1
|
Robson T, Worthington J, McKeown SR, Hirst DG. Radiogenic Therapy: Novel Approaches for Enhancing Tumor Radiosensitivity. Technol Cancer Res Treat 2016; 4:343-61. [PMID: 16029055 DOI: 10.1177/153303460500400404] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy (RT) is a well established modality for treating many forms of cancer. However, despite many improvements in treatment planning and delivery, the total radiation dose is often too low for tumor cure, because of the risk of normal tissue damage. Gene therapy provides a new adjunctive strategy to enhance the effectiveness of RT, offering the potential for preferential killing of cancer cells and sparing of normal tissues. This specificity can be achieved at several levels including restricted vector delivery, transcriptional targeting and specificity of the transgene product. This review will focus on those gene therapy strategies that are currently being evaluated in combination with RT, including the use of radiation sensitive promoters to control the timing and location of gene expression specifically within tumors. Therapeutic transgenes chosen for their radiosensitizing properties will also be reviewed, these include: gene correction therapy, in which normal copies of genes responsible for radiation-induced apoptosis are transfected to compensate for the deletions or mutated variants in tumor cells (p53 is the most widely studied example). enzymes that synergize the radiation effect, by generation of a toxic species from endogenous precursors ( e.g., inducible nitric oxide synthase) or by activation of non toxic prodrugs to toxic species ( e.g., herpes simplex virus thymidine kinase/ganciclovir) within the target tissue. conditionally replicating oncolytic adenoviruses that synergize the radiation effect. membrane transport proteins ( e.g., sodium iodide symporter) to facilitate uptake of cytotoxic radionuclides. The evidence indicates that many of these approaches are successful for augmenting radiation induced tumor cell killing with clinical trials currently underway.
Collapse
Affiliation(s)
- T Robson
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| | | | | | | |
Collapse
|
2
|
Kawashita Y, Deb NJ, Garg M, Kabarriti R, Alfieri A, Takahashi M, Roy-Chowdhury J, Guha C. An autologous in situ tumor vaccination approach for hepatocellular carcinoma. 1. Flt3 ligand gene transfer increases antitumor effects of a radio-inducible suicide gene therapy in an ectopic tumor model. Radiat Res 2014; 182:191-200. [PMID: 24972258 DOI: 10.1667/rr13594.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Hepatocellular carcinoma (HCC) often presents as a diffuse or multifocal tumor making it difficult to control by surgery or radiation. Radio-inducible herpes simplex virus thymidine kinase (HSV-TK) gene therapy has been shown to enhance local tumor control after radiation therapy (RT), while limiting the expression of the transgene in the irradiated tumor tissues. To prevent liver tumor recurrence and control systemic disease while limiting the potential bystander toxicity of HSV-TK therapy, we proposed to stimulate endogenous dendritic cell (DC) proliferation with systemic adenovirus Flt3 ligand (Adeno-Flt3L) gene therapy, followed by primary tumor radiation therapy combined with a radio-inducible HSV-TK gene therapy. We hypothesized that adenovirus-expressing Flt3L gene therapy will stimulate DC proliferation, allowing the upregulated DCs to locally harness tumor antigens released from HSV-TK/RT-treated HCC cells, thereby converting irradiated tumors to an autologous in situ tumor vaccine in mice with primary liver tumors. To test this hypothesis, an expression vector of HSV-TK was constructed under the control of a radio-inducible promoter early-growth response (Egr-TK) and a recombinant adenovirus-expressing human Flt3L was constructed. The Adeno-Flt3L [10(9) plaque forming units (pfu)] was administered intravenously on days 1 and 8 after radiation therapy. The murine hepatoma cell line (BNL1ME) was stably transfected by Egr-TK or Egr-Null (encoding no therapeutic gene). Palpable tumors in BALB/c mice were treated with a localized dose of 25 Gy of radiation followed by ganciclovir (GCV, 100 mg/kg, 14 days). Four treatment cohorts were compared: Egr-Null/GCV + RT + Adeno-LacZ; Egr-Null/GCV + RT + Adeno-Flt3L; Egr-TK/GCV + RT + Adeno-LacZ; and Egr-TK/GCV + RT + Adeno-Flt3L. There was no primary tumor regression in the Egr-Null tumors after radiation therapy alone. In contrast, Egr-TK tumors had nearly complete tumor regression for 3 weeks after radiation therapy (P < 0.01), however, long-term follow-up demonstrated primary tumor recurrence and death secondary to pulmonary metastasis. Flt3L expression was confirmed by serum bioassay (mean = 88 ng/mL) in these animals and Western blotting of tissue culture medium in Adeno-Flt3L-infected BaF/huFlt3L cells. Radiation therapy with Adeno-Flt3L gene therapy effectively retarded primary tumor growth when compared to radiation therapy alone. The trimodality therapy (Egr-TK/GCV + RT + Adeno-Flt3L) was the most efficacious with 40% complete tumor regression (>100 days) and <20% pulmonary metastases, indicating the development of sustained antitumor immune response. These studies provide a rationale for triple modality therapies with radiation-inducible HSV-TK gene therapy and Adeno-Flt3L when used in combination with primary tumor radiation therapy for improved local and systemic control of HCC.
Collapse
Affiliation(s)
- Yujo Kawashita
- a Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Mirecka A, Paszkowska-Szczur K, Scott RJ, Górski B, van de Wetering T, Wokołorczyk D, Gromowski T, Serrano-Fernandez P, Cybulski C, Kashyap A, Gupta S, Gołąb A, Słojewski M, Sikorski A, Lubiński J, Dębniak T. Common variants of xeroderma pigmentosum genes and prostate cancer risk. Gene 2014; 546:156-61. [PMID: 24933002 DOI: 10.1016/j.gene.2014.06.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 06/10/2014] [Accepted: 06/12/2014] [Indexed: 01/07/2023]
Abstract
The genetic basis of prostate cancer (PC) is complex and appears to involve multiple susceptibility genes. A number of studies have evaluated a possible correlation between several NER gene polymorphisms and PC risk, but most of them evaluated only single SNPs among XP genes and the results remain inconsistent. Out of 94 SNPs located in seven XP genes (XPA-XPG) a total of 15 SNPs were assayed in 720 unselected patients with PC and compared to 1121 healthy adults. An increased risk of disease was associated with the XPD SNP, rs1799793 (Asp312Asn) AG genotype (OR=2.60; p<0.001) and with the AA genotype (OR=531; p<0.0001) compared to the control population. Haplotype analysis of XPD revealed one protective haplotype and four associated with an increased disease risk, which showed that the A allele (XPD rs1799793) appeared to drive the main effect on promoting prostate cancer risk. Polymorphism in XPD gene appears to be associated with the risk of prostate cancer.
Collapse
Affiliation(s)
- Aneta Mirecka
- Department of Genetics and Pathology, Pomeranian Medical University, Poland.
| | | | - Rodney J Scott
- School of Biomedical Sciences, University of Newcastle, Australia
| | - Bohdan Górski
- Department of Genetics and Pathology, Pomeranian Medical University, Poland
| | | | | | - Tomasz Gromowski
- Department of Genetics and Pathology, Pomeranian Medical University, Poland
| | | | - Cezary Cybulski
- Department of Genetics and Pathology, Pomeranian Medical University, Poland
| | - Aniruddh Kashyap
- Department of Genetics and Pathology, Pomeranian Medical University, Poland
| | - Satish Gupta
- Department of Genetics and Pathology, Pomeranian Medical University, Poland; Postgraduate School of Molecular Medicine, Warsaw Medical University, Poland
| | - Adam Gołąb
- Department of Urology, Pomeranian Medical University, Poland
| | | | | | - Jan Lubiński
- Department of Genetics and Pathology, Pomeranian Medical University, Poland
| | - Tadeusz Dębniak
- Department of Genetics and Pathology, Pomeranian Medical University, Poland
| |
Collapse
|
4
|
Chang JW, Park KH, HWANG HS, Shin YS, Oh YT, Kim CH. Protective effects of Korean red ginseng against radiation-induced apoptosis in human HaCaT keratinocytes. JOURNAL OF RADIATION RESEARCH 2014; 55:245-256. [PMID: 24078877 PMCID: PMC3951072 DOI: 10.1093/jrr/rrt109] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 07/28/2013] [Accepted: 08/15/2013] [Indexed: 06/02/2023]
Abstract
Radiation-induced oral mucositis is a dose-limiting toxic side effect for patients with head and neck cancer. Numerous attempts at improving radiation-induced oral mucositis have not produced a qualified treatment. Ginseng polysaccharide has multiple immunoprotective effects. Our aim was to investigate the effectiveness of Korean red ginseng (KRG) on radiation-induced damage in the human keratinocyte cell line HaCaT and in an in vivo zebrafish model. Radiation inhibited HaCaT cell proliferation and migration in a cell viability assay and wound healing assay, respectively. KRG protected against these effects. KRG attenuated the radiation-induced embryotoxicity in the zebrafish model. Irradiation of HaCaT cells caused apoptosis and changes in mitochondrial membrane potential (MMP). KRG inhibited the radiation-induced apoptosis and intracellular generation of reactive oxygen species (ROS), and stabilized the radiation-induced loss of MMP. Western blots revealed KRG-mediated reduced expression of ataxia telangiectasia mutated protein (ATM), p53, c-Jun N-terminal kinase (JNK), p38 and cleaved caspase-3, compared with their significant increase after radiation treatment. The collective results suggest that KRG protects HaCaT cells by blocking ROS generation, inhibiting changes in MMP, and inhibiting the caspase, ATM, p38 and JNK pathways.
Collapse
Affiliation(s)
- Jae Won Chang
- Department of Otolaryngology, School of Medicine, Ajou University, 5 Wonchon-Dong, Yeongtong-Gu, Suwon, 442-749, Korea
- Center for Cell Death Regulating Biodrugs, School of Medicine, Ajou University, 5 Wonchon-Dong, Yeongtong-Gu, Suwon, 442-749, Korea
| | - Keun Hyung Park
- Department of Otolaryngology, School of Medicine, Ajou University, 5 Wonchon-Dong, Yeongtong-Gu, Suwon, 442-749, Korea
- Center for Cell Death Regulating Biodrugs, School of Medicine, Ajou University, 5 Wonchon-Dong, Yeongtong-Gu, Suwon, 442-749, Korea
| | - Hye Sook HWANG
- Department of Otolaryngology, School of Medicine, Ajou University, 5 Wonchon-Dong, Yeongtong-Gu, Suwon, 442-749, Korea
- Center for Cell Death Regulating Biodrugs, School of Medicine, Ajou University, 5 Wonchon-Dong, Yeongtong-Gu, Suwon, 442-749, Korea
| | - Yoo Seob Shin
- Department of Otolaryngology, School of Medicine, Ajou University, 5 Wonchon-Dong, Yeongtong-Gu, Suwon, 442-749, Korea
- Center for Cell Death Regulating Biodrugs, School of Medicine, Ajou University, 5 Wonchon-Dong, Yeongtong-Gu, Suwon, 442-749, Korea
| | - Young-Taek Oh
- Department of Radiation Oncology, School of Medicine, Ajou University, 5 Wonchon-Dong, Yeongtong-Gu, Suwon, 442-749, Korea
| | - Chul-Ho Kim
- Department of Otolaryngology, School of Medicine, Ajou University, 5 Wonchon-Dong, Yeongtong-Gu, Suwon, 442-749, Korea
- Center for Cell Death Regulating Biodrugs, School of Medicine, Ajou University, 5 Wonchon-Dong, Yeongtong-Gu, Suwon, 442-749, Korea
| |
Collapse
|
5
|
Fokas E, Prevo R, Hammond EM, Brunner TB, McKenna WG, Muschel RJ. Targeting ATR in DNA damage response and cancer therapeutics. Cancer Treat Rev 2014; 40:109-17. [PMID: 23583268 DOI: 10.1016/j.ctrv.2013.03.002] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 03/06/2013] [Accepted: 03/06/2013] [Indexed: 12/12/2022]
Abstract
The ataxia telangiectasia and Rad3-related (ATR) plays an important role in maintaining genome integrity during DNA replication through the phosphorylation and activation of Chk1 and regulation of the DNA damage response. Preclinical studies have shown that disruption of ATR pathway can exacerbate the levels of replication stress in oncogene-driven murine tumors to promote cell killing. Additionally, inhibition of ATR can sensitise tumor cells to radiation or chemotherapy. Accumulating evidence suggests that targeting ATR can selectively sensitize cancer cells but not normal cells to DNA damage. Furthermore, in hypoxic conditions, ATR blockade results in overloading replication stress and DNA damage response causing cell death. Despite the attractiveness of ATR inhibition in the treatment of cancer, specific ATR inhibitors have remained elusive. In the last two years however, selective ATR inhibitors suitable for in vitro and - most recently - in vivo studies have been identified. In this article, we will review the literature on ATR function, its role in DDR and the potential of ATR inhibition to enhance the efficacy of radiation and chemotherapy.
Collapse
Affiliation(s)
- Emmanouil Fokas
- Gray Institute for Radiation Oncology and Biology, Department of Oncology, Oxford University, Oxford, United Kingdom; Department of Radiation Therapy and Oncology, Johann Wolfgang Goethe University, Frankfurt, Germany.
| | | | | | | | | | | |
Collapse
|
6
|
Palacios DA, Miyake M, Rosser CJ. Radiosensitization in prostate cancer: mechanisms and targets. BMC Urol 2013; 13:4. [PMID: 23351141 PMCID: PMC3583813 DOI: 10.1186/1471-2490-13-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 12/05/2012] [Indexed: 01/05/2023] Open
Abstract
Prostate cancer is the second most commonly diagnosed cancer in American men over the age of 45 years and is the third most common cause of cancer related deaths in American men. In 2012 it is estimated that 241,740 men will be diagnosed with prostate cancer and 28,170 men will succumb to prostate cancer. Currently, radiation therapy is one of the most common definitive treatment options for localized prostate cancer. However, significant number of patients undergoing radiation therapy will develop locally persistent/recurrent tumours. The varying response rates to radiation may be due to 1) tumor microenvironment, 2) tumor stage/grade, 3) modality used to deliver radiation, and 4) dose of radiation. Higher doses of radiation has not always proved to be effective and have been associated with increased morbidity. Compounds designed to enhance the killing effects of radiation, radiosensitizers, have been extensively investigated over the past decade. The development of radiosensitizing agents could improve survival, improve quality of life and reduce costs, thus benefiting both patients and healthcare systems. Herin, we shall review the role and mechanisms of various agents that can sensitize tumours, specifically prostate cancer.
Collapse
Affiliation(s)
- Diego A Palacios
- Section of Urologic Oncology, MD Anderson Cancer Center Orlando, Orlando, FL 32806, USA
| | | | | |
Collapse
|
7
|
Upon the tightrope in prostate cancer: two acrobats on the same tightrope to cross the finishline. Mol Cell Biochem 2011; 364:53-7. [PMID: 22200977 DOI: 10.1007/s11010-011-1204-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 12/15/2011] [Indexed: 01/03/2023]
Abstract
Prostate cancer is a multifactorial, multistep progressive disorder that is undruggable to date because of stumbling blocks in the standardization of therapy. It is triggered by a broad range of proteins, signaling networks and DNA damage response modulators. It is becoming increasingly apparent that DNA repair mediators have split personalities, as they are instrumental in suppressing and promoting carcinogenesis. In this article, we discuss on post-transcriptional processing of regulators of DNA damage response, and how DNA repair proteins trigger shuttling of androgen receptor. Substantial fraction of information has been added into the existing literature of ATM biology; however, the particular area of post-transcriptional processing errors and gene therapy for reprogramming of ATM has been left unaddressed in prostate cancer. It is therefore noteworthy that the facet of targeting strategy, antisense morpholino oligonucleotides chemistry, and systematic delivery of AOs has promising outlook in splice-targeted antisense-mediated therapy.
Collapse
|
8
|
Zou J, Qiao X, Ye H, Zhang Y, Xian J, Zhao H, Liu S. Inhibition of ataxia-telangiectasia mutated by antisense oligonucleotide nanoparticles induces radiosensitization of head and neck squamous-cell carcinoma in mice. Cancer Biother Radiopharm 2009; 24:339-46. [PMID: 19435407 DOI: 10.1089/cbr.2008.0557] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Ataxia-telangiectasia-mutated (ATM) is a radiosensitization gene. In the present study, we investigated the efficacy of poly(D,L-lactide-co-glycolide) (PLGA) nanoparticles containing ATM antisense oligonucleotides (ASOs) for the radiosensitization of head and neck squamous-cell carcinoma in mice, using the SCCVII cell line. Nanoparticles containing ATM ASOs were prepared with PLGA by using a double-emulsion solvent evaporation method. The results showed that the nanoparticles were suitable for intracellular uptake, and ATM ASOs inhibited ATM expression when delivered by using nanoparticles or lipofectin, but not in their free form. Meanwhile, we found that ATM reduction sensitized SCCVII cells in vitro and tumors in vivo to irradiation. In conclusion, biodegradable PLGA nanoparticles, used as a delivery carrier, enhanced intracellular uptake of ATM ASOs into SCCVII cells and the inhibitory effect of ATM ASOs. These results demonstrated that antisense ATM therapy, using PLGA nanoparticles, might provide a therapeutic benefit to patients undergoing radiation therapy for head and neck squamous-cell carcinoma.
Collapse
Affiliation(s)
- Jian Zou
- Department of Otolaryngology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
9
|
Abstract
Although tumor gene therapy falls behind its clinical use, the combination of irradiation and gene therapy is full of promise in cancer therapy based on traditional radiotherapy, chemotherapy and surgery. We have termed it as radiogenic therapy. This review focuses on the following aspects of radiogenic therapy in recent years: improvement of gene transfer efficiency by irradiation, radiotherapy combined with cytokine gene delivery or enhancement of the immunity of tumor cells by transgene, direct stimulation by radiation to produce cytotoxic agents, increase of tumor cell radiosensitivity in gene therapy by controlling the radiosensitivity genes and adjusting the fraction dose and interval of radiation so as to achieve the optimum antitumor effect while reducing the normal tissue damage, radioprotective gene therapy enhancing radiation tumor killing effect while protecting the normal tissue and organs with transgene using transfer vectors.
Collapse
Affiliation(s)
- Feng-Ling Min
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, Gansu Province, China.
| | | | | |
Collapse
|
10
|
Collis SJ, Khater K, DeWeese TL. Novel therapeutic strategies in prostate cancer management using gene therapy in combination with radiation therapy. World J Urol 2003; 21:275-89. [PMID: 12920560 DOI: 10.1007/s00345-003-0363-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2003] [Accepted: 07/07/2003] [Indexed: 12/01/2022] Open
Affiliation(s)
- Spencer J Collis
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | |
Collapse
|
11
|
Fernández PL, Hernández L, Farré X, Campo E, Cardesa A. Alterations of cell cycle-regulatory genes in prostate cancer. Pathobiology 2003; 70:1-10. [PMID: 12415186 DOI: 10.1159/000065998] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Deregulated proliferation is one of the main events in neoplastic transformation, and this has prompted increased attention being given to the understanding of the mechanisms involved in cell cycle regulation and its alterations. The 'retinoblastoma pathway', a key effector controlling G1-S phase transition, includes several oncogenes and tumour suppressor genes which display a wide range of abnormalities with potential usefulness as markers of evolution or treatment response in prostate cancer. Among these, the existence of p53 mutations seems to predict resistance to radiotherapy or systemic treatment, and p16 overexpression or p27 downregulation seems to serve as markers of poor evolution. The well-established existence of a critical hormonal role in prostate carcinogenesis coupled with the relationship of androgenic activity and regulation of several cell cycle modulators forces cell cycle control in the prostate to be envisioned as a highly complex steroid-influenced system, which will undoubtedly have critical implications in the future management of prostate cancer patients.
Collapse
Affiliation(s)
- Pedro L Fernández
- Department of Anatomical Pathology, Hospital Clínic, University of Barcelona, Barcelona, Spain.
| | | | | | | | | |
Collapse
|
12
|
Dong X, Wang L, Taniguchi K, Wang X, Cunningham JM, McDonnell SK, Qian C, Marks AF, Slager SL, Peterson BJ, Smith DI, Cheville JC, Blute ML, Jacobsen SJ, Schaid DJ, Tindall DJ, Thibodeau SN, Liu W. Mutations in CHEK2 associated with prostate cancer risk. Am J Hum Genet 2003; 72:270-80. [PMID: 12533788 PMCID: PMC379222 DOI: 10.1086/346094] [Citation(s) in RCA: 224] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2002] [Accepted: 10/29/2002] [Indexed: 01/10/2023] Open
Abstract
The DNA-damage-signaling pathway has been implicated in all human cancers. However, the genetic defects and the mechanisms of this pathway in prostate carcinogenesis remain poorly understood. In this study, we analyzed CHEK2, the upstream regulator of p53 in the DNA-damage-signaling pathway, in several groups of patients with prostate cancer. A total of 28 (4.8%) germline CHEK2 mutations (16 of which were unique) were found among 578 patients. Additional screening for CHEK2 mutations in 149 families with familial prostate cancer revealed 11 mutations (5 unique) in nine families. These mutations included two frameshift and three missense mutations. Importantly, 16 of 18 unique CHEK2 mutations identified in both sporadic and familial cases were not detected among 423 unaffected men, suggesting a pathological effect of CHEK2 mutations in prostate cancer development. Analyses of the two frameshift mutations in Epstein Barr virus-transformed cell lines, using reverse-transcriptase polymerase chain reaction and western blot analysis, revealed abnormal splicing for one mutation and dramatic reduction of CHEK2 protein levels in both cases. Overall, our data suggest that mutations in CHEK2 may contribute to prostate cancer risk and that the DNA-damage-signaling pathway may play an important role in the development of prostate cancer.
Collapse
Affiliation(s)
- Xiangyang Dong
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Liang Wang
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Ken Taniguchi
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Xianshu Wang
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Julie M. Cunningham
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Shannon K. McDonnell
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Chiping Qian
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Angela F. Marks
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Susan L. Slager
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Brett J. Peterson
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - David I. Smith
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - John C. Cheville
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Michael L. Blute
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Steve J. Jacobsen
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Daniel J. Schaid
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Donald J. Tindall
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Stephen N. Thibodeau
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| | - Wanguo Liu
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, and Departments of Health Sciences Research and Urology, Mayo Clinic/Mayo Medical School, Rochester, MN
| |
Collapse
|
13
|
Abstract
One of the cornerstones of the web of signaling pathways governing cellular life and differentiation is the DNA damage response. It spans a complex network of pathways, ranging from DNA repair to modulation of numerous processes in the cell. DNA double-strand breaks (DSBs), which are formed as a result of genotoxic stress or normal recombinational processes, are extremely lethal lesions that rapidly mobilize this intricate defense system. The master controller that pilots cellular responses to DSBs is the ATM protein kinase, which turns on this network by phosphorylating key players in its various branches. ATM is the protein product of the gene mutated in the human genetic disorder ataxia-telangiectasia (A-T), which is characterized by neuronal degeneration, immunodeficiency, sterility, genomic instability, cancer predisposition, and radiation sensitivity. The clinical and cellular phenotype of A-T attests to the numerous roles of ATM, on the one hand, and to the link between the DNA damage response and developmental processes on the other hand. Recent studies of this protein and its effectors, combined with a thorough investigation of animal models of A-T, have led to new insights into the mode of action of this master controller of the DNA damage response. The evidence that ATM is involved in signaling pathways other than those related to damage response, particularly ones relating to cellular growth and differentiation, reinforces the multifaceted nature of this protein, in which genome stability, developmental processes, and cancer cross paths.
Collapse
Affiliation(s)
- Y Shiloh
- Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Israel
| | | |
Collapse
|
14
|
Kaminski JM, Nguyen K, Buyyounouski M, Pollack A. Prostate cancer gene therapy and the role of radiation. Cancer Treat Rev 2002; 28:49-64. [PMID: 12027414 DOI: 10.1053/ctrv.2002.0250] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Even though prostate cancer is detected earlier than in the pre-PSA era, prostate cancer is the second leading cause of cancer mortality in the American male. Prostate cancer therapy is not ideal, especially for high-risk localized and metastatic cancer; therefore, investigators have sought new therapeutic modalities such as angiogenesis inhibitors, inhibitors of the cell signaling pathway, vaccines, and gene therapy. Gene therapy has emerged as potential therapy for both localized and systemic prostate cancer. Gene therapy has been shown to work supra-additively with radiation in controlling prostate cancer in vivo. With further technological advances in radiation therapy, gene therapy, and the understanding of prostate cancer biology, gene therapy will potentially have an important role in prostate cancer therapy.
Collapse
Affiliation(s)
- J M Kaminski
- Department of Radiation Oncology, Fox Chase Cancer Center, 7701 Burolme Avenue, Philadelphia, PA 19111, USA.
| | | | | | | |
Collapse
|