1
|
Kopecka J, Salaroglio IC, Perez-Ruiz E, Sarmento-Ribeiro AB, Saponara S, De Las Rivas J, Riganti C. Hypoxia as a driver of resistance to immunotherapy. Drug Resist Updat 2021; 59:100787. [PMID: 34840068 DOI: 10.1016/j.drup.2021.100787] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023]
Abstract
Hypoxia, a hallmark of solid tumors, determines the selection of invasive and aggressive malignant clones displaying resistance to radiotherapy, conventional chemotherapy or targeted therapy. The recent introduction of immunotherapy, based on immune checkpoint inhibitors (ICPIs) and chimeric antigen receptor (CAR) T-cells, has markedly transformed the prognosis in some tumors but also revealed the existence of intrinsic or acquired drug resistance. In the current review we highlight hypoxia as a culprit of immunotherapy failure. Indeed, multiple metabolic cross talks between tumor and stromal cells determine the prevalence of immunosuppressive populations within the hypoxic tumor microenvironment and confer upon tumor cells resistance to ICPIs and CAR T-cells. Notably, hypoxia-triggered angiogenesis causes immunosuppression, adding another piece to the puzzle of hypoxia-induced immunoresistance. If these factors concurrently contribute to the resistance to immunotherapy, they also unveil an unexpected Achille's heel of hypoxic tumors, providing the basis for innovative combination therapies that may rescue the efficacy of ICPIs and CAR T-cells. Although these treatments reveal both a bright side and a dark side in terms of efficacy and safety in clinical trials, they represent the future solution to enhance the efficacy of immunotherapy against hypoxic and therapy-resistant solid tumors.
Collapse
Affiliation(s)
| | | | - Elizabeth Perez-Ruiz
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, IBIMA, Málaga, Spain
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology and University Clinic of Hematology and Coimbra Institute for Clinical and Biomedical Research - Group of Environment Genetics and Oncobiology (iCBR/CIMAGO), Faculty of Medicine, University of Coimbra (FMUC), Center for Innovative Biomedicine and Biotechnology (CIBB) and Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | | | - Javier De Las Rivas
- Cancer Research Center (CiC-IBMCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca (USAL), and Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | | |
Collapse
|
2
|
Akman M, Belisario DC, Salaroglio IC, Kopecka J, Donadelli M, De Smaele E, Riganti C. Hypoxia, endoplasmic reticulum stress and chemoresistance: dangerous liaisons. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:28. [PMID: 33423689 PMCID: PMC7798239 DOI: 10.1186/s13046-020-01824-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023]
Abstract
Solid tumors often grow in a micro-environment characterized by < 2% O2 tension. This condition, together with the aberrant activation of specific oncogenic patwhays, increases the amount and activity of the hypoxia-inducible factor-1α (HIF-1α), a transcription factor that controls up to 200 genes involved in neoangiogenesis, metabolic rewiring, invasion and drug resistance. Hypoxia also induces endoplasmic reticulum (ER) stress, a condition that triggers cell death, if cells are irreversibly damaged, or cell survival, if the stress is mild.Hypoxia and chronic ER stress both induce chemoresistance. In this review we discuss the multiple and interconnected circuitries that link hypoxic environment, chronic ER stress and chemoresistance. We suggest that hypoxia and ER stress train and select the cells more adapted to survive in unfavorable conditions, by activating pleiotropic mechanisms including apoptosis inhibition, metabolic rewiring, anti-oxidant defences, drugs efflux. This adaptative process unequivocally expands clones that acquire resistance to chemotherapy.We believe that pharmacological inhibitors of HIF-1α and modulators of ER stress, although characterized by low specificty and anti-cancer efficacy when used as single agents, may be repurposed as chemosensitizers against hypoxic and chemorefractory tumors in the next future.
Collapse
Affiliation(s)
- Muhlis Akman
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino, Italy
| | | | | | - Joanna Kopecka
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino, Italy
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Enrico De Smaele
- Department of Experimental Medicine, Sapienza University of Roma, Roma, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino, Italy.
| |
Collapse
|
3
|
Roy S, Kumaravel S, Sharma A, Duran CL, Bayless KJ, Chakraborty S. Hypoxic tumor microenvironment: Implications for cancer therapy. Exp Biol Med (Maywood) 2020; 245:1073-1086. [PMID: 32594767 DOI: 10.1177/1535370220934038] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
IMPACT STATEMENT Hypoxia contributes to tumor aggressiveness and promotes growth of many solid tumors that are often resistant to conventional therapies. In order to achieve successful therapeutic strategies targeting different cancer types, it is necessary to understand the molecular mechanisms and signaling pathways that are induced by hypoxia. Aberrant tumor vasculature and alterations in cellular metabolism and drug resistance due to hypoxia further confound this problem. This review focuses on the implications of hypoxia in an inflammatory TME and its impact on the signaling and metabolic pathways regulating growth and progression of cancer, along with changes in lymphangiogenic and angiogenic mechanisms. Finally, the overarching role of hypoxia in mediating therapeutic resistance in cancers is discussed.
Collapse
Affiliation(s)
- Sukanya Roy
- Department of Medical Physiology, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807, USA
| | - Subhashree Kumaravel
- Department of Medical Physiology, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807, USA
| | - Ankith Sharma
- Department of Medical Physiology, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807, USA
| | - Camille L Duran
- Department of Molecular & Cellular Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Kayla J Bayless
- Department of Molecular & Cellular Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Sanjukta Chakraborty
- Department of Medical Physiology, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807, USA
| |
Collapse
|
4
|
Zhang H, Liang F, Yue J, Liu P, Wang J, Wang Z, Li H, Cheng D, Du J, Zhang K, Du P. MicroRNA‑137 regulates hypoxia‑mediated migration and epithelial‑mesenchymal transition in prostate cancer by targeting LGR4 via the EGFR/ERK signaling pathway. Int J Oncol 2020; 57:540-549. [PMID: 32626928 DOI: 10.3892/ijo.2020.5064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 04/15/2020] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs (miRs) serve an integral role in prostate cancer. The present study aimed to investigate the effects and mechanisms of miR‑137 in hypoxia‑mediated migration and epithelial‑mesenchymal transition (EMT). PC3 and DU145 prostate cancer cells were exposed to hypoxia for 24 h, after which the expression of miR‑137 was determined by reverse transcription‑quantitative PCR (RT‑qPCR). The cells were transfected with a miR‑137 mimic or inhibitor, followed by hypoxia exposure. The results demonstrated that hypoxia reduced miR‑137 expression. Further results from the Cell Counting Kit‑8, Cell Death Detection ELISA plus kit, Transwell assay, RT‑qPCR and western blotting assays revealed that the miR‑137 mimic prevented cell proliferation, facilitated apoptosis and repressed cell migration, invasiveness, and expression of N‑cadherin, vimentin and matrix metalloproteinase 2; the miR‑137 inhibitor exerted the opposite effects. A dual‑-luciferase reporter assay determined that miR‑137 directly targeted leucine‑rich repeat‑containing G protein‑coupled receptor 4 (LGR4). Additionally, miR‑137 negatively regulated the epidermal growth factor receptor/extracellular signal‑-regulated kinase (EGFR/ERK) signaling pathway by targeting LGR4. LGR4 silencing or EGFR/ERK inhibition abolished the effects of miR‑137 inhibitor on cell migration and EMT. In conclusion, by targeting LGR4 via the EGFR/ERK signaling pathway, miR‑137 inhibited prostate cancer cell migration and EMT.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Urology, Zhengzhou Central Hospital Affiliated to Zhengzhou University (Zhengzhou Central Hospital), Zhengzhou, Henan 450007, P.R. China
| | - Fang Liang
- Department of Oncology, Zhengzhou Central Hospital Affiliated to Zhengzhou University (Zhengzhou Central Hospital), Zhengzhou, Henan 450007, P.R. China
| | - Junmin Yue
- Department of Urology, Zhengzhou Central Hospital Affiliated to Zhengzhou University (Zhengzhou Central Hospital), Zhengzhou, Henan 450007, P.R. China
| | - Peng Liu
- Department of Urology, Zhengzhou Central Hospital Affiliated to Zhengzhou University (Zhengzhou Central Hospital), Zhengzhou, Henan 450007, P.R. China
| | - Junyong Wang
- Department of Urology, Zhengzhou Central Hospital Affiliated to Zhengzhou University (Zhengzhou Central Hospital), Zhengzhou, Henan 450007, P.R. China
| | - Zhaoyang Wang
- Department of Urology, Zhengzhou Central Hospital Affiliated to Zhengzhou University (Zhengzhou Central Hospital), Zhengzhou, Henan 450007, P.R. China
| | - Hongxing Li
- Department of Urology, Zhengzhou Central Hospital Affiliated to Zhengzhou University (Zhengzhou Central Hospital), Zhengzhou, Henan 450007, P.R. China
| | - Duo Cheng
- Department of Oncology, Zhengzhou Central Hospital Affiliated to Zhengzhou University (Zhengzhou Central Hospital), Zhengzhou, Henan 450007, P.R. China
| | - Jie Du
- Department of Oncology, Zhengzhou Central Hospital Affiliated to Zhengzhou University (Zhengzhou Central Hospital), Zhengzhou, Henan 450007, P.R. China
| | - Kai Zhang
- Department of Urology, Zhengzhou Central Hospital Affiliated to Zhengzhou University (Zhengzhou Central Hospital), Zhengzhou, Henan 450007, P.R. China
| | - Peng Du
- Department of Urology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing ), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| |
Collapse
|
5
|
Effect of optimal combination of Huangqi (Radix Astragali Mongolici) and Ezhu (Rhizoma Curcumae Phaeocaulis) on proliferation and apoptosis of A549 lung cancer cells. J TRADIT CHIN MED 2018. [DOI: 10.1016/s0254-6272(18)30625-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
6
|
Doktorova H, Hrabeta J, Khalil MA, Eckschlager T. Hypoxia-induced chemoresistance in cancer cells: The role of not only HIF-1. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2015; 159:166-77. [PMID: 26001024 DOI: 10.5507/bp.2015.025] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/07/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The aim of this review is to provide the information about molecular basis of hypoxia-induced chemoresistance, focusing on the possibility of diagnostic and therapeutic use. RESULTS Hypoxia is a common feature of tumors and represents an independent prognostic factor in many cancers. It is the result of imbalances in the intake and consumption of oxygen caused by abnormal vessels in the tumor and the rapid proliferation of cancer cells. Hypoxia-induced resistance to cisplatin, doxorubicin, etoposide, melphalan, 5-flouoruracil, gemcitabine, and docetaxel has been reported in a number of experiments. Adaptation of tumor cells to hypoxia has important biological effects. The most studied factor responsible for these effects is hypoxia-inducible factor-1 (HIF-1) that significantly contributes to the aggressiveness and chemoresistance of different tumors. The HIF-1 complex, induced by hypoxia, binds to target genes, thereby increasing the expression of many genes. In addition, the expression of hundreds of genes can be also decreased in response to hypoxia in HIF-1 dependent manner, but without the detection of HIF-1 in these genes' promoters. HIF-1 independent mechanisms for drug resistance in hypoxia have been described, however, they are still rarely reported. The first clinical studies focusing on diagnosis of hypoxia and on inhibition of hypoxia-induced changes in cancer cells are starting to yield results. CONCLUSIONS The adaptation to hypoxia requires many genetic and biochemical responses that regulate one another. Hypoxia-induced resistance is a very complex field and we still know very little about it. Different approaches to circumvent hypoxia in tumors are under development.
Collapse
Affiliation(s)
- Helena Doktorova
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Jan Hrabeta
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Mohamed Ashraf Khalil
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Tomas Eckschlager
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
7
|
Neschadim A, Pritzker LB, Pritzker KPH, Branch DR, Summerlee AJS, Trachtenberg J, Silvertown JD. Relaxin receptor antagonist AT-001 synergizes with docetaxel in androgen-independent prostate xenografts. Endocr Relat Cancer 2014; 21:459-71. [PMID: 24812057 DOI: 10.1530/erc-14-0088] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Androgen hormones and the androgen receptor (AR) pathway are the main targets of anti-hormonal therapies for prostate cancer. However, resistance inevitably develops to treatments aimed at the AR pathway resulting in androgen-independent or hormone-refractory prostate cancer (HRPC). Therefore, there is a significant unmet need for new, non-androgen anti-hormonal strategies for the management of prostate cancer. We demonstrate that a relaxin hormone receptor antagonist, AT-001, an analog of human H2 relaxin, represents a first-in-class anti-hormonal candidate treatment designed to significantly curtail the growth of androgen-independent human prostate tumor xenografts. Chemically synthesized AT-001, administered subcutaneously, suppressed PC3 xenograft growth by up to 60%. AT-001 also synergized with docetaxel, standard first-line chemotherapy for HRPC, to suppress tumor growth by more than 98% in PC3 xenografts via a mechanism involving the downregulation of hypoxia-inducible factor 1 alpha and the hypoxia-induced response. Our data support developing AT-001 for clinical use as an anti-relaxin hormonal therapy for advanced prostate cancer.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Binding, Competitive
- Blotting, Western
- Cell Proliferation/drug effects
- Docetaxel
- Drug Synergism
- Humans
- Immunoenzyme Techniques
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Neoplasms, Hormone-Dependent/drug therapy
- Neoplasms, Hormone-Dependent/metabolism
- Neoplasms, Hormone-Dependent/pathology
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Receptors, Androgen/metabolism
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, Peptide/antagonists & inhibitors
- Taxoids/pharmacology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Anton Neschadim
- Armour Therapeutics Inc., 124 Orchard View Boulevard, Toronto, Ontario, Canada Rna Diagnostics Inc., 595 Bay Street, Suite 1204, Toronto, Ontario, Canada Departments of Laboratory Medicine and Pathobiology Surgery, University of Toronto, Toronto, Ontario, Canada Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada Departments of Medicine Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada Centre for Innovation, Canadian Blood Services, Toronto, Ontario, Canada Division of Advanced Diagnostics - Infection and Immunity, Toronto General Research Institute (TGRI), University Health Network, Toronto, Ontario, Canada Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada Departments of Surgery and Medical Imaging, University of Toronto, Toronto, Ontario, Canada Division of Urology, Department of Surgical Oncology Prostate Centre, Princess Margaret Hospital Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
8
|
Terraneo L, Bianciardi P, Caretti A, Ronchi R, Samaja M. Chronic systemic hypoxia promotes LNCaP prostate cancer growth in vivo. Prostate 2010; 70:1243-54. [PMID: 20333700 DOI: 10.1002/pros.21160] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Solid tumors contain underperfused regions where hypoxia-inducible factor-1alpha (HIF-1alpha) over-expression induces hypoxia adaptation and cell proliferation. We test the hypothesis that systemic hypoxia promotes prostate cancer growth in vivo and examine HIF-1alpha centrality in this effect. METHODS Male athymic mice were xenografted with 3 x 10(6) LNCaP cells per each flank and exposed for 28 days to either chronic hypoxia (CH, 10% O(2)) or CH with reoxygenation (CHReox, 3 times/week for 1 hr), with normoxia as control (n = 17, 9, and 20, respectively). At the end of the observation, mice were euthanized and tumors harvested for analyses. RESULTS The successful xenografts grew faster in CH and CHReox than in normoxia (first-order rate constants 0.15 +/- 0.01, 0.18 +/- 0.03, and 0.09 +/- 0.01 day(-1), P < 0.05, n = 18, 15, and 25, respectively). Furthermore, the tumor masses at the end were 4.09 +/- 0.58, 3.42 +/- 0.55, and 1.86 +/- 0.25 mg/g bw (P < 0.05), respectively. HIF-1alpha, assayed by Western blot and immunofluorescence, was slightly increased in CH with respect to normoxia, but markedly over-expressed (5-10 times) in CHReox (P < 0.001). The tumor hemoglobin content, higher in CH and CHReox than in normoxia, reflected the higher blood hemoglobin concentration, not neovascularization, as supported by similar expression levels of vascular endothelial growth factor (VEGF) in the three groups. By contrast, protein kinase B (Akt) was more phosphorylated in both hypoxic groups than in normoxia (P < 0.01). CONCLUSION In vivo systemic hypoxia promotes prostate cancer growth regardless of HIF-1alpha expression level and neovascularization, suggesting an important role for hypoxia-dependent pathways that do not involve HIF-1alpha, as the phosphatidyl inositol-3-phosphate signaling cascade.
Collapse
Affiliation(s)
- Laura Terraneo
- Department of Medicine, Surgery and Dentistry, San Paolo Hospital, University of Milan, Milan, Italy
| | | | | | | | | |
Collapse
|
9
|
Walsh S, Gill C, O'Neill A, Fitzpatrick JM, Watson RWG. Hypoxia increases normal prostate epithelial cell resistance to receptor-mediated apoptosisviaAKT activation. Int J Cancer 2009; 124:1871-8. [DOI: 10.1002/ijc.24145] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
10
|
Marignol L, Foley R, Southgate TD, Coffey M, Hollywood D, Lawler M. Hypoxia response element-driven cytosine deaminase/5-fluorocytosine gene therapy system: a highly effective approach to overcome the dynamics of tumour hypoxia and enhance the radiosensitivity of prostate cancer cellsin vitro. J Gene Med 2009; 11:169-79. [DOI: 10.1002/jgm.1281] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
11
|
Daly PJ, Docherty NG, Healy DA, McGuire BB, Fitzpatrick JM, Watson RWG. The single insult of hypoxic preconditioning induces an antiapoptotic response in human proximal tubular cells, in vitro, across cold storage. BJU Int 2008; 103:254-9. [PMID: 18782307 DOI: 10.1111/j.1464-410x.2008.08010.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To examine whether hypoxia (one of the many components of ischaemic preconditioning) can induce a protective response in culture renal tubular cells, and thus determine if non-lethal periods of hypoxia could confer protection against apoptotic injury to human proximal tubular cells during cold storage and subsequent cytotoxic insult, and establish the cellular mechanisms by which this protection is induced. MATERIALS AND METHODS Human proximal tubular cells (HK-2) were pre-incubated for 24 h in normoxic or hypoxic conditions and then incubated at 4 degrees C for 6 h to mimic cold storage, before being returned to normal conditions and exposed to varying concentrations of cyclosporine A (CSA). Cell viability and apoptosis were measured using propidium iodide staining and flow cytometry. The expression of heat-shock protein (HSP)-70 was determined by Western blotting. RESULTS Hypoxia had no effect on cell viability or apoptosis. Pre-exposure of cells to hypoxia significantly protected against CSA-induced damage even after a period of cold storage. Western blotting analysis showed that hypoxia up-regulated the anti-apoptotic protein HSP-70. HK-2 cells over-expressing HSP-70 mimicked hypoxia preconditioning, in that they were protected during cold storage and CSA-induced apoptosis. CONCLUSION Exposure of renal tubular cells to a sequential model of cold storage, reperfusion and incubation with CSA resulted in apoptotic cell death. Preconditioning these cells with hypoxia induced a protective response and up-regulation of the anti-apoptotic protein HSP-70. There was a similar response in non-preconditioned cells over-expressing HSP-70. Further understanding of the cellular changes occurring during this period of preconditioning will allow the development of more targeted, clinically relevant methods of preconditioning in renal transplantation.
Collapse
Affiliation(s)
- Pádraig J Daly
- UCD School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | | | | | | | | | | |
Collapse
|
12
|
Chan N, Milosevic M, Bristow RG. Tumor hypoxia, DNA repair and prostate cancer progression: new targets and new therapies. Future Oncol 2007; 3:329-41. [PMID: 17547528 DOI: 10.2217/14796694.3.3.329] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Increasingly, the tumor microenvironment and hypoxia are being studied as potential prognostic factors in prostate cancer given their effects on the hypoxia inducible factor-1α and vascular endothelial growth factor signaling pathways. Based on immunohistochemical studies using hypoxic cell markers and direct oxygen-electrode measurements, clinically relevant levels of hypoxia are detected in 30–90% of prostate cancers. Exciting new data suggest that hypoxia can alter cell-cycle checkpoints and DNA repair within the prostate epithelium, thereby driving genetic instability and tumor aggression. Novel therapies designed to target the hypoxic response and resulting defective DNA repair may therefore be effective as chemoprevention agents or as adjuncts to surgery, radiotherapy and chemotherapy to improve clinical outcome.
Collapse
Affiliation(s)
- Norman Chan
- Departments of Medical Biophysics & Radiation Oncology, University of Toronto, Princess Margaret Hospital (University Health Network), Toronto, Ontario, Canada.
| | | | | |
Collapse
|
13
|
Maxwell PJ, Gallagher R, Seaton A, Wilson C, Scullin P, Pettigrew J, Stratford IJ, Williams KJ, Johnston PG, Waugh DJJ. HIF-1 and NF-κB-mediated upregulation of CXCR1 and CXCR2 expression promotes cell survival in hypoxic prostate cancer cells. Oncogene 2007; 26:7333-45. [PMID: 17533374 DOI: 10.1038/sj.onc.1210536] [Citation(s) in RCA: 158] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Hypoxic cancer cells are resistant to treatment, leading to the selection of cells with a more malignant phenotype. The expression of interleukin-8 (IL-8) plays an important role in the tumorigenesis and metastasis of solid tumors including prostate cancer. Recently, we detected elevated expression of IL-8 and IL-8 receptors in human prostate cancer tissue. The objective of the current study was to determine whether hypoxia increases IL-8 and IL-8 receptor expression in prostate cancer cells and whether this contributes to a survival advantage in hypoxic cells. IL-8, CXCR1 and CXCR2 messenger RNA (mRNA) expression in PC3 cells was upregulated in response to hypoxia in a time-dependent manner. Elevated IL-8 secretion following hypoxia was detected by enzyme-linked immunosorbent assay, while immunoblotting confirmed elevated receptor expression. Attenuation of hypoxia-inducible factor (HIF-1) and nuclear factor-kappaB (NF-kappaB) transcriptional activity using small interfering RNA (siRNA), a HIF-1 dominant-negative and pharmacological inhibitors, abrogated hypoxia-induced transcription of CXCR1 and CXCR2 in PC3 cells. Furthermore, chromatin-IP analysis demonstrated binding of HIF-1 and NF-kappaB to CXCR1. Finally, inhibition of IL-8 signaling potentiated etoposide-induced cell death in hypoxic PC3 cells. These results suggest that IL-8 signaling confers a survival advantage to hypoxic prostate cancer cells, and therefore, strategies to inhibit IL-8 signaling may sensitize hypoxic tumor cells to conventional treatments.
Collapse
MESH Headings
- Cell Survival
- Chromatin Immunoprecipitation
- Enzyme-Linked Immunosorbent Assay
- Flow Cytometry
- Humans
- Hypoxia
- Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Immunoblotting
- Immunoprecipitation
- Interleukin-8/metabolism
- Male
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Receptors, Interleukin-8A/antagonists & inhibitors
- Receptors, Interleukin-8A/genetics
- Receptors, Interleukin-8A/metabolism
- Receptors, Interleukin-8B/antagonists & inhibitors
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Transcription, Genetic
- Up-Regulation
Collapse
Affiliation(s)
- P J Maxwell
- Centre for Cancer Research and Cell Biology, Queens University Belfast, Belfast, Northern Ireland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Lekas AG, Lazaris AC, Chrisofos M, Papatsoris AG, Lappas D, Patsouris E, Deliveliotis C. Finasteride effects on hypoxia and angiogenetic markers in benign prostatic hyperplasia. Urology 2006; 68:436-41. [PMID: 16904480 DOI: 10.1016/j.urology.2006.03.038] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2005] [Revised: 02/11/2006] [Accepted: 03/21/2006] [Indexed: 10/24/2022]
Abstract
OBJECTIVES To assess the effects of finasteride on angiogenetic and hypoxia markers in benign prostatic hyperplasia. METHODS A total of 178 patients aged 51 to 85 years (mean 68.7) with benign prostatic hyperplasia and awaiting transurethral prostate resection were prospectively randomized into a group of patients receiving finasteride (group 1; 88 patients) and a group of patients who received no medication until transurethral prostate resection (group 2; 90 patients). Tissue specimens were immunohistochemically stained with monoclonal antibodies against CD34 for microvessel density (MVD), vascular endothelial growth factor (VEGF), and hypoxia inducible factor-1alpha (HIF-1alpha). RESULTS Blood loss during transurethral prostate resection was significantly higher in group 2 compared with group 1 (P <0.001). The distribution of CD34 immunostaining was mainly at the suburethral prostate. MVD, VEGF, and HIF-1alpha values were significantly lower statistically (P <0.001) in group 1 compared with group 2. In the finasteride group (group 1), the positive correlation of the immunoreactivity of CD34 and HIF-1alpha, VEGF and HIF-1alpha, and VEGF and CD34 was statistically significant (P <0.001). In the same group, MVD and VEGF and HIF-1alpha expression correlated statistically with the treatment duration. CONCLUSIONS Finasteride administration in benign prostatic hyperplasia results in statistically significant suppression of MVD, VEGF, and HIF-1alpha in a time-dependent manner.
Collapse
|
15
|
O'Connor K, Gill C, Tacke M, Rehmann FJK, Strohfeldt K, Sweeney N, Fitzpatrick JM, Watson RWG. Novel titanocene anti-cancer drugs and their effect on apoptosis and the apoptotic pathway in prostate cancer cells. Apoptosis 2006; 11:1205-14. [PMID: 16699961 DOI: 10.1007/s10495-006-6796-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Advanced prostate cancer is not curable by current treatment strategies indicating a significant need for new chemotherapeutic options. Highly substituted ansa-titanocene compounds have shown promising cytotoxic activity in a range of cancers. The objectives of this study are to examine the effects of these titanocene compounds on prostate cancer cells. Prostate cell lines were treated with three novel titanocene compounds and compared to titanocene dichloride and cisplatin. Percent apoptosis, viability and cell cycle were assessed using propidium iodide DNA incorporation with flow cytometry. Cytochrome C was assessed by western blotting of mitochondrial and cytoplasmic fractions. Apoptosis Inducing Factor was assessed by confocal microscopy. These novel compounds induced more apoptosis compared to cisplatin in a dose dependent manner. Compound Y had the most significant effect on cell cycle and apoptosis. Despite the release of cytochrome C from the mitochondrial fraction there was no inhibition of apoptosis with the pan caspase inhibitor, ZVAD-FMK. AIF was shown to translocate from the cytosol to the nucleus mediating a caspase independent cell death. Bcl-2 over expressing PC-3 cells, which were resistant to cisplatin induced apoptosis, underwent apoptosis following treatment with all the titanocene compounds. This study demonstrates possible mechanisms by which these novel titanocene compounds can mediate their apoptotic effect in vitro. The fact that they can induce more apoptosis than cisplatin in advanced cancer cell lines would confer an advantage over cisplatin. They represent exciting new agents with future potential for the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- K O'Connor
- UCD School of Medicine and Medical Sciences, Mater Misericordiae University Hospital, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | | | | | | | |
Collapse
|