1
|
Cunningham ML, Schiewer MJ. PARP-ish: Gaps in Molecular Understanding and Clinical Trials Targeting PARP Exacerbate Racial Disparities in Prostate Cancer. Cancer Res 2024; 84:743102. [PMID: 38635890 PMCID: PMC11217733 DOI: 10.1158/0008-5472.can-23-3458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/25/2024] [Accepted: 04/02/2024] [Indexed: 04/20/2024]
Abstract
PARP is a nuclear enzyme with a major function in the DNA damage response. PARP inhibitors (PARPi) have been developed for treating tumors harboring homologous recombination repair (HRR) defects that lead to a dependency on PARP. There are currently three PARPi approved for use in advanced prostate cancer (PCa), and several others are in clinical trials for this disease. Recent clinical trial results have reported differential efficacy based on the specific PARPi utilized as well as patient race. There is a racial disparity in PCa, where African American (AA) males are twice as likely to develop and die from the disease compared to European American (EA) males. Despite the disparity, there continues to be a lack of diversity in clinical trial cohorts for PCa. In this review, PARP nuclear functions, inhibition, and clinical relevance are explored through the lens of racial differences. This review will touch on the biological variations that have been explored thus far between AA and EA males with PCa to offer rationale for investigating PARPi response in the context of race at both the basic science and the clinical development levels.
Collapse
Affiliation(s)
- Moriah L. Cunningham
- Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.
| | - Matthew J. Schiewer
- Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania.
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
2
|
Tam A, Mercier BD, Thomas RM, Tizpa E, Wong IG, Shi J, Garg R, Hampel H, Gray SW, Williams T, Bazan JG, Li YR. Moving the Needle Forward in Genomically-Guided Precision Radiation Treatment. Cancers (Basel) 2023; 15:5314. [PMID: 38001574 PMCID: PMC10669735 DOI: 10.3390/cancers15225314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/06/2023] [Accepted: 10/13/2023] [Indexed: 11/26/2023] Open
Abstract
Radiation treatment (RT) is a mainstay treatment for many types of cancer. Recommendations for RT and the radiation plan are individualized to each patient, taking into consideration the patient's tumor pathology, staging, anatomy, and other clinical characteristics. Information on germline mutations and somatic tumor mutations is at present rarely used to guide specific clinical decisions in RT. Many genes, such as ATM, and BRCA1/2, have been identified in the laboratory to confer radiation sensitivity. However, our understanding of the clinical significance of mutations in these genes remains limited and, as individual mutations in such genes can be rare, their impact on tumor response and toxicity remains unclear. Current guidelines, including those from the National Comprehensive Cancer Network (NCCN), provide limited guidance on how genetic results should be integrated into RT recommendations. With an increasing understanding of the molecular underpinning of radiation response, genomically-guided RT can inform decisions surrounding RT dose, volume, concurrent therapies, and even omission to further improve oncologic outcomes and reduce risks of toxicities. Here, we review existing evidence from laboratory, pre-clinical, and clinical studies with regard to how genetic alterations may affect radiosensitivity. We also summarize recent data from clinical trials and explore potential future directions to utilize genetic data to support clinical decision-making in developing a pathway toward personalized RT.
Collapse
Affiliation(s)
- Andrew Tam
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.); (B.D.M.); (R.M.T.); (E.T.); (I.G.W.); (J.S.); (R.G.); (T.W.)
| | - Benjamin D. Mercier
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.); (B.D.M.); (R.M.T.); (E.T.); (I.G.W.); (J.S.); (R.G.); (T.W.)
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (H.H.); (S.W.G.)
| | - Reeny M. Thomas
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.); (B.D.M.); (R.M.T.); (E.T.); (I.G.W.); (J.S.); (R.G.); (T.W.)
| | - Eemon Tizpa
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.); (B.D.M.); (R.M.T.); (E.T.); (I.G.W.); (J.S.); (R.G.); (T.W.)
| | - Irene G. Wong
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.); (B.D.M.); (R.M.T.); (E.T.); (I.G.W.); (J.S.); (R.G.); (T.W.)
| | - Juncong Shi
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.); (B.D.M.); (R.M.T.); (E.T.); (I.G.W.); (J.S.); (R.G.); (T.W.)
| | - Rishabh Garg
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.); (B.D.M.); (R.M.T.); (E.T.); (I.G.W.); (J.S.); (R.G.); (T.W.)
| | - Heather Hampel
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (H.H.); (S.W.G.)
| | - Stacy W. Gray
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (H.H.); (S.W.G.)
| | - Terence Williams
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.); (B.D.M.); (R.M.T.); (E.T.); (I.G.W.); (J.S.); (R.G.); (T.W.)
| | - Jose G. Bazan
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.); (B.D.M.); (R.M.T.); (E.T.); (I.G.W.); (J.S.); (R.G.); (T.W.)
| | - Yun R. Li
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd., Duarte, CA 91010, USA; (A.T.); (B.D.M.); (R.M.T.); (E.T.); (I.G.W.); (J.S.); (R.G.); (T.W.)
- Department of Cancer Genetics and Epigenetics, City of Hope National Medical Center, Duarte, CA 91010, USA
- Division of Quantitative Medicine & Systems Biology, Translational Genomics Research Institute, 445 N. Fifth Street, Phoenix, AZ 85022, USA
| |
Collapse
|
3
|
Sorrentino C, Di Carlo E. Molecular Targeted Therapies in Metastatic Prostate Cancer: Recent Advances and Future Challenges. Cancers (Basel) 2023; 15:2885. [PMID: 37296848 PMCID: PMC10251915 DOI: 10.3390/cancers15112885] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Prostate cancer is the most frequent malignant tumor in men, and, despite the great improvements in survival in patients with localized cancer, the prognosis for metastatic disease remains poor. Novel molecular targeted therapies, which block specific molecules or signaling pathways in tumor cells or in their microenvironment, have shown encouraging results in metastatic castration-resistant prostate cancer. Among these therapeutic approaches, prostate-specific membrane antigen-targeted radionuclide therapies and DNA repair inhibitors represent the most promising ones, with some therapeutic protocols already approved by the FDA, whereas therapies targeting tumor neovascularization and immune checkpoint inhibitors have not yet demonstrated clear clinical benefits. In this review, the most relevant studies and clinical trials on this topic are illustrated and discussed, together with future research directions and challenges.
Collapse
Affiliation(s)
- Carlo Sorrentino
- Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Emma Di Carlo
- Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
4
|
Kallenbach J, Atri Roozbahani G, Heidari Horestani M, Baniahmad A. Distinct mechanisms mediating therapy-induced cellular senescence in prostate cancer. Cell Biosci 2022; 12:200. [PMID: 36522745 PMCID: PMC9753376 DOI: 10.1186/s13578-022-00941-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Prostate cancer (PCa) is an age-related malignancy in men with a high incidence rate. PCa treatments face many obstacles due to cancer cell resistance and many bypassing mechanisms to escape therapy. According to the intricacy of PCa, many standard therapies are being used depending on PCa stages including radical prostatectomy, radiation therapy, androgen receptor (AR) targeted therapy (androgen deprivation therapy, supraphysiological androgen, and AR antagonists) and chemotherapy. Most of the aforementioned therapies have been implicated to induce cellular senescence. Cellular senescence is defined as a stable cell cycle arrest in the G1 phase and is one of the mechanisms that prevent cancer proliferation. RESULTS In this review, we provide and analyze different mechanisms of therapy-induced senescence (TIS) in PCa and their effects on the tumor. Interestingly, it seems that different molecular pathways are used by cancer cells for TIS. Understanding the complexity and underlying mechanisms of cellular senescence is very critical due to its role in tumorigenesis. The most prevalent analyzed pathways in PCa as TIS are the p53/p21WAF1/CIP1, the p15INK4B/p16INK4A/pRb/E2F/Cyclin D, the ROS/ERK, p27Kip1/CDK/pRb, and the p27Kip1/Skp2/C/EBP β signaling. Despite growth inhibition, senescent cells are highly metabolically active. In addition, their secretome, which is termed senescence-associated secretory phenotype (SASP), affects within the tumor microenvironment neighboring non-tumor and tumor cells and thereby may regulate the growth of tumors. Induction of cancer cell senescence is therefore a double-edged sword that can lead to reduced or enhanced tumor growth. CONCLUSION Thus, dependent on the type of senescence inducer and the specific senescence-induced cellular pathway, it is useful to develop pathway-specific senolytic compounds to specifically targeting senescent cells in order to evict senescent cells and thereby to reduce SASP side effects.
Collapse
Affiliation(s)
- Julia Kallenbach
- grid.9613.d0000 0001 1939 2794Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07740 Jena, Germany
| | - Golnaz Atri Roozbahani
- grid.9613.d0000 0001 1939 2794Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07740 Jena, Germany
| | - Mehdi Heidari Horestani
- grid.9613.d0000 0001 1939 2794Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07740 Jena, Germany
| | - Aria Baniahmad
- grid.9613.d0000 0001 1939 2794Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07740 Jena, Germany
| |
Collapse
|
5
|
Wang H, Xie H, Wang S, Zhao J, Gao Y, Chen J, Zhao Y, Guo G. PARP-1 genetic polymorphism associated with radiation sensitivity of non-small cell lung cancer. Pathol Oncol Res 2022; 28:1610751. [PMID: 36590386 PMCID: PMC9795517 DOI: 10.3389/pore.2022.1610751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022]
Abstract
About 70% of non-small cell lung cancer (NSCLC) patients require radiotherapy. However, due to the difference in radiation sensitivity, the treatment outcome may differ for the same pathology and choice of treatment. Poly (ADP-ribose) polymerase 1 (PARP-1) is a key gene responsible for DNA repair and is involved in base excision repair as well as repair of single strand break induced by ionizing radiation and oxidative damage. In order to investigate the relationship between PARP-1 gene polymorphism and radiation sensitivity in NSCLC, we collected 141 primary NSCLC patients undergoing three-dimensional conformal radiotherapy. For each case, the gross tumor volumes (GTV) before radiation and that after 40 Gy radiation were measured to calculate the tumor regression rate. TaqMan real-time polymerase chain reaction was performed to genotype the single-nucleotide polymorphisms (SNPs). Genotype frequencies for PARP-1 genotypes were 14.2% for C/C, 44.7% for C/G and 41.1% for G/G. The average tumor regression rate after 40 Gy radiation therapy was 35.1% ± 0.192. Tumor regression rate of mid-term RT of C/C genotype was 44.6% ± 0.170, which was higher than that of genotype C/G and G/G (32.4% ± 0.196 and 34.8% ± 0.188, respectively) with statistical significance (F = 3.169 p = 0.045). The higher tumor regression rate in patients with C/C genotype suggested that G allele was a protective factor against radiation therapy. Using the median tumor regression rate of 34%, we divided the entire cohort into two groups, and found that the frequency distribution of PARP-1 gene rs3219073 had significant difference between these two groups (p < 0.05). These results showed that PARP-1 gene polymorphism may affect patient radiation sensitivity and predict the efficacy of radiotherapy. It therefore presents an opportunity for developing new therapeutic targets to improve radiotherapy outcome.
Collapse
Affiliation(s)
- Hetong Wang
- Department of Radiation Oncology, The Tenth People’s Hospital of Shenyang, Shenyang, China,Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Haitao Xie
- Department of Radiation Oncology, Liaoning Cancer Hospital, Shenyang, China
| | | | - Jiaying Zhao
- Department of Radiation Oncology, Qingdao United Family Healthcare, Qingdao, China
| | - Ya Gao
- Department of Oncology, Kailuan Hospital, Tangshan, Hebei, China
| | - Jun Chen
- Department of Radiation Oncology, The Tenth People’s Hospital of Shenyang, Shenyang, China
| | - Yuxia Zhao
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Genyan Guo
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China,*Correspondence: Genyan Guo,
| |
Collapse
|
6
|
Moreira-Silva F, Henrique R, Jerónimo C. From Therapy Resistance to Targeted Therapies in Prostate Cancer. Front Oncol 2022; 12:877379. [PMID: 35686097 PMCID: PMC9170957 DOI: 10.3389/fonc.2022.877379] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is the second most common malignancy among men worldwide. Although early-stage disease is curable, advanced stage PCa is mostly incurable and eventually becomes resistant to standard therapeutic options. Different genetic and epigenetic alterations are associated with the development of therapy resistant PCa, with specific players being particularly involved in this process. Therefore, identification and targeting of these molecules with selective inhibitors might result in anti-tumoral effects. Herein, we describe the mechanisms underlying therapy resistance in PCa, focusing on the most relevant molecules, aiming to enlighten the current state of targeted therapies in PCa. We suggest that selective drug targeting, either alone or in combination with standard treatment options, might improve therapeutic sensitivity of resistant PCa. Moreover, an individualized analysis of tumor biology in each PCa patient might improve treatment selection and therapeutic response, enabling better disease management.
Collapse
Affiliation(s)
- Filipa Moreira-Silva
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (He-alth Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (He-alth Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences of the University of Porto (ICBAS-UP), Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (He-alth Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), Porto, Portugal.,Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences of the University of Porto (ICBAS-UP), Porto, Portugal
| |
Collapse
|
7
|
Al-Jumayli M, Brown SL, Chetty IJ, Extermann M, Movsas B. The Biological Process of Aging and the Impact of Ionizing Radiation. Semin Radiat Oncol 2022; 32:172-178. [DOI: 10.1016/j.semradonc.2021.11.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
8
|
Seo Y, Tamari K, Takahashi Y, Minami K, Tatekawa S, Isohashi F, Suzuki O, Akino Y, Ogawa K. Poly (ADP-ribose) polymerase inhibitors sensitize cancer cells to hypofractionated radiotherapy through altered selection of DNA double-strand break repair pathways. Int J Radiat Biol 2022; 98:1222-1234. [PMID: 34919022 DOI: 10.1080/09553002.2022.2020357] [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: 10/19/2022]
Abstract
PURPOSE Poly (ADP-ribose) polymerase inhibitors (PARPi) are known to induce radiosensitization. However, the exact mechanisms of radiosensitization remain unclear. We previously reported that PARPi may have a unique radiosensitizing effect to enhance β-components of the linear-quadratic model. The aim of this study was to evaluate PARPi in combination with high-dose-per-fraction radiotherapy and to elucidate the underlying mechanisms of its radiosensitization. MATERIALS AND METHODS Radiosensitizing effects of PARPi PJ34, olaparib, and veliparib were measured using a colony-forming assay in the human cancer cell lines, HCT116, NCI-H460, and HT29. Six different radiation dose fractionation schedules were examined by tumor regrowth assay using three-dimensional multicellular spheroids of HCT116, NCI-H460, SW620, and HCT15. The mechanisms of radiosensitization were analyzed by measuring DNA double-strand breaks (DSB), DNA damage responses, chromosomal translocations, cellular senescence, and cell cycle analysis. RESULTS Olaparib and PJ34 were found to show radiosensitization preferentially at higher radiation doses per fraction. Similar results were obtained using a mouse model bearing human tumor xenografts. A kinetic analysis of DNA damage responses and repairs showed that olaparib and PJ34 reduced the homologous recombination activity. However, a neutral comet assay showed that PJ34 treatment did not affect the physical rejoining of DNA-DSBs induced by ionizing radiation. Cell cycle analysis revealed that olaparib and PJ34 strikingly increased G1 tetraploid cells following irradiation, leading to premature senescence. The C-banding analysis of metaphase spreads showed that olaparib and PJ34 significantly increased ionizing radiation-induced dicentric chromosomes. The data suggests that PARPi olaparib and PJ34 altered the choice of DNA-DSB repair pathways rather than reducing the total amount of DNA-DSB repair, which resulted in increased repair errors. Increased quadratic misrepair was one of the mechanisms of PARP-mediated radiosensitization, preferentially at the higher dose range compared to the lower dose range. CONCLUSION PARPi may be a promising candidate to combine with stereotactic hypofractionated radiotherapy, aiming at high-dose region-directed radiosensitization.
Collapse
Affiliation(s)
- Yuji Seo
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Keisuke Tamari
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yutaka Takahashi
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kazumasa Minami
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shotaro Tatekawa
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Fumiaki Isohashi
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Osamu Suzuki
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuichi Akino
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kazuhiko Ogawa
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
9
|
Macedo-Silva C, Benedetti R, Ciardiello F, Cappabianca S, Jerónimo C, Altucci L. Epigenetic mechanisms underlying prostate cancer radioresistance. Clin Epigenetics 2021; 13:125. [PMID: 34103085 PMCID: PMC8186094 DOI: 10.1186/s13148-021-01111-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/02/2021] [Indexed: 12/24/2022] Open
Abstract
Radiotherapy (RT) is one of the mainstay treatments for prostate cancer (PCa), a highly prevalent neoplasm among males worldwide. About 30% of newly diagnosed PCa patients receive RT with a curative intent. However, biochemical relapse occurs in 20–40% of advanced PCa treated with RT either alone or in combination with adjuvant-hormonal therapy. Epigenetic alterations, frequently associated with molecular variations in PCa, contribute to the acquisition of a radioresistant phenotype. Increased DNA damage repair and cell cycle deregulation decreases radio-response in PCa patients. Moreover, the interplay between epigenome and cell growth pathways is extensively described in published literature. Importantly, as the clinical pattern of PCa ranges from an indolent tumor to an aggressive disease, discovering specific targetable epigenetic molecules able to overcome and predict PCa radioresistance is urgently needed. Currently, histone-deacetylase and DNA-methyltransferase inhibitors are the most studied classes of chromatin-modifying drugs (so-called ‘epidrugs’) within cancer radiosensitization context. Nonetheless, the lack of reliable validation trials is a foremost drawback. This review summarizes the major epigenetically induced changes in radioresistant-like PCa cells and describes recently reported targeted epigenetic therapies in pre-clinical and clinical settings. ![]()
Collapse
Affiliation(s)
- Catarina Macedo-Silva
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy.,Cancer Biology and Epigenetics Group, Research Center at Portuguese Oncology Institute of Porto, F Bdg, 1st Floor, Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Rosaria Benedetti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center at Portuguese Oncology Institute of Porto, F Bdg, 1st Floor, Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal. .,Department of Pathology and Molecular Immunology at School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal.
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naplei, Italy.
| |
Collapse
|
10
|
Abstract
The use of DNA-damaging agents such as radiotherapy and chemotherapy has been a mainstay treatment protocol for many cancers, including lung and prostate. Recently, FDA approval of inhibitors of DNA repair, and targeting innate immunity to enhance the efficacy of DNA-damaging agents have gained much attention. Yet, inherent or acquired resistance against DNA-damaging therapies persists as a fundamental drawback. While cancer eradication by causing cancer cell death through induction of apoptosis is the ultimate goal of anti-cancer treatments, autophagy and senescence are two major cellular responses induced by clinically tolerable doses of DNA-damaging therapies. Unlike apoptosis, autophagy and senescence can act as both pro-tumorigenic as well as tumor suppressive mechanisms. DNA damage-induced senescence is associated with a pro-inflammatory secretory phenotype, which contributes to reshaping the tumor- immune microenvironment. Moreover, PTEN (phosphatase and tensin homolog) is a tumor supressor deleted in many tumors, and has been implicated in both senescence and autophagy. This review presents an overview of the literature on the regulation and consequences of DNA damage- induced senescence in cancer cells, with a specific focus on autophagy and PTEN. Both autophagy and senescence occur concurrently in the same cells in response to DNA damaging agents. However, a deterministic relationship between these fundamental processes has been controversial. We present experimental evidence obtained with tumor cells, with a prime focus on two models of cancer, prostate and lung. A better understanding of mechanisms associated with DNA damage-induced cellular senescence is central to fully exploit the potential of DNA-damaging agents against cancer.
Collapse
Affiliation(s)
- Arishya Sharma
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States.
| | - Alexandru Almasan
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States; Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States.
| |
Collapse
|
11
|
Bryson BL, Tamagno I, Taylor SE, Parameswaran N, Chernosky NM, Balasubramaniam N, Jackson MW. Aberrant Induction of a Mesenchymal/Stem Cell Program Engages Senescence in Normal Mammary Epithelial Cells. Mol Cancer Res 2020; 19:651-666. [PMID: 33443106 DOI: 10.1158/1541-7786.mcr-19-1181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 10/23/2020] [Accepted: 12/15/2020] [Indexed: 11/16/2022]
Abstract
Although frequently associated with tumor progression, inflammatory cytokines initially restrain transformation by inducing senescence, a key tumor-suppressive barrier. Here, we demonstrate that the inflammatory cytokine, oncostatin M, activates a mesenchymal/stem cell (SC) program that engages cytokine-induced senescence (CIS) in normal human epithelial cells. CIS is driven by Snail induction and requires cooperation between STAT3 and the TGFβ effector, SMAD3. Importantly, as cells escape CIS, they retain the mesenchymal/SC program and are thereby bestowed with a set of cancer SC (CSC) traits. Of therapeutic importance, cells that escape CIS can be induced back into senescence by CDK4/6 inhibition, confirming that the mechanisms allowing cells to escape senescence are targetable and reversible. Moreover, by combining CDK4/6 inhibition with a senolytic therapy, mesenchymal/CSCs can be efficiently killed. Our studies provide insight into how the CIS barriers that prevent tumorigenesis can be exploited as potential therapies for highly aggressive cancers. IMPLICATIONS: These studies reveal how a normal cell's arduous escape from senescence can bestow aggressive features early in the transformation process, and how this persistent mesenchymal/SC program can create a novel potential targetability following tumor development.
Collapse
Affiliation(s)
- Benjamin L Bryson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Ilaria Tamagno
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Sarah E Taylor
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Neetha Parameswaran
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Noah M Chernosky
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Nikhila Balasubramaniam
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Mark W Jackson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
12
|
Song J, Chen W, Zhu G, Wang W, Sun F, Zhu J. Immunogenomic Profiling and Classification of Prostate Cancer Based on HIF-1 Signaling Pathway. Front Oncol 2020; 10:1374. [PMID: 32850440 PMCID: PMC7425731 DOI: 10.3389/fonc.2020.01374] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 06/30/2020] [Indexed: 12/24/2022] Open
Abstract
The HIF-1 signaling pathway plays an important role in the pathogenesis of cancer. Many studies have explored the progression of prostate cancer (PCa) under hypoxic conditions based on transcriptome data; few have uncovered the immunogenomic profiling and prostate cancer classification based on the HIF-1 signaling pathway. This pathway may help to identify the optimal subset of PCa patients responsive to immunotherapy/chemotherapy. The immunogenomic PCa subsets were classified based on profiling of the HIF-1 signaling pathway, using four publicly available PCa datasets. Three PCa subtypes that named as HIF-1 High (HIF-1_H), HIF-1 Medium (HIF-1_M), and HIF-1 Low (HIF-1_L) were identified. Functional enrichment was analyzed in each subtype. Several cancer-associated and immune-related pathways were hyperactivated in the HIF-1_H subtypes. In contrast, HIF-1_L subtypes were enriched in cell cycle and cell repair. Compared with other subtypes, HIF-1_H subtypes have greater immune cell infiltration, anti-tumor immune activity, and better survival prognosis. The submap and TIDE algorithm were used to predict the clinical response to immune checkpoint blockade, and GDSC was employed to screen potential chemotherapeutic targets for the treatment of PCa. Several chemotherapy drugs were identified in the GDSC dataset, including ABT 888, Temsirolimus, and EHT 1864. Meanwhile, HIF-1_H was defined as an early PCa marker, which is more likely to respond to immunotherapy. The identification of immunogenomic PCa subtypes based on the HIF-1 signaling pathway has potential clinical implications for PCa treatment. Immunopositive PCa subtypes will help to explore the reasons for the poor response of PCa to immunotherapy, and it is expected that immunotherapy will guide the personalized treatment of PCa patients.
Collapse
Affiliation(s)
- Jukun Song
- Medical College, Guizhou University, Guiyang, China
- Department of Oral and Maxillofacial Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Weiming Chen
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Guohua Zhu
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Wei Wang
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Fa Sun
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jianguo Zhu
- Medical College, Guizhou University, Guiyang, China
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
13
|
de Bono JS, Guo C, Gurel B, De Marzo AM, Sfanos KS, Mani RS, Gil J, Drake CG, Alimonti A. Prostate carcinogenesis: inflammatory storms. Nat Rev Cancer 2020; 20:455-469. [PMID: 32546840 DOI: 10.1038/s41568-020-0267-9] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/24/2020] [Indexed: 02/06/2023]
Abstract
Prostate cancer is a major cause of cancer morbidity and mortality. Intra-prostatic inflammation is a risk factor for prostate carcinogenesis, with diet, chemical injury and an altered microbiome being causally implicated. Intra-prostatic inflammatory cell recruitment and expansion can ultimately promote DNA double-strand breaks and androgen receptor activation in prostate epithelial cells. The activation of the senescence-associated secretory phenotype fuels further 'inflammatory storms', with free radicals leading to further DNA damage. This drives the overexpression of DNA repair and tumour suppressor genes, rendering these genes susceptible to mutagenic insults, with carcinogenesis accelerated by germline DNA repair gene defects. We provide updates on recent advances in elucidating prostate carcinogenesis and explore novel therapeutic and prevention strategies harnessing these discoveries.
Collapse
Affiliation(s)
- Johann S de Bono
- The Institute of Cancer Research, London, UK.
- The Royal Marsden NHS Foundation Trust, Sutton, UK.
| | - Christina Guo
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Bora Gurel
- The Institute of Cancer Research, London, UK
| | | | - Karen S Sfanos
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ram S Mani
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jesús Gil
- MRC London Institute of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | | | - Andrea Alimonti
- Institute of Oncology Research, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
14
|
Hacker BC, Rafat M. Organoids as Complex In Vitro Models for Studying Radiation-Induced Cell Recruitment. Cell Mol Bioeng 2020; 13:341-357. [PMID: 32952734 PMCID: PMC7479086 DOI: 10.1007/s12195-020-00625-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 06/10/2020] [Indexed: 01/01/2023] Open
Abstract
Patients with triple negative breast cancer (TNBC) typically receive chemotherapy, surgery, and radiation therapy. Although this treatment improves prognosis for most patients, some patients continue to experience recurrence within 5 years. Preclinical studies have shown that immune cell infiltration at the irradiated site may play a significant role in tumor cell recruitment; however, little is known about the mechanisms that govern this process. This lack of knowledge highlights the need to evaluate radiation-induced cell infiltration with models that have controllable variables and maintain biological integrity. Mammary organoids are multicellular three-dimensional (3D) in vitro models, and they have been used to examine many aspects of mammary development and tumorigenesis. Organoids are also emerging as a powerful tool to investigate normal tissue radiation damage. In this review, we evaluate recent advances in mammary organoid technology, consider the advantages of using organoids to study radiation response, and discuss future directions for the applications of this technique.
Collapse
Affiliation(s)
- Benjamin C. Hacker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN USA
| | - Marjan Rafat
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN USA
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN USA
| |
Collapse
|
15
|
Zhang J, Si J, Gan L, Zhou R, Guo M, Zhang H. Harnessing the targeting potential of differential radiobiological effects of photon versus particle radiation for cancer treatment. J Cell Physiol 2020; 236:1695-1711. [PMID: 32691425 DOI: 10.1002/jcp.29960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/09/2020] [Indexed: 01/04/2023]
Abstract
Radiotherapy is one of the major modalities for malignancy treatment. High linear energy transfer (LET) charged-particle beams, like proton and carbon ions, exhibit favourable depth-dose distributions and radiobiological enhancement over conventional low-LET photon irradiation, thereby marking a new era in high precision medicine. Tumour cells have developed multicomponent signal transduction networks known as DNA damage responses (DDRs), which initiate cell-cycle checkpoints and induce double-strand break (DSB) repairs in the nucleus by nonhomologous end joining or homologous recombination pathways, to manage ionising radiation (IR)-induced DNA lesions. DNA damage induction and DSB repair pathways are reportedly dependent on the quality of radiation delivered. In this review, we summarise various types of DNA lesion and DSB repair mechanisms, upon irradiation with low and high-LET radiation, respectively. We also analyse factors influencing DNA repair efficiency. Inhibition of DNA damage repair pathways and dysfunctional cell-cycle checkpoint sensitises tumour cells to IR. Radio-sensitising agents, including DNA-PK inhibitors, Rad51 inhibitors, PARP inhibitors, ATM/ATR inhibitors, chk1 inhibitors, wee1 kinase inhibitors, Hsp90 inhibitors, and PI3K/AKT/mTOR inhibitors have been found to enhance cell killing by IR through interference with DDRs, cell-cycle arrest, or other cellular processes. The cotreatment of these inhibitors with IR may represent a promising therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Jinhua Zhang
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jing Si
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Lu Gan
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Rong Zhou
- Research Center for Ecological Impacts and Environmental Health Effects of Toxic and Hazardous Chemicals, Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment of the People's Republic of China, Nanjing, China
| | - Menghuan Guo
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Hong Zhang
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
16
|
Ghorai A, Mahaddalkar T, Thorat R, Dutt S. Sustained inhibition of PARP-1 activity delays glioblastoma recurrence by enhancing radiation-induced senescence. Cancer Lett 2020; 490:44-53. [PMID: 32645394 DOI: 10.1016/j.canlet.2020.06.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 06/08/2020] [Accepted: 06/26/2020] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) is the most common primary brain tumor and is highly aggressive with a median survival of 15 months. We have previously shown that residual cells of GBM form multinucleated giant cells (MNGCs) showing a senescent phenotype, but eventually escape from therapy induced senescence (TIS), resulting in GBM recurrence. Here we demonstrate the role of PARP-1 in TIS and its recovery. We show that genetic and pharmacological inhibition of PARP-1 has an anti-proliferative effect on GBM cell lines and primary cultures derived from patient samples. Furthermore, the PARP-1 inhibitor olaparib, in combination with radiation increased MNGCs formation and senescence as assessed by β-galactosidase activity, and macroH2A1 levels in residual cells. Additionally, we found that reduced PARP-1 activity and not protein levels in residual cells was crucial for MNGCs formation and their maintenance in the senescent state. PARP-1 activity was restored to higher levels in recurrent cells that escaped from TIS. Importantly, olaparib + radiation treatment significantly delayed recurrence in vitro as well in vivo in orthotopic GBM mouse models with a significant increase in overall survival of mice. Overall, this study demonstrates that sustained inhibition of PARP-1 activity during radiation treatment significantly delays GBM recurrence.
Collapse
Affiliation(s)
- Atanu Ghorai
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Tejashree Mahaddalkar
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Rahul Thorat
- Laboratory Animal Facility, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, India
| | - Shilpee Dutt
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India.
| |
Collapse
|
17
|
DNA Damage- But Not Enzalutamide-Induced Senescence in Prostate Cancer Promotes Senolytic Bcl-xL Inhibitor Sensitivity. Cells 2020; 9:cells9071593. [PMID: 32630281 PMCID: PMC7408442 DOI: 10.3390/cells9071593] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 12/16/2022] Open
Abstract
Cellular senescence is a natural tumor suppression mechanism defined by a stable proliferation arrest. In the context of cancer treatment, cancer cell therapy-induced senescence (TIS) is emerging as an omnipresent cell fate decision that can be pharmacologically targeted at the molecular level to enhance the beneficial aspects of senescence. In prostate cancer (PCa), TIS has been reported using multiple different model systems, and a more systematic analysis would be useful to identify relevant senescence manipulation molecular targets. Here we show that a spectrum of PCa senescence phenotypes can be induced by clinically relevant therapies. We found that DNA damage inducers like irradiation and poly (ADP-ribose) polymerase1 (PARP) inhibitors triggered a stable PCa-TIS independent of the p53 status. On the other hand, enzalutamide triggered a reversible senescence-like state that lacked evidence of cell death or DNA damage. Using a small senolytic drug panel, we found that senescence inducers dictated senolytic sensitivity. While Bcl-2 family anti-apoptotic inhibitor were lethal for PCa-TIS cells harboring evidence of DNA damage, they were ineffective against enzalutamide-TIS cells. Interestingly, piperlongumine, which was described as a senolytic, acted as a senomorphic to enhance enzalutamide-TIS proliferation arrest without promoting cell death. Overall, our results suggest that TIS phenotypic hallmarks need to be evaluated in a context-dependent manner because they can vary with senescence inducers, even within identical cancer cell populations. Defining this context-dependent spectrum of senescence phenotypes is key to determining subsequent molecular strategies that target senescent cancer cells.
Collapse
|
18
|
Tran Chau V, Liu W, Gerbé de Thoré M, Meziani L, Mondini M, O'Connor MJ, Deutsch E, Clémenson C. Differential therapeutic effects of PARP and ATR inhibition combined with radiotherapy in the treatment of subcutaneous versus orthotopic lung tumour models. Br J Cancer 2020; 123:762-771. [PMID: 32546832 PMCID: PMC7463250 DOI: 10.1038/s41416-020-0931-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 04/30/2020] [Accepted: 05/21/2020] [Indexed: 11/09/2022] Open
Abstract
Background Subcutaneous mouse tumour models are widely used for the screening of novel antitumour treatments, although these models are poor surrogate models of human cancers. Methods We compared the antitumour efficacy of the combination of ionising radiation (IR) with two DNA damage response inhibitors, the PARP inhibitor olaparib and the ATR inhibitor AZD6738 (ceralasertib), in subcutaneous versus orthotopic cancer models. Results Olaparib delayed the growth of irradiated Lewis lung carcinoma (LL2) subcutaneous tumours, in agreement with previous reports in human cell lines. However, the olaparib plus IR combination showed a very narrow therapeutic window against LL2 lung orthotopic tumours, with nearly no additional antitumour effect compared with that of IR alone, and tolerability issues emerged at high doses. The addition of AZD6738 greatly enhanced the efficacy of the olaparib plus IR combination treatment against subcutaneous but not orthotopic LL2 tumours. Moreover, olaparib plus AZD6738 administration concomitant with IR even worsened the response to radiation of head and neck orthotopic tumours and induced mucositis. Conclusions These major differences in the responses to treatments between subcutaneous and orthotopic models highlight the importance of using more pathologically relevant models, such as syngeneic orthotopic models, to determine the most appropriate therapeutic approaches for translation to the clinic.
Collapse
Affiliation(s)
- Vanessa Tran Chau
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.,Labex LERMIT, DHU TORINO, SIRIC SOCRATE, Villejuif, France
| | - Winchygn Liu
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.,Labex LERMIT, DHU TORINO, SIRIC SOCRATE, Villejuif, France
| | - Marine Gerbé de Thoré
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.,Labex LERMIT, DHU TORINO, SIRIC SOCRATE, Villejuif, France
| | - Lydia Meziani
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.,Labex LERMIT, DHU TORINO, SIRIC SOCRATE, Villejuif, France
| | - Michele Mondini
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.,Labex LERMIT, DHU TORINO, SIRIC SOCRATE, Villejuif, France
| | - Mark J O'Connor
- Oncology Innovative Medicines and Early Clinical Development, AstraZeneca, Cambridge, UK
| | - Eric Deutsch
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France. .,Labex LERMIT, DHU TORINO, SIRIC SOCRATE, Villejuif, France. .,Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France.
| | - Céline Clémenson
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France. .,Labex LERMIT, DHU TORINO, SIRIC SOCRATE, Villejuif, France.
| |
Collapse
|
19
|
Jannetti SA, Zeglis BM, Zalutsky MR, Reiner T. Poly(ADP-Ribose)Polymerase (PARP) Inhibitors and Radiation Therapy. Front Pharmacol 2020; 11:170. [PMID: 32194409 PMCID: PMC7062869 DOI: 10.3389/fphar.2020.00170] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/07/2020] [Indexed: 12/15/2022] Open
Abstract
Poly(ADP-ribose)polymerase-1 (PARP1) is a DNA repair enzyme highly expressed in the nuclei of mammalian cells, with a structure and function that have attracted interest since its discovery. PARP inhibitors, moreover, can be used to induce synthetic lethality in cells where the homologous recombination (HR) pathway is deficient. Several small molecule PARP inhibitors have been approved by the FDA for multiple cancers bearing this deficiency These PARP inhibitors also act as radiosensitizing agents by delaying single strand break (SSB) repair and causing subsequent double strand break (DSB) generation, a concept that has been leveraged in various preclinical models of combination therapy with PARP inhibitors and ionizing radiation. Researchers have determined the efficacy of various PARP inhibitors at sub-cytotoxic concentrations in radiosensitizing multiple human cancer cell lines to ionizing radiation. Furthermore, several groups have begun evaluating combination therapy strategies in mouse models of cancer, and a fluorescent imaging agent that allows for subcellular imaging in real time has been developed from a PARP inhibitor scaffold. Other PARP inhibitor scaffolds have been radiolabeled to create PET imaging agents, some of which have also entered clinical trials. Most recently, these highly targeted small molecules have been radiolabeled with therapeutic isotopes to create radiotherapeutics and radiotheranostics in cancers whose primary interventions are surgical resection and whole-body radiotherapy. In this review we discuss the utilization of these small molecules in combination therapies and in scaffolds for imaging agents, radiotherapeutics, and radiotheranostics. Development of these radiolabeled PARP inhibitors has presented promising results for new interventions in the fight against some of the most intractable cancers.
Collapse
Affiliation(s)
- Stephen A. Jannetti
- Department of Biochemistry, Hunter College, New York, NY, United States
- Ph.D. Program in Biochemistry, CUNY Graduate Center, New York, NY, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Brian M. Zeglis
- Department of Biochemistry, Hunter College, New York, NY, United States
- Ph.D. Program in Biochemistry, CUNY Graduate Center, New York, NY, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Ph.D. Program in Chemistry, CUNY Graduate Center, New York, NY, United States
| | - Michael R. Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, NC, United States
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
20
|
Zhang W, van Gent DC, Incrocci L, van Weerden WM, Nonnekens J. Role of the DNA damage response in prostate cancer formation, progression and treatment. Prostate Cancer Prostatic Dis 2020; 23:24-37. [PMID: 31197228 PMCID: PMC8076026 DOI: 10.1038/s41391-019-0153-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/05/2019] [Accepted: 04/09/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Clinical and preclinical studies have revealed that alterations in DNA damage response (DDR) pathways may play an important role in prostate cancer (PCa) etiology and progression. These alterations can influence PCa responses to radiotherapy and anti-androgen treatment. The identification of DNA repair gene aberrations in PCa has driven the interest for further evaluation whether these genetic changes may serve as biomarkers for patient stratification. METHODS In this review, we summarize the current knowledge on DDR alterations in PCa, their potential impact on clinical interventions and prospects for improved management of PCa. We particularly focus on the influence of DDR gene mutations on PCa initiation and progression and describe the underlying mechanisms. RESULTS AND CONCLUSIONS A better understanding of these mechanisms, will contribute to better disease management as treatment strategies can be chosen based on the specific disease properties, since a growing number of treatments are targeting DDR pathway alterations (such as Poly(ADP-ribose) polymerase inhibitors). Furthermore, the recently discovered crosstalk between the DDR and androgen receptor signaling opens a new array of possible strategies to optimize treatment combinations. We discuss how these recent and ongoing studies will help to improve diagnostic, prognostic and therapeutic approaches for PCa management.
Collapse
Affiliation(s)
- Wenhao Zhang
- grid.5645.2000000040459992XDepartment of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Dik C. van Gent
- grid.5645.2000000040459992XDepartment of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands ,grid.5645.2000000040459992XOncode Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Luca Incrocci
- grid.508717.c0000 0004 0637 3764Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Wytske M. van Weerden
- grid.5645.2000000040459992XDepartment of Experimental Urology, Erasmus MC, Rotterdam, The Netherlands
| | - Julie Nonnekens
- grid.5645.2000000040459992XDepartment of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands ,grid.5645.2000000040459992XDepartment of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
21
|
Lee TW, Wong WW, Dickson BD, Lipert B, Cheng GJ, Hunter FW, Hay MP, Wilson WR. Radiosensitization of head and neck squamous cell carcinoma lines by DNA-PK inhibitors is more effective than PARP-1 inhibition and is enhanced by SLFN11 and hypoxia. Int J Radiat Biol 2019; 95:1597-1612. [PMID: 31490091 DOI: 10.1080/09553002.2019.1664787] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Background and purpose: Poly(ADP-ribose)polymerase-1 (PARP1) and DNA-dependent protein kinase (DNA-PK) play key roles in the repair of radiation-induced DNA double strand breaks, but it is unclear which is the preferred therapeutic target in radiotherapy. Here we compare small molecule inhibitors of both as radiosensitizers of head and neck squamous cell carcinoma (HNSCC) cell lines.Methods: Two PARP1 inhibitors (olaparib, veliparib) and two DNA-PK inhibitors (KU57788, IC87361) were tested in 14 HNSCC cell lines and two non-tumorigenic lines (HEK-293 and WI-38/Va-13), with drug exposure for 6 or 24 h post-irradiation, using regrowth assays. For three lines (UT-SCC-54C, -74B, -76B), radiosensitization was also assessed by clonogenic assay under oxia and acute (6 h) anoxia, and for 54C cells under chronic hypoxia (0.2% O2 for 48 h). Relationships between sensitizer enhancement ratios (SER) and gene expression, assessed by RNA sequencing, were evaluated.Results: The inhibitors were minimally cytotoxic in the absence of radiation, with 74B and 54C cells the most sensitive to both olaparib and KU57788. Median SER values for each inhibitor at 1.1 µM were 1.12 (range 1.02-1.24) for olaparib, 1.08 (1.04-1.13) for veliparib, 1.35 (1.10-1.64) for IC87361 and 1.77 (1.41-2.38) for KU57788. The higher SER values for the DNA-PK inhibitors were observed with all cell lines (except HEK-293) and all concentrations tested and were confirmed by clonogenic assay. Radiosensitization by the DNA-PK inhibitors correlated with expression of SLFN11 mRNA. Radiosensitization by IC87361 and olaparib was significantly enhanced under acute anoxia and chronic hypoxia.Conclusions: The DNA-PK inhibitors KU57788 and IC87361 are more effective radiosensitizers than the PARP-1 inhibitors olaparib and veliparib at non-cytotoxic concentrations in HNSCC cell cultures and their activity is enhanced by SLFN11 and hypoxia.
Collapse
Affiliation(s)
- Tet Woo Lee
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Way Wua Wong
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Benjamin D Dickson
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Barbara Lipert
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Gary J Cheng
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Francis W Hunter
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Michael P Hay
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - William R Wilson
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| |
Collapse
|
22
|
Smigiel JM, Taylor SE, Bryson BL, Tamagno I, Polak K, Jackson MW. Cellular plasticity and metastasis in breast cancer: a pre- and post-malignant problem. JOURNAL OF CANCER METASTASIS AND TREATMENT 2019; 5:47. [PMID: 32355893 PMCID: PMC7192216 DOI: 10.20517/2394-4722.2019.26] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
As a field we have made tremendous strides in treating breast cancer, with a decline in the past 30 years of overall breast cancer mortality. However, this progress is met with little affect once the disease spreads beyond the primary site. With a 5-year survival rate of 22%, 10-year of 13%, for those patients with metastatic breast cancer (mBC), our ability to effectively treat wide spread disease is minimal. A major contributing factor to this ineffectiveness is the complex make-up, or heterogeneity, of the primary site. Within a primary tumor, secreted factors, malignant and pre-malignant epithelial cells, immune cells, stromal fibroblasts and many others all reside alongside each other creating a dynamic environment contributing to metastasis. Furthermore, heterogeneity contributes to our lack of understanding regarding the cells' remarkable ability to undergo epithelial/non-cancer stem cell (CSC) to mesenchymal/CSC (E-M/CSC) plasticity. The enhanced invasion & motility, tumor-initiating potential, and acquired therapeutic resistance which accompanies E-M/CSC plasticity implicates a significant role in metastasis. While most work trying to understand E-M/CSC plasticity has been done on malignant cells, recent evidence is emerging concerning the ability for pre-malignant cells to undergo E-M/CSC plasticity and contribute to the metastatic process. Here we will discuss the importance of E-M/CSC plasticity within malignant and pre-malignant populations of the tumor. Moreover, we will discuss how one may potentially target these populations, ultimately disrupting the metastatic cascade and increasing patient survival for those with mBC.
Collapse
Affiliation(s)
- Jacob M. Smigiel
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Sarah E. Taylor
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Benjamin L. Bryson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ilaria Tamagno
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Kelsey Polak
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Mark W. Jackson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
23
|
Majera D, Skrott Z, Bouchal J, Bartkova J, Simkova D, Gachechiladze M, Steigerova J, Kurfurstova D, Gursky J, Korinkova G, Cwiertka K, Hodny Z, Mistrik M, Bartek J. Targeting genotoxic and proteotoxic stress-response pathways in human prostate cancer by clinically available PARP inhibitors, vorinostat and disulfiram. Prostate 2019; 79:352-362. [PMID: 30499118 DOI: 10.1002/pros.23741] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 10/24/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Castration-resistant prostate cancer (PCa) represents a serious health challenge. Based on mechanistically-supported rationale we explored new therapeutic options based on clinically available drugs with anticancer effects, including inhibitors of PARP1 enzyme (PARPi), and histone deacetylases (vorinostat), respectively, and disulfiram (DSF, known as alcohol-abuse drug Antabuse) and its copper-chelating metabolite CuET that inhibit protein turnover. METHODS Drugs and their combination with ionizing radiation (IR) were tested in various cytotoxicity assays in three human PCa cell lines including radio-resistant stem-cell like derived cells. Mechanistically, DNA damage repair, heat shock and unfolded protein response (UPR) pathways were assessed by immunofluorescence and immunoblotting. RESULTS We observed enhanced sensitivity to PARPi/IR in PC3 cells consistent with lower homologous recombination (HR) repair. Vorinostat sensitized DU145 cells to PARPi/IR and decreased mutant p53. Vorinostat also impaired HR-mediated DNA repair, as determined by Rad51 foci formation and downregulation of TOPBP1 protein, and overcame radio-resistance of stem-cell like DU145-derived cells. All PCa models responded well to CuET or DSF combined with copper. We demonstrated that DSF interacts with copper in the culture media and forms adequate levels of CuET indicating that DSF/copper and CuET may be considered as comparable treatments. Both DSF/copper and CuET evoked hallmarks of UPR in PCa cells, documented by upregulation of ATF4, CHOP and phospho-eIF2α, with ensuing heat shock response encompassing activation of HSF1 and HSP70. Further enhancing the cytotoxicity of CuET, combination with an inhibitor of the anti-apoptotic protein survivin (YM155, currently undergoing clinical trials) promoted the UPR-induced toxicity, yielding synergistic effects of CuET and YM155. CONCLUSIONS We propose that targeting genotoxic and proteotoxic stress responses by combinations of available drugs could inspire innovative strategies to treat castration-resistant PCa.
Collapse
Affiliation(s)
- Dusana Majera
- Laboratory of Genome Integrity, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Zdenek Skrott
- Laboratory of Genome Integrity, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Jan Bouchal
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Jirina Bartkova
- Danish Cancer Society Research Center, Copenhagen, Denmark
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| | - Dana Simkova
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Mariam Gachechiladze
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Jana Steigerova
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Daniela Kurfurstova
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Jan Gursky
- Laboratory of Genome Integrity, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Gabriela Korinkova
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Karel Cwiertka
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University, University Hospital, Olomouc, Czech Republic
| | - Zdenek Hodny
- Department of Genome Integrity, Institute of Molecular Genetics of the CAS, v.v.i., Prague, Czech Republic
| | - Martin Mistrik
- Laboratory of Genome Integrity, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Jiri Bartek
- Laboratory of Genome Integrity, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
- Danish Cancer Society Research Center, Copenhagen, Denmark
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
- Department of Genome Integrity, Institute of Molecular Genetics of the CAS, v.v.i., Prague, Czech Republic
| |
Collapse
|
24
|
Zhong H, Chen C, Tammali R, Breen S, Zhang J, Fazenbaker C, Kennedy M, Conway J, Higgs BW, Holoweckyj N, Raja R, Harper J, Pierce AJ, Herbst R, Tice DA. Improved Therapeutic Window in BRCA-mutant Tumors with Antibody-linked Pyrrolobenzodiazepine Dimers with and without PARP Inhibition. Mol Cancer Ther 2018; 18:89-99. [DOI: 10.1158/1535-7163.mct-18-0314] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 07/31/2018] [Accepted: 10/16/2018] [Indexed: 11/16/2022]
|
25
|
Laird JH, Lok BH, Ma J, Bell A, de Stanchina E, Poirier JT, Rudin CM. Talazoparib Is a Potent Radiosensitizer in Small Cell Lung Cancer Cell Lines and Xenografts. Clin Cancer Res 2018; 24:5143-5152. [PMID: 29945991 DOI: 10.1158/1078-0432.ccr-18-0401] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/09/2018] [Accepted: 06/22/2018] [Indexed: 12/12/2022]
Abstract
Purpose: Small cell lung cancer (SCLC) is an aggressive malignancy with a critical need for novel therapies. Our goal was to determine whether PARP inhibition could sensitize SCLC cells to ionizing radiation (IR) and if so, to determine the contribution of PARP trapping to radiosensitization.Experimental Design: Short-term viability assays and clonogenic survival assays (CSA) were used to assess radiosensitization in 6 SCLC cell lines. Doses of veliparib and talazoparib with equivalent enzymatic inhibitory activity but differing PARP trapping activity were identified and compared in CSAs. Talazoparib, IR, and their combination were tested in three patient-derived xenograft (PDX) models.Results: Talazoparib radiosensitized 5 of 6 SCLC cell lines in short-term viability assays and confirmed in 3 of 3 cell lines by CSAs. Concentrations of 200 nmol/L talazoparib and 1,600 nmol/L veliparib similarly inhibited PAR polymerization; however, talazoparib exhibited greater PARP trapping activity that was associated with superior radiosensitization. This observation further correlated with an increased number of double-stranded DNA breaks induced by talazoparib as compared with veliparib. Finally, a dose of 0.2 mg/kg talazoparib in vivo caused tumor growth inhibition in combination with IR but not as a single agent in 3 SCLC PDX models.Conclusions: PARP inhibition effectively sensitizes SCLC cell lines and PDXs to IR, and PARP trapping activity enhances this effect. PARP inhibitors, especially those with high PARP trapping activity, may provide a powerful tool to improve the efficacy of radiotherapy in SCLC. Clin Cancer Res; 24(20); 5143-52. ©2018 AACR.
Collapse
Affiliation(s)
- James H Laird
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York.,New York University School of Medicine, New York, New York
| | - Benjamin H Lok
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jennifer Ma
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.,Albert Einstein College of Medicine, Bronx, New York
| | - Andrew Bell
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John T Poirier
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York. .,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Charles M Rudin
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York. .,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| |
Collapse
|
26
|
Labay E, Mauceri HJ, Efimova EV, Flor AC, Sutton HG, Kron SJ, Weichselbaum RR. Repurposing cephalosporin antibiotics as pro-senescent radiosensitizers. Oncotarget 2017; 7:33919-33. [PMID: 27129153 PMCID: PMC5085128 DOI: 10.18632/oncotarget.8984] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/13/2016] [Indexed: 12/11/2022] Open
Abstract
Radiation therapy remains a significant therapeutic modality in the treatment of cancer. An attractive strategy would be to enhance the benefits of ionizing radiation (IR)with radiosensitizers. A high-content drug repurposing screen of approved and investigational agents, natural products and other small molecules has identified multiple candidates that blocked repair of IR damage in vitro. Here, we validated a subset of these hits in vitro and then examined effects on tumor growth after IR in a murine tumor model. Based on robust radiosensitization in vivo and other favorable properties of cephalexin, we conducted additional studies with other beta-lactam antibiotics. When combined with IR, each cephalosporin tested increased DNA damage and slowed tumor growth without affecting normal tissue toxicity. Our data implicate reactive oxygen species in the mechanism by which cephalosporins augment the effects of IR. This work provides a rationale for using commonly prescribed beta-lactam antibiotics as non-toxic radiosensitizers to enhance the therapeutic ratio of radiotherapy.
Collapse
Affiliation(s)
- Edwardine Labay
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
| | - Helena J Mauceri
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
| | - Elena V Efimova
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL, USA
| | - Amy C Flor
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL, USA
| | - Harold G Sutton
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
| | - Stephen J Kron
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL, USA.,Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA.,Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
27
|
Lourenco LM, Jiang Y, Drobnitzky N, Green M, Cahill F, Patel A, Shanneik Y, Moore J, Ryan AJ. PARP Inhibition Combined With Thoracic Irradiation Exacerbates Esophageal and Skin Toxicity in C57BL6 Mice. Int J Radiat Oncol Biol Phys 2017; 100:767-775. [PMID: 29413287 DOI: 10.1016/j.ijrobp.2017.10.051] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/23/2017] [Accepted: 10/30/2017] [Indexed: 12/17/2022]
Abstract
PURPOSE Poly (ADP-ribose) polymerase (PARP) inhibitors have been shown to enhance the radiosensitivity of cancer cells in vitro in a replication-dependent manner. Their in vivo radiosensitizing effects have also been demonstrated in preclinical tumor models. However, whether PARP inhibition can enhance the response to radiation therapy in normal tissues has been largely neglected. We hypothesized that PARP inhibition might also potentiate the response of replicating normal tissues to radiation therapy. In this study, we examined the normal tissue response in mice treated with PARP inhibitors (BMN673 or AZD2281) in combination with thoracic irradiation. METHODS AND MATERIALS The antitumor effects of fractionated irradiation (5 Gy × 4) in combination with BMN673 were evaluated in nude mice bearing established Calu-6 human lung cancer xenografts. The normal tissue response was evaluated in C57BL6 mice that were treated with BMN673 or AZD2281 combined with fractionated irradiation, 5 Gy × 4, delivered to the whole thorax. Body weight and histology of the esophagus and skin in the field of irradiation were examined. The DNA damage response in the esophagus and skin was assessed by γH2AX immunohistochemistry. RESULTS While PARP inhibition enhanced irradiation-induced tumor growth inhibition in nude mice, it was also associated with significant body weight loss and increased damage to the esophagus and skin within the field of irradiation in C57BL6 mice. PARP inhibition compromised the repair of irradiation-induced DNA damage in the esophagus and skin. CONCLUSIONS Although PARP inhibition enhanced the antitumor response to fractionated irradiation, it also enhanced the irradiation response in replicating normal tissues. Therefore, our study suggests that additional caution may be warranted in the clinical development of combination therapies using PARP inhibitors and radiation therapy, in particular where the field of irradiation includes the esophagus.
Collapse
Affiliation(s)
- Luiza Madia Lourenco
- Cancer Research UK and UK Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Yanyan Jiang
- Cancer Research UK and UK Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Neele Drobnitzky
- Cancer Research UK and UK Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Marcus Green
- Cancer Research UK and UK Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Fiona Cahill
- Cancer Research UK and UK Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Agata Patel
- Cancer Research UK and UK Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Yasmin Shanneik
- Cancer Research UK and UK Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - John Moore
- Cancer Research UK and UK Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Anderson J Ryan
- Cancer Research UK and UK Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK.
| |
Collapse
|
28
|
Lesueur P, Chevalier F, Austry JB, Waissi W, Burckel H, Noël G, Habrand JL, Saintigny Y, Joly F. Poly-(ADP-ribose)-polymerase inhibitors as radiosensitizers: a systematic review of pre-clinical and clinical human studies. Oncotarget 2017; 8:69105-69124. [PMID: 28978184 PMCID: PMC5620324 DOI: 10.18632/oncotarget.19079] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 06/19/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Poly-(ADP-Ribose)-Polymerase (PARP) inhibitors are becoming important actors of anti-neoplasic agents landscape, with recent but narrow FDA's approvals for ovarian BRCA mutated cancers and prostatic cancer. Nevertheless, PARP inhibitors are also promising drugs for combined treatments particularly with radiotherapy. More than seven PARP inhibitors have been currently developed. Central Role of PARP in DNA repair, makes consider PARP inhibitor as potential radiosensitizers, especially for tumors with DNA repair defects, such as BRCA mutation, because of synthetic lethality. Furthermore the replication-dependent activity of PARP inhibitor helps to maintain the differential effect between tumoral and healthy tissues. Inhibition of chromatin remodeling, G2/M arrest, vasodilatory effect induced by PARP inhibitor, also participate to their radio-sensitization effect. MATERIALS AND METHODS Here, after highlighting mechanisms of PARP inhibitors radiosensitization we methodically searched PubMed, Google Scholar, Cochrane Databases and meeting proceedings for human pre-clinical and clinical studies that evaluated PARP inhibitor radiosensitizing effect. Enhancement ratio, when available, was systematically reported. RESULTS Sixty four studies finally met our selection criteria and were included in the analysis. Only three pre-clinical studies didn't find any radiosensitizing effect. Median enhancement ratio vary from 1,3 for prostate tumors to 1,5 for lung cancers. Nine phase I or II trials assessed safety data. CONCLUSION PARP inhibitors are promising radiosensitizers, but need more clinical investigation. The next ten years will be determining for judging their real potential.
Collapse
Affiliation(s)
- Paul Lesueur
- Laboratoire d'Accueil et de Recherche avec les Ions Accélérés, CEA, CIMAP-GANIL, 14000 Caen, France.,Centre Francois Baclesse Centre de Lutte Contre le Cancer, Radiotherapy Unit, 14000 Caen, France
| | - François Chevalier
- Laboratoire d'Accueil et de Recherche avec les Ions Accélérés, CEA, CIMAP-GANIL, 14000 Caen, France
| | - Jean-Baptiste Austry
- Laboratoire d'Accueil et de Recherche avec les Ions Accélérés, CEA, CIMAP-GANIL, 14000 Caen, France
| | - Waisse Waissi
- EA 3430, Laboratoire de Radiobiologie, Centre Paul Strauss, 67000 Strasbourg, France
| | - Hélène Burckel
- EA 3430, Laboratoire de Radiobiologie, Centre Paul Strauss, 67000 Strasbourg, France
| | - Georges Noël
- EA 3430, Laboratoire de Radiobiologie, Centre Paul Strauss, 67000 Strasbourg, France
| | - Jean-Louis Habrand
- Centre Francois Baclesse Centre de Lutte Contre le Cancer, Radiotherapy Unit, 14000 Caen, France
| | - Yannick Saintigny
- Laboratoire d'Accueil et de Recherche avec les Ions Accélérés, CEA, CIMAP-GANIL, 14000 Caen, France
| | - Florence Joly
- Centre Francois Baclesse Centre de Lutte Contre le Cancer, Clinical Research Unit, 14000 Caen, France
| |
Collapse
|
29
|
Lohse I, Kumareswaran R, Cao P, Pitcher B, Gallinger S, Bristow RG, Hedley DW. Effects of Combined Treatment with Ionizing Radiation and the PARP Inhibitor Olaparib in BRCA Mutant and Wild Type Patient-Derived Pancreatic Cancer Xenografts. PLoS One 2016; 11:e0167272. [PMID: 28033382 PMCID: PMC5199060 DOI: 10.1371/journal.pone.0167272] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 10/28/2016] [Indexed: 02/07/2023] Open
Abstract
Background The BRCA2 gene product plays an important role in DNA double strand break repair. Therefore, we asked whether radiation sensitivity of pancreatic cancers developing in individuals with germline BRCA2 mutations can be enhanced by agents that inhibit poly (ADP-ribose) polymerase (PARP). Methods We compared the sensitivity of two patient-derived pancreatic cancer xenografts, expressing a truncated or wild type BRCA 2, to ionizing radiation alone or in combination with olaparib (AZD-2281). Animals were treated with either a single dose of 12Gy, 7 days of olaparib or 7 days of olaparib followed by a single dose of 12Gy. Response was assessed by tumour growth delay and the activation of damage response pathways. Results The BRCA2 mutated and wild type tumours showed similar radiation sensitivity, and treatment with olaparib did not further sensitize either model when compared to IR alone. Conclusions While PARP inhibition has been shown to be effective in BRCA-mutated breast and ovarian cancers, it is less well established in pancreatic cancer patients. Our results show no radiosensitization in a germline BRCA 2 mutant and suggest that combining PARP inhibition and IR may not be beneficial in BRCA 2 related pancreatic tumors.
Collapse
Affiliation(s)
- Ines Lohse
- Ontario Cancer Institute and Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ramya Kumareswaran
- Ontario Cancer Institute and Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Pinjiang Cao
- Ontario Cancer Institute and Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Bethany Pitcher
- Ontario Cancer Institute and Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Steven Gallinger
- Mount Sinai Hospital, Joseph and Wolf Lebovic Health Complex, Toronto, Ontario, Canada
- Translational Research Initiative in Pancreas Cancer, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Robert G. Bristow
- Ontario Cancer Institute and Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - David W. Hedley
- Ontario Cancer Institute and Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Oncology and Haematology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
30
|
Somnay Y, Lubner S, Gill H, Matsumura JB, Chen H. The PARP inhibitor ABT-888 potentiates dacarbazine-induced cell death in carcinoids. Cancer Gene Ther 2016; 23:348-354. [PMID: 27632933 PMCID: PMC5083201 DOI: 10.1038/cgt.2016.39] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 08/12/2016] [Indexed: 02/07/2023]
Abstract
Monoagent DNA-alkylating chemotherapies like dacarbazine are among a paucity of medical treatments for advanced carcinoid tumors, but are limited by host toxicity and intrinsic chemoresistance through the base excision repair (BER) pathway via poly (ADP-ribose) polymerase (PARP). Hence, inhibitors of PARP may potentiate DNA-damaging agents by blocking BER and DNA restoration. We show that the PARP inhibitor ABT-888 (Veliparib) enhances the cytotoxic effects of dacarbazine in carcinoids. Two human carcinoid cell lines (BON and H727) treated with a combination of ABT-888 and dacarbazine resulted in synergistic growth inhibition signified by combination indices <1 on the Chou-Talalay scale. ABT-888 administered prior to varying dacarbazine doses promoted the suppression of neuroendocrine biomarkers of malignancy ASCL1 and CgA, shown by Western analysis. ATM phosphorylation and p21Waf1/Cip1 activation, indicative of DNA damage, were increased by ABT-888 when combined with dacarbazine treatment, suggesting BER pathway attenuation by ABT-888. PE Annexin V/7-AAD staining and sorting revealed a profound induction of apoptosis following combination treatment, which was further confirmed by increased PARP cleavage. These results demonstrate that ABT-888 synergizes dacarbazine treatment in carcinoids. Therefore, ABT-888 may help treat carcinoids unresponsive or refractory to mainstay therapies.
Collapse
Affiliation(s)
- Y Somnay
- Endocrine Surgery Research Laboratories, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - S Lubner
- Division of Hematology and Medical Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - H Gill
- Endocrine Surgery Research Laboratories, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - J B Matsumura
- Endocrine Surgery Research Laboratories, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - H Chen
- Endocrine Surgery Research Laboratories, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Surgery, University of Alabama- Birmingham, Birmingham, AL, USA
| |
Collapse
|
31
|
Gewirtz DA, Alotaibi M, Yakovlev VA, Povirk LF. Tumor Cell Recovery from Senescence Induced by Radiation with PARP Inhibition. Radiat Res 2016; 186:327-332. [PMID: 27588595 DOI: 10.1667/rr14437.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Inhibitors of poly(ADP-ribose) polymerase (PARP) are clinically used as single-agent therapy for tumors with BRCA1 or BRCA2 mutations. One approach to expanding the use of PARP inhibitors to a wider range of tumors is to combine them with cytotoxic chemotherapy or radiotherapy. Preclinical studies in experimental animals and tumor cells in culture indicate that PARP inhibition modestly sensitizes most tumor cells to ionizing radiation. Studies of cell behavior after these combined treatments show that radiosensitization is manifested predominantly in an increase in prolonged growth arrest and senescence, with little or no contribution from apoptosis. The secretory phenotype associated with senescence can target these tumor cells for immune surveillance, and therefore increased senescence can effectively contribute to tumor control. However, the possible recovery of senescent cells and re-entry into cell cycle after prolonged arrest also needs to be considered. Such recovery could lead to tumor recurrence, yet may not be reflected in short-term assays commonly used to assess radiosensitization.
Collapse
Affiliation(s)
| | - Moureq Alotaibi
- Department of a Pharmacology, Toxicology and Medicine and.,c College of Pharmacy, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Vasily A Yakovlev
- b Radiation Oncology, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298; and
| | | |
Collapse
|
32
|
Jiang Y, Verbiest T, Devery AM, Bokobza SM, Weber AM, Leszczynska KB, Hammond EM, Ryan AJ. Hypoxia Potentiates the Radiation-Sensitizing Effect of Olaparib in Human Non-Small Cell Lung Cancer Xenografts by Contextual Synthetic Lethality. Int J Radiat Oncol Biol Phys 2016; 95:772-81. [PMID: 27020103 PMCID: PMC4856738 DOI: 10.1016/j.ijrobp.2016.01.035] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/22/2015] [Accepted: 01/18/2016] [Indexed: 12/13/2022]
Abstract
PURPOSE Poly(ADP-ribose) polymerase (PARP) inhibitors potentiate radiation therapy in preclinical models of human non-small cell lung cancer (NSCLC) and other types of cancer. However, the mechanisms underlying radiosensitization in vivo are incompletely understood. Herein, we investigated the impact of hypoxia on radiosensitization by the PARP inhibitor olaparib in human NSCLC xenograft models. METHODS AND MATERIALS NSCLC Calu-6 and Calu-3 cells were irradiated in the presence of olaparib or vehicle under normoxic (21% O2) or hypoxic (1% O2) conditions. In vitro radiosensitivity was assessed by clonogenic survival assay and γH2AX foci assay. Established Calu-6 and Calu-3 subcutaneous xenografts were treated with olaparib (50 mg/kg, daily for 3 days), radiation (10 Gy), or both. Tumors (n=3/group) were collected 24 or 72 hours after the first treatment. Immunohistochemistry was performed to assess hypoxia (carbonic anhydrase IX [CA9]), vessels (CD31), DNA double strand breaks (DSB) (γH2AX), and apoptosis (cleaved caspase 3 [CC3]). The remaining xenografts (n=6/group) were monitored for tumor growth. RESULTS In vitro, olaparib showed a greater radiation-sensitizing effect in Calu-3 and Calu-6 cells in hypoxic conditions (1% O2). In vivo, Calu-3 tumors were well-oxygenated, whereas Calu-6 tumors had extensive regions of hypoxia associated with down-regulation of the homologous recombination protein RAD51. Olaparib treatment increased unrepaired DNA DSB (P<.001) and apoptosis (P<.001) in hypoxic cells of Calu-6 tumors following radiation, whereas it had no significant effect on radiation-induced DNA damage response in nonhypoxic cells of Calu-6 tumors or in the tumor cells of well-oxygenated Calu-3 tumors. Consequently, olaparib significantly increased radiation-induced growth inhibition in Calu-6 tumors (P<.001) but not in Calu-3 tumors. CONCLUSIONS Our data suggest that hypoxia potentiates the radiation-sensitizing effects of olaparib by contextual synthetic killing, and that tumor hypoxia may be a potential biomarker for selecting patients who may get the greatest benefit from the addition of olaparib to radiation therapy.
Collapse
Affiliation(s)
- Yanyan Jiang
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Tom Verbiest
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Aoife M Devery
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Sivan M Bokobza
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Anika M Weber
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Katarzyna B Leszczynska
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Ester M Hammond
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Anderson J Ryan
- Cancer Research UK/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
33
|
Alotaibi M, Sharma K, Saleh T, Povirk LF, Hendrickson EA, Gewirtz DA. Radiosensitization by PARP Inhibition in DNA Repair Proficient and Deficient Tumor Cells: Proliferative Recovery in Senescent Cells. Radiat Res 2016; 185:229-45. [PMID: 26934368 PMCID: PMC4821451 DOI: 10.1667/rr14202.1] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Radiotherapy continues to be a primary modality in the treatment of cancer. In addition to promoting apoptosis, radiation-induced DNA damage can promote autophagy and senescence, both of which can theoretically function to prolong tumor survival. In this work, we tested the hypothesis that autophagy and/or senescence could be permissive for DNA repair, thereby facilitating tumor cell recovery from radiation-induced growth arrest and/or cell death. In addition, studies were designed to elucidate the involvement of autophagy and senescence in radiosensitization by PARP inhibitors and the re-emergence of a proliferating tumor cell population. In the context of this work, the relationship between radiation-induced autophagy and senescence was also determined. Studies were performed using DNA repair-proficient HCT116 colon carcinoma cells and a repair-deficient ligase IV(-/-) isogenic cell line. Exposure to radiation promoted a parallel induction of autophagy and senescence that was strongly correlated with the extent of persistent H2AX phosphorylation in both cell lines, however, inhibition of autophagy failed to suppress senescence, indicating that the two responses were dissociable. Exposure to radiation resulted in a transient arrest in the HCT116 cells while arrest was prolonged in the ligase IV(-/-) cells, however, both cell lines ultimately recovered proliferative function, which may reflect maintenance of DNA repair capacity. The PARP inhibitors, olaparib and niraparib, increased the extent of persistent DNA damage induced by radiation exposure as well as the extent of both autophagy and senescence. Neither cell line underwent significant apoptosis by radiation exposure alone or in the presence of the PARP inhibitors. Inhibition of autophagy failed to attenuate radiosensitization, indicating that autophagy was not involved in the action of the PARP inhibitors. As with radiation alone, despite sensitization by PARP inhibition, proliferative recovery was evident within a period of 10-20 days. While inhibition of DNA repair via PARP inhibition may initially sensitize tumor cells to radiation via the promotion of senescence, this strategy does not appear to interfere with proliferative recovery, which could ultimately contribute to disease recurrence.
Collapse
Affiliation(s)
- Moureq Alotaibi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University,
P.O. Box 2457, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Khushboo Sharma
- Department of Pharmacology and Toxicology, Virginia Commonwealth University
| | - Tareq Saleh
- Department of Pharmacology and Toxicology, Virginia Commonwealth University
| | - Lawrence F. Povirk
- Department of Pharmacology and Toxicology, Virginia Commonwealth University
| | - Eric A. Hendrickson
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis MN 55455
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University
- Department of Medicine, Massey Cancer Center
| |
Collapse
|
34
|
Nassour J, Martien S, Martin N, Deruy E, Tomellini E, Malaquin N, Bouali F, Sabatier L, Wernert N, Pinte S, Gilson E, Pourtier A, Pluquet O, Abbadie C. Defective DNA single-strand break repair is responsible for senescence and neoplastic escape of epithelial cells. Nat Commun 2016; 7:10399. [PMID: 26822533 PMCID: PMC4740115 DOI: 10.1038/ncomms10399] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 12/08/2015] [Indexed: 12/22/2022] Open
Abstract
The main characteristic of senescence is its stability which relies on the persistence of DNA damage. We show that unlike fibroblasts, senescent epithelial cells do not activate an ATM-or ATR-dependent DNA damage response (DDR), but accumulate oxidative-stress-induced DNA single-strand breaks (SSBs). These breaks remain unrepaired because of a decrease in PARP1 expression and activity. This leads to the formation of abnormally large and persistent XRCC1 foci that engage a signalling cascade involving the p38MAPK and leading to p16 upregulation and cell cycle arrest. Importantly, the default in SSB repair also leads to the emergence of post-senescent transformed and mutated precancerous cells. In human-aged skin, XRCC1 foci accumulate in the epidermal cells in correlation with a decline of PARP1, whereas DDR foci accumulate mainly in dermal fibroblasts. These findings point SSBs as a DNA damage encountered by epithelial cells with aging which could fuel the very first steps of carcinogenesis.
Collapse
Affiliation(s)
- Joe Nassour
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Sébastien Martien
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Nathalie Martin
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Emeric Deruy
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Elisa Tomellini
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Nicolas Malaquin
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Fatima Bouali
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Laure Sabatier
- Commissariat à l'Energie Atomique (CEA), Laboratoire de Radiobiologie et Oncologie (LRO), 18 route du Panorama - BP6, 92265 Fontenay-aux-Roses 53011, France
| | - Nicolas Wernert
- Institute of Pathology, University of Bonn, 53011 Bonn, Germany
| | - Sébastien Pinte
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France.,Institute for Research on Cancer and Aging, Nice (IRCAN), University of Nice Sophia Antipolis, CNRS, UMR7284, INSERM U108, Faculty of Medecine of Nice; CHU of Nice, Nice, France
| | - Eric Gilson
- Institute for Research on Cancer and Aging, Nice (IRCAN), University of Nice Sophia Antipolis, CNRS, UMR7284, INSERM U108, Faculty of Medecine of Nice; CHU of Nice, Nice, France
| | - Albin Pourtier
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Olivier Pluquet
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Corinne Abbadie
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| |
Collapse
|
35
|
In vivo studies of the PARP inhibitor, AZD-2281, in combination with fractionated radiotherapy: An exploration of the therapeutic ratio. Radiother Oncol 2015; 116:486-94. [PMID: 26277432 DOI: 10.1016/j.radonc.2015.08.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 07/29/2015] [Accepted: 08/01/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND PURPOSE Pre-clinical data have shown that PARP inhibitors (PARPi) may increase the efficacy of radiotherapy in prostate cancer. However, it is uncertain as to whether PARPi lead to clonogenic kill when combined with radiotherapy (RT). MATERIAL AND METHODS We tested the PARP inhibitor AZD-2281 as a radiosensitizing agent under oxic and hypoxic conditions for clonogenic survival in vitro and in vivo using the human prostate cancer cell line, 22Rv1. In addition, the effects of PARPi+RT on normal tissue were investigated using a crypt clonogenic assay. RESULTS AZD-2281 inhibited cellular PARP activity under both oxic and hypoxic conditions. The addition of AZD-2281 radiosensitized 22Rv1 cells under oxia, acute hypoxia and chronic hypoxia in vitro. The combination of AZD-2281 with fractionated radiotherapy resulted in a significant growth delay and clonogenic kill in vivo. No increased gut toxicity was observed using this combined PARPi+radiotherapy regimen. CONCLUSIONS This is the first preclinical study to demonstrate direct clonogenic kill in vivo by the addition of AZD-2281 to radiotherapy. As we did not observe gut toxicity, the use of PARPi in the context of prostate cancer radiotherapy warrants further investigation in clinical trials.
Collapse
|
36
|
Malyuchenko NV, Kotova EY, Kulaeva OI, Kirpichnikov MP, Studitskiy VM. PARP1 Inhibitors: antitumor drug design. Acta Naturae 2015; 7:27-37. [PMID: 26483957 PMCID: PMC4610162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
The poly (ADP-ribose) polymerase 1 (PARP1) enzyme is one of the promising molecular targets for the discovery of antitumor drugs. PARP1 is a common nuclear protein (1-2 million molecules per cell) serving as a "sensor" for DNA strand breaks. Increased PARP1 expression is sometimes observed in melanomas, breast cancer, lung cancer, and other neoplastic diseases. The PARP1 expression level is a prognostic indicator and is associated with a poor survival prognosis. There is evidence that high PARP1 expression and treatment-resistance of tumors are correlated. PARP1 inhibitors are promising antitumor agents, since they act as chemo- and radiosensitizers in the conventional therapy of malignant tumors. Furthermore, PARP1 inhibitors can be used as independent, effective drugs against tumors with broken DNA repair mechanisms. Currently, third-generation PARP1 inhibitors are being developed, many of which are undergoing Phase II clinical trials. In this review, we focus on the properties and features of the PARP1 inhibitors identified in preclinical and clinical trials. We also describe some problems associated with the application of PARP1 inhibitors. The possibility of developing new PARP1 inhibitors aimed at DNA binding and transcriptional activity rather than the catalytic domain of the protein is discussed.
Collapse
Affiliation(s)
- N. V. Malyuchenko
- Lomonosov Moscow State University, Leninskie Gory, 1/12, Moscow, 119991, Russia
| | - E. Yu. Kotova
- Fox Chase Cancer Center, Philadelphia, PA, 19111-2497, USA
| | - O. I. Kulaeva
- Lomonosov Moscow State University, Leninskie Gory, 1/12, Moscow, 119991, Russia
- Fox Chase Cancer Center, Philadelphia, PA, 19111-2497, USA
| | - M. P. Kirpichnikov
- Lomonosov Moscow State University, Leninskie Gory, 1/12, Moscow, 119991, Russia
| | - V. M. Studitskiy
- Lomonosov Moscow State University, Leninskie Gory, 1/12, Moscow, 119991, Russia
- Fox Chase Cancer Center, Philadelphia, PA, 19111-2497, USA
| |
Collapse
|
37
|
Bridges KA, Toniatti C, Buser CA, Liu H, Buchholz TA, Meyn RE. Niraparib (MK-4827), a novel poly(ADP-Ribose) polymerase inhibitor, radiosensitizes human lung and breast cancer cells. Oncotarget 2015; 5:5076-86. [PMID: 24970803 PMCID: PMC4148123 DOI: 10.18632/oncotarget.2083] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The aim of this study was to assess niraparib (MK-4827), a novel poly(ADP-Ribose) polymerase (PARP) inhibitor, for its ability to radiosensitize human tumor cells. Human tumor cells derived from lung, breast and prostate cancers were tested for radiosensitization by niraparib using clonogenic survival assays. Both p53 wild-type and p53-defective lines were included. The ability of niraparib to alter the repair of radiation-induced DNA double strand breaks (DSBs) was determined using detection of γ-H2AX foci and RAD51 foci. Clonogenic survival analyses indicated that micromolar concentrations of niraparib radiosensitized tumor cell lines derived from lung, breast, and prostate cancers independently of their p53 status but not cell lines derived from normal tissues. Niraparib also sensitized tumor cells to H2O2 and converted H2O2-induced single strand breaks (SSBs) into DSBs during DNA replication. These results indicate that human tumor cells are significantly radiosensitized by the potent and selective PARP-1 inhibitor, niraparib, in the in vitro setting. The mechanism of this effect appears to involve a conversion of sublethal SSBs into lethal DSBs during DNA replication due to the inhibition of base excision repair by the drug. Taken together, our findings strongly support the clinical evaluation of niraparib in combination with radiation.
Collapse
Affiliation(s)
- Kathleen A Bridges
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - Huifeng Liu
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Thomas A Buchholz
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Raymond E Meyn
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
38
|
Barboro P, Ferrari N, Capaia M, Petretto A, Salvi S, Boccardo S, Balbi C. Expression of nuclear matrix proteins binding matrix attachment regions in prostate cancer. PARP-1: New player in tumor progression. Int J Cancer 2015; 137:1574-86. [DOI: 10.1002/ijc.29531] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/11/2015] [Accepted: 03/17/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Paola Barboro
- IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro, Dipartimento Diagnostica della Patologia e delle Cure ad Alta Complessità Tecnologica; Genoa Italy
| | - Nicoletta Ferrari
- IRCCs AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro, Dipartimento Terapie Oncologiche Integrate; Genoa Italy
| | - Matteo Capaia
- IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro, Dipartimento Diagnostica della Patologia e delle Cure ad Alta Complessità Tecnologica; Genoa Italy
| | - Andrea Petretto
- Laboratorio di Spettrometria di Massa, Core Facility, Istituto Giannina Gaslini; Genoa Italy
| | - Sandra Salvi
- IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro, Dipartimento Diagnostica della Patologia e delle Cure ad Alta Complessità Tecnologica; Genoa Italy
| | - Simona Boccardo
- IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro, Dipartimento Diagnostica della Patologia e delle Cure ad Alta Complessità Tecnologica; Genoa Italy
| | - Cecilia Balbi
- IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro, Dipartimento Diagnostica della Patologia e delle Cure ad Alta Complessità Tecnologica; Genoa Italy
| |
Collapse
|
39
|
Krajewska M, Fehrmann RSN, de Vries EGE, van Vugt MATM. Regulators of homologous recombination repair as novel targets for cancer treatment. Front Genet 2015; 6:96. [PMID: 25852742 PMCID: PMC4367534 DOI: 10.3389/fgene.2015.00096] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 02/23/2015] [Indexed: 12/20/2022] Open
Abstract
To cope with DNA damage, cells possess a complex signaling network called the ‘DNA damage response’, which coordinates cell cycle control with DNA repair. The importance of this network is underscored by the cancer predisposition that frequently goes along with hereditary mutations in DNA repair genes. One especially important DNA repair pathway in this respect is homologous recombination (HR) repair. Defects in HR repair are observed in various cancers, including hereditary breast, and ovarian cancer. Intriguingly, tumor cells with defective HR repair show increased sensitivity to chemotherapeutic reagents, including platinum-containing agents. These observations suggest that HR-proficient tumor cells might be sensitized to chemotherapeutics if HR repair could be therapeutically inactivated. HR repair is an extensively regulated process, which depends strongly on the activity of various other pathways, including cell cycle pathways, protein-control pathways, and growth factor-activated receptor signaling pathways. In this review, we discuss how the mechanistic wiring of HR is controlled by cell-intrinsic or extracellular pathways. Furthermore, we have performed a meta-analysis on available genome-wide RNA interference studies to identify additional pathways that control HR repair. Finally, we discuss how these HR-regulatory pathways may provide therapeutic targets in the context of radio/chemosensitization.
Collapse
Affiliation(s)
- Małgorzata Krajewska
- Department of Medical Oncology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen Groningen, Netherlands
| | - Rudolf S N Fehrmann
- Department of Medical Oncology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen Groningen, Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen Groningen, Netherlands
| | - Marcel A T M van Vugt
- Department of Medical Oncology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen Groningen, Netherlands
| |
Collapse
|
40
|
Abstract
The development of poly (adenosine diphosphate [ADP]) ribose polymerase (PARP) inhibitors (PARPi) has progressed greatly over the last few years and has shown encouraging results in the BRCA1/2 mutation–related cancers. This article attempts to summarize the rationale and theory behind PARPi, the clinical trials already reported, as well as ongoing studies designed to determine the role of PARPi in patients with and without germline mutations of BRCA genes. Future plans for PARPi both as monotherapy and in combination with standard cytotoxics, other biological agents, and as radiosensitizers are also covered. The widening scope of PARPi adds another important targeted agent to the growing list of molecular inhibitors; future and ongoing trials will identify the most effective role for PARPi, including for patients other than BRCA germline mutation carriers.
Collapse
Affiliation(s)
- Sarah Benafif
- Mount Vernon Cancer Centre, Northwood, Middlesex, UK
| | - Marcia Hall
- Mount Vernon Cancer Centre, Northwood, Middlesex, UK
| |
Collapse
|
41
|
Biss M, Xiao W. Selective tumor killing based on specific DNA-damage response deficiencies. Cancer Biol Ther 2014; 13:239-46. [DOI: 10.4161/cbt.18921] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
42
|
Radiation oncology in vitro: trends to improve radiotherapy through molecular targets. BIOMED RESEARCH INTERNATIONAL 2014; 2014:461687. [PMID: 25302298 PMCID: PMC4180203 DOI: 10.1155/2014/461687] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 07/16/2014] [Indexed: 12/17/2022]
Abstract
Much has been investigated to improve the beneficial effects of radiotherapy especially in that case where radioresistant behavior is observed. Beyond simple identification of resistant phenotype the discovery and development of specific molecular targets have demonstrated therapeutic potential in cancer treatment including radiotherapy. Alterations on transduction signaling pathway related with MAPK cascade are the main axis in cancer cellular proliferation even as cell migration and invasiveness in irradiated tumor cell lines; then, for that reason, more studies are in course focusing on, among others, DNA damage enhancement, apoptosis stimulation, and growth factors receptor blockages, showing promising in vitro results highlighting molecular targets associated with ionizing radiation as a new radiotherapy strategy to improve clinical outcome. In this review we discuss some of the main molecular targets related with tumor cell proliferation and migration as well as their potential contributions to radiation oncology improvements.
Collapse
|
43
|
Pu H, Horbinski C, Hensley PJ, Matuszak EA, Atkinson T, Kyprianou N. PARP-1 regulates epithelial-mesenchymal transition (EMT) in prostate tumorigenesis. Carcinogenesis 2014; 35:2592-601. [PMID: 25173886 DOI: 10.1093/carcin/bgu183] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) is involved in key cellular processes such as DNA replication and repair, gene transcription, cell proliferation and apoptosis. The role of PARP-1 in prostate cancer development and progression is not fully understood. The present study investigated the function of PARP-1 in prostate growth and tumorigenesis in vivo. Functional inactivation of PARP-1 by gene-targeted deletion led to a significant reduction in the prostate gland size in young PARP-1-/- mice (6 weeks) compared with wild-type (WT) littermates. To determine the effect of PARP-1 functional loss on prostate cancer onset, PARP-1-/- mice were crossed with the transgenic adenocarcinoma of the mouse prostate (TRAMP) mice. Pathological assessment of prostate tumors revealed that TRAMP+/-, PARP-1-/- mice exhibited higher grade prostate tumors compared with TRAMP+/- PARP-1+/+ (16-28 weeks) that was associated with a significantly increased proliferative index and decreased apoptosis among the epithelial cells in TRAMP+/- PARP-1-/- prostate tumors. Furthermore tumors harboring PARP-1 loss, exhibited a downregulation of nuclear androgen receptor. Impairing PARP-1 led to increased levels of transforming growth factor-β (TGF-β) and Smads that correlated with induction of epithelial-mesenchymal transition (EMT), as established by loss of E-cadherin and β-catenin and upregulation of N-cadherin and ZEB-1. Our findings suggest that impaired PARP-1 function promotes prostate tumorigenesis in vivo via TGF-β-induced EMT. Defining the EMT control by PARP-1 during prostate cancer progression is of translational significance for optimizing PARP-1 therapeutic targeting and predicting response in metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
| | | | | | - Emily A Matuszak
- Department of Toxicology, University of Kentucky College of Medicine, Lexington, KY 40535, USA
| | | | - Natasha Kyprianou
- Departments of Urology, Pathology, Biochemistry, The Markey Cancer Center and Department of Toxicology, University of Kentucky College of Medicine, Lexington, KY 40535, USA
| |
Collapse
|
44
|
Su JM, Thompson P, Adesina A, Li XN, Kilburn L, Onar-Thomas A, Kocak M, Chyla B, McKeegan E, Warren KE, Goldman S, Pollack IF, Fouladi M, Chen A, Giranda V, Boyett J, Kun L, Blaney SM. A phase I trial of veliparib (ABT-888) and temozolomide in children with recurrent CNS tumors: a pediatric brain tumor consortium report. Neuro Oncol 2014; 16:1661-8. [PMID: 24908656 DOI: 10.1093/neuonc/nou103] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND A phase I trial of veliparib (ABT-888), an oral poly(ADP-ribose) polymerase (PARP) inhibitor, and temozolomide (TMZ) was conducted in children with recurrent brain tumors to (i) estimate the maximum tolerated doses (MTDs) or recommended phase II doses (RP2Ds) of veliparib and TMZ; (ii) describe the toxicities of this regimen; and (iii) evaluate the plasma pharmacokinetic parameters and extent of PARP inhibition in peripheral blood mononuclear cells (PBMCs) following veliparib. METHODS TMZ was given once daily and veliparib twice daily for 5 days every 28 days. Veliparib concentrations and poly(ADP-ribose) (PAR) levels in PBMCs were measured on days 1 and 4. Analysis of pharmacokinetic and PBMC PAR levels were performed twice during study conduct to rationally guide dose modifications and to determine biologically optimal MTD/RP2D. RESULTS Twenty-nine evaluable patients were enrolled. Myelosuppression (grade 4 neutropenia and thrombocytopenia) were dose limiting. The RP2Ds are veliparib 25 mg/m(2) b.i.d. and TMZ 135 mg/m(2)/d. Only 2 out of 12 patients treated at RP2Ds experienced dose-limiting toxicities. Although no objective response was observed, 4 patients had stable disease >6 months in duration, including 1 with glioblastoma multiforme and 1 with ependymoma. At the RP2D of veliparib, pediatric pharmacokinetic parameters were similar to those in adults. CONCLUSIONS Veliparib and TMZ at the RP2D were well tolerated in children with recurrent brain tumors. A phase I/II trial to evaluate the tolerability and efficacy of veliparib, TMZ, and radiation in children with newly diagnosed brainstem gliomas is in progress.
Collapse
Affiliation(s)
- Jack M Su
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Patrick Thompson
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Adekunle Adesina
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Xiao-Nan Li
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Lindsay Kilburn
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Arzu Onar-Thomas
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Mehmet Kocak
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Brenda Chyla
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Evelyn McKeegan
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Katherine E Warren
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Stewart Goldman
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Ian F Pollack
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Maryam Fouladi
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Alice Chen
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Vincent Giranda
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - James Boyett
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Larry Kun
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| | - Susan M Blaney
- Texas Children's Cancer Center, Baylor College of Medicine (J.M.S., P.T., A.A., X-N.L., S.M.B.); Children's National Medical Center (L.K.); St. Jude Children's Research Hospital (A.O-T., J.B., L.K.); University of Tennessee Health Science Center (M.K.); AbbVie Pharmaceuticals (B.C., E.M., V.G.); National Cancer Institute, Pediatric Oncology Branch (K.E.W.); Children's Hospital of Chicago (S.G.); Children's Hospital of Pittsburgh (I.F.P.); Cincinnati Children's Hospital Medical Center (M.F.); Cancer Therapy Evaluation Program, National Cancer Institute (A.C.)
| |
Collapse
|
45
|
Targeting DNA repair with combination veliparib (ABT-888) and temozolomide in patients with metastatic castration-resistant prostate cancer. Invest New Drugs 2014; 32:904-12. [PMID: 24764124 DOI: 10.1007/s10637-014-0099-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 03/30/2014] [Indexed: 12/22/2022]
Abstract
Androgen receptor-mediated transcription is directly coupled with the induction of DNA damage, and castration-resistant tumor cells exhibit increased activity of poly (ADP-ribose) polymerase (PARP)-1, a DNA repair enzyme. This study assessed the efficacy and safety of low dose oral PARP inhibitor veliparib (ABT-888) and temozolomide (TMZ) in docetaxel-pretreated patients with metastatic castration-resistant prostate cancer (mCRPC) in a single-arm, open-label, pilot study. Patients with mCRPC progressing on at least one docetaxel-based therapy and prostate specific antigen (PSA) ≥ 2 ng/mL were treated with veliparib 40 mg twice daily on days 1-7 and TMZ once daily (150 mg/m(2)/day cycle 1; if well tolerated then 200 mg/m(2)/day cycle 2 onwards) on days 1-5 q28 days. Patients received 2 (median) treatment cycles (range, 1-9). The primary endpoint was confirmed PSA response rate (decline ≥ 30 %). Twenty-six eligible patients were enrolled, 25 evaluable for PSA response. Median baseline PSA was 170 ng/mL. Two patients had a confirmed PSA response (8.0 %; 95 % CI: 1.0-26.0), 13 stable PSA, and 10 PSA progression. The median progression-free survival was 9 weeks (95 % CI: 7.9-17) and median overall survival 39.6 weeks (95 % CI: 26.6-not estimable). The most frequent treatment-emergent adverse events (AEs) were thrombocytopenia (77 %), anemia (69 %), fatigue (50 %), neutropenia (42 %), nausea (38 %), and constipation (23 %). Grade 3/4 AEs occurring in > 10 % of patients were thrombocytopenia (23 %) and anemia (15 %). Veliparib and TMZ combination was well tolerated but with modest activity. Biomarker analysis supported the proof of concept that this combination has some antitumor activity in mCRPC.
Collapse
|
46
|
Azad A, Bukczynska P, Jackson S, Haupt Y, Haput Y, Cullinane C, McArthur GA, Solomon B. Co-targeting deoxyribonucleic acid-dependent protein kinase and poly(adenosine diphosphate-ribose) polymerase-1 promotes accelerated senescence of irradiated cancer cells. Int J Radiat Oncol Biol Phys 2014; 88:385-94. [PMID: 24411611 DOI: 10.1016/j.ijrobp.2013.10.043] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 10/29/2013] [Accepted: 10/31/2013] [Indexed: 01/11/2023]
Abstract
PURPOSE To examine the effects of combined blockade of DNA-dependent protein kinase (DNA-PK) and poly(adenosine diphosphate-ribose) polymerase-1 (PARP-1) on accelerated senescence in irradiated H460 and A549 non-small cell lung cancer cells. METHODS AND MATERIALS The effects of KU5788 and AG014699 (inhibitors of DNA-PK and PARP-1, respectively) on clonogenic survival, DNA double-strand breaks (DSBs), apoptosis, mitotic catastrophe, and accelerated senescence in irradiated cells were examined in vitro. For in vivo experiments, H460 xenografts established in athymic nude mice were treated with BEZ235 (a DNA-PK, ATM, and phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor) and AG014699 to determine effects on proliferation, DNA DSBs, and accelerated senescence after radiation. RESULTS Compared with either inhibitor alone, combination treatment with KU57788 and AG014699 reduced postradiation clonogenic survival and significantly increased persistence of Gamma-H2AX (γH2AX) foci in irradiated H460 and A549 cells. Notably, these effects coincided with the induction of accelerated senescence in irradiated cells as reflected by positive β-galactosidase staining, G2-M cell-cycle arrest, enlarged and flattened cellular morphology, increased p21 expression, and senescence-associated cytokine secretion. In irradiated H460 xenografts, concurrent therapy with BEZ235 and AG014699 resulted in sustained Gamma-H2AX (γH2AX) staining and prominent β-galactosidase activity. CONCLUSION Combined DNA-PK and PARP-1 blockade increased tumor cell radiosensitivity and enhanced the prosenescent properties of ionizing radiation in vitro and in vivo. These data provide a rationale for further preclinical and clinical testing of this therapeutic combination.
Collapse
Affiliation(s)
- Arun Azad
- Division of Cancer Research, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Pathology, St. Vincent's Hospital, University of Melbourne, Parkville, Victoria, Australia.
| | - Patricia Bukczynska
- Division of Cancer Research, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Susan Jackson
- Division of Cancer Research, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Ygal Haupt
- Division of Cancer Research, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Ygal Haput
- Division of Cancer Research, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Carleen Cullinane
- Division of Cancer Research, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Grant A McArthur
- Division of Cancer Research, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Division of Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Parkville, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Benjamin Solomon
- Division of Cancer Research, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Division of Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Parkville, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
47
|
Af Hällström TM, Zhao H, Tian J, Rantanen V, Reese SW, Nolley R, Laiho M, Peehl DM. A tissue graft model of DNA damage response in the normal and malignant human prostate. J Urol 2014; 191:842-9. [PMID: 24035881 PMCID: PMC4009951 DOI: 10.1016/j.juro.2013.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2013] [Indexed: 12/26/2022]
Abstract
PURPOSE DNA damage responses are relevant to prostate cancer initiation, progression and treatment. Few models of the normal and malignant human prostate that maintain stromal-epithelial interactions in vivo exist in which to study DNA damage responses. We evaluated the feasibility of maintaining tissue slice grafts at subcutaneous vs subrenal capsular sites in RAG2(-/-)γC(-/-) mice to study the DNA damage responses of normal and malignant glands. MATERIALS AND METHODS We compared the take rate and histology of tissue slice grafts from fresh, precision cut surgical specimens that were maintained for 1 to 4 weeks in subcutaneous vs subrenal capsular sites. Induction of γH2AX, p53, ATM and apoptosis was evaluated as a measure of the DNA damage response after irradiation. RESULTS The take rate of subcutaneous tissue slice grafts was higher than typically reported but lower than at the subrenal capsular site. Subcutaneous tissue slice grafts frequently showed basal cell hyperplasia, squamous metaplasia and cystic atrophy, and cancer did not survive. In contrast, normal and malignant histology was well maintained in subrenal capsular tissue slice grafts. Regardless of implantation site the induction of γH2AX and ATM occurred in tissue slice graft epithelium 1 hour after irradiation and decreased to basal level by 24 hours, indicating DNA damage recognition and repair. As observed previously in prostatic ex vivo models, p53 was not activated. Notably, tumor but not normal cells responded to irradiation by undergoing apoptosis. CONCLUSIONS To our knowledge this is the first study of DNA damage responses in a patient derived prostate tissue graft model. The subrenal capsular site of RAG2(-/-)γC(-/-) mice optimally maintains normal and malignant histology and function, permitting novel studies of DNA damage responses in a physiological context.
Collapse
Affiliation(s)
- Taija M Af Hällström
- Department of Urology, Stanford University School of Medicine, Stanford, California; Molecular Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Finland
| | - Hongjuan Zhao
- Department of Urology, Stanford University School of Medicine, Stanford, California
| | - Junqiang Tian
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Ville Rantanen
- Department of Virology, Haartman Institute and Molecular Imaging Unit and Computational Systems Biology Laboratory, Institute of Biomedicine and Genome-Scale Biology Program, University of Helsinki, Finland
| | - Stephen W Reese
- Department of Urology, Stanford University School of Medicine, Stanford, California
| | - Rosalie Nolley
- Department of Urology, Stanford University School of Medicine, Stanford, California
| | - Marikki Laiho
- Molecular Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Finland; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Donna M Peehl
- Department of Urology, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
48
|
Curtin NJ. Inhibiting the DNA damage response as a therapeutic manoeuvre in cancer. Br J Pharmacol 2014; 169:1745-65. [PMID: 23682925 DOI: 10.1111/bph.12244] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 03/20/2013] [Indexed: 01/05/2023] Open
Abstract
UNLABELLED The DNA damage response (DDR), consisting of an orchestrated network of proteins effecting repair and signalling to cell cycle arrest, to allow time to repair, is essential for cell viability and to prevent DNA damage being passed on to daughter cells. The DDR is dysregulated in cancer with some pathways up-regulated and others down-regulated or lost. Up-regulated pathways can confer resistance to anti-cancer DNA damaging agents. Therefore, inhibitors of key components of these pathways have the potential to prevent this therapeutic resistance. Conversely, defects in a particular DDR pathway may lead to dependence on a complementary pathway. Inhibition of this complementary pathway may result in tumour-specific cell killing. Thus, inhibitors of the DDR have the potential to increase the efficacy of DNA damaging chemotherapy and radiotherapy and have single-agent activity against tumours with a specific DDR defect. This review describes the compounds that have been designed to inhibit specific DDR targets and summarizes the pre-clinical and clinical evaluation of these inhibitors of DNA damage signalling and repair. LINKED ARTICLES This article is part of a themed section on Emerging Therapeutic Aspects in Oncology. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.169.issue-8.
Collapse
Affiliation(s)
- N J Curtin
- Northern Institute for Cancer Research, Medical School, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
49
|
|
50
|
Wu W, Zhu H, Liang Y, Kong Z, Duan X, Li S, Zhao Z, Yang D, Zeng G. Expression of PARP-1 and its active polymer PAR in prostate cancer and benign prostatic hyperplasia in Chinese patients. Int Urol Nephrol 2014; 46:1345-9. [PMID: 24436031 DOI: 10.1007/s11255-014-0642-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 01/02/2014] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND AIMS Aberrant expression of PARP-1 has been reported in various human malignancies and was involved in the progression and metastasis of cancers. However, little is known about PARP-1 expression in prostate cancer (PCa). This study aimed to investigate the expression of PARP-1 and its active polymer poly(ADP-ribose) (PAR) in PCa and benign prostatic hyperplasia (BPH) tissues from Chinese patients. METHODS The expression of PARP-1 and PAR in PCa and benign prostate hyperplasia tissues was assessed by immunohistochemistry in 78 PCa patients and 49 BPH patients. The relationship between the expression of PARP-1 or PAR and clinicopathological parameters in PCa patients was also analyzed. RESULTS Both the positive and strong positive expression rates of PARP-1 in PCa tissues were significantly higher than those in BPH tissues. Although spearman correlations analysis showed the over-expression of PARP-1 and PAR in PCa tissues was not correlated with age, serum PSA level and Gleason scores (GS), an increasing trend was observed between over-expression of PARP-1 or PAR and the PSA levels (TPSA >20 vs TPSA ≤20) or GS grade (GS ≥8 vs GS ≤6). CONCLUSION PARP-1 and PAR expression is markedly elevated in PCa than that in BPH tissues, which may implicate that PARP-1 and PAR are involved in the development of PCa, and the possible expansion in the use of poly(ADP-ribose) polymerase inhibitors for targeting therapy of PCa in select patients alone or combined with chemotherapy or radiation.
Collapse
Affiliation(s)
- Wenqi Wu
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Kangda Road 1#, Haizhu District, Guangzhou, 510230, Guangdong, China,
| | | | | | | | | | | | | | | | | |
Collapse
|