 95% CI = 86% to 205%, P < .001). Administration of the DLL4 antibody or DBZ after ionizing radiation resulted in a supra-additive growth delay compared with vehicle (vehicle vs DLL4 antibody + ionizing radiation: 6.8 vs 44.3 days, difference = 37.5 days, 95% CI = 32 to 43 days, P < .001; vehicle vs DBZ + ionizing radiation: 7.1 vs 24.4 days, difference = 17.3 days, 95% CI = 15.9 to 18.6 days, P < .001). Treatment of mice with the DLL4 antibody alone or in combination with ionizing radiation increased tumor vessel density but reduced tumor blood flow. Combination therapy with DLL4 antibody and ionizing radiation resulted in extensive tumor necrosis in LS174T xenografts and enhanced tumor growth delay in FaDu xenografts. CONCLUSION The combination of specific DLL4-Notch blockade and ionizing radiation impairs tumor growth by promoting nonfunctional tumor angiogenesis and extensive tumor necrosis, independent of tumor DLL4 expression.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Adenocarcinoma/blood supply
- Adenocarcinoma/metabolism
- Adenocarcinoma/radiotherapy
- Amyloid Precursor Protein Secretases/antagonists & inhibitors
- Animals
- Antibodies, Monoclonal/pharmacology
- Antineoplastic Agents/pharmacology
- Blood Flow Velocity
- Calcium-Binding Proteins
- Carcinoma, Squamous Cell/blood supply
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/radiotherapy
- Cell Line, Tumor
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/radiotherapy
- Combined Modality Therapy
- Dibenzazepines/pharmacology
- Humans
- Hypopharyngeal Neoplasms/metabolism
- Hypopharyngeal Neoplasms/radiotherapy
- Immunoblotting
- Immunohistochemistry
- Intercellular Signaling Peptides and Proteins/immunology
- Luminescent Measurements
- Mice
- Mice, Nude
- Necrosis
- Neoplasms/blood supply
- Neoplasms/metabolism
- Neoplasms/radiotherapy
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/prevention & control
- Neovascularization, Pathologic/radiotherapy
- Radiation, Ionizing
- Receptors, Notch/antagonists & inhibitors
- Signal Transduction
- Transplantation, Heterologous
- Ultrasonography, Doppler
Collapse
Affiliation(s)
- Stanley K Liu
- DPhil, Molecular Oncology Unit, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Firlej V, Mathieu JRR, Gilbert C, Lemonnier L, Nakhlé J, Gallou-Kabani C, Guarmit B, Morin A, Prevarskaya N, Delongchamps NB, Cabon F. Thrombospondin-1 triggers cell migration and development of advanced prostate tumors. Cancer Res 2011; 71:7649-58. [PMID: 22037878 DOI: 10.1158/0008-5472.can-11-0833] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The antitumor effects of pharmacologic inhibitors of angiogenesis are hampered in patients by the rapid development of tumor resistance, notably through increased invasiveness and accelerated metastasis. Here, we reevaluated the role of the endogenous antiangiogenic thrombospondin 1 (TSP1) in prostate carcinomas in which angiogenesis is an active process. In xenografted tumors, we observed that TSP1 altogether inhibited angiogenesis and fostered tumor development. Our results show that TSP1 is a potent stimulator of prostate tumor cell migration. This effect required CD36, which also mediates TSP1 antiangiogenic activity, and was mimicked by an antiangiogenic TSP1-derived peptide. As suspected for pharmacologic inhibitors of angiogenesis, the TSP1 capacities to increase hypoxia and to trigger cell migration are thus inherently linked. Importantly, although antiangiogenic TSP1 increases hypoxia in vivo, our data show that, in turn, hypoxia induced TSP1, thus generating a vicious circle in prostate tumors. In radical prostatectomy specimens, we found TSP1 expression significantly associated with invasive tumors and with tumors which eventually recurred. TSP1 may thus help select patients at risk of prostate-specific antigen relapse. Together, the data suggest that intratumor disruption of the hypoxic cycle through TSP1 silencing will limit tumor invasion.
Collapse
|
47
|
Investigation on tumor hypoxia in resectable primary prostate cancer as demonstrated by 18F-FAZA PET/CT utilizing multimodality fusion techniques. Eur J Nucl Med Mol Imaging 2011; 38:1816-23. [PMID: 21833840 DOI: 10.1007/s00259-011-1876-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 06/26/2011] [Indexed: 02/04/2023]
Abstract
PURPOSE As hypoxia is believed to play an important role in the development and progression of prostate cancer, we evaluated whether 18F-labeled fluoroazomycin arabinoside (18F-FAZA) would be useful to identify tumor hypoxia in resectable prostate cancer. METHODS Positron emission tomography (PET)/CT was performed on 14 patients with untreated localized primary prostate cancer 3 h post-injection of approximately 390 MBq of 18F-FAZA using forced diuresis to decrease radioactivity in the urinary bladder. Anatomical trans-pelvic coil and pre- and post-contrast 1.5 T MRI with endorectal coil were performed on the same day. Patients underwent radical prostatectomy and ex vivo 3 T MRI of the prostatectomy specimen within 14 days following in vivo imaging. Imaging results were verified by whole mount histopathology plus tissue microarray (TMA) immunohistochemical (IHC) analysis for carbonic anhydrase IX (CAIX) and hypoxia-inducible factor 1α (HIF-1α). Registration of in vivo imaging with histology was achieved using mutual information software and performing ex vivo MRI of the prostatectomy specimen and whole mount sectioning with block face photography as intermediate steps. RESULTS Whole mount histology identified 43 tumor nodules, 19 of them larger than 1 ml as determined on coregistered volumes featuring 18F-FAZA, MRI, and histological 3-D image information. None of these lesions was found to be positive for CAIX or visualized by 18F-FAZA PET/CT while IHC for HIF-1α showed variable staining of tumor tissues. Accordingly, no correlation was found between 18F-FAZA uptake and Gleason scores. CONCLUSION Our data based on 18F-FAZA PET/CT and CAIX IHC do not support the presence of clinically relevant hypoxia in localized primary prostate cancer including high-grade disease. Activation of HIF-1α may be independent of tissue hypoxia in primary prostate cancer.
Collapse
|
48
|
Bentzen SM, Gregoire V. Molecular imaging-based dose painting: a novel paradigm for radiation therapy prescription. Semin Radiat Oncol 2011; 21:101-10. [PMID: 21356478 PMCID: PMC3052283 DOI: 10.1016/j.semradonc.2010.10.001] [Citation(s) in RCA: 217] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dose painting is the prescription of a nonuniform radiation dose distribution to the target volume based on functional or molecular images shown to indicate the local risk of relapse. Two prototypical strategies for implementing this novel paradigm in radiation oncology are reviewed: subvolume boosting and dose painting by numbers. Subvolume boosting involves the selection of a "target within the target," defined by image segmentation on the basis of the quantitative information in the image or morphologically, and this is related to image-based target volume selection and delineation. Dose painting by numbers is a voxel-level prescription of dose based on a mathematical transformation of the image intensity of individual pixels. The quantitative use of images to decide both where and how to delivery radiation therapy in an individual case is also called theragnostic imaging. Dose painting targets are imaging surrogates for cellular or microenvironmental phenotypes associated with poor radioresponsiveness. In this review, the focus is on the following positron emission tomography tracers: FDG and choline as surrogates for tumor burden, fluorothymidine as a surrogate for proliferation (or cellular growth fraction) and hypoxia-sensitive tracers, including [(18)F] fluoromisonidazole, EF3, EF5, and (64)Cu-labeled copper(II) diacetyl-di(N(4)-methylthiosemicarbazone) as surrogates of cellular hypoxia. Research advances supporting the clinicobiological rationale for dose painting are reviewed as are studies of the technical feasibility of optimizing and delivering realistic dose painted radiation therapy plans. Challenges and research priorities in this exciting research field are defined and a possible design for a randomized clinical trial of dose painting is presented.
Collapse
Affiliation(s)
- Søren M Bentzen
- Departments of Human Oncology, Medical Physics, Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI 53792, USA.
| | | |
Collapse
|
49
|
Cozzarini C, Fiorino C, Da Pozzo LF, Alongi F, Berardi G, Bolognesi A, Briganti A, Broggi S, Deli A, Guazzoni G, Perna L, Pasetti M, Salvadori G, Montorsi F, Rigatti P, Di Muzio N. Clinical factors predicting late severe urinary toxicity after postoperative radiotherapy for prostate carcinoma: a single-institute analysis of 742 patients. Int J Radiat Oncol Biol Phys 2010; 82:191-9. [PMID: 21109361 DOI: 10.1016/j.ijrobp.2010.09.027] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 09/14/2010] [Accepted: 09/24/2010] [Indexed: 11/19/2022]
Abstract
PURPOSE To investigate the clinical factors independently predictive of long-term severe urinary sequelae after postprostatectomy radiotherapy. PATIENTS AND METHODS Between 1993 and 2005, 742 consecutive patients underwent postoperative radiotherapy with either adjuvant (n = 556; median radiation dose, 70.2 Gy) or salvage (n = 186; median radiation dose, 72 Gy) intent. RESULTS After a median follow-up of 99 months, the 8-year risk of Grade 2 or greater and Grade 3 late urinary toxicity was almost identical (23.9% vs. 23.7% and 12% vs. 10%) in the adjuvant and salvage cohorts, respectively. On univariate analysis, acute toxicity was significantly predictive of late Grade 2 or greater sequelae in both subgroups (p <.0001 in both cases), and hypertension (p = .02) and whole-pelvis radiotherapy (p = .02) correlated significantly in the adjuvant cohort only. The variables predictive of late Grade 3 sequelae were acute Grade 2 or greater toxicity in both groups and whole-pelvis radiotherapy (8-year risk of Grade 3 events, 21% vs. 11%, p = .007), hypertension (8-year risk, 18% vs. 10%, p = .005), age ≤ 62 years at RT (8-year risk, 16% vs. 11%, p = .04) in the adjuvant subset, and radiation dose >72 Gy (8-year risk, 19% vs. 6%, p = .007) and age >71 years (8-year risk, 16% vs. 6%, p = .006) in the salvage subgroup. Multivariate analysis confirmed the independent predictive role of all the covariates indicated as statistically significant on univariate analysis. CONCLUSIONS The risk of late Grade 2 or greater and Grade 3 urinary toxicity was almost identical, regardless of the RT intent. In the salvage cohort, older age and greater radiation doses resulted in a worse toxicity profile, and younger, hypertensive patients experienced a greater rate of severe late sequelae in the adjuvant setting. The causes of this latter correlation and apparently different etiopathogenesis of chronic damage in the two subgroups were unclear and deserve additional investigation.
Collapse
Affiliation(s)
- Cesare Cozzarini
- Department of Radiotherapy, San Raffaele Scientific Institute, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Morphology of hypoxia following cryoablation in a prostate cancer murine model: Its relationship to necrosis, apoptosis and, microvessel density. Cryobiology 2010; 61:148-54. [DOI: 10.1016/j.cryobiol.2010.06.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 06/22/2010] [Indexed: 11/20/2022]
|