1
|
Kim DM, Lee SY, Lim JC, Cho EH, Park UJ. RUNX3 regulates the susceptibility against EGFR-targeted non-small cell lung cancer therapy using 47Sc-conjugated cetuximab. BMC Cancer 2023; 23:652. [PMID: 37438719 DOI: 10.1186/s12885-023-11161-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 07/07/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Radioimmunotherapy with cetuximab and conjugates with various radioisotopes is a feasible treatment option for different tumor models. Scandium-47 (47Sc), one of several β--particle-emitting radioisotopes, displays favorable physical and chemical properties for conjugation to monoclonal antibodies. However, the therapeutic efficacy of 47Sc in preclinical and clinical studies is largely unknown. Given that intrinsic alterations in tumors greatly contribute to resistance to anti-epidermal growth factor receptor (EGFR)-targeted therapy, research on overcoming resistance to radioimmunotherapy using cetuximab is required. METHODS 47Sc was produced by irradiation of a CaCO3 target at the HANARO research reactor in KAERI (Korea Atomic Energy Research Institute) and prepared by chromatographic separation of the irradiated target. Cetuximab was conjugated with 47Sc using the bifunctional chelating agent DTPA. Radiochemical purity was determined using instant thin-layer chromatography. The immunoreactivity of 47Sc-DTPA-cetuximab was evaluated using the Lindmo method and an in vitro cell-binding assay. The inhibitory effects of cetuximab and 47Sc-DTPA-cetuximab were confirmed using cell growth inhibition and BrdU cell proliferation assays. Differences in protein expression levels between cetuximab- and 47Sc-DTPA-cetuximab-treated cells were confirmed using western blotting. Complex formation between RUNX3 and DNA repair components was confirmed using immunoprecipitation and western blotting. RESULTS Cetuximab induces cell cycle arrest and cell death in EGFR-overexpressing NSCLC cells. Radiolabeling of cetuximab with 47Sc led to increased therapeutic efficacy relative to cetuximab alone. Application of 47Sc-DTPA-cetuximab induced DNA damage responses, and activation of RUNX3 significantly enhanced the therapeutic efficacy of 47Sc-DTPA-cetuximab. RUNX3 mediated susceptibility to EGFR-targeted NSCLC therapy using 47Sc-DTPA-cetuximab via interaction with components of the DNA damage and repair machinery. CONCLUSIONS 47Sc-DTPA-cetuximab promoted cell death in EGFR-overexpressing NSCLC cells by targeting EGFR and inducing DNA damage as a result of β irradiation emitted from the conjugated 47Sc. Activation of RUNX3 played a key role in DNA damage and repair processes in response to the ionizing radiation and inhibited cell growth, thus leading to more effective tumor suppression. RUNX3 can potentially moderate susceptibility to 47Sc-conjugated cetuximab by modulating DNA damage and repair process mechanisms.
Collapse
Affiliation(s)
- Da-Mi Kim
- Radioisotope Research Division, Korea Atomic Energy Research Institute, Daejeon, 34057, Republic of Korea.
| | - So-Young Lee
- Radioisotope Research Division, Korea Atomic Energy Research Institute, Daejeon, 34057, Republic of Korea
| | - Jae-Cheong Lim
- Radioisotope Research Division, Korea Atomic Energy Research Institute, Daejeon, 34057, Republic of Korea
| | - Eun-Ha Cho
- Radioisotope Research Division, Korea Atomic Energy Research Institute, Daejeon, 34057, Republic of Korea
| | - Ul-Jae Park
- Radioisotope Research Division, Korea Atomic Energy Research Institute, Daejeon, 34057, Republic of Korea
| |
Collapse
|
2
|
Tesfay L, Paul BT, Hegde P, Brewer M, Habbani S, Jellison E, Moore T, Wu H, Torti SV, Torti FM. Complementary anti-cancer pathways triggered by inhibition of sideroflexin 4 in ovarian cancer. Sci Rep 2022; 12:19936. [PMID: 36402786 PMCID: PMC9675821 DOI: 10.1038/s41598-022-24391-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022] Open
Abstract
DNA damaging agents are a mainstay of standard chemotherapy for ovarian cancer. Unfortunately, resistance to such DNA damaging agents frequently develops, often due to increased activity of DNA repair pathways. Sideroflexin 4 (SFXN4) is a little-studied inner mitochondrial membrane protein. Here we demonstrate that SFXN4 plays a role in synthesis of iron sulfur clusters (Fe-S) in ovarian cancer cells and ovarian cancer tumor-initiating cells, and that knockdown of SFXN4 inhibits Fe-S biogenesis in ovarian cancer cells. We demonstrate that this has two important consequences that may be useful in anti-cancer therapy. First, inhibition of Fe-S biogenesis triggers the accumulation of excess iron, leading to oxidative stress. Second, because enzymes critical to multiple DNA repair pathways require Fe-S clusters for their function, DNA repair enzymes and DNA repair itself are inhibited by reduction of SFXN4. Through this dual mechanism, SFXN4 inhibition heightens ovarian cancer cell sensitivity to DNA-damaging drugs and DNA repair inhibitors used in ovarian cancer therapy, such as cisplatin and PARP inhibitors. Sensitization is achieved even in drug resistant ovarian cancer cells. Further, knockout of SFXN4 decreases DNA repair and profoundly inhibits tumor growth in a mouse model of ovarian cancer metastasis. Collectively, these results suggest that SFXN4 may represent a new target in ovarian cancer therapy.
Collapse
Affiliation(s)
- Lia Tesfay
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Bibbin T Paul
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Poornima Hegde
- Department of Pathology, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Molly Brewer
- Department of Obstetrics and Gynecology, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Samrin Habbani
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, IN, 47907, USA
| | - Evan Jellison
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Timothy Moore
- Statistical Consulting Services, Center for Open Research Resources, University of Connecticut, Storrs, CT, 06269, USA
| | - Hao Wu
- Department of Statistics, University of Connecticut, Storrs, CT, 06269, USA
| | - Suzy V Torti
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA.
| | - Frank M Torti
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| |
Collapse
|
3
|
Malakoti F, Targhazeh N, Abadifard E, Zarezadeh R, Samemaleki S, Asemi Z, Younesi S, Mohammadnejad R, Hadi Hossini S, Karimian A, Alemi F, Yousefi B. DNA repair and damage pathways in mesothelioma development and therapy. Cancer Cell Int 2022; 22:176. [PMID: 35501851 PMCID: PMC9063177 DOI: 10.1186/s12935-022-02597-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/18/2022] [Indexed: 12/30/2022] Open
Abstract
Malignant mesothelioma (MMe) is an aggressive neoplasm that occurs through the transformation of mesothelial cells. Asbestos exposure is the main risk factor for MMe carcinogenesis. Other important etiologies for MMe development include DNA damage, over-activation of survival signaling pathways, and failure of DNA damage response (DDR). In this review article, first, we will describe the most important signaling pathways that contribute to MMe development and their interaction with DDR. Then, the contribution of DDR failure in MMe progression will be discussed. Finally, we will review the latest MMe therapeutic strategies that target the DDR pathway.
Collapse
Affiliation(s)
- Faezeh Malakoti
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Niloufar Targhazeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Erfan Abadifard
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Zarezadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Samemaleki
- Department of Immunology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Simin Younesi
- Schoole of Health and Biomedical Sciences, RMIT University, Melbourne, Vic, Australia
| | - Reza Mohammadnejad
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Hadi Hossini
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ansar Karimian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| | - Forough Alemi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Bahman Yousefi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Differential gene expression in cisplatin-resistant and -sensitive testicular germ cell tumor cell lines. Oncotarget 2020; 11:4735-4753. [PMID: 33473258 PMCID: PMC7771712 DOI: 10.18632/oncotarget.27844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
Testicular germ cell tumors (TGCTs) represent a well curable malignity due to their exceptional response to cisplatin (CDDP). Despite remarkable treatment results, approximately 5% of TGCT patients develop CDDP resistance and die. Exceptional curability makes TGCTs a highly valuable model system for studying the molecular mechanisms of CDDP sensitivity. Our study was aimed at revealing difference in gene expression between the CDDP-resistant and -sensitive TGCT cell lines, and hence at identifying candidate genes that could serve as potential biomarkers of CDDP response. Using gene expression array, we identified 281 genes that are differentially expressed in CDDP-resistant compared to -sensitive TGCT cell lines. The expression of 25 genes with the highest fold change was validated by RT-qPCR. Of them, DNMT3L, GAL, IGFBP2, IGFBP7, L1TD1, NANOG, NTF3, POU5F1, SOX2, WNT6, ZFP42, ID2, PCP4, SLC40A1 and TRIB3, displayed comparable expression change in gene expression array and RT-qPCR, when all CDDP-resistant TGCT cell lines were pairwise combined with all -sensitive ones. Products of the identified genes are pluripotency factors, or are involved in processes, such as cell metabolism, proliferation or migration. We propose that, after clinical validation, these genes could serve as prognostic biomarkers for early detection of CDDP response in TGCT patients.
Collapse
|
5
|
Raturi VP, Wu C, Mohammad S, Hojo H, Bei Y, Nakamura M, Okumura M, Rachi T, Singh R, Gupta R, Parmar D, Hasan F, Gaur J, Kishan D, Kumar S, Badajena A, Katepogu P, Shigematsu N. Could excision repair cross‐complementing group‐1 mRNA expression from peripheral blood lymphocytes predict locoregional failure with cisplatin chemoradiation for locally advanced laryngeal cancer? Asia Pac J Clin Oncol 2020; 16:e19-e26. [DOI: 10.1111/ajco.13239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 08/07/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Vijay Parshuram Raturi
- Department of Radiation OncologyKing George's Medical University Lucknow India
- Division of Radiation Oncology and Particle TherapyNational Cancer Center Hospital East Kashiwa Japan
| | - Chen‐Ta Wu
- Department of Radiation OncologyGraduate School of Medicine, Keio University Tokyo Japan
| | - Suhel Mohammad
- Department of Radiation OncologyKing George's Medical University Lucknow India
| | - Hidehiro Hojo
- Division of Radiation Oncology and Particle TherapyNational Cancer Center Hospital East Kashiwa Japan
| | - Yanping Bei
- Division of Radiation Oncology and Particle TherapyNational Cancer Center Hospital East Kashiwa Japan
| | - Masaki Nakamura
- Division of Radiation Oncology and Particle TherapyNational Cancer Center Hospital East Kashiwa Japan
| | - Masayuki Okumura
- Division of Radiation Oncology and Particle TherapyNational Cancer Center Hospital East Kashiwa Japan
| | - Toshiya Rachi
- Division of Radiation Oncology and Particle TherapyNational Cancer Center Hospital East Kashiwa Japan
| | - Rahul Singh
- Department of Radiation OncologyKing George's Medical University Lucknow India
| | - Rajeev Gupta
- Department of Radiation OncologyKing George's Medical University Lucknow India
| | | | - Feza Hasan
- Indian Institute of Toxicology and Research Lucknow India
| | - Jalaj Gaur
- Department of Radiation OncologyKing George's Medical University Lucknow India
| | - Dewesh Kishan
- Department of Radiation OncologyKing George's Medical University Lucknow India
| | - Saurabh Kumar
- Department of RadiologyKing George's Medical University Lucknow India
| | - Avinash Badajena
- Department of Radiation OncologyKing George's Medical University Lucknow India
| | - Pranay Katepogu
- Department of Radiation OncologyKing George's Medical University Lucknow India
| | | |
Collapse
|
6
|
Klinakis A, Karagiannis D, Rampias T. Targeting DNA repair in cancer: current state and novel approaches. Cell Mol Life Sci 2020; 77:677-703. [PMID: 31612241 PMCID: PMC11105035 DOI: 10.1007/s00018-019-03299-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/06/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022]
Abstract
DNA damage response, DNA repair and genomic instability have been under study for their role in tumor initiation and progression for many years now. More recently, next-generation sequencing on cancer tissue from various patient cohorts have revealed mutations and epigenetic silencing of various genes encoding proteins with roles in these processes. These findings, together with the unequivocal role of DNA repair in therapeutic response, have fueled efforts toward the clinical exploitation of research findings. The successful example of PARP1/2 inhibitors has also supported these efforts and led to numerous preclinical and clinical trials with a large number of small molecules targeting various components involved in DNA repair singularly or in combination with other therapies. In this review, we focus on recent considerations related to DNA damage response and new DNA repair inhibition agents. We then discuss how immunotherapy can collaborate with these new drugs and how epigenetic drugs can rewire the activity of repair pathways and sensitize cancer cells to DNA repair inhibition therapies.
Collapse
Affiliation(s)
- Apostolos Klinakis
- Biomedical Research Foundation of the Academy of Athens, 11527, Athens, Greece.
| | - Dimitris Karagiannis
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, 10032, USA
| | - Theodoros Rampias
- Biomedical Research Foundation of the Academy of Athens, 11527, Athens, Greece.
| |
Collapse
|
7
|
Cierna Z, Miskovska V, Roska J, Jurkovicova D, Pulzova LB, Sestakova Z, Hurbanova L, Machalekova K, Chovanec M, Rejlekova K, Svetlovska D, Kalavska K, Kajo K, Babal P, Mardiak J, Ward TA, Mego M, Chovanec M. Increased levels of XPA might be the basis of cisplatin resistance in germ cell tumours. BMC Cancer 2020; 20:17. [PMID: 31906898 PMCID: PMC6945513 DOI: 10.1186/s12885-019-6496-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 12/23/2019] [Indexed: 11/12/2022] Open
Abstract
Background Germ cell tumours (GCTs) represent a highly curable malignity as they respond well to cisplatin (CDDP)-based chemotherapy. Nevertheless, a small proportion of GCT patients relapse or do not respond to therapy. As this might be caused by an increased capacity to repair CDDP-induced DNA damage, identification of DNA repair biomarkers predicting inadequate or aberrant response to CDDP, and thus poor prognosis for GCT patients, poses a challenge. The objective of this study is to examine the expression levels of the key nucleotide excision repair (NER) factors, XPA, ERCC1 and XPF, in GCT patients and cell lines. Methods Two hundred seven GCT patients’ specimens with sufficient follow-up clinical-pathological data and pairwise combinations of CDDP-resistant and -sensitive GCT cell lines were included. Immunohistochemistry was used to detect the ERCC1, XPF and XPA protein expression levels in GCT patients’ specimen and Western blot and qRT-PCR examined the protein and mRNA expression levels in GCT cell lines. Results GCT patients with low XPA expression had significantly better overall survival than patients with high expression (hazard ratio = 0.38, 95% confidence interval: 0.12–1.23, p = 0.0228). In addition, XPA expression was increased in the non-seminomatous histological subtype, IGCCCG poor prognosis group, increasing S stage, as well as the presence of lung, liver and non-pulmonary visceral metastases. Importantly, a correlation between inadequate or aberrant CDDP response and XPA expression found in GCT patients was also seen in GCT cell lines. Conclusions XPA expression is an additional independent prognostic biomarker for stratifying GCT patients, allowing for improvements in decision-making on treatment for those at high risk of refractoriness or relapse. In addition, it could represent a novel therapeutic target in GCTs.
Collapse
Affiliation(s)
- Zuzana Cierna
- Department of Pathology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Vera Miskovska
- 1st Department of Oncology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,St. Elisabeth Cancer Institute, Bratislava, Slovakia
| | - Jan Roska
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Dana Jurkovicova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lucia Borszekova Pulzova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Zuzana Sestakova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lenka Hurbanova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | - Michal Chovanec
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,National Cancer Institute, Bratislava, Slovakia
| | - Katarina Rejlekova
- St. Elisabeth Cancer Institute, Bratislava, Slovakia.,2nd Department of Oncology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Daniela Svetlovska
- St. Elisabeth Cancer Institute, Bratislava, Slovakia.,Translational Research Unit, Comenius University, Bratislava, Slovakia
| | - Katarina Kalavska
- St. Elisabeth Cancer Institute, Bratislava, Slovakia.,Translational Research Unit, Comenius University, Bratislava, Slovakia.,Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Karol Kajo
- St. Elisabeth Cancer Institute, Bratislava, Slovakia
| | - Pavel Babal
- Department of Pathology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,Faculty Hospital with Policlinics Skalica a.s., Skalica, Slovakia
| | - Jozef Mardiak
- St. Elisabeth Cancer Institute, Bratislava, Slovakia.,2nd Department of Oncology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Thomas A Ward
- Department of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, UK
| | - Michal Mego
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia.,2nd Department of Oncology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.,National Cancer Institute, Bratislava, Slovakia.,Translational Research Unit, Comenius University, Bratislava, Slovakia
| | - Miroslav Chovanec
- Department of Genetics, Cancer Research Institute, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia.
| |
Collapse
|
8
|
Gong Y, Ju H, Ren G, Wu Y. Cisplatin based induction chemotherapy modified by ERCC1 improved the outcome of young adults with locally advanced oral squamous cell carcinoma. J Cancer 2019; 10:2083-2090. [PMID: 31205569 PMCID: PMC6548157 DOI: 10.7150/jca.28959] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 03/07/2019] [Indexed: 12/12/2022] Open
Abstract
Objective: To evaluate the efficacy of induction chemotherapy in young adults with locally advanced oral squamous cell carcinoma (OSCC) and the usefulness of ERCC1 as a prognostic indicator. Methods: A total of 156 young adults with locally advanced OSCC were retrospectively analyzed from May 2007 to May 2017. Cisplatin based induction chemotherapy followed by surgery and upfront surgery were the primary treatment options for locally advanced OSCC. ERCC1 was evaluated by immunohistochemistry. Multivariate analysis was performed to identify significant prognostic factors for the overall survival (OS) in young adults with locally advanced OSCC. Results: Extracapsular spread (ECS) (p<0.0001) and UICC staging (p<0.0001) were critical prognostic factors for OS in young adults with locally advanced OSCC. The 5-year OS was 83.2% in N0 patients received induction chemotherapy and 61.7% in N0 patients received upfront surgery (p<0.05). Patients with a low ERCC1 expression were more likely to benefit from induction chemotherapy, as the 5-year OS was 22.4% in patients with a high ERCC1 expression and 84.7% in patients with a low ERCC1 expression, respectively (p<0.0001). However, induction chemotherapy resulted in a higher 5-year OS (84.7%) than upfront surgery (59.1%) in patients with a low ERCC1 expression (p=0.03). Conclusions: Induction chemotherapy can improve the outcome of N0 patients. However, the ERCC1 expression should be determined in young patients with locally advanced OSCC prior to induction chemotherapy, as it is a useful biomarker for predicting the outcome after induction chemotherapy.
Collapse
Affiliation(s)
- Yin Gong
- Dept. of of stomatology, First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Houyu Ju
- Dept. of Oral & Maxillofacial -head and neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Guoxin Ren
- Dept. of Oral & Maxillofacial -head and neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Yunteng Wu
- Dept. of Oral & Maxillofacial -head and neck Oncology, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China
| |
Collapse
|
9
|
Exosomal miR-196a derived from cancer-associated fibroblasts confers cisplatin resistance in head and neck cancer through targeting CDKN1B and ING5. Genome Biol 2019; 20:12. [PMID: 30642385 PMCID: PMC6332863 DOI: 10.1186/s13059-018-1604-0] [Citation(s) in RCA: 300] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/04/2018] [Indexed: 01/19/2023] Open
Abstract
Background Cisplatin resistance is a major challenge for advanced head and neck cancer (HNC). Understanding the underlying mechanisms and developing effective strategies against cisplatin resistance are highly desired in the clinic. However, how tumor stroma modulates HNC growth and chemoresistance is unclear. Results We show that cancer-associated fibroblasts (CAFs) are intrinsically resistant to cisplatin and have an active role in regulating HNC cell survival and proliferation by delivering functional miR-196a from CAFs to tumor cells via exosomes. Exosomal miR-196a then binds novel targets, CDKN1B and ING5, to endow HNC cells with cisplatin resistance. Exosome or exosomal miR-196a depletion from CAFs functionally restored HNC cisplatin sensitivity. Importantly, we found that miR-196a packaging into CAF-derived exosomes might be mediated by heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1). Moreover, we also found that high levels of plasma exosomal miR-196a are clinically correlated with poor overall survival and chemoresistance. Conclusions The present study finds that CAF-derived exosomal miR-196a confers cisplatin resistance in HNC by targeting CDKN1B and ING5, indicating miR-196a may serve as a promising predictor of and potential therapeutic target for cisplatin resistance in HNC. Electronic supplementary material The online version of this article (10.1186/s13059-018-1604-0) contains supplementary material, which is available to authorized users.
Collapse
|
10
|
Lockhart AC, Bauer TM, Aggarwal C, Lee CB, Harvey RD, Cohen RB, Sedarati F, Nip TK, Faessel H, Dash AB, Dezube BJ, Faller DV, Dowlati A. Phase Ib study of pevonedistat, a NEDD8-activating enzyme inhibitor, in combination with docetaxel, carboplatin and paclitaxel, or gemcitabine, in patients with advanced solid tumors. Invest New Drugs 2018; 37:87-97. [PMID: 29781056 PMCID: PMC6510847 DOI: 10.1007/s10637-018-0610-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/11/2018] [Indexed: 11/29/2022]
Abstract
Purpose This phase Ib study (NCT01862328) evaluated the maximum tolerated dose (MTD), safety, and efficacy of pevonedistat in combination with standard-of-care chemotherapies in patients with solid tumors. Methods Patients received pevonedistat with docetaxel (arm 1, n = 22), carboplatin plus paclitaxel (arm 2, n = 26), or gemcitabine (arm 3, n = 10) in 21-days (arms 1 and 2) or 28-days (arm 3) cycles. A lead-in cohort (arm 2a, n = 6) determined the arm 2 carboplatin dose. Dose escalation proceeded via continual modified reassessment. Results Pevonedistat MTD was 25 mg/m2 (arm 1) or 20 mg/m2 (arm 2); arm 3 was discontinued due to poor tolerability. Fifteen (23%) patients experienced dose-limiting toxicities during cycle 1 (grade ≥3 liver enzyme elevations, febrile neutropenia, and thrombocytopenia), managed with dose holds or reductions. Drug-related adverse events (AEs) occurred in 95% of patients. Most common AEs included fatigue (56%) and nausea (50%). One drug-related death occurred in arm 3 (febrile neutropenia). Pevonedistat exposure increased when co-administered with carboplatin plus paclitaxel; no obvious changes were observed when co-administered with docetaxel or gemcitabine. Among 54 response-evaluable patients, two had complete responses (arm 2) and 10 had partial responses (three in arm 1, one in arm 2a, six in arm 2); overall response rates were 16% (arm 1) and 35% (arm 2). High ERCC1 expression correlated with clinical benefit in arm 2. Conclusion Pevonedistat with docetaxel or with carboplatin plus paclitaxel was tolerable without cumulative toxicity. Sustained clinical responses were observed in pretreated patients receiving pevonedistat with carboplatin and paclitaxel. ClinicalTrials.gov identifier: NCT01862328.
Collapse
Affiliation(s)
- A Craig Lockhart
- Division of Medical Oncology, University of Miami/Sylvester Comprehensive Cancer Center, 1120 NW 14th Street, Suite 650L, Miami, FL, USA.
| | - Todd M Bauer
- Drug Development, Sarah Cannon Research Institute/Tennessee Oncology PLLC., Nashville, TN, USA
| | - Charu Aggarwal
- Division of Hematology-Oncology, Department of Medicine, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carrie B Lee
- Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - R Donald Harvey
- Departments of Hematology and Medical Oncology and Pharmacology, Emory University School of Medicine, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Roger B Cohen
- Department of Medicine, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Farhad Sedarati
- Oncology Clinical Research, Millennium Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Tsz Keung Nip
- Biostatistics, Takeda Development Centre EU, London, UK
| | - Hélène Faessel
- Quantitative Clinical Pharmacology, Millennium Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Ajeeta B Dash
- Translational and Biomarker Research, Millennium Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Bruce J Dezube
- Oncology Clinical Research, Millennium Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Douglas V Faller
- Oncology Clinical Research, Millennium Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Afshin Dowlati
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
11
|
Xu S, Yu Y, Rong J, Hu D, Zhang L, Fu S, Yang H, Fan J, Yang L, Wu J. Expression of BRCA1 and ERCC1 as predictive clinical outcome after radiochemotherapy in patients with locoregionally moderate-advanced nasopharyngeal carcinoma. Oncotarget 2018; 8:31355-31367. [PMID: 28404895 PMCID: PMC5458213 DOI: 10.18632/oncotarget.15565] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 01/26/2017] [Indexed: 01/18/2023] Open
Abstract
In this study, we examined ERCC1 and BRCA1 expression and clinical outcome of 201 phase-III-IV nasopharyngeal carcinoma patients who were treated with cisplatin-based induced chemotherapy and concurrent radiochemotherapy. The chemotherapy response rate of BRCA1– and BRCA1+ patients was 73.6% and 55.8%, respectively. In addition, the chemotherapy response rate of ERCC1– and ERCC1+ patients was 76.9% and 56.6%, respectively. In patients’ tissues, ERCC1 expression associated with BRCA1 expression. The chemotherapy response rate of BRCA1– and ERCC1– patients was (82.1%) and higher than that of other groups (range 52.4-73.1%). The radiochemotherapy response rate of BRCA1– and ERCC1– patients was higher than that BRCA1+ and ERCC1+ patients. BRCA1– and ERCC1– patients showed higher 3-year overall survival, failure-free survival, locoregional failure-free survival and distant failure-free survival compared to BRCA1+ or ERCC1+ patients. Moreover, the 3-year overall survival, failure-free survival and distant failure-free survival of the BRCA1– and ERCC1– group were higher than that of other groups. TNM stage, ERCC1 expression and the correlation between BRCA1 and ERCC1 expression seemed significant overall survival factors. In conclusion, in nasopharyngeal carcinoma patients, ERCC1 and BRCA1 may be a predictor of response to platinum-based chemotherapy and concurrent radiochemotherapy.
Collapse
Affiliation(s)
- Shan Xu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P. R. China
| | - Yanxin Yu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P. R. China
| | - Jinfeng Rong
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P. R. China
| | - Defeng Hu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P. R. China
| | - LiJun Zhang
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P. R. China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P. R. China
| | - Hongru Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P. R. China
| | - Juan Fan
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P. R. China
| | - Linglin Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P. R. China
| | - Jingbo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P. R. China
| |
Collapse
|
12
|
Abstract
The identification of certain genomic alterations (EGFR, ALK, ROS1, BRAF) or immunological markers (PD-L1) in tissues or cells has led to targeted treatment for patients presenting with late stage or metastatic lung cancer. These biomarkers can be detected by immunohistochemistry (IHC) and/or by molecular biology (MB) techniques. These approaches are often complementary but depending on, the quantity and quality of the biological material, the urgency to get the results, the access to technological platforms, the financial resources and the expertise of the team, the choice of the approach can be questioned. The possibility of detecting simultaneously several molecular targets, and of analyzing the degree of tumor mutation burden and of the micro-satellite instability, as well as the recent requirement to quantify the expression of PD-L1 in tumor cells, has led to case by case development of algorithms and international recommendations, which depend on the quality and quantity of biological samples. This review will highlight the different predictive biomarkers detected by IHC for treatment of lung cancer as well as the present advantages and limitations of this approach. A number of perspectives will be considered.
Collapse
|
13
|
Zhang Z, Jiang C, Hu L. Low Expression of Excision Repair Cross-complementation Group-1 Protein Predicts Better Outcome in Patients with Locally Advanced Nasopharyngeal Cancer Treated with Concurrent Chemoradiotherapy. TUMORI JOURNAL 2018. [DOI: 10.1177/1578.17218] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Zhongxin Zhang
- Department of Oncology, Qilu Hospital, Shandong University, Jinan
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| | - Changqing Jiang
- Department of Pathology, Qingdao Municipal Hospital, Qingdao, China
| | - Likuan Hu
- Department of Oncology, Qilu Hospital, Shandong University, Jinan
| |
Collapse
|
14
|
Dual inhibition of ATR and ATM potentiates the activity of trabectedin and lurbinectedin by perturbing the DNA damage response and homologous recombination repair. Oncotarget 2017; 7:25885-901. [PMID: 27029031 PMCID: PMC5041952 DOI: 10.18632/oncotarget.8292] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 03/04/2016] [Indexed: 12/27/2022] Open
Abstract
Trabectedin (Yondelis®, ecteinascidin-743, ET-743) is a marine-derived natural product approved for treatment of advanced soft tissue sarcoma and relapsed platinum-sensitive ovarian cancer. Lurbinectedin is a novel anticancer agent structurally related to trabectedin. Both ecteinascidins generate DNA double-strand breaks that are processed through homologous recombination repair (HRR), thereby rendering HRR-deficient cells particularly sensitive. We here characterize the DNA damage response (DDR) to trabectedin and lurbinectedin in HeLa cells. Our results show that both compounds activate the ATM/Chk2 (ataxia-telangiectasia mutated/checkpoint kinase 2) and ATR/Chk1 (ATM and RAD3-related/checkpoint kinase 1) pathways. Interestingly, pharmacological inhibition of Chk1/2, ATR or ATM is not accompanied by any significant improvement of the cytotoxic activity of the ecteinascidins while dual inhibition of ATM and ATR strongly potentiates it. Accordingly, concomitant inhibition of both ATR and ATM is an absolute requirement to efficiently block the formation of γ-H2AX, MDC1, BRCA1 and Rad51 foci following exposure to the ecteinascidins. These results are not restricted to HeLa cells, but are shared by cisplatin-sensitive and -resistant ovarian carcinoma cells. Together, our data identify ATR and ATM as central coordinators of the DDR to ecteinascidins and provide a mechanistic rationale for combining these compounds with ATR and ATM inhibitors.
Collapse
|
15
|
An HJ, Jo H, Jung CK, Kang JH, Kim MS, Sun DI, Cho KJ, Cho JH, Won HS, Sun DS, Ko YH. Prognostic implication of ERCC1 protein expression in resected oropharynx and oral cavity cancer. Pathol Res Pract 2017. [PMID: 28645807 DOI: 10.1016/j.prp.2017.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Excision repair cross complement group 1 (ERCC1) expression is a predictive biomarker for platinum-containing treatment in squamous cell carcinoma of head and neck (SCCHN). However, the prognostic significance after surgical resection is not well understood. METHODS Oropharynx (n=143) or oral cavity (n=61) SCCHN patients undergoing surgery were included. ERCC1 protein expression and HPV status were assessed by ERCC1 and p16 immunohistochemistry. RESULTS The ERCC1, over-expressed in 66.7% of patients, was associated with oral cavity cancer (P<0.001), well differentiation (P=0.036), and HPV negativity (P<0.001). In TCGA database, ERCC1 mRNA upregulation was enriched in HPV-negative and oral cavity cancers, and associated with HRAS mutation (P<0.001). The prognostic role of ERCC1 was not different according to HPV status. High ERCC1 expression showed a trend toward poor prognosis in patients with an advanced stage (P=0.079) with marginal significance. CONCLUSIONS The ERCC1 expression was not prognostic in surgically resected oropharynx/oral cavity SCCHN, irrespective of HPV status. However, it could provide additional prognostic information for advanced stage patients.
Collapse
Affiliation(s)
- Ho Jung An
- Division of Oncology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Republic of Korea
| | - Heejoon Jo
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Chan Kwon Jung
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin Hyoung Kang
- Division of Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Min Sik Kim
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong-Il Sun
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kwang Jae Cho
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung-Hae Cho
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hye Sung Won
- Division of Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Der Sheng Sun
- Division of Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoon Ho Ko
- Division of Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
16
|
EL Baiomy MA, El Kashef WF. ERCC1 Expression in Metastatic Triple Negative Breast Cancer Patients Treated with Platinum-Based Chemotherapy. Asian Pac J Cancer Prev 2017; 18:507-513. [PMID: 28345838 PMCID: PMC5454751 DOI: 10.22034/apjcp.2017.18.2.507] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Possible targeted therapies for metastatic triple negative breast cancer (TNBC) include cytotoxic
chemotherapy that causes interstrand breaks (platinum-based drugs). The excision repair cross-complementation 1
(ERCC1) enzyme plays an essential role in the nucleotide excision repair pathway, removing platinum-induced DNA
adducts and contributing to cisplatin resistance. Detecting ERCC1 overexpression is important in considering treatment
options for metastatic TNBC, including individualized approaches to therapy, and may facilitate improved responses or
reduction of unnecessary toxicity. We hypothesized that assigning cisplatin based on pretreatment ERCC1 expression
would improve response and survival. This study was conducted to assess the impact of ERCC1 expression on PFS,
OS and response rates in metastatic triple negative breast cancer patients treated with platinum-based chemotherapy.
Methods: From June 2012 to November 2013, 52 metastatic triple negative breast cancer patients were enrolled.
ERCC1 protein expression was detected from pretreatment biopsies by Immunohistochemistry. All patients received
cisplatin plus paclitaxel. The primary end point was the impact of ERCC1 expression on PFS and OS. Results: 34
patients (65.4%) showed positive ERCC1 expression while 18 (34.6%) proved negative. Positive ERCC1 expression
was associated with short PFS (median, 5 months vs. 7 months; P = 0.043), short OS (median, 9 months vs. 11 months;
P = 0.033) and poor response to cisplatin based chemotherapy (P = 0.046). Conclusions: This prospective study further
validated ERCC1 as a reliable biomarker for customized chemotherapy in metastatic triple negative breast cancer
patients. High expression of ERCC1 was thereby fond to be significantly associated with poor outcome in patients
treated with platinum based chemotherapy.
Collapse
|
17
|
Martens-de Kemp SR, Brink A, van der Meulen IH, de Menezes RX, te Beest DE, Leemans CR, van Beusechem VW, Braakhuis BJ, Brakenhoff RH. The FA/BRCA Pathway Identified as the Major Predictor of Cisplatin Response in Head and Neck Cancer by Functional Genomics. Mol Cancer Ther 2016; 16:540-550. [DOI: 10.1158/1535-7163.mct-16-0457] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/18/2016] [Accepted: 12/06/2016] [Indexed: 11/16/2022]
|
18
|
Swick AD, Stein AP, McCulloch TM, Hartig GK, Ong IM, Sampene E, Prabakaran PJ, Liu CZ, Kimple RJ. Defining the boundaries and expanding the utility of head and neck cancer patient derived xenografts. Oral Oncol 2016; 64:65-72. [PMID: 28024726 DOI: 10.1016/j.oraloncology.2016.11.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 10/11/2016] [Accepted: 11/27/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND Patient derived xenografts (PDXs) represent an essential tool in oncologic research, and we sought to further expand our repertoire of head and neck squamous cell carcinoma (HNSCC) while determining potential boundaries for this system. METHODS We consented new patients for PDX development and determined if a 24-h time delay from tumor excision to xenograft implantation affected PDX establishment. We developed a tissue microarray (TMA) from formalin fixed, paraffin embedded PDXs and their subsequent passages and carried out quantitative immunohistochemistry for EGFR, pEGFR, pAkt, pERK and ERCC1. First and last passaged PDXs were compared via a paired t-test to examine for the stability of protein expression across passages. We performed a similar comparison of the mutational profile of the patient tumor and resulting xenografts using a targeted sequencing approach. RESULTS No patient/tumor characteristics influenced PDX take rate and the 24-h time delay from tumor excision to xenograft implantation did not affect PDX establishment, growth or histology. There was no significant difference in biomarker expression between the first and last passaged PDXs for EGFR, pEGFR, pAkt, and ERCC1. For pERK there was a significant difference (p=0.002), but further analysis demonstrated this only arose in three of 15 PDXs. Targeted sequencing revealed striking stability of passenger and likely driver mutations from patient to xenograft. CONCLUSIONS The stability of protein expression across PDX passages will hopefully allow greater investigation of predictive biomarkers in order to identify ones for further pre-clinical and clinical investigation.
Collapse
Affiliation(s)
- Adam D Swick
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Andrew P Stein
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Timothy M McCulloch
- Department of Otolaryngology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Gregory K Hartig
- Department of Otolaryngology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Irene M Ong
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Emmanuel Sampene
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Prashanth J Prabakaran
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Cheng Z Liu
- Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA.
| |
Collapse
|
19
|
Bahamon BN, Gao F, Danaee H. Development and Validation of an ERCC1 Immunohistochemistry Assay for Solid Tumors. Arch Pathol Lab Med 2016; 140:1397-1403. [PMID: 27610644 DOI: 10.5858/arpa.2016-0006-oa] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT - Excision repair cross-complementation 1 (ERCC1) is a key enzyme in nuclear excision repair pathway and has a critical role in helping remove DNA adducts caused by cross-linking agents, such as platinum-containing cancer chemotherapies and other DNA-damaging therapeutic modalities. ERCC1 expression, evaluated by techniques such as immunohistochemistry, has been associated with clinical response; ERCC1+ tumors are more resistant to cisplatin treatment than are ERCC1- tumors. Although several immunohistochemistry, anti-ERCC1 antibodies are available, the 8F1 clone, in particular, has been used in many studies. Recent evidence has suggested that the 8F1 antibody cross-reacts with at least one other protein, raising concerns about the specificity of this clone. OBJECTIVE - To design an immunohistochemistry assay to detect ERCC1 levels that show dynamic range and consistent analytic performance. DESIGN - Two different primary antibodies to ERCC1, clones 4F9 and D6G6, were evaluated on formalin-fixed, paraffin-embedded tissue. We then performed a fit-for-purpose assay validation with the 4F9 clone, which included sensitivity assessment across several solid tumor types and evaluation of analytic parameters, such as precision and reproducibility. RESULTS - The 4F9 clone was consistently superior to the D6G6 clone in the optimization phase. A range of expression was seen in ovarian, head and neck, non-small cell lung, and esophageal cancer samples when tested with the 4F9 clone. The antibody showed acceptable reproducibility (31.02%) and precision (16.06%). CONCLUSIONS - This assay can be used to assess ERCC1 levels during clinical studies of patient tumors from a variety of tumor types.
Collapse
Affiliation(s)
- Brittany N Bahamon
- From the Departments of Translational Medicine (Ms Bahamon and Dr Danaee) and Biostatistics (Dr Gao), Takeda Pharmaceuticals Inc, Cambridge, Massachusetts. Dr Gao is now with the Department of Biostatistics, Biogen, Cambridge
| | | | | |
Collapse
|
20
|
Unique features of trabectedin mechanism of action. Cancer Chemother Pharmacol 2015; 77:663-71. [DOI: 10.1007/s00280-015-2918-1] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 11/13/2015] [Indexed: 12/12/2022]
|
21
|
Bišof V, Zajc Petranović M, Rakušić Z, Samardžić KR, Juretić A. The prognostic and predictive value of excision repair cross-complementation group 1 (ERCC1) protein in 1288 patients with head and neck squamous cell carcinoma treated with platinum-based therapy: a meta-analysis. Eur Arch Otorhinolaryngol 2015; 273:2305-17. [PMID: 26179868 DOI: 10.1007/s00405-015-3710-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/30/2015] [Indexed: 02/02/2023]
Abstract
Excision repair cross-complementation group 1 (ERCC1) protein has been extensively investigated as a prognostic and predictive factor for platinum-based treatment in head and neck squamous cell carcinoma (HNSCC) but with inconsistent results. We performed the present meta-analysis to better elucidate this issue in advanced HNSCC. A literature search was conducted using the PubMed and Web of Science databases. The inclusion criteria were head and neck cancer patients with platinum-based treatment and evaluation of the correlation between ERCC1 expression and clinical outcomes [objective response rate (ORR), progression-free survival (PFS), and overall survival (OS), both unadjusted and adjusted estimates]. In high vs. low pooled analyses, high ERCC1 expression was associated with unfavorable OS [hazard ratio (HR) = 1.95, 95 % confidence interval (CI) 1.18-3.21, p = 0.009], PFS (HR = 2.39, 95 % CI 1.74-3.28, p = 0.000) and ORR (odds ratio = 0.48, 95 % CI 0.23-0.98, p = 0.044). In the subgroup analysis of adjusted OS estimates, ERCC1 was a predictor of shorter survival in Asians (HR = 3.13, 95 % CI 2.09-4.70, p = 0.000) and Caucasians (HR = 2.02, 95 % CI 1.32-3.07, p = 0.001) but of longer survival in South Americans (HR = 0.17, 95 % CI 0.07-0.40, p = 0.000). Immunohistochemistry proved to be of predictive value irrespective of used antibody (p = 0.009). In the stratified analysis according to the tumor site, ERCC1 expression was associated with OS in nasopharyngeal cancer (HR = 2.72, 95 % CI 1.79-4.13, p = 0.000). ERCC1 has a potential to become predictive and prognostic factor enabling treatment tailoring in HNSCC patients.
Collapse
Affiliation(s)
- Vesna Bišof
- Department of Oncology, University Hospital Centre Zagreb, Kišpatićeva 12, Zagreb, Croatia. .,School of Medicine, University of Osijek, Osijek, Croatia.
| | | | - Zoran Rakušić
- Department of Oncology, University Hospital Centre Zagreb, Kišpatićeva 12, Zagreb, Croatia
| | | | - Antonio Juretić
- Department of Oncology, University Hospital Centre Zagreb, Kišpatićeva 12, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
22
|
Ciaparrone M, Caspiani O, Bicciolo G, Signorelli D, Simonelli I, de Campora L, Mazzarella G, Mecozzi A, Pianelli C, Camaioni A, Catalano P, Pasqualetti P, Fabiano A, Radici M, Marmiroli L, Corsi DC. Predictive Role of ERCC1 Expression in Head and Neck Squamous Cell Carcinoma Patients Treated with Surgery and Adjuvant Cisplatin-Based Chemoradiation. Oncology 2015; 89:227-34. [DOI: 10.1159/000430447] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/13/2015] [Indexed: 11/19/2022]
|
23
|
Xuelei M, Jingwen H, Wei D, Hongyu Z, Jing Z, Changle S, Lei L. ERCC1 plays an important role in predicting survival outcomes and treatment response for patients with HNSCC: A meta-analysis. Oral Oncol 2015; 51:483-92. [DOI: 10.1016/j.oraloncology.2015.02.094] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 02/03/2015] [Accepted: 02/16/2015] [Indexed: 10/23/2022]
|
24
|
Gao Y, Liu D. The roles of excision repair cross-complementation group1 in objective response after cisplatin-based concurrent chemoradiotherapy and survival in head and neck cancers: a systematic review and meta-analysis. Oral Oncol 2015; 51:570-7. [PMID: 25857670 DOI: 10.1016/j.oraloncology.2015.03.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/14/2015] [Accepted: 03/20/2015] [Indexed: 12/26/2022]
Abstract
Our aim of the study was to investigate the precise relationship of repair cross-complementation group 1 (ERCC1) expression and the survival as well as objective response rate to cisplatin-based concurrent chemoradiotherapy (CCRT) and a meta-analysis was conducted to analysis ERCC1's prognostic roles in head and neck cancer. A search based on published articles in PubMed, Embase and CKNI database (up to Oct 15, 2014) to find eligible studies meeting eligibility criteria and then a meta-analysis was conducted to assess the outcomes in head and neck squamous cell carcinomas (HNSCC) patients with different ERCC1 expression. The principle outcomes were hazard ratio (HR) for survival analysis and relative risks (RR) for objective response. Fixed or random model was used for calculation according to the heterogeneity. The results showed that 9 studies involving 568 patients met the inclusion criteria. Low/negative expression of ERCC1 was associated with longer overall survival (OS) and profession-free survival (PFS) after receiving cisplatin-based CCRT therapy (HR 0.38; 95% confidence interval (CI) 0.21-0.63; P<0.001 and HR 0.37; 95%CI 0.21-0.63; P<0.001). And there was no significant difference discovered in objective response rate between low/negative and high/positive ERCC1 expression (RR 1.19; 95%CI 1.00-1.43; P=0.06). Evidence of modest heterogeneity was found between ERCC1 expression and OS (I(2)=48.8%, P<0.05) and subgroup analysis was performed based on ethnicity, variable methods and primary tumor location. The conclusion is that ERCC1 might be the one of adverse prognostic factors affecting the survival time and objective response to cisplatin-based chemoradiotherap due to its drug-resistance characteristics.
Collapse
Affiliation(s)
- Yang Gao
- Human Biology, University of Toronto, 300 Huron Street, Room 105, Toronto, Ontario M5S3J6, Canada
| | - Dong Liu
- Department of Otorhinolaryngology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Jinan 250012, China; Key Laboratory of Otolaryngology, Ministry of Health, 44 West Wenhua Road, Jinan 250012, China.
| |
Collapse
|
25
|
Stein AP, Swick AD, Smith MA, Blitzer GC, Yang RZ, Saha S, Harari PM, Lambert PF, Liu CZ, Kimple RJ. Xenograft assessment of predictive biomarkers for standard head and neck cancer therapies. Cancer Med 2015; 4:699-712. [PMID: 25619980 PMCID: PMC4430263 DOI: 10.1002/cam4.387] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 11/16/2014] [Accepted: 11/18/2014] [Indexed: 12/27/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains a challenging cancer to treat with overall 5-year survival on the order of 50–60%. Therefore, predictive biomarkers for this disease would be valuable to provide more effective and individualized therapeutic approaches for these patients. While prognostic biomarkers such as p16 expression correlate with outcome; to date, no predictive biomarkers have been clinically validated for HNSCC. We generated xenografts in immunocompromised mice from six established HNSCC cell lines and evaluated response to cisplatin, cetuximab, and radiation. Tissue microarrays were constructed from pre- and posttreatment tumor samples derived from each xenograft experiment. Quantitative immunohistochemistry was performed using a semiautomated imaging and analysis platform to determine the relative expression of five potential predictive biomarkers: epidermal growth factor receptor (EGFR), phospho-EGFR, phospho-Akt, phospho-ERK, and excision repair cross-complementation group 1 (ERCC1). Biomarker levels were compared between xenografts that were sensitive versus resistant to a specific therapy utilizing a two-sample t-test with equal standard deviations. Indeed the xenografts displayed heterogeneous responses to each treatment, and we linked a number of baseline biomarker levels to response. This included low ERCC1 being associated with cisplatin sensitivity, low phospho-Akt correlated with cetuximab sensitivity, and high total EGFR was related to radiation resistance. Overall, we developed a systematic approach to identifying predictive biomarkers and demonstrated several connections between biomarker levels and treatment response. Despite these promising initial results, this work requires additional preclinical validation, likely involving the use of patient-derived xenografts, prior to moving into the clinical realm for confirmation among patients with HNSCC.
Collapse
Affiliation(s)
- Andrew P Stein
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Adam D Swick
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Molly A Smith
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Grace C Blitzer
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Robert Z Yang
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Sandeep Saha
- Department of Biostatistics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Paul M Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Paul F Lambert
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Cheng Z Liu
- Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706
| |
Collapse
|
26
|
Quantitative methodology is critical for assessing DNA methylation and impacts on correlation with patient outcome. Clin Epigenetics 2014; 6:22. [PMID: 25859283 PMCID: PMC4391486 DOI: 10.1186/1868-7083-6-22] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 10/17/2014] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND DNA hypermethylation is reported as a frequent event and prognostic marker in head and neck squamous cell carcinomas (HNSCC). Methylation has been commonly assessed with non-quantitative methodologies, such as methylation-specific PCR (MSP). We investigated previously reported hypermethylated genes with quantitative methodology in oral tongue squamous cell carcinomas (OTSCC). RESULTS The methylation status of 12 genes in 115 OTSCC samples was assessed by one or more of three quantitative analyses: methylation sensitive high resolution melting (MS-HRM), sensitive-melting analysis after real time-methylation specific PCR (SMART-MSP), and bisulfite pyrosequencing. In contrast to much of the literature, either no or infrequent locus-specific methylation was identified by MS-HRM for DAPK1, RASSF1A, MGMT, MLH1, APC, CDH1, CDH13, BRCA1, ERCC1, and ATM. The most frequently methylated loci were RUNX3 (18/108 methylated) and ABO (22/107 methylated). Interrogation of the Cancer Genome Atlas (TCGA) HNSCC cohort confirmed the frequency of significant methylation for the loci investigated. Heterogeneous methylation of RUNX3 (18/108) and ABO (22/107) detected by MS-HRM, conferred significantly worse survival (P = 0.01, and P = 0.03). However, following quantification of methylation levels using pyrosequencing, only four tumors had significant quantities (>15%) of RUNX3 methylation which correlated with a worse patient outcome (P <0.001), while the prognostic significance of ABO hypermethylation was lost. RUNX3 methylation was not prognostic for the TCGA cohort (P = 0.76). CONCLUSIONS We demonstrated the critical need for quantification of methylation levels and its impact on correlative analyses. In OTSCC, we found little evidence of significant or frequent hypermethylation of many loci reported to be commonly methylated. It is likely that previous reports have overestimated the frequency of significant methylation events as a consequence of the use of non-quantitative methodology.
Collapse
|
27
|
Bharthuar A, Saif Ur Rehman S, Black JD, Levea C, Malhotra U, Mashtare TL, Iyer R. Breast cancer resistance protein (BCRP) and excision repair cross complement-1 (ERCC1) expression in esophageal cancers and response to cisplatin and irinotecan based chemotherapy. J Gastrointest Oncol 2014; 5:253-8. [PMID: 25083297 DOI: 10.3978/j.issn.2078-6891.2014.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 04/02/2014] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Esophageal cancer patients face a dismal outcome despite tri-modality management and median survival remains 15-18 months. Breast cancer resistance protein (BCRP) is an ATP-dependent efflux protein associated with chemotherapy resistance. The role of BCRP expression in esophageal cancer and normal esophageal cells is not known. Excision repair cross complement-1 (ERCC1) overexpression has been correlated with poorer response to cisplatin based chemotherapy. We examined the expression of BCRP and ERCC1 in patients with esophageal cancer and correlated it with survival in patients receiving irinotecan and cisplatin based chemotherapy. METHODS With IRB approval, 40 cases of esophageal cancer diagnosed from 2004-2008, were stained for BCRP and ERCC1 expression by immunohistochemistry and scored by a pathologist blinded to clinical data. Baseline demographics, therapy given and survival data were collected and correlated with BCRP and ERCC1 expression. Fisher's exact test was used to determine association between BCRP and ERCC1 expression and demographics. Cox proportional hazards model was used for association of BCRP and ERCC1 with survival. RESULTS On immunohistochemistry, 30/40 cancers (75%) expressed BCRP. Interestingly, down-regulation of BCRP expression in tumor compared with normal cells was seen in 40% of patients. ERCC1 positivity was seen in 15/30 cases (50%). Median overall survival (OS) was 19 months with no difference in survival between BCRP positive and negative patients (P=0.13) or ERCC1 positive and negative patients (P=0.85). Estimated hazard ratio (HR) of death for BRCP positive patients was 2.29 (95% CI: 0.79-6.64) and for ERCC1 positive patients was 1.09 (95% CI: 0.46-2.56). There was no association of BCRP and ERCC1 expression with disease stage, age, gender or histology. For patients who received cisplatin and irinotecan as first line chemotherapy, there was no difference in survival based on BCRP or ERCC1 status. CONCLUSIONS BCRP expression is seen in a majority of esophageal cancers and normal esophageal mucosa. ERCC1 expression is seen in about half of the patients with esophageal cancer. Irinotecan based studies with esophageal and gastric cancer suggest response rates of 14-65%. Whether the 40% of tumors in our study found with down regulation of BCRP expression, constitute a majority of these responders needs to be prospectively validated in a larger data set. It should include markers such as ERCC1 predicting response to 5-fluorouracil and platinum based chemotherapy, to enable individualizing therapy for this cancer.
Collapse
Affiliation(s)
- Anubha Bharthuar
- 1 Department of Medicine, Roswell Park Cancer Institute and University at Buffalo New York, USA ; 2 Department of Pathology, Roswell Park Cancer Institute, Buffalo New York, USA ; 3 Department of Biostatistics, Roswell Park Cancer Institute and University at Buffalo New York, USA
| | - Sana Saif Ur Rehman
- 1 Department of Medicine, Roswell Park Cancer Institute and University at Buffalo New York, USA ; 2 Department of Pathology, Roswell Park Cancer Institute, Buffalo New York, USA ; 3 Department of Biostatistics, Roswell Park Cancer Institute and University at Buffalo New York, USA
| | - Jennifer D Black
- 1 Department of Medicine, Roswell Park Cancer Institute and University at Buffalo New York, USA ; 2 Department of Pathology, Roswell Park Cancer Institute, Buffalo New York, USA ; 3 Department of Biostatistics, Roswell Park Cancer Institute and University at Buffalo New York, USA
| | - Charles Levea
- 1 Department of Medicine, Roswell Park Cancer Institute and University at Buffalo New York, USA ; 2 Department of Pathology, Roswell Park Cancer Institute, Buffalo New York, USA ; 3 Department of Biostatistics, Roswell Park Cancer Institute and University at Buffalo New York, USA
| | - Usha Malhotra
- 1 Department of Medicine, Roswell Park Cancer Institute and University at Buffalo New York, USA ; 2 Department of Pathology, Roswell Park Cancer Institute, Buffalo New York, USA ; 3 Department of Biostatistics, Roswell Park Cancer Institute and University at Buffalo New York, USA
| | - Terry L Mashtare
- 1 Department of Medicine, Roswell Park Cancer Institute and University at Buffalo New York, USA ; 2 Department of Pathology, Roswell Park Cancer Institute, Buffalo New York, USA ; 3 Department of Biostatistics, Roswell Park Cancer Institute and University at Buffalo New York, USA
| | - Renuka Iyer
- 1 Department of Medicine, Roswell Park Cancer Institute and University at Buffalo New York, USA ; 2 Department of Pathology, Roswell Park Cancer Institute, Buffalo New York, USA ; 3 Department of Biostatistics, Roswell Park Cancer Institute and University at Buffalo New York, USA
| |
Collapse
|
28
|
Galluzzi L, Vitale I, Michels J, Brenner C, Szabadkai G, Harel-Bellan A, Castedo M, Kroemer G. Systems biology of cisplatin resistance: past, present and future. Cell Death Dis 2014; 5:e1257. [PMID: 24874729 PMCID: PMC4047912 DOI: 10.1038/cddis.2013.428] [Citation(s) in RCA: 551] [Impact Index Per Article: 55.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 09/23/2013] [Accepted: 09/26/2013] [Indexed: 12/16/2022]
Abstract
The platinum derivative cis-diamminedichloroplatinum(II), best known as cisplatin, is currently employed for the clinical management of patients affected by testicular, ovarian, head and neck, colorectal, bladder and lung cancers. For a long time, the antineoplastic effects of cisplatin have been fully ascribed to its ability to generate unrepairable DNA lesions, hence inducing either a permanent proliferative arrest known as cellular senescence or the mitochondrial pathway of apoptosis. Accumulating evidence now suggests that the cytostatic and cytotoxic activity of cisplatin involves both a nuclear and a cytoplasmic component. Despite the unresolved issues regarding its mechanism of action, the administration of cisplatin is generally associated with high rates of clinical responses. However, in the vast majority of cases, malignant cells exposed to cisplatin activate a multipronged adaptive response that renders them less susceptible to the antiproliferative and cytotoxic effects of the drug, and eventually resume proliferation. Thus, a large fraction of cisplatin-treated patients is destined to experience therapeutic failure and tumor recurrence. Throughout the last four decades great efforts have been devoted to the characterization of the molecular mechanisms whereby neoplastic cells progressively lose their sensitivity to cisplatin. The advent of high-content and high-throughput screening technologies has accelerated the discovery of cell-intrinsic and cell-extrinsic pathways that may be targeted to prevent or reverse cisplatin resistance in cancer patients. Still, the multifactorial and redundant nature of this phenomenon poses a significant barrier against the identification of effective chemosensitization strategies. Here, we discuss recent systems biology studies aimed at deconvoluting the complex circuitries that underpin cisplatin resistance, and how their findings might drive the development of rational approaches to tackle this clinically relevant problem.
Collapse
Affiliation(s)
- L Galluzzi
- 1] Gustave Roussy, Villejuif, France [2] Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France [3] Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
| | - I Vitale
- 1] Regina Elena National Cancer Institute, Rome, Italy [2] National Institute of Health, Rome, Italy
| | - J Michels
- 1] Gustave Roussy, Villejuif, France [2] Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France [3] INSERM, U848, Villejuif, France
| | - C Brenner
- 1] INSERM, UMRS 769; LabEx LERMIT, Châtenay Malabry, France [2] Faculté de Pharmacie, Université de Paris Sud/Paris XI, Châtenay Malabry, France
| | - G Szabadkai
- 1] Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK [2] Department of Biomedical Sciences, Università Degli Studi di Padova, Padova, Italy
| | - A Harel-Bellan
- 1] Laboratoire Epigenetique et Cancer, Université de Paris Sud/Paris XI, Gif-Sur-Yvette, France [2] CNRS, FRE3377, Gif-Sur-Yvette, France [3] Commissariat à l'Energie Atomique (CEA), Saclay, France
| | - M Castedo
- 1] Gustave Roussy, Villejuif, France [2] Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France [3] INSERM, U848, Villejuif, France
| | - G Kroemer
- 1] Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France [2] Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France [3] INSERM, U848, Villejuif, France [4] Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France [5] Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| |
Collapse
|
29
|
Cell cycle association and hypoxia regulation of excision repair cross complementation group 1 protein (ERCC1) in tumor cells of head and neck cancer. Tumour Biol 2014; 35:7807-19. [PMID: 24817012 PMCID: PMC4158184 DOI: 10.1007/s13277-014-2001-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 04/22/2014] [Indexed: 12/18/2022] Open
Abstract
Excision repair cross complementation group 1 (ERCC1) is a key component of homologous recombination-based repair of interstrand DNA cross-links (ICLs). As a consequence, ERCC1 mediates resistance to mitomycin C (MMC) and platinum chemotherapeutic agents and may predict treatment failure. Clinical response to MMC or cisplatin (CDDP)-based radiochemotherapy (RCT) was assessed in 106 head and neck squamous cell carcinoma (HNSCC) patients and correlated with cell nuclear immunoreactivity of the mouse monoclonal (clone: 8 F1) ERCC1 antibody in tumor tissue samples. BEAS-2B epithelial and Detroit 562 pharyngeal squamous carcinoma cells were treated with CDDP, MMC, and 5-fluorouracil (5-FU) at 50 % growth inhibitory (IC-50) concentrations. ERCC1 protein synthesis was compared with cell cycle distribution using combined immunocytochemistry and flow cytometry. ERCC1 messenger RNA (mRNA) and protein expression was investigated in normoxic and hypoxic conditions in Detroit 562 cells. Clinically, the nonresponder revealed significantly lower HNSCC tissue ERCC1 immunoreactivity than the responder (p = 0.0064) or control normal mucosa, which led to further mechanistic investigations. In vitro, control cells and cells treated with cytotoxic agents showed increasing ERCC1 levels from the G1 through S and G2 phases of the cell cycle. In CDDP-treated cells, ERCC1 mRNA and protein expression increased. Under hypoxic conditions, ERCC1 gene expression significantly decreased. Although ERCC1+ cells show increased chemoresistance, they might be particularly radiosensitive, representing G2 cell cycle phase and less hypoxic. ERCC1 expression might be indirectly related with some conditions important for RCT treatment, but it is not a clear predictor for its failure in HNSCC patients.
Collapse
|
30
|
Joseph B, Vishwanath L, Venugopal BK. Radiosensitization in head and neck cancer: do we have an alternative to platins? Role of taxanes. Oral Surg Oral Med Oral Pathol Oral Radiol 2014; 117:324-8. [DOI: 10.1016/j.oooo.2013.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 10/08/2013] [Accepted: 10/13/2013] [Indexed: 10/26/2022]
|
31
|
Tan XG, Yang ZL, Yang LP, Miao XY. Expression of DNA-repair proteins and their significance in pancreatic cancer and non-cancerous pancreatic tissues of Sprague-Dawley rats. World J Surg Oncol 2014; 12:32. [PMID: 24502441 PMCID: PMC3931407 DOI: 10.1186/1477-7819-12-32] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Accepted: 01/28/2014] [Indexed: 12/30/2022] Open
Abstract
Background To establish a model of pancreatic cancer induced by 7,12-dimethylbenzantracene (DMBA) in Sprague–Dawley (SD) rats, and detect the expression of DNA-repair proteins (MGMT, ERCC1, hMSH2, and hMLH1) and their significance in pancreatic cancer and non-cancerous pancreatic tissues of SD rats. Methods DMBA was directly implanted into the parenchyma of rat pancreas (group A and group B), and group B rats were then treated with trichostatin A (TSA). The rats in both groups were executed within 3 to 5 months, and their pancreatic tissues were observed by macrography and under microscopy. Meanwhile, the rats in the control group (group C) were executed at 5 months. Immunohistochemistry was used to assay the expression of MGMT, ERCC1, hMSH2, and hMLH1. Results The incidence of pancreatic cancer in group A within 3 to 5 months was 48.7% (18/37), including 1 case of fibrosarcoma. The incidence of pancreatic cancer in group B was 33.3% (12/36), including 1 case of fibrosarcoma. The mean of maximal diameters of tumors in group A was higher than that in group B (P <0.05). No pathological changes were found in pancreas of group C and other main organs (except pancreas) of group A and group B. No statistical differences were found among the positive rates of MGMT, ERCC1, hMSH2, and hMLH1 in ductal adenocarcinoma and non-cancerous pancreatic tissues of group A (P >0.05). The positive rates of MGMT, ERCC1, hMSH2, and hMLH1 were significantly lower in ductal adenocarcinoma than those in non-cancerous tissues of group B (P ≤0.05). All pancreas of group C had positive expression of MGMT, ERCC1, hMSH2, and hMLH1 and two cases of fibrosarcoma showed a negative expression. Conclusions DMBA, directly implanted into the parenchyma of pancreas, creates an ideal pancreatic cancer model within a short time. TSA might restrain DNA damage related to the genesis and growth of pancreatic cancer in rats. The DNA-repair proteins, including MGMT, ERCC1, hMSH2, and hMLH1, might play an important role in the genesis of pancreatic cancer induced by DMBA in rats.
Collapse
Affiliation(s)
| | - Zhu-lin Yang
- Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| | | | | |
Collapse
|
32
|
Li FY, Ren XB, Xie XY, Zhang J. Meta-analysis of Excision Repair Cross-complementation Group 1 (ERCC1) Association with Response to Platinum-based Chemotherapy in Ovarian Cancer. Asian Pac J Cancer Prev 2013; 14:7203-6. [DOI: 10.7314/apjcp.2013.14.12.7203] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
33
|
Weiss J, Hayes DN. Classifying squamous cell carcinoma of the head and neck: prognosis, prediction and implications for therapy. Expert Rev Anticancer Ther 2013; 14:229-36. [PMID: 24313652 DOI: 10.1586/14737140.2014.863154] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Traditionally, squamous cell cancers of the head and neck (SCCHN) have been classified by their anatomic location and stage. This system has been unsatisfactory in that it leaves substantial heterogeneity in prognosis and inadequate definition of optimal therapy. The most promising novel marker for superior prognosis in SCCHN is human papillomavirus (HPV). Overexpression of the EGFR bears an adverse prognosis; no marker provides clear predictive power for benefit from EGFR inhibition. Low expression of the DNA repair enzymes and excision repair cross-complementing rodent repair deficiency (ERCC1) and x-ray repair complementing defective repair in Chinese hamster cells 1 (XRCC1) may predict chemotherapy and chemoradiotherapy sensitivity. Tumors expressing cyclin D1 have a poor prognosis. Genomic characterization has subdivided SCCHN into four categories with clear biologic themes. Basal cancers express high levels of TGF-α and have other perturbations of the EGFR axis. Mesenchymal cancers show evidence for epithelial to mesenchymal transition. Atypical cancers lack both EGFR amplification and deletion of 9p; they also have a higher rate of HPV positivity than the other groups. Classical tumors demonstrate gene signatures similar to those previously associated with exposure to cigarette smoke; patients with this signature had a greater smoking history than patients in the other groups.
Collapse
Affiliation(s)
- Jared Weiss
- The Lineberger Comprehensive Cancer Center at the University of North Carolina, Chapel Hill, NC, USA
| | | |
Collapse
|
34
|
Genome stability pathways in head and neck cancers. Int J Genomics 2013; 2013:464720. [PMID: 24364026 PMCID: PMC3834617 DOI: 10.1155/2013/464720] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 09/19/2013] [Accepted: 09/20/2013] [Indexed: 12/12/2022] Open
Abstract
Genomic instability underlies the transformation of host cells toward malignancy, promotes development of invasion and metastasis and shapes the response of established cancer to treatment. In this review, we discuss recent advances in our understanding of genomic stability in squamous cell carcinoma of the head and neck (HNSCC), with an emphasis on DNA repair pathways. HNSCC is characterized by distinct profiles in genome stability between similarly staged cancers that are reflected in risk, treatment response and outcomes. Defective DNA repair generates chromosomal derangement that can cause subsequent alterations in gene expression, and is a hallmark of progression toward carcinoma. Variable functionality of an increasing spectrum of repair gene polymorphisms is associated with increased cancer risk, while aetiological factors such as human papillomavirus, tobacco and alcohol induce significantly different behaviour in induced malignancy, underpinned by differences in genomic stability. Targeted inhibition of signalling receptors has proven to be a clinically-validated therapy, and protein expression of other DNA repair and signalling molecules associated with cancer behaviour could potentially provide a more refined clinical model for prognosis and treatment prediction. Development and expansion of current genomic stability models is furthering our understanding of HNSCC pathophysiology and uncovering new, promising treatment strategies.
Collapse
|
35
|
Vassalou H, Stathopoulos E, Fiolitaki G, Koutsopoulos A, Voutsina A, Georgoulias V, Mavroudis D. Excision-repair-cross-complement-1 protein as a prognostic factor in patients with advanced non-small cell lung cancer treated with platinum-based first-line chemotherapy. Lung Cancer 2013; 82:324-9. [DOI: 10.1016/j.lungcan.2013.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 07/21/2013] [Accepted: 08/01/2013] [Indexed: 12/20/2022]
|
36
|
Takahashi Y, Bell D, Agarwal G, Roberts D, Xie TX, El-Naggar A, Myers JN, Hanna EY. Comprehensive assessment of prognostic markers for sinonasal squamous cell carcinoma. Head Neck 2013; 36:1094-102. [PMID: 23836481 DOI: 10.1002/hed.23423] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 06/11/2013] [Accepted: 06/19/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Little is known about the molecular signature of the rare tumor sinonasal squamous cell carcinoma (SNSCC). The purpose of this study was to comprehensively assess various molecular biomarkers in SNSCC. METHODS We chose 13 markers for this study, which have been known as prognostic markers in head and neck squamous cell carcinoma. Expression of these markers was examined by either in situ hybridization or immunohistochemical methods on tissue microarrays made from 70 SNSCC specimens and 28 matched-pair normal tissues from patients who underwent surgical resection at our institution. Expression data were correlated with patient clinicopathologic parameters and survival. RESULTS Of the 13 markers, only epidermal growth factor receptor (EGFR) protein expression was associated with significantly shorter disease-free survival (DFS; p = .01307). EGFR expression was also associated with shorter overall survival (OS), but the difference was not statistically significant. CONCLUSION Targeted inhibition of tumor EGFR expression may be a new approach to treating SNSCC.
Collapse
Affiliation(s)
- Yoko Takahashi
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | | | | | | |
Collapse
|
37
|
A systematic review of current and emerging approaches in the field of larynx preservation. Radiother Oncol 2013; 110:16-24. [PMID: 24139733 DOI: 10.1016/j.radonc.2013.08.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 08/11/2013] [Accepted: 08/12/2013] [Indexed: 11/20/2022]
Abstract
Treatment options targeting laryngeal preservation include conservative surgery, concurrent chemo-radiotherapy, induction chemotherapy (IC) followed by radiotherapy (RT), and alternating chemo-radiation. The goal of this paper was to perform a systematic review of randomized clinical trials (RCTs) on current and emerging approaches in the field of larynx preservation. The search identified 36 papers of which 27 did not fall within the inclusion criteria (i.e. non-RCTs). IC followed by RT has been shown to allow laryngeal preservation in about two-thirds of pts with locally advanced laryngeal or hypopharyngeal cancer without compromising survival. IC is regarded as the landmark treatment of non-surgical larynx preservation approaches. Concomitant and alternating chemoradiotherapy treatments are also acceptable in larynx preservation.
Collapse
|
38
|
Mehra R, Zhu F, Yang DH, Cai KQ, Weaver J, Singh MK, Nikonova AS, Golemis EA, Flieder DB, Cooper HS, Lango M, Ridge JA, Burtness B. Quantification of excision repair cross-complementing group 1 and survival in p16-negative squamous cell head and neck cancers. Clin Cancer Res 2013; 19:6633-43. [PMID: 24088734 DOI: 10.1158/1078-0432.ccr-13-0152] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE Multimodality treatment of squamous cell carcinoma of the head and neck (SCCHN) often involves radiotherapy and cisplatin-based therapy. Elevated activity of DNA repair mechanisms, such as the nucleotide excision repair (NER) pathway, of which ERCC1 is a rate-limiting element, are associated with cisplatin and possibly RT resistance. We have determined excision repair cross-complementing group 1 (ERCC1) expression in human papillomavirus (HPV)-negative SCCHN treated with surgery [± adjuvant radiotherapy/chemoradiation (CRT)]. EXPERIMENTAL DESIGN We assessed ERCC1 protein expression in archival tumors using immunofluorescence staining and automatic quantitative analysis (AQUA) with three antibodies to ERCC1 (8F1, FL297, and HPA029773). Analysis with Classification and Regression Tree (CART) methods ascertained the cutoff points between high/low ERCC1 expression. Multivariable analysis adjusted for age, T, and N stage. Kaplan-Meier curves determined median survival. ERCC1 expression at initial tumor presentation and in recurrent disease were compared. Performance characteristics of antibodies were assessed. RESULTS ERCC1 low/high groups were defined on the basis of AQUA analysis with 8F1/2009, FL297, and HPA029773. Among patients treated with surgery plus adjuvant radiotherapy/CRT, longer median survival was observed in ERCC1-low versus ERCC1-high tumors (64 vs. 29 months; P = 0.02; HPA029773). Data obtained with HPA029773 indicated no survival difference among patients treated only with surgery. Recurrent cancers had lower ERCC1 AQUA scores than tumors from initial presentation. Extensive characterization indicated optimal specificity and performance by the HPA029773 antibody. CONCLUSIONS Using AQUA, with the specific ERCC1 antibody HPA029773, we found a statistical difference in survival among high/low-ERCC1 tumors from patients treated with surgery and adjuvant radiotherapy.
Collapse
Affiliation(s)
- Ranee Mehra
- Authors' Affiliation: Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
ERCC1 is a prognostic biomarker in locally advanced head and neck cancer: results from a randomised, phase II trial. Br J Cancer 2013; 109:2096-105. [PMID: 24064970 PMCID: PMC3798971 DOI: 10.1038/bjc.2013.576] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 08/20/2013] [Accepted: 08/26/2013] [Indexed: 12/12/2022] Open
Abstract
Background: Cisplatin-radiotherapy is a preferred standard for locally advanced, head and neck squamous cell carcinoma (HNSCC). However, the cisplatin-attributable survival benefit is small and toxicity substantial. A biomarker of cisplatin resistance could guide treatment selection and spare morbidity. The ERCC1-XPF nuclease is critical to DNA repair pathways resolving cisplatin-induced lesions. Methods: In a phase II trial, patients with untreated Stage III-IVb HNSCC were randomised to cisplatin-radiotherapy with/without erlotinib. Archived primary tumours were available from 90 of 204 patients for this planned substudy. Semi-quantitative ERCC1 protein expression (H-score) was determined using the FL297, 4F9, and 8F1 antibodies. The primary analysis evaluated the relationship between continuous ERCC1 protein expression and progression-free survival (PFS). Secondary analyses included two pre-specified ERCC1 cutpoints and performance in HPV-associated disease. Results: Higher ERCC1 expression was associated with inferior PFS, as measured by the specific antibodies FL297 (HR=2.5, 95% CI=1.1–5.9, P=0.03) and 4F9 (HR=3.0, 95% CI=1.2–7.8, P=0.02). Patients with increased vs decreased/normal ERCC1 expression experienced inferior PFS (HR=4.8 for FL297, P=0.003; HR=5.5 for 4F9, P=0.007). This threshold remained prognostic in HPV-associated disease. Conclusion: ERCC1-XPF protein expression by the specific FL297 and 4F9 antibodies is prognostic in patients undergoing definitive cisplatin-radiotherapy for HNSCC, irrespective of HPV status.
Collapse
|
40
|
Patel MR, Zhao N, Ang MK, Stadler ME, Fritchie K, Weissler MC, Zanation AM, Harris SL, Funkhouser WK, Olshan AF, Shores CG, Hayes DN. ERCC1 Protein Expression Is Associated with Differential Survival in Oropharyngeal Head and Neck Squamous Cell Carcinoma. Otolaryngol Head Neck Surg 2013; 149:587-95. [DOI: 10.1177/0194599813496522] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Objective To investigate ERCC1 protein expression and its relationship to clinical factors and treatment outcomes in patients with head and neck squamous cell carcinoma (HNSCC). Design Case series. Setting Tertiary care academic center. Subjects One hundred and seventy-six patients diagnosed with HNSCC and treated with intent to cure between 2002 and 2008 were analyzed with respect to clinical data and tumor pathology. Main Outcome Measures Tissue microarrays were constructed from tumor blocks and immunohistochemical staining for ERCC1 performed. ERCC1 expression status was dichotomized into high and low using the Allred score. Clinical characteristics of patients with high versus low ERCC1 expression were compared. Distributions of overall survival (OS) and progression-free survival (PFS) were analyzed using the Kaplan-Meier method. Results Of 176 patients, ERCC1 showed baseline nuclear staining in 148 patients (84.1%). Lower staining intensity ERCC1 expression was prominent in parabasal cells in the lower half of the epithelium, while at high staining intensity, ERCC1 expression was present throughout the epithelium. The median H-score was 50. No significant differences in age, gender, smoking status, tumor site, or stage were seen between the high and low ERCC1 expression groups. Expression of ERCC1 stratified by tumor site correlates with OS. Patients with oropharyngeal HNSCC and high ERCC1 expression (H-score > 120) were more likely to survive ( P < .01) and remain disease free when compared to non-oropharyngeal squamous cell carcinoma (SCCa) patients with high ERCC1 expression despite treatment modality and human papillomavirus virus (HPV) status. Conclusion Patients with oropharyngeal SCCa and high ERCC1 expression may have better outcomes despite HPV status.
Collapse
Affiliation(s)
- Mihir R. Patel
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Ni Zhao
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mei-Kim Ang
- University of North Carolina Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael E. Stadler
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Karen Fritchie
- Department of Pathology and Lab Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Mark C. Weissler
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Adam M. Zanation
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Stephen L. Harris
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - William K. Funkhouser
- Department of Pathology and Lab Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Andrew F. Olshan
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Carol G. Shores
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - D. Neil Hayes
- University of North Carolina Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
41
|
Gan Y, Li XR, Chen DJ, Wu JH. Association between polymorphisms of XRCC1 Arg399Gln and XPD Lys751Gln genes and prognosis of colorectal cancer in a Chinese population. Asian Pac J Cancer Prev 2013; 13:5721-4. [PMID: 23317245 DOI: 10.7314/apjcp.2012.13.11.5721] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We conducted this study to detect associations between XRCC1 Arg399Gln and XPD Lys751Gln genotypes and survival of colorectal cancer patients treated with 5-FU/oxalipatin chemotherapy. We included 289 Chinese patients with advanced colorectal cancer, who had received 5-FU/oxalipatin chemotherapy as first-line treatment from January 2005 to January 2007. All patients were followed up till Nov. 2011. Genotyping for XRCC1 Arg399Gln and XPD Lys751Gln polymorphisms was based upon duplex polymerase-chain-reaction with the PCR-RFLP method. In our study, we found the XRCC1 399 Gln/Gln genotype to confer significantly higher rates of response to chemotherapy when compared to the Arg/Arg genotype [OR (95% CI)=2.56(1.57-2.55)]. patients with the XPD 751 Gln/Gln genotype had significantly higher rates of response to chemotherapy [OR (95% CI)=1.54(0.87-2.65)] and those with the XRCC1 399 Gln/Gln genotype had a longer average survival time and significantly lower risk of death than did those with the Arg/Arg genotype [HR (95% CI)=0.66(0.36-0.95)]. Similarly, those carrying the XPD 751Gln/Gln genotype had 0.51-fold the risk of death of those with XPD 751Lys/Lys [HR (95% CI)=0.51(0.33-0.94)]. In conclusion, it is suggested that the XRCC1 Arg399Gln and XPD Lys751Gln polymorphisms should be routinely assessed to determine colorectal patients who are more likely to benefit from 5-FU/oxalipatin chemotherapy.
Collapse
Affiliation(s)
- Yi Gan
- Department of General Surgery, 3rd Xiang-Ya Hospital of Central South University, Changsha, China
| | | | | | | |
Collapse
|
42
|
Association between ERCC1 and XPA expression and polymorphisms and the response to cisplatin in testicular germ cell tumours. Br J Cancer 2013; 109:68-75. [PMID: 23807173 PMCID: PMC3708571 DOI: 10.1038/bjc.2013.303] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/15/2013] [Accepted: 05/23/2013] [Indexed: 02/07/2023] Open
Abstract
Background: Cisplatin cures over 80% of testicular germ cell tumours (TGCTs), and nucleotide-excision repair (NER) modifies the sensitivity to cisplatin. We explored the association between NER proteins and their polymorphisms with cisplatin sensitivity (CPS) and overall survival (OS) of patients with non-seminomatous (ns)-TGCTs. Methods: The expression of ERCC1 and XPA and the presence of γH2AX were evaluated in cancer cell lines and in fresh ns-TGCTs. The ERCC1 protein was also determined in ns-TGCTs. The differences between CPS and non-CPS cell lines and patients were analysed by Student's t- or χ2-tests. The differences in OS were analysed using the log-rank test, and the hazard ratios (HRs) were calculated using the Cox model. Results: High ERCC1 expression was observed in the non-CPS cells, and both ERCC1 and γH2AX expressions were augmented after cisplatin treatment. Increased ERCC1 expression was also identified in non-CPS patients. Neither polymorphism was associated with either CPS or OS. The presence of ERCC1 was associated with non-CPS (P=0.05) and adjusted in the prognosis groups. The HR in ERCC1-negative and non-CPS patients was >14.43, and in ERCC1-positive and non-CPS patients the HR was >11.86 (P<0.001). Conclusions: High levels of ERCC1 were associated with non-CPS, suggesting that ERCC1 could be used as a potential indicator of the response to cisplatin and prognosis in ns-TGCTs.
Collapse
|
43
|
Chu H, Gu D, Xu M, Xu Z, Gong Y, Gong W, Tang Y, Zhou J, Tong N, Zhang Z, Chen J, Wang M. A genetic variant in ERCC2 is associated with gastric cancer prognosis in a Chinese population. Mutagenesis 2013; 28:441-6. [PMID: 23680703 DOI: 10.1093/mutage/get023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Endogenous and exogenous factors can induce DNA damage, leading to increased risk of cancer. Nucleotide excision repair (NER) is considered as the most versatile DNA repair pathway to deal with a variety of different DNA lesions. ERCC1 and ERCC2 are the two important proteins in NER pathway. In this study, we investigated the association of three functional single nucleotide polymorphisms (SNPs) (ERCC1 rs11615, ERCC2 rs13181 and ERCC2 rs1799793) with the clinical outcome of 940 gastric cancer patients in a Chinese population. Multiplex SNaPshot technology was used to genotype these three SNPs. Our results revealed that individuals with ERCC2 rs13181TG/GG genotypes had a decreased risk of death compared with those with TT genotype [log-rank P = 0.008; adjusted hazard ratio = 0.68, 95% confidence interval = 0.51-0.91] and this protective effect was more pronounced among the subgroups of patients with tumour size ≤ 5 cm (0.59, 0.39-0.89), non-cardia gastric tumour (0.69, 0.48-0.98), no lymph node metastasis (0.55, 0.32-0.96), no distant metastasis (0.70, 0.52-0.95) and chemotherapy (0.39, 0.21-0.72). We conclude that ERCC2 rs13181 polymorphism could play different roles in the overall survival of gastric cancer. Further larger studies should be conducted to validate our findings.
Collapse
Affiliation(s)
- Haiyan Chu
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University, 818 East Tianyuan Road, Nanjing 211166, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Martens-de Kemp SR, Dalm SU, Wijnolts FMJ, Brink A, Honeywell RJ, Peters GJ, Braakhuis BJM, Brakenhoff RH. DNA-bound platinum is the major determinant of cisplatin sensitivity in head and neck squamous carcinoma cells. PLoS One 2013; 8:e61555. [PMID: 23613873 PMCID: PMC3629194 DOI: 10.1371/journal.pone.0061555] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 03/11/2013] [Indexed: 01/21/2023] Open
Abstract
PURPOSE The combination of systemic cisplatin with local and regional radiotherapy as primary treatment of head and neck squamous cell carcinoma (HNSCC) leads to cure in approximately half of the patients. The addition of cisplatin has significant effects on outcome, but despite extensive research the mechanism underlying cisplatin response is still not well understood. METHODS We examined 19 HNSCC cell lines with variable cisplatin sensitivity. We determined the TP53 mutational status of each cell line and investigated the expression levels of 11 potentially relevant genes by quantitative real-time PCR. In addition, we measured cisplatin accumulation and retention, as well as the level of platinum-DNA adducts. RESULTS We found that the IC50 value was significantly correlated with the platinum-DNA adduct levels that accumulated during four hours of cisplatin incubation (p = 0.002). We could not find a significant correlation between cisplatin sensitivity and any of the other parameters tested, including the expression levels of established cisplatin influx and efflux transporters. Furthermore, adduct accumulation did not correlate with mRNA expression of the investigated influx pumps (CTR1 and OCT3) nor with that of the examined DNA repair genes (ATR, ATM, BRCA1, BRCA2 and ERCC1). CONCLUSION Our findings suggest that the cisplatin-DNA adduct level is the most important determinant of cisplatin sensitivity in HNSCC cells. Imaging with radio-labeled cisplatin might have major associations with outcome.
Collapse
MESH Headings
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cisplatin/metabolism
- Cisplatin/pharmacology
- Cisplatin/therapeutic use
- DNA Adducts/metabolism
- DNA Adducts/pharmacology
- DNA, Neoplasm/metabolism
- Drug Screening Assays, Antitumor
- Gene Expression Regulation, Neoplastic/drug effects
- Genes, Neoplasm/genetics
- Head and Neck Neoplasms/drug therapy
- Head and Neck Neoplasms/genetics
- Head and Neck Neoplasms/pathology
- Humans
- Inhibitory Concentration 50
- Mutation/genetics
- Platinum/pharmacology
- Platinum/therapeutic use
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Squamous Cell Carcinoma of Head and Neck
- Statistics, Nonparametric
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
- Sanne R Martens-de Kemp
- Department of Otolaryngology/Head-Neck Surgery, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Gerhard R, Carvalho A, Carneiro V, Bento RS, Uemura G, Gomes M, Albergaria A, Schmitt F. Clinicopathological significance of ERCC1 expression in breast cancer. Pathol Res Pract 2013; 209:331-6. [PMID: 23702380 DOI: 10.1016/j.prp.2013.02.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 01/22/2013] [Accepted: 02/11/2013] [Indexed: 10/27/2022]
Abstract
The excision repair cross-complementation 1 (ERCC1) enzyme plays an essential role in the nucleotide excision repair pathway and is associated with resistance to platinum-based chemotherapy in different types of cancer. The aim of the present study was to evaluate the clinicopathological significance of ERCC1 expression in breast cancer patients. We analyzed the immunohistochemical expression of ERCC1 in a tissue microarray from 135 primary breast carcinomas and correlated the immunohistochemical findings with clinicopathological factors and outcome data. ERCC1 expression analysis was available for 109 cases. In this group, 58 (53.2%) were positive for ERCC1. ERCC1-positive expression was correlated with smaller tumor size (P=0.007) and with positivity for estrogen receptor (P=0.040), but no correlation was found with other clinicopathological features. Although not statistically significant, triple negative breast cancers were more frequently negative for ERCC1 (61.5% of the cases) compared to the non-triple negative breast cancer cases (41.5%). In conclusion, ERCC1 expression correlated significantly with favorable prognostic factors, such as smaller tumor size and ER-positivity, suggesting a possible role for ERCC1 as a predictive and/or prognostic marker in breast cancer.
Collapse
Affiliation(s)
- Renê Gerhard
- IPATIMUP - Institute of Molecular Pathology and Immunology of Porto University, Porto, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Choi IJ, Kim DW, Kim DY, Lee CH, Rhee CS. Predictive markers for neoadjuvant chemotherapy in advanced squamous cell carcinoma of maxillary sinus: Preliminary report. Acta Otolaryngol 2013; 133:291-6. [PMID: 23146027 DOI: 10.3109/00016489.2012.734928] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONCLUSION ERCC 1 seems to be promising as a predictive marker for response to neoadjuvant chemotherapy (NAC) and early decision for surgery in advanced squamous cell carcinoma (SCC) of the maxillary sinus. OBJECTIVES This study aimed to find a possible relation of ERCC1 or XRCC1 expression with response to NAC and prognosis in advanced SCC of the maxillary sinus. METHODS From 1998 to 2006, 17 patients with advanced SCC of the maxillary sinus received NAC at the Seoul National University Hospital. The expression of ERCC1 and XRCC1 was assessed by immunohistochemistry. Complete and partial remissions were categorized as the chemo-sensitive group. On the other hand, stable and progressive diseases were categorized as the chemo-resistant group. RESULTS Of these 17 patients, 1 had complete remission, 6 had partial remission, 4 had stable disease, and 6 had progression of disease. The 5-year survival rate was 40% for all 17 patients. The expression of ERCC1 and XRCC1 was not correlated with nodal or distant metastasis. With a cut-off value of 65%, patients with higher ERCC1 scores showed chemo-resistance and survival disadvantage over those with lower ERCC1 scores. However, XRCC1 did not show a significant effect on the response to NAC.
Collapse
Affiliation(s)
- Ik Joon Choi
- Departments of Otorhinolaryngology, Korea Cancer Center Hospital, Korea
| | | | | | | | | |
Collapse
|
47
|
Bai ZL, Wang YY, Zhe H, He JL, Hai P. ERCC1 mRNA levels can predict the response to cisplatin-based concurrent chemoradiotherapy of locally advanced cervical squamous cell carcinoma. Radiat Oncol 2012; 7:221. [PMID: 23259415 PMCID: PMC3536645 DOI: 10.1186/1748-717x-7-221] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 12/17/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The purpose of this study was to investigate whether the excision repair cross-complementation group 1 (ERCC1) mRNA expression could predict treatment response of patients with locally advanced cervical squamous cell carcinoma (LACSCC) who underwent cisplatin-based concurrent chemoradiotherapy (CCCRT). METHODS A total of sixty LACSCC patients, treated with radical CCCRT from a single institution were evaluated. ERCC1 mRNA expression was determined by quantitative real-time RT-PCR in pre-treatment tumor tissues. The association of ERCC1 status with clinicopathological characteristics (age, histological grade, tumor size, parametrial invasion, lymph node metastasis and FIGO stage) and treatment response were analyzed. RESULTS No significant association between ERCC1 mRNA expression and clinicopathological characteristics were observed. Patients with low ERCC1 mRNA level had a significantly higher rate of complete response (86.21%) than patients with high level of ERCC1 expression (19.36%; p < 0.001). In the logistic regression analysis, low ERCC1 mRNA level retained an independent role in predicting complete response to CCCRT (P < 0.001). An ERCC1 expression level of 0.0901 was determined as an optimal cutoff value to identify complete response patients to CCCRT treatment. The sensitivity for detection of a complete response was 81.48% with a specificity of 96.97% (area under the curve, 0.893; 95% confidence interval, 0.804-0.983). CONCLUSIONS This is the first analysis of the association between ERCC1 mRNA levels and treatment response in patients with LACSCC. Low ERCC1 mRNA level appears to be a highly specific predictor of response to CCCRT in LACSCC.
Collapse
Affiliation(s)
- Zhou-lan Bai
- Graduate School, Ningxia Medical University, No.1160 Shengli Str, Yinchuan, Ningxia 750004, China
| | | | | | | | | |
Collapse
|
48
|
|
49
|
Barakat KH, Jordheim LP, Perez-Pineiro R, Wishart D, Dumontet C, Tuszynski JA. Virtual screening and biological evaluation of inhibitors targeting the XPA-ERCC1 interaction. PLoS One 2012; 7:e51329. [PMID: 23272099 PMCID: PMC3522735 DOI: 10.1371/journal.pone.0051329] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2012] [Accepted: 10/10/2012] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Nucleotide excision repair (NER) removes many types of DNA lesions including those induced by UV radiation and platinum-based therapy. Resistance to platinum-based therapy correlates with high expression of ERCC1, a major element of the NER machinery. The interaction between ERCC1 and XPA is essential for a successful NER function. Therefore, one way to regulate NER is by inhibiting the activity of ERCC1 and XPA. METHODOLOGY/PRINCIPAL FINDINGS Here we continued our earlier efforts aimed at the identification and characterization of novel inhibitors of the ERCC1-XPA interaction. We used a refined virtual screening approach combined with a biochemical and biological evaluation of the compounds for their ability to interact with ERCC1 and to sensitize cells to UV radiation. Our findings reveal a new validated ERCC1-XPA inhibitor that significantly sensitized colon cancer cells to UV radiation indicating a strong inhibition of the ERCC1-XPA interaction. CONCLUSIONS NER is a major factor in acquiring resistance to platinum-based therapy. Regulating the NER pathway has the potential of improving the efficacy of platinum treatments. One approach that we followed is to inhibit the essential interaction between the two NER elements, ERCC1 and XPA. Here, we performed virtual screening against the ERCC1-XPA interaction and identified novel inhibitors that block the XPA-ERCC1 binding. The identified inhibitors significantly sensitized colon cancer cells to UV radiation indicating a strong inhibition of the ERCC1-XPA interaction.
Collapse
Affiliation(s)
- Khaled H. Barakat
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada
- Department of Engineering Mathematics and Physics, Fayoum University, Fayoum, Egypt
| | - Lars P. Jordheim
- Université de Lyon, Lyon, France
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | | | - David Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Computing Science, University of Alberta, Edmonton, Alberta, Canada
| | - Charles Dumontet
- Université de Lyon, Lyon, France
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Jack A. Tuszynski
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
- * E-mail:
| |
Collapse
|
50
|
Worsham MJ, Ali H, Dragovic J, Schweitzer VP. Molecular characterization of head and neck cancer: how close to personalized targeted therapy? Mol Diagn Ther 2012; 16:209-22. [PMID: 22873739 DOI: 10.2165/11635330-000000000-00000] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Molecular targeted therapy in head and neck squamous cell carcinoma (HNSCC) continues to make strides, and holds much promise. Cetuximab remains the sole US FDA-approved molecular targeted therapy available for HNSCC, though several new biologic agents targeting the epidermal growth factor receptor (EGFR) and other pathways are currently in the regulatory approval pipeline. While targeted therapies have the potential to be personalized, their current use in HNSCC is not personalized. This is illustrated for EGFR-targeted drugs, where EGFR as a molecular target has yet to be individualized for HNSCC. Future research needs to identify factors that correlate with response (or lack of one) and the underlying genotype-phenotype relationship that dictates this response. Comprehensive exploration of genetic and epigenetic landscapes in HNSCC is opening new frontiers to further enlighten and mechanistically inform newer as well as existing molecular targets, and to set a course for eventually translating these discoveries into therapies for patients. This opinion offers a snapshot of the evolution of molecular subtyping in HNSCC and its current clinical applicability, as well as new emergent paradigms with implications for controlling this disease in the future.
Collapse
Affiliation(s)
- Maria J Worsham
- Department of OtolaryngologyHead and Neck Surgery, Henry Ford Health System, Detroit, MI 48202, USA.
| | | | | | | |
Collapse
|