1
|
Cros-Perrial E, Beaumel S, Gimbert M, Camus N, Vicente C, Sekiou I, Figuet L, Duruisseaux M, Dumontet C, Jordheim LP. SLX4 and XPF are involved in cell migration and EMT in a cell-specific manner. Biochem Pharmacol 2025; 236:116885. [PMID: 40118290 DOI: 10.1016/j.bcp.2025.116885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
SLX4 and XPF are two proteins involved in DNA repair, but very little is known about their potential roles in other processes of cancer cell biology. We developed original cell models with CRISPR-Cas9-mediated knock-out of SLX4 and/or XPF using five different cell lines (A549, NCI-H1703, COLO-357, HT-29 and HEK-293 T), and performed characterization with cell biology experiments including migration assays, drug sensitivity testing, cell proliferation assessment and Western blots for relevant proteins. Results showed decreased migration of all models in HT-29 cells, of XPF-deficient COLO-357 cells and of SLX4-deficient HEK-293 T cells. Modified cell models had overall increased sensitivity to cisplatin and mitomycine C, and some models showed an increased frequency of double-stranded DNA damages. One NCI-H1703 cell model showed major karyotypic modifications, and epithelial to mesenchymal transition (EMT)-related proteins were modified in several models. Finally, knocking out one or both proteins in A549 cells had not the same impact on in vivo growth in mice. These original cell models allowed us to identify new and DNA repair-unrelated cellular roles of SLX4 and XPF in cancer cell biology. Our results should be considered within work on Nucleotide Excision Repair (NER) inhibition targeting SLX, XPF or other related proteins.
Collapse
Affiliation(s)
- Emeline Cros-Perrial
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France
| | - Sabine Beaumel
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France
| | - Manon Gimbert
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France
| | - Ninon Camus
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France
| | - Clara Vicente
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France
| | - Imane Sekiou
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France
| | - Léa Figuet
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France
| | - Michaël Duruisseaux
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France; Hospices Civils de Lyon, Lyon, France
| | - Charles Dumontet
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France; Hospices Civils de Lyon, Lyon, France
| | - Lars Petter Jordheim
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France.
| |
Collapse
|
2
|
Li L, Xiao M, Zhang L, Zhao S, Zhang G, Wu S, Jin C, Yang J, Lu X. Excision repair cross complementation group 1 gene exon 3 skipping isoform presents selective cGAS-STING activation in platinum-sensitive lung adenocarcinoma. Free Radic Biol Med 2025; 235:73-85. [PMID: 40258523 DOI: 10.1016/j.freeradbiomed.2025.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/23/2025]
Abstract
Platinum-based chemotherapy is widely used as a frontline therapy for lung adenocarcinoma, while its efficacy is limited by agent resistance and severe toxicity. Recently the immunotherapy represents an alternative or complement to Platinum-based chemotherapy. Interestingly, the sensitivity to platinum is known as a relevant phenotypical biomarker of valid immunotherapy due to the defects in DNA damage response (DDR) in cancer cells. The cGAS/STING pathway detects cytosolic DNA to activate innate immune response, which seems to become a bridge linking DDR and cancer immunogenicity. This study aimed to investigate the effect of ERCC1 splicing isoforms on the cGAS/STING pathway. Besides, the association of ERCC1 splicing isoforms with cGAS/STING signaling in cisplatin-treated cells was analyzed, and the modulation of PRPF8 on ERCC1 exon skipping splicing was elucidated by RNA immunoprecipitation. Finally, we also explored the potential role of an herbal extract β-elemene as chemosensitizer and activator of cGAS/STING signaling. We demonstrated that ERCC1 exon 3 inclusion was of equal importance to exon 8 and endowed ERCC1 with an elevated DNA repair activity, which was linked with cisplatin resistance and cGAS-STING suppression. Mechanistically, PRPF8 was identified to be directly interacted with modulating ERCC1 exon 3 skipping, while β-elemene was found to be involved in the activation of cGAS-STING signaling as an inhibitor of PRPF8. Our data reveal that the ERCC1 exon 3 skipping isoform is associated with DDR and the cGAS/STING innate immune pathway, which provide preclinical rationale for using alternative or complement immunotherapy in Platinum-sensitive NSCLC patients.
Collapse
Affiliation(s)
- Liuli Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China; Department of Pathophysiology College of High Altitude Military Medicine Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
| | - Mingyang Xiao
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China
| | - Liang Zhang
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Shenyang, 110042, People's Republic of China
| | - Shuang Zhao
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China
| | - Guopei Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China
| | - Shengwen Wu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China
| | - Cuihong Jin
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China
| | - Jinghua Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China
| | - Xiaobo Lu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, People's Republic of China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, People's Republic of China.
| |
Collapse
|
3
|
Zhou M, Tian M, Li Z, Wang C, Guo Z. Overview of splicing variation in ovarian cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189288. [PMID: 39993511 DOI: 10.1016/j.bbcan.2025.189288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025]
Abstract
Ovarian cancer remains one of the deadliest gynecological malignancies, with a persistently high mortality rate despite promising advancements in immunotherapy. Aberrant splicing events play a crucial role in cancer heterogeneity and treatment resistance. Many splicing variants, especially those involving key molecular markers such as BRCA1/2, are closely linked to disease progression and treatment outcomes. These variants and related splicing factors hold significant clinical value as diagnostic and prognostic biomarkers and therapeutic targets. This review provides a comprehensive overview of splicing variants in ovarian cancer, emphasizing their role in metastasis and resistance, and offers insights to advance biomarker development and treatment strategies.
Collapse
Affiliation(s)
- Min Zhou
- From the Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mengdie Tian
- From the Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhuoer Li
- From the Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chunli Wang
- From the Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhiqiang Guo
- From the Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
4
|
Rakkannan G, Mohanty AK, Das II, Nayak S, Sahoo L, Kumar R, Rasal A, Rather MA, Ahmad I, Sundaray JK. Triflumezopyrim induced oxidative stress, DNA damage and apoptosis on Labeo rohita: Insights from Bioinformatics, Histopathological and Molecular approaches. Int J Biol Macromol 2025; 304:140911. [PMID: 39938845 DOI: 10.1016/j.ijbiomac.2025.140911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 01/14/2025] [Accepted: 02/09/2025] [Indexed: 02/14/2025]
Abstract
Triflumezopyrim is a novel mesoionic pesticide used in Asian rice fields to control brown plant hopper, which can reach aquatic environments through drift during application or surface runoff. Thus, this study aimed to investigate the effect of chronic exposure of sub-lethal concentrations of triflumezopyrim (2.468 ppm, 1.480 ppm, and 0.740 ppm) on freshwater carp, Labeo rohita.This study encompassed a multi-pronged approach, including molecular docking studies to elucidate the interactions between triflumezopyrim and selected proteins, an assessment of 8-OHdG (8-hydroxy-2-deoxyguanosine) activity to gauge DNA damage, histopathological examinations to identify cellular alterations, and expression of genes involved in oxidative stress, DNA repair, and apoptosis in L. rohita. We observed dose-dependent responses in 8-OHdG activity and the expression of select genes, with higher concentrations of triflumezopyrim yielding more pronounced transcriptional alterations. Notably, histopathological examinations of liver and brain tissues vividly portrayed the impact of triflumezopyrim on L. rohita. These findings contribute to the assessment of biological toxicity and the environmental footprint left by chemical pollutants such as triflumezopyrim. The study emphasizes the crucial role of monitoring histopathological alterations, 8-OHdG activity and gene expression changes as potential biomarkers for assessing exposures to triflumezopyrim. This research provides valuable insights into the ecological implications of triflumezopyrim in aquatic ecosystems.
Collapse
Affiliation(s)
- Gowrimanohari Rakkannan
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneshwar 751002, India
| | - Aman Kumar Mohanty
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneshwar 751002, India
| | - Ipsita Iswari Das
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneshwar 751002, India
| | - Sipra Nayak
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneshwar 751002, India
| | - Lakshman Sahoo
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneshwar 751002, India
| | - Rajesh Kumar
- Aquaculture Production and Environment Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751002, India
| | - Avinash Rasal
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneshwar 751002, India
| | - Mohd Ashraf Rather
- Division of Fish Genetics and Biotechnology, Faculty of Fisheries Rangil-Ganderbal, SKUAST, Kashmir 190006, India.
| | - Ishtiyaq Ahmad
- Division of Fish Genetics and Biotechnology, Faculty of Fisheries Rangil-Ganderbal, SKUAST, Kashmir 190006, India
| | - Jitendra Kumar Sundaray
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneshwar 751002, India.
| |
Collapse
|
5
|
Duan M, Leng S, Mao P. Cisplatin in the era of PARP inhibitors and immunotherapy. Pharmacol Ther 2024; 258:108642. [PMID: 38614254 DOI: 10.1016/j.pharmthera.2024.108642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/21/2024] [Accepted: 03/29/2024] [Indexed: 04/15/2024]
Abstract
Platinum compounds such as cisplatin, carboplatin and oxaliplatin are widely used in chemotherapy. Cisplatin induces cytotoxic DNA damage that blocks DNA replication and gene transcription, leading to arrest of cell proliferation. Although platinum therapy alone is effective against many tumors, cancer cells can adapt to the treatment and gain resistance. The mechanisms for cisplatin resistance are complex, including low DNA damage formation, high DNA repair capacity, changes in apoptosis signaling pathways, rewired cell metabolisms, and others. Drug resistance compromises the clinical efficacy and calls for new strategies by combining cisplatin with other therapies. Exciting progress in cancer treatment, particularly development of poly (ADP-ribose) polymerase (PARP) inhibitors and immune checkpoint inhibitors, opened a new chapter to combine cisplatin with these new cancer therapies. In this Review, we discuss how platinum synergizes with PARP inhibitors and immunotherapy to bring new hope to cancer patients.
Collapse
Affiliation(s)
- Mingrui Duan
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Shuguang Leng
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.
| | - Peng Mao
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
6
|
Conway PJ, Dao J, Kovalskyy D, Mahadevan D, Dray E. Polyploidy in Cancer: Causal Mechanisms, Cancer-Specific Consequences, and Emerging Treatments. Mol Cancer Ther 2024; 23:638-647. [PMID: 38315992 PMCID: PMC11174144 DOI: 10.1158/1535-7163.mct-23-0578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/19/2023] [Accepted: 01/30/2024] [Indexed: 02/07/2024]
Abstract
Drug resistance is the major determinant for metastatic disease and fatalities, across all cancers. Depending on the tissue of origin and the therapeutic course, a variety of biological mechanisms can support and sustain drug resistance. Although genetic mutations and gene silencing through epigenetic mechanisms are major culprits in targeted therapy, drug efflux and polyploidization are more global mechanisms that prevail in a broad range of pathologies, in response to a variety of treatments. There is an unmet need to identify patients at risk for polyploidy, understand the mechanisms underlying polyploidization, and to develop strategies to predict, limit, and reverse polyploidy thus enhancing efficacy of standard-of-care therapy that improve better outcomes. This literature review provides an overview of polyploidy in cancer and offers perspective on patient monitoring and actionable therapy.
Collapse
Affiliation(s)
- Patrick J Conway
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas
- Department of Molecular Immunology & Microbiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Jonathan Dao
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Dmytro Kovalskyy
- Greehey Children's Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas
| | - Daruka Mahadevan
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas
- Department of Molecular Immunology & Microbiology, University of Texas Health San Antonio, San Antonio, Texas
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
| | - Eloise Dray
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
7
|
Marima R, Mosoane B, Mtshali N, Basera A, Kgatle M, Grech G, Dlamini Z. Mechanisms of chemotherapy resistance in cervical cancer. STRATEGIES FOR OVERCOMING CHEMOTHERAPY RESISTANCE IN CERVICAL CANCER 2024:53-70. [DOI: 10.1016/b978-0-443-28985-9.00012-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
8
|
George IA, Chauhan R, Dhawale R, Iyer R, Limaye S, Sankaranarayanan R, Venkataramanan R, Kumar P. Insights into therapy resistance in cervical cancer. ADVANCES IN CANCER BIOLOGY - METASTASIS 2022; 6:100074. [DOI: 10.1016/j.adcanc.2022.100074] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
9
|
Shah SM, Demidova EV, Lesh RW, Hall MJ, Daly MB, Meyer JE, Edelman MJ, Arora S. Therapeutic implications of germline vulnerabilities in DNA repair for precision oncology. Cancer Treat Rev 2022; 104:102337. [PMID: 35051883 PMCID: PMC9016579 DOI: 10.1016/j.ctrv.2021.102337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022]
Abstract
DNA repair vulnerabilities are present in a significant proportion of cancers. Specifically, germline alterations in DNA repair not only increase cancer risk but are associated with treatment response and clinical outcomes. The therapeutic landscape of cancer has rapidly evolved with the FDA approval of therapies that specifically target DNA repair vulnerabilities. The clinical success of synthetic lethality between BRCA deficiency and poly(ADP-ribose) polymerase (PARP) inhibition has been truly revolutionary. Defective mismatch repair has been validated as a predictor of response to immune checkpoint blockade associated with durable responses and long-term benefit in many cancer patients. Advances in next generation sequencing technologies and their decreasing cost have supported increased genetic profiling of tumors coupled with germline testing of cancer risk genes in patients. The clinical adoption of panel testing for germline assessment in high-risk individuals has generated a plethora of genetic data, particularly on DNA repair genes. Here, we highlight the therapeutic relevance of germline aberrations in DNA repair to identify patients eligible for precision treatments such as PARP inhibitors (PARPis), immune checkpoint blockade, chemotherapy, radiation therapy and combined treatment. We also discuss emerging mechanisms that regulate DNA repair.
Collapse
Affiliation(s)
- Shreya M Shah
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, United States; Science Scholars Program, Temple University, Philadelphia, PA, United States
| | - Elena V Demidova
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, United States; Kazan Federal University, Kazan, Russian Federation
| | - Randy W Lesh
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, United States; Geisinger Commonwealth School of Medicine, Scranton, PA, United States
| | - Michael J Hall
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, United States; Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Mary B Daly
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, United States; Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Joshua E Meyer
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, United States; Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Martin J Edelman
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, United States.
| | - Sanjeevani Arora
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, United States; Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, United States.
| |
Collapse
|
10
|
Circulating Tumor Cells in Breast Cancer Patients: A Balancing Act between Stemness, EMT Features and DNA Damage Responses. Cancers (Basel) 2022; 14:cancers14040997. [PMID: 35205744 PMCID: PMC8869884 DOI: 10.3390/cancers14040997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/11/2022] [Accepted: 02/13/2022] [Indexed: 02/04/2023] Open
Abstract
Circulating tumor cells (CTCs) traverse vessels to travel from the primary tumor to distant organs where they adhere, transmigrate, and seed metastases. To cope with these challenges, CTCs have reached maximal flexibility to change their differentiation status, morphology, migratory capacity, and their responses to genotoxic stress caused by metabolic changes, hormones, the inflammatory environment, or cytostatic treatment. A significant percentage of breast cancer cells are defective in homologous recombination repair and other mechanisms that protect the integrity of the replication fork. To prevent cell death caused by broken forks, alternative, mutagenic repair, and bypass pathways are engaged but these increase genomic instability. CTCs, arising from such breast tumors, are endowed with an even larger toolbox of escape mechanisms that can be switched on and off at different stages during their journey according to the stress stimulus. Accumulating evidence suggests that DNA damage responses, DNA repair, and replication are integral parts of a regulatory network orchestrating the plasticity of stemness features and transitions between epithelial and mesenchymal states in CTCs. This review summarizes the published information on these regulatory circuits of relevance for the design of biomarkers reflecting CTC functions in real-time to monitor therapeutic responses and detect evolving chemoresistance mechanisms.
Collapse
|
11
|
Oishi T, Sasaki Y, Tong Y, Chen L, Onodera T, Iwasa S, Udo E, Furusato B, Fujimori H, Imamichi S, Honda T, Bessho T, Fukuoka J, Ashizawa K, Yanagihara K, Nakao K, Yamada Y, Hiraoka N, Masutani M. A newly established monoclonal antibody against ERCC1 detects major isoforms of ERCC1 in gastric cancer. Glob Health Med 2021; 3:226-235. [PMID: 34532603 DOI: 10.35772/ghm.2021.01001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/25/2021] [Accepted: 05/10/2021] [Indexed: 11/08/2022]
Abstract
Identifying patients resistant to cisplatin treatment is expected to improve cisplatin-based chemotherapy for various types of cancers. Excision repair cross-complementing group 1 (ERCC1) is involved in several repair processes of cisplatin-induced DNA crosslinks. ERCC1 overexpression is reported as a candidate prognostic factor and considered to cause cisplatin resistance in major solid cancers. However, anti-ERCC1 antibodies capable of evaluating expression levels of ERCC1 in clinical specimens were not fully optimized. A mouse monoclonal antibody against human ERCC1 was generated in this study. The developed antibody 9D11 specifically detected isoforms of 201, 202, 203 but not 204, which lacks the exon 3 coding region. To evaluate the diagnostic usefulness of this antibody, we have focused on gastric cancer because it is one of the major cancers in Japan. When ERCC1 expression was analyzed in seventeen kinds of human gastric cancer cell lines, all the cell lines were found to express either 201, 202, and/or 203 as major isoforms of ERCC1, but not 204 by Western blotting analysis. Immunohistochemical staining showed that ERCC1 protein was exclusively detected in nuclei of the cells and a moderate level of constant positivity was observed in nuclei of vascular endothelial cells. It showed a clear staining pattern in clinical specimens of gastric cancers. Antibody 9D11 may thus be useful for estimating expression levels of ERCC1 in clinical specimens.
Collapse
Affiliation(s)
- Takayuki Oishi
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Laboratory of Collaborative Research, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan.,Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Frontier Life Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yuka Sasaki
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Laboratory of Collaborative Research, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan.,Department of Frontier Life Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Ying Tong
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Lichao Chen
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Frontier Life Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takae Onodera
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Laboratory of Collaborative Research, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan.,Department of Frontier Life Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Satoru Iwasa
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Emiko Udo
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Cancer Genomics Unit, Clinical Genomics Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Bungo Furusato
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Cancer Genomics Unit, Clinical Genomics Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Hiroaki Fujimori
- Department of Frontier Life Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shoji Imamichi
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Laboratory of Collaborative Research, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Clinical Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tadayoshi Bessho
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Junya Fukuoka
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazuto Ashizawa
- Cancer Genomics Unit, Clinical Genomics Center, Nagasaki University Hospital, Nagasaki, Japan.,Department of Clinical Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazuyoshi Yanagihara
- Division of Biomarker Discovery, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan
| | - Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yasuhide Yamada
- Department of Medical Oncology, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Comprehensive Cancer Center, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Nobuyoshi Hiraoka
- Division of Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Mitsuko Masutani
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Laboratory of Collaborative Research, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan.,Department of Frontier Life Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
12
|
Iwata H, Umeyama Y, Liu Y, Zhang Z, Schnell P, Mori Y, Fletcher O, Marshall JC, Johnson JG, Wood LS, Toi M, Finn RS, Turner NC, Bartlett CH, Cristofanilli M. Evaluation of the Association of Polymorphisms With Palbociclib-Induced Neutropenia: Pharmacogenetic Analysis of PALOMA-2/-3. Oncologist 2021; 26:e1143-e1155. [PMID: 33955129 PMCID: PMC8265363 DOI: 10.1002/onco.13811] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/20/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND The most frequently reported treatment-related adverse event in clinical trials with the cyclin-dependent kinase 4/6 (CDK4/6) inhibitor palbociclib is neutropenia. Allelic variants in ABCB1 and ERCC1 might be associated with early occurrence (i.e., end of week 2 treatment) of grade 3/4 neutropenia. Pharmacogenetic analyses were performed to uncover associations between single nucleotide polymorphisms (SNPs) in these genes, patient baseline characteristics, and early occurrence of grade 3/4 neutropenia. MATERIALS AND METHODS ABCB1 (rs1045642, rs1128503) and ERCC1 (rs3212986, rs11615) were analyzed in germline DNA from palbociclib-treated patients from PALOMA-2 (n = 584) and PALOMA-3 (n = 442). SNP, race, and cycle 1 day 15 (C1D15) absolute neutrophil count (ANC) data were available for 652 patients. Univariate and multivariable analyses evaluated associations between SNPs, patient baseline characteristics, and early occurrence of grade 3/4 neutropenia. Analyses were stratified by Asian (n = 122) and non-Asian (n = 530) ethnicity. Median progression-free survival (mPFS) was estimated using the Kaplan-Meier method. The effect of genetic variants on palbociclib pharmacokinetics was analyzed. RESULTS ABCB1 and ERCC1_rs11615 SNP frequencies differed between Asian and non-Asian patients. Multivariable analysis showed that low baseline ANC was a strong independent risk factor for C1D15 grade 3/4 neutropenia regardless of race (Asians: odds ratio [OR], 6.033, 95% confidence interval [CI], 2.615-13.922, p < .0001; Non-Asians: OR, 6.884, 95% CI, 4.138-11.451, p < .0001). ABCB1_rs1128503 (C/C vs. T/T: OR, 0.57, 95% CI, 0.311-1.047, p = .070) and ERCC1_rs11615 (A/A vs. G/G: OR, 1.75, 95% CI, 0.901-3.397, p = .098) were potential independent risk factors for C1D15 grade 3/4 neutropenia in non-Asian patients. Palbociclib mPFS was consistent across genetic variants; exposure was not associated with ABCB1 genotype. CONCLUSION This is the first comprehensive assessment of pharmacogenetic data in relationship to exposure to a CDK4/6 inhibitor. Pharmacogenetic testing may inform about potentially increased likelihood of patients developing severe neutropenia (NCT01740427, NCT01942135). IMPLICATIONS FOR PRACTICE Palbociclib plus endocrine therapy improves hormone receptor-positive/human epidermal growth factor receptor 2-negative advanced breast cancer outcomes, but is commonly associated with neutropenia. Genetic variants in ABCB1 may influence palbociclib exposure, and in ERCC1 are associated with chemotherapy-induced severe neutropenia. Here, the associations of single nucleotide polymorphisms in these genes and baseline characteristics with neutropenia were assessed. Low baseline absolute neutrophil count was a strong risk factor (p < .0001) for grade 3/4 neutropenia. There was a trend indicating that ABCB1_rs1128503 and ERCC1_rs11615 were potential risk factors (p < .10) for grade 3/4 neutropenia in non-Asian patients. Pharmacogenetic testing could inform clinicians about the likelihood of severe neutropenia with palbociclib.
Collapse
Affiliation(s)
| | | | - Yuan Liu
- Pfizer Inc, San Diego, California, USA
| | - Zhe Zhang
- Pfizer Inc, San Diego, California, USA
| | | | | | - Olivia Fletcher
- Breast Cancer Now Toby Robins Research Centre, The Institute for Cancer Research, London, United Kingdom
| | | | | | | | - Masakazu Toi
- Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Richard S Finn
- David Geffen School of Medicine, University of California Los Angeles, Santa Monica, California, USA
| | - Nicholas C Turner
- Breast Cancer Now Toby Robins Research Centre, The Institute for Cancer Research, London, United Kingdom.,Royal Marsden Hospital, London, United Kingdom
| | | | | |
Collapse
|
13
|
Sreekumar R, Al-Saihati H, Emaduddin M, Moutasim K, Mellone M, Patel A, Kilic S, Cetin M, Erdemir S, Navio MS, Lopez MA, Curtis N, Yagci T, Primrose JN, Price BD, Berx G, Thomas GJ, Tulchinsky E, Mirnezami A, Sayan AE. The ZEB2-dependent EMT transcriptional programme drives therapy resistance by activating nucleotide excision repair genes ERCC1 and ERCC4 in colorectal cancer. Mol Oncol 2021; 15:2065-2083. [PMID: 33931939 PMCID: PMC8333771 DOI: 10.1002/1878-0261.12965] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 02/16/2021] [Accepted: 04/12/2021] [Indexed: 12/18/2022] Open
Abstract
Resistance to adjuvant chemotherapy is a major clinical problem in the treatment of colorectal cancer (CRC). The aim of this study was to elucidate the role of an epithelial to mesenchymal transition (EMT)‐inducing protein, ZEB2, in chemoresistance of CRC, and to uncover the underlying mechanism. We performed IHC for ZEB2 and association analyses with clinical outcomes on primary CRC and matched CRC liver metastases in compliance with observational biomarker study guidelines. ZEB2 expression in primary tumours was an independent prognostic marker of reduced overall survival and disease‐free survival in patients who received adjuvant FOLFOX chemotherapy. ZEB2 expression was retained in 96% of liver metastases. The ZEB2‐dependent EMT transcriptional programme activated nucleotide excision repair (NER) pathway largely via upregulation of the ERCC1 gene and other components in NER pathway, leading to enhanced viability of CRC cells upon oxaliplatin treatment. ERCC1‐overexpressing CRC cells did not respond to oxaliplatin in vivo, as assessed using a murine orthotopic model in a randomised and blinded preclinical study. Our findings show that ZEB2 is a biomarker of tumour response to chemotherapy and risk of recurrence in CRC patients. We propose that the ZEB2‐ERCC1 axis is a key determinant of chemoresistance in CRC.
Collapse
Affiliation(s)
| | - Hajir Al-Saihati
- Cancer Sciences Division, University of Southampton, UK.,College of Applied Medical Sciences, University of Hafr Al-Batin, Saudi Arabia
| | | | | | | | - Ashish Patel
- Cancer Sciences Division, University of Southampton, UK
| | - Seval Kilic
- Cancer Sciences Division, University of Southampton, UK
| | - Metin Cetin
- Department of Molecular Biology and Genetics, Gebze Technical University, Turkey
| | - Sule Erdemir
- Department of Molecular Biology and Genetics, Gebze Technical University, Turkey
| | | | | | - Nathan Curtis
- Department of Surgery, Southampton University Hospital NHS Trust, UK
| | - Tamer Yagci
- Department of Molecular Biology and Genetics, Gebze Technical University, Turkey
| | - John N Primrose
- Cancer Sciences Division, University of Southampton, UK.,Department of Surgery, Southampton University Hospital NHS Trust, UK
| | - Brendan D Price
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Geert Berx
- Molecular Cellular Oncology Lab, Department for Biomedical Molecular Biology, Ghent University, Belgium.,Cancer Research Institute Ghent (CRIG), Belgium
| | | | - Eugene Tulchinsky
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana, Kazakhstan
| | - Alex Mirnezami
- Cancer Sciences Division, University of Southampton, UK.,College of Applied Medical Sciences, University of Hafr Al-Batin, Saudi Arabia
| | - A Emre Sayan
- Cancer Sciences Division, University of Southampton, UK
| |
Collapse
|
14
|
Hua L, Chen S, Wei M, Shen Y, Long J, Lin Z, Meng Y, Guo C, Huang H, Tu X, Yao M. Predictive Value of ERCC1 mRNA Level from Receiver-Operator Characteristic and Pretreatment EBV-DNA Virus Load in Stage II Nasopharyngeal Carcinoma Patients Receiving Intensity-Modulated Radiotherapy with Concurrent Cisplatin. Cancer Biother Radiopharm 2021; 37:2-10. [PMID: 33764811 DOI: 10.1089/cbr.2020.4474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: The molecular mechanisms underlying chemoresistance are still poorly understood in nasopharyngeal cancer; the protein expression of ERCC1 in DNA repair genes has been reported related to resistance platinum and predicting treatment outcomes in various malignant carcinomas, but the benefit for predicting outcomes with optimal cutoff value of ERCC1mRNA is controversial. The level of plasma Epstein-Barr virus (EBV) DNA is positively correlated with clinical stages of nasopharyngeal carcinoma (NPC). The predictive value of ERCC1mRNA from receiver-operator characteristic (ROC) and EBV-DNA level for stratified treatment with stage II NPC is exactly unclear. This study aims to assess the predictive value of combined EBV-DNA and ERCC1 in stage II nasopharyngeal cancer (NPC) patients treated with intensity-modulated radiotherapy (IMRT) with concurrent cisplatin, and provide guidance for future stratified treatment. Methods: A total of 86 stage II NPC patients who received IMRT and concurrent cisplatin-based chemotherapy with or without cisplatin-based adjuvant chemotherapy had measurements of ERCC1 mRNA, and pretreatment EBV-DNA levels were analyzed by real-time PCR (RT-PCR). Associations of ERCC1 mRNA and pretreatment EBV-DNA levels with clinical characteristics and survivals were evaluated. Results: Cutoff value of ERCC1 mRNA obtained from ROC curve was used, and there were significant differences in progression-free survival (PFS) and overall survival (OS) and overall response rate (ORR) between high expression group and low expression group (p = 0.021 and 0.030 and 0.000, respectively). Patients with pretreatment EBV-DNA <2000 copies/mL had significantly better PFS and ORR (p = 0.024 and 0.043, respectively) and a marginally significant impact on OS (p = 0.062) than those with pretreatment EBV-DNA ≥2000 copies/mL. Patients were divided into three groups by combination of ERCC1 mRNA and EBV-DNA level: ERCC1 mRNA low expression/pre-EBV-DNA <2000 copies/mL, ERCC1 mRNA low expression/pre-EBV-DNA ≥2000 copies/mL, and ERCC1 mRNA high expression/pre-EBV-DNA ≥2000 copies/mL. There were significant differences in ORR among the three groups (p = 0.005). The median follow-up was 62 months (range 22-84) with a follow-up rate of 90.70%. In these groups by combination of ERCC1 mRNA and EBV-DNA level, 1, 3, 5-year OS were 100%, 100%, 100%; 100%, 94.1%, 90.9%; and 100%, 85%, 72.9%, respectively (p = 0.038); 1, 3, 5-year PFS were 100%, 100%, 100%; 97.1%, 91.2%, 84.8%; and 95%, 85%, 71.4%, respectively (p = 0.028). Multivariate analysis showed that combination of ERCC1 mRNA and EBV-DNA levels remained independent prognostic factor but not ERCC1 mRNA and EBV-DNA alone. Conclusions: Combined ERCC1 mRNA and pre-EBV-DNA is a better prognostic biomarker in stage II NPC patients treated with concurrent chemoradiation. Patients with ERCC1 mRNA high expression/pre-EBV-DNA ≥2000 copies/mL may benefit from more aggressive treatment.
Collapse
Affiliation(s)
- Li Hua
- Department of Oncology, the Forth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Shaojun Chen
- Department of Oncology, the Forth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Mengzhuan Wei
- Department of Oncology, the Forth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Yongqi Shen
- Department of Oncology, The Liuzhou Railway Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Jianxin Long
- Department of Oncology, Qinzhou Hospital of Traditional Chinese Medicine, Qinzhou, China
| | - Zhan Lin
- Department of Oncology, The Yulin First People's Hospital, Yulin, China
| | - Yiliang Meng
- Department of Oncology, The Baishe People's Hospital, Baishe, China
| | - Chengxian Guo
- Clinical Pharmacology Center, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Haixin Huang
- Department of Oncology, the Forth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Xiaoning Tu
- Department of Head and Neck Surgery, the Forth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Min Yao
- Department of Radiation Onclogy, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
15
|
Was H, Borkowska A, Olszewska A, Klemba A, Marciniak M, Synowiec A, Kieda C. Polyploidy formation in cancer cells: How a Trojan horse is born. Semin Cancer Biol 2021; 81:24-36. [PMID: 33727077 DOI: 10.1016/j.semcancer.2021.03.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/29/2021] [Accepted: 03/03/2021] [Indexed: 01/04/2023]
Abstract
Ploidy increase has been shown to occur in different type of tumors and participate in tumor initiation and resistance to the treatment. Polyploid giant cancer cells (PGCCs) are cells with multiple nuclei or a single giant nucleus containing multiple complete sets of chromosomes. The mechanism leading to formation of PGCCs may depend on: endoreplication, mitotic slippage, cytokinesis failure, cell fusion or cell cannibalism. Polyploidy formation might be triggered in response to various genotoxic stresses including: chemotherapeutics, radiation, hypoxia, oxidative stress or environmental factors like: air pollution, UV light or hyperthermia. A fundamental feature of polyploid cancer cells is the generation of progeny during the reversal of the polyploid state (depolyploidization) that may show high aggressiveness resulting in the formation of resistant disease and tumor recurrence. Therefore, we propose that modern anti-cancer therapies should be designed taking under consideration polyploidization/ depolyploidization processes, which confer the polyploidization a hidden potential similar to a Trojan horse delayed aggressiveness. Various mechanisms and stress factors leading to polyploidy formation in cancer cells are discussed in this review.
Collapse
Affiliation(s)
- Halina Was
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Szaserow 128 Street, Warsaw, Poland.
| | - Agata Borkowska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Szaserow 128 Street, Warsaw, Poland; Postgraduate School of Molecular Medicine, Zwirki i Wigury 61 Street, Warsaw, Poland
| | - Aleksandra Olszewska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Szaserow 128 Street, Warsaw, Poland; Postgraduate School of Molecular Medicine, Zwirki i Wigury 61 Street, Warsaw, Poland
| | - Aleksandra Klemba
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Szaserow 128 Street, Warsaw, Poland; College of Inter-Faculty Individual Studies in Mathematics and Natural Sciences, University of Warsaw, Banacha 2c Street, Warsaw, Poland
| | - Marta Marciniak
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Szaserow 128 Street, Warsaw, Poland
| | - Agnieszka Synowiec
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Szaserow 128 Street, Warsaw, Poland
| | - Claudine Kieda
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Szaserow 128 Street, Warsaw, Poland
| |
Collapse
|
16
|
Ganzinelli M, Linardou H, Alvisi MF, Caiola E, Lo Russo G, Cecere FL, Bettini AC, Psyrri A, Milella M, Rulli E, Fabbri A, De Maglie M, Romanelli P, Murray S, Broggini M, Marabese M, Garassino MC. Single-arm, open label prospective trial to assess prediction of the role of ERCC1/XPF complex in the response of advanced NSCLC patients to platinum-based chemotherapy. ESMO Open 2021; 6:100034. [PMID: 33422766 PMCID: PMC7809372 DOI: 10.1016/j.esmoop.2020.100034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/24/2020] [Accepted: 12/06/2020] [Indexed: 11/05/2022] Open
Abstract
Background Platinum-based therapy, combined or not with immune checkpoint inhibitors, represents a front-line choice for patients with non-small-cell lung cancer (NSCLC). Despite the improved outcomes in the last years for this malignancy, only a sub-group of patients have long-term benefit. Excision repair cross-complementation group 1 (ERCC1) has been considered a potential biomarker to predict the outcome of platinum-based chemotherapy in NSCLC. However, the ERCC1 gene is transcribed in four splice variants where the isoform 202 was described as the only one active and able to complex Xeroderma pigmentosum group F-complementing protein (XPF). Here, we prospectively investigated if the active form of ERCC1, as assessed by the ERCC1/XPF complex (ERCC1/XPF), could predict the sensitivity to platinum compounds. Patients and methods Prospectively enrolled, patients with advanced NSCLC treated with a first-line regimen containing platinum were centrally evaluated for ERCC1/XPF by a proximity ligation assay. Overall survival (OS), progression-free survival (PFS) and objective response rate (ORR) were analyzed. Results The absence of the ERCC1/XPF in the tumor suggested a trend of worst outcomes in terms of both OS [hazard ratio (HR) 1.41, 95% confidence interval (CI) 0.67-2.94, P = 0.373] and PFS (HR 1.61, 95% CI 0.88-3.03, P = 0.123). ORR was marginally influenced in ERCC1/XPF-negative and -positive groups [odds ratio (stable disease + progressive disease versus complete response + partial response) 0.87, 95% CI 0.25-3.07, P = 0.832]. Conclusion The lack of ERCC1/XPF complex in NSCLC tumor cells might delineate a group of patients with poor outcomes when treated with platinum compounds. ERCC1/XPF absence might well identify patients for whom a different therapeutic approach could be necessary. This is the first study investigating the ERCC1/XPF complex as a platinum-based therapy response biomarker in NSCLC. The lack of ERCC1/XPF complex might delineate a group of patients with poor outcomes when treated with platinum compounds. ERCC1/XPF absence might identify tumors for whom a different therapeutic approach than platinum compounds could be necessary.
Collapse
Affiliation(s)
- M Ganzinelli
- Unit of Thoracic Oncology, Medical Oncology Department 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - H Linardou
- 4th Oncology Department, Metropolitan Hospital, Athens, Greece
| | - M F Alvisi
- Laboratory of Methodology for Clinical Research, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - E Caiola
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - G Lo Russo
- Unit of Thoracic Oncology, Medical Oncology Department 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - F L Cecere
- Division of Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - A C Bettini
- UO Oncologia Medica, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - A Psyrri
- Section of Oncology, Department of Internal Medicine, Attikon Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - M Milella
- Department of Medicine, Section of Medical Oncology, University and Hospital Trust of Verona, Verona, Italy
| | - E Rulli
- Laboratory of Methodology for Clinical Research, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - A Fabbri
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - M De Maglie
- Mouse and Animal Pathology Lab, Fondazione Filarete, Milan, Italy; Department of Veterinary Medicine, University of Milan, Milan, Italy
| | - P Romanelli
- Mouse and Animal Pathology Lab, Fondazione Filarete, Milan, Italy; Department of Veterinary Medicine, University of Milan, Milan, Italy
| | - S Murray
- Biomarker Solutions Ltd, London, UK
| | - M Broggini
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| | - M Marabese
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - M C Garassino
- Unit of Thoracic Oncology, Medical Oncology Department 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
17
|
Role of Nucleotide Excision Repair in Cisplatin Resistance. Int J Mol Sci 2020; 21:ijms21239248. [PMID: 33291532 PMCID: PMC7730652 DOI: 10.3390/ijms21239248] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
Cisplatin is a chemotherapeutic drug used for the treatment of a number of cancers. The efficacy of cisplatin relies on its binding to DNA and the induction of cytotoxic DNA damage to kill cancer cells. Cisplatin-based therapy is best known for curing testicular cancer; however, treatment of other solid tumors with cisplatin has not been as successful. Pre-clinical and clinical studies have revealed nucleotide excision repair (NER) as a major resistance mechanism against cisplatin in tumor cells. NER is a versatile DNA repair system targeting a wide range of helix-distorting DNA damage. The NER pathway consists of multiple steps, including damage recognition, pre-incision complex assembly, dual incision, and repair synthesis. NER proteins can recognize cisplatin-induced DNA damage and remove the damage from the genome, thereby neutralizing the cytotoxicity of cisplatin and causing drug resistance. Here, we review the molecular mechanism by which NER repairs cisplatin damage, focusing on the recent development of genome-wide cisplatin damage mapping methods. We also discuss how the expression and somatic mutations of key NER genes affect the response of cancer cells to cisplatin. Finally, small molecules targeting NER factors provide important tools to manipulate NER capacity in cancer cells. The status of research on these inhibitors and their implications in cancer treatment will be discussed.
Collapse
|
18
|
Koutsoukos K, Andrikopoulou A, Dedes N, Zagouri F, Bamias A, Dimopoulos MA. Clinical Perspectives of ERCC1 in Bladder Cancer. Int J Mol Sci 2020; 21:E8829. [PMID: 33266377 PMCID: PMC7700570 DOI: 10.3390/ijms21228829] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/12/2020] [Accepted: 11/18/2020] [Indexed: 12/28/2022] Open
Abstract
ERCC1 is a key regulator of nucleotide excision repair (NER) pathway that repairs bulky DNA adducts, including intrastrand DNA adducts and interstrand crosslinks (ICLs). Overexpression of ERCC1 has been linked to increased DNA repair capacity and platinum resistance in solid tumors. Multiple single nucleotide polymorphisms (SNPs) have been detected in ERCC1 gene that may affect ERCC1 protein expression. Platinum-based treatment remains the cornerstone of urothelial cancer treatment. Given the expanding application of neoadjuvant and adjuvant chemotherapy in locally advanced bladder cancer, there is an emerging need for biomarkers that could distinguish potential responders to cisplatin treatment. Extensive research has been done regarding the prognostic and predictive role of ERCC1 gene expression and polymorphisms in bladder cancer. Moreover, novel compounds have been recently developed to target ERCC1 protein function in order to maximize sensitivity to cisplatin. We aim to review all the existing literature regarding the role of the ERCC1 gene in bladder cancer and address future perspectives for its clinical application.
Collapse
Affiliation(s)
- Konstantinos Koutsoukos
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, 11528 Athens, Greece; (K.K.); (A.A.); (N.D.); (F.Z.)
| | - Angeliki Andrikopoulou
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, 11528 Athens, Greece; (K.K.); (A.A.); (N.D.); (F.Z.)
| | - Nikos Dedes
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, 11528 Athens, Greece; (K.K.); (A.A.); (N.D.); (F.Z.)
| | - Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, 11528 Athens, Greece; (K.K.); (A.A.); (N.D.); (F.Z.)
| | - Aristotelis Bamias
- 2nd Propaedeutic Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, “ATTIKON” University Hospital, Rimini 1, 12462 Chaidari, Greece;
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, 11528 Athens, Greece; (K.K.); (A.A.); (N.D.); (F.Z.)
| |
Collapse
|
19
|
Guffanti F, Alvisi MF, Caiola E, Ricci F, De Maglie M, Soldati S, Ganzinelli M, Decio A, Giavazzi R, Rulli E, Damia G. Impact of ERCC1, XPF and DNA Polymerase β Expression on Platinum Response in Patient-Derived Ovarian Cancer Xenografts. Cancers (Basel) 2020; 12:cancers12092398. [PMID: 32847049 PMCID: PMC7564949 DOI: 10.3390/cancers12092398] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/12/2020] [Accepted: 08/20/2020] [Indexed: 12/17/2022] Open
Abstract
Platinum resistance is an unmet medical need in ovarian carcinoma. Molecular biomarkers to predict the response to platinum-based therapy could allow patient stratification and alternative therapeutic strategies early in clinical management. Sensitivity and resistance to platinum therapy are partially determined by the tumor’s intrinsic DNA repair activities, including nucleotide excision repair (NER) and base excision repair (BER). We investigated the role of the NER proteins—ERCC1, XPF, ERCC1/XPF complex—and of the BER protein DNA polymerase β, as possible biomarkers of cisplatin (DDP) response in a platform of recently established patient-derived ovarian carcinoma xenografts (OC-PDXs). ERCC1 and DNA polymerase β protein expressions were measured by immunohistochemistry, the ERCC1/XPF foci number was detected by proximity ligation assay (PLA) and their mRNA levels by real-time PCR. We then correlated the proteins, gene expression and ERCC1/XPF complexes with OC-PDXs’ response to platinum. To the best of our knowledge, this is the first investigation of the role of the ERCC1/XPF complex, detected by PLA, in relation to the response to DDP in ovarian carcinoma. None of the proteins in the BER and NER pathways studied predicted platinum activity in this panel of OC-PDXs, nor did the ERCC1/XPF foci number. These results were partially explained by the experimental evidence that the ERCC1/XPF complex increases after DDP treatment and this possibly better associates with the cancer cells’ abilities to activate the NER pathway to repair platinum-induced damage than its basal level. Our findings highlight the need for DNA functional assays to predict the response to platinum-based therapy.
Collapse
Affiliation(s)
- Federica Guffanti
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (F.G.); (E.C.); (F.R.)
| | - Maria Francesca Alvisi
- Laboratory of Methodology for Clinical Research, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (M.F.A.); (E.R.)
| | - Elisa Caiola
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (F.G.); (E.C.); (F.R.)
| | - Francesca Ricci
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (F.G.); (E.C.); (F.R.)
| | - Marcella De Maglie
- Mouse and Animal Pathology Lab (MAPLab), Filarete Foundation, Department of Veterinary Medicine, University of Milan, 20139 Milan, Italy;
| | - Sabina Soldati
- Department of Veterinary Pathology, University of Milan, 20133 Milan, Italy;
| | - Monica Ganzinelli
- Unit of Thoracic Oncology, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Alessandra Decio
- Laboratory of Cancer Metastasis Therapeutics, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (A.D.); (R.G.)
| | - Raffaella Giavazzi
- Laboratory of Cancer Metastasis Therapeutics, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (A.D.); (R.G.)
| | - Eliana Rulli
- Laboratory of Methodology for Clinical Research, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (M.F.A.); (E.R.)
| | - Giovanna Damia
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (F.G.); (E.C.); (F.R.)
- Correspondence: ; Tel.: +39-0239014234
| |
Collapse
|
20
|
Kulkarni S, Solomon M, Pankaj D, Carnelio S, Chandrashekar C, Shetty N. Elucidating the role of excision repair cross-complement group 1 in oral epithelial dysplasia and early invasive squamous cell carcinoma: An immunohistochemical study. J Oral Maxillofac Pathol 2020; 24:20-25. [PMID: 32508443 PMCID: PMC7269280 DOI: 10.4103/jomfp.jomfp_60_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 07/09/2019] [Accepted: 07/26/2019] [Indexed: 11/17/2022] Open
Abstract
Objectives: Oral epithelial dysplasia (OED) is characterized by cellular alterations which have the proclivity of progressing to squamous cell carcinoma. Excision repair cross-complement group 1 (ERCC1) is one of the key proteins involved in nucleotide excision repair (NER) pathway. The expression of ERCC1 has been studied in colorectal, esophageal, ovarian and oral squamous cell carcinoma; but, very few studies have been done to apprehend the expression of ERCC1 in OED and early invasive squamous cell carcinoma (EISCC). The goal of this study is to evaluate the role of ERCC1 in OED and EISCC. Materials and Methods: Histopathologically diagnosed cases of moderate dysplasia (n = 10), severe dysplasia (n = 10) and EISCC (n = 10) were retrieved. 4 μ thick sections were cut from the formalin-fixed paraffin-embedded tissue blocks. The sections were immunohistochemically stained for ERCC1 following standard protocols. The expression of ERCC1 was evaluated semiquantitatively. Statistical analysis was carried out using Fischer's exact t-test. Results: The expression of ERCC1 was found to be strong (+3) in EISCC, moderate (+2) in severe dysplasia and mild (+1) in moderate dysplasia. Thus, the results were statistically significant between the three groups (P < 0.001). Conclusion: Disruption in the mechanisms that regulate cell cycle checkpoints and DNA repair mechanism results in genomic instability; these alterations might contribute to carcinoma. ERCC1 is essential to repair the DNA damage induced by various carcinogens. The present study shows significant difference in the expression of ERCC1 between EISCC and OED, which suggests ERCC1 could be used as one of the predictive markers.
Collapse
Affiliation(s)
- Spoorti Kulkarni
- Department of Oral Pathology and Microbiology, Manipal College of Dental Science's, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Monica Solomon
- Department of Oral Pathology and Microbiology, Manipal College of Dental Science's, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Deepthi Pankaj
- Department of Oral Pathology and Microbiology, Manipal College of Dental Science's, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sunitha Carnelio
- Department of Oral Pathology and Microbiology, Manipal College of Dental Science's, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Chetana Chandrashekar
- Department of Oral Pathology and Microbiology, Manipal College of Dental Science's, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Nisha Shetty
- Department of Oral Pathology and Microbiology, Manipal College of Dental Science's, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
21
|
Gonzalez-Hunt CP, Sanders LH. DNA damage and repair in Parkinson's disease: Recent advances and new opportunities. J Neurosci Res 2020; 99:180-189. [PMID: 32048327 DOI: 10.1002/jnr.24592] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/13/2020] [Accepted: 01/26/2020] [Indexed: 12/17/2022]
Abstract
Parkinson's disease (PD) is the most common movement neurodegenerative disorder. Although our understanding of the underlying mechanisms of pathogenesis in PD has greatly expanded, this knowledge thus far has failed to translate into disease-modifying therapies. Therefore, it is of the utmost urgency to interrogate further the multifactorial etiology of PD. DNA repair defects cause many neurodegenerative diseases. An exciting new PD research avenue is the role that DNA damage and repair may play in neuronal death. The goal of this mini-review was to discuss the evidence for the types of DNA damage that accumulates in PD, which has provided clues for which DNA repair pathways, such as DNA double-strand break repair, are dysfunctional. We further highlight compelling data for activation of the DNA damage response in familial and idiopathic PD. The significance of DNA damage and repair is emerging in the PD field and linking these insights to PD pathogenesis may provide new insights into PD pathophysiology and consequently lead to new therapies.
Collapse
Affiliation(s)
| | - Laurie H Sanders
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
22
|
Interplay between BRCA1 and GADD45A and Its Potential for Nucleotide Excision Repair in Breast Cancer Pathogenesis. Int J Mol Sci 2020; 21:ijms21030870. [PMID: 32013256 PMCID: PMC7037490 DOI: 10.3390/ijms21030870] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
A fraction of breast cancer cases are associated with mutations in the BRCA1 (BRCA1 DNA repair associated, breast cancer type 1 susceptibility protein) gene, whose mutated product may disrupt the repair of DNA double-strand breaks as BRCA1 is directly involved in the homologous recombination repair of such DNA damage. However, BRCA1 can stimulate nucleotide excision repair (NER), the most versatile system of DNA repair processing a broad spectrum of substrates and playing an important role in the maintenance of genome stability. NER removes carcinogenic adducts of diol-epoxy derivatives of benzo[α]pyrene that may play a role in breast cancer pathogenesis as their accumulation is observed in breast cancer patients. NER deficiency was postulated to be intrinsic in stage I of sporadic breast cancer. BRCA1 also interacts with GADD45A (growth arrest and DNA damage-inducible protein GADD45 alpha) that may target NER machinery to actively demethylate genome sites in order to change the expression of genes that may be important in breast cancer. Therefore, the interaction between BRCA1 and GADD45 may play a role in breast cancer pathogenesis through the stimulation of NER, increasing the genomic stability, removing carcinogenic adducts, and the local active demethylation of genes important for cancer transformation.
Collapse
|
23
|
Xiao M, Cui S, Zhang L, Yu T, Zhang G, Zhang Q, Li L, Cai Y, Jin C, Yang J, Wu S, Lu X. AC138128.1 an Intronic lncRNA originating from ERCC1 Implies a Potential Application in Lung Cancer Treatment. J Cancer 2019; 10:3608-3617. [PMID: 31333777 PMCID: PMC6636308 DOI: 10.7150/jca.31832] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 05/08/2019] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is one of the most devastating tumors with a high incidence and mortality worldwide. Polymorphisms and expression of ERCC1 commonly predicted the occurrence and prognosis of lung cancer. However, few studies have focused on long non-coding RNAs related to ERCC1 though some studies reminded the importance of its post-transcriptional regulation. In the present study, an intronic lncRNA AC138128.1 originated from ERCC1 was firstly identified in microarray chip and database, and its possibility as a novel biomarker to predict lung cancer treatment was further discussed. Firstly, the qRT-PCR data showed that AC138128.1 expression was much lower in lung cancer comparing with its para-cancer tissues, which further analyzed by ROC curve. Similarly, the difference was also verified in 16HBE, A549 and LK2 cells. Then AC138128.1 expression was found to have an increasing trend in a dose or time-dependent manner after cisplatin treatment. Finally, the subcellular distribution of AC138128.1 reminded that AC138128.1 was mainly expressed in the nucleus. Interestingly a positive relationship between AC138128.1 and ERCC1 expression was only found in cancer tissues, which reminded AC138128.1 may be involved in the regulation of ERCC1. Therefore, as a preliminary exploration of the lncRNA originated from ERCC1, the present study suggested AC138128.1 is of potential value in predicting platinum analogue benefit in lung cancer.
Collapse
Affiliation(s)
- Mingyang Xiao
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, P.R. China
| | - Su Cui
- Dept. of Thoracic Surgery Ward 2, The first Hospital of China Medical University, Shenyang, P.R. China
| | - Liang Zhang
- Dept. of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Shenyang, P.R. China
| | - Tao Yu
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, P.R. China
| | - Guopei Zhang
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, P.R. China
| | - Qianye Zhang
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, P.R. China
| | - Liuli Li
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, P.R. China
| | - Yuan Cai
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, P.R. China
| | - Cuihong Jin
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, P.R. China
| | - Jinghua Yang
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, P.R. China
| | - Shengwen Wu
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, P.R. China
| | - Xiaobo Lu
- Dept. of Toxicology, School of Public Health, China Medical University, Shenyang, P.R. China
| |
Collapse
|
24
|
Hernandez AL, Young CD, Wang JH, Wang XJ. Lessons learned from SMAD4 loss in squamous cell carcinomas. Mol Carcinog 2019; 58:1648-1655. [PMID: 31140647 DOI: 10.1002/mc.23049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/30/2019] [Accepted: 05/10/2019] [Indexed: 12/13/2022]
Abstract
SMAD4 is a potent tumor suppressor and a central mediator of the TGFß signaling pathway. SMAD4 genetic loss is frequent in squamous cell carcinomas (SCCs). Reports of SMAD4 expression in SCCs vary significantly possibly due to inter-tumor heterogeneity or technical reasons. SMAD4 loss is an initiation event for SCCs. In tumor epithelial cells, SMAD4 loss causes increased proliferation, decreased apoptosis, and "Brca-like" genomic instability associated with DNA repair defects. SMAD4 loss also plays a role in the expansion of cancer stem cells. Epithelial SMAD4 loss causes overexpression of TGFß that is released into the tumor microenvironment and contributes to SCC progression through proinflammatory and immune evasive mechanisms. SMAD4 loss, while not a direct therapeutic target, is associated with multiple targetable pathways that require further therapeutic studies. Altogether, SMAD4 loss is a potential biomarker in SCCs that should be further studied for its values in prognostic and therapeutic predictions. Such information will potentially guide future biomarker-driven clinical trial designs and improve SCC patient outcomes.
Collapse
Affiliation(s)
- Ariel L Hernandez
- Department of Pathology, School of Medicine, University of Colorado, Aurora, Colorado
| | - Christian D Young
- Department of Pathology, School of Medicine, University of Colorado, Aurora, Colorado
| | - Jing H Wang
- Department of Immunology and Microbiology, University of Colorado, Aurora, Colorado.,Department of Biomedical Research, National Jewish Health, Denver, Colorado
| | - Xiao-Jing Wang
- Department of Pathology, School of Medicine, University of Colorado, Aurora, Colorado.,Research Service, Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, Colorado
| |
Collapse
|
25
|
Platinum Resistance in Ovarian Cancer: Role of DNA Repair. Cancers (Basel) 2019. [PMID: 30669514 DOI: 10.3390/cancers11010119]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological cancer. It is initially responsive to cisplatin and carboplatin, two DNA damaging agents used in first line therapy. However, almost invariably, patients relapse with a tumor resistant to subsequent treatment with platinum containing drugs. Several mechanisms associated with the development of acquired drug resistance have been reported. Here we focused our attention on DNA repair mechanisms, which are fundamental for recognition and removal of platinum adducts and hence for the ability of these drugs to exert their activity. We analyzed the major DNA repair pathways potentially involved in drug resistance, detailing gene mutation, duplication or deletion as well as polymorphisms as potential biomarkers for drug resistance development. We dissected potential ways to overcome DNA repair-associated drug resistance thanks to the development of new combinations and/or drugs directly targeting DNA repair proteins or taking advantage of the vulnerability arising from DNA repair defects in EOCs.
Collapse
|
26
|
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological cancer. It is initially responsive to cisplatin and carboplatin, two DNA damaging agents used in first line therapy. However, almost invariably, patients relapse with a tumor resistant to subsequent treatment with platinum containing drugs. Several mechanisms associated with the development of acquired drug resistance have been reported. Here we focused our attention on DNA repair mechanisms, which are fundamental for recognition and removal of platinum adducts and hence for the ability of these drugs to exert their activity. We analyzed the major DNA repair pathways potentially involved in drug resistance, detailing gene mutation, duplication or deletion as well as polymorphisms as potential biomarkers for drug resistance development. We dissected potential ways to overcome DNA repair-associated drug resistance thanks to the development of new combinations and/or drugs directly targeting DNA repair proteins or taking advantage of the vulnerability arising from DNA repair defects in EOCs.
Collapse
|
27
|
Platinum Resistance in Ovarian Cancer: Role of DNA Repair. Cancers (Basel) 2019; 11:cancers11010119. [PMID: 30669514 PMCID: PMC6357127 DOI: 10.3390/cancers11010119] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/15/2019] [Accepted: 01/17/2019] [Indexed: 12/22/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological cancer. It is initially responsive to cisplatin and carboplatin, two DNA damaging agents used in first line therapy. However, almost invariably, patients relapse with a tumor resistant to subsequent treatment with platinum containing drugs. Several mechanisms associated with the development of acquired drug resistance have been reported. Here we focused our attention on DNA repair mechanisms, which are fundamental for recognition and removal of platinum adducts and hence for the ability of these drugs to exert their activity. We analyzed the major DNA repair pathways potentially involved in drug resistance, detailing gene mutation, duplication or deletion as well as polymorphisms as potential biomarkers for drug resistance development. We dissected potential ways to overcome DNA repair-associated drug resistance thanks to the development of new combinations and/or drugs directly targeting DNA repair proteins or taking advantage of the vulnerability arising from DNA repair defects in EOCs.
Collapse
|
28
|
Heyza JR, Lei W, Watza D, Zhang H, Chen W, Back JB, Schwartz AG, Bepler G, Patrick SM. Identification and Characterization of Synthetic Viability with ERCC1 Deficiency in Response to Interstrand Crosslinks in Lung Cancer. Clin Cancer Res 2018; 25:2523-2536. [PMID: 30538112 DOI: 10.1158/1078-0432.ccr-18-3094] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/14/2018] [Accepted: 12/06/2018] [Indexed: 12/20/2022]
Abstract
PURPOSE ERCC1/XPF is a DNA endonuclease with variable expression in primary tumor specimens, and has been investigated as a predictive biomarker for efficacy of platinum-based chemotherapy. The failure of clinical trials utilizing ERCC1 expression to predict response to platinum-based chemotherapy suggests additional mechanisms underlying the basic biology of ERCC1 in the response to interstrand crosslinks (ICLs) remain unknown. We aimed to characterize a panel of ERCC1 knockout (Δ) cell lines, where we identified a synthetic viable phenotype in response to ICLs with ERCC1 deficiency. EXPERIMENTAL DESIGN We utilized the CRISPR-Cas9 system to create a panel of ERCC1Δ lung cancer cell lines which we characterized. RESULTS We observe that loss of ERCC1 hypersensitizes cells to cisplatin when wild-type (WT) p53 is retained, whereas there is only modest sensitivity in cell lines that are p53mutant/null. In addition, when p53 is disrupted by CRISPR-Cas9 (p53*) in ERCC1Δ/p53WT cells, there is reduced apoptosis and increased viability after platinum treatment. These results were recapitulated in 2 patient data sets utilizing p53 mutation analysis and ERCC1 expression to assess overall survival. We also show that kinetics of ICL-repair (ICL-R) differ between ERCC1Δ/p53WT and ERCC1Δ/p53* cells. Finally, we provide evidence that cisplatin tolerance in the context of ERCC1 deficiency relies on DNA-PKcs and BRCA1 function. CONCLUSIONS Our findings implicate p53 as a potential confounding variable in clinical assessments of ERCC1 as a platinum biomarker via promoting an environment in which error-prone mechanisms of ICL-R may be able to partially compensate for loss of ERCC1.See related commentary by Friboulet et al., p. 2369.
Collapse
Affiliation(s)
- Joshua R Heyza
- Department of Oncology, Karmanos Cancer Institute and Wayne State University, Detroit, Michigan
| | - Wen Lei
- Department of Oncology, Karmanos Cancer Institute and Wayne State University, Detroit, Michigan
| | - Donovan Watza
- Department of Oncology, Karmanos Cancer Institute and Wayne State University, Detroit, Michigan
| | - Hao Zhang
- Department of Oncology, Karmanos Cancer Institute and Wayne State University, Detroit, Michigan
| | - Wei Chen
- Department of Oncology, Karmanos Cancer Institute and Wayne State University, Detroit, Michigan.,Biostatistics Core, Karmanos Cancer Institute and Wayne State University, Detroit, Michigan
| | - Jessica B Back
- Department of Oncology, Karmanos Cancer Institute and Wayne State University, Detroit, Michigan
| | - Ann G Schwartz
- Department of Oncology, Karmanos Cancer Institute and Wayne State University, Detroit, Michigan
| | - Gerold Bepler
- Department of Oncology, Karmanos Cancer Institute and Wayne State University, Detroit, Michigan.
| | - Steve M Patrick
- Department of Oncology, Karmanos Cancer Institute and Wayne State University, Detroit, Michigan.
| |
Collapse
|
29
|
Touat M, Sourisseau T, Dorvault N, Chabanon RM, Garrido M, Morel D, Krastev DB, Bigot L, Adam J, Frankum JR, Durand S, Pontoizeau C, Souquère S, Kuo MS, Sauvaigo S, Mardakheh F, Sarasin A, Olaussen KA, Friboulet L, Bouillaud F, Pierron G, Ashworth A, Lombès A, Lord CJ, Soria JC, Postel-Vinay S. DNA repair deficiency sensitizes lung cancer cells to NAD+ biosynthesis blockade. J Clin Invest 2018; 128:1671-1687. [PMID: 29447131 PMCID: PMC5873862 DOI: 10.1172/jci90277] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 02/01/2018] [Indexed: 01/04/2023] Open
Abstract
Synthetic lethality is an efficient mechanism-based approach to selectively target DNA repair defects. Excision repair cross-complementation group 1 (ERCC1) deficiency is frequently found in non-small-cell lung cancer (NSCLC), making this DNA repair protein an attractive target for exploiting synthetic lethal approaches in the disease. Using unbiased proteomic and metabolic high-throughput profiling on a unique in-house-generated isogenic model of ERCC1 deficiency, we found marked metabolic rewiring of ERCC1-deficient populations, including decreased levels of the metabolite NAD+ and reduced expression of the rate-limiting NAD+ biosynthetic enzyme nicotinamide phosphoribosyltransferase (NAMPT). We also found reduced NAMPT expression in NSCLC samples with low levels of ERCC1. These metabolic alterations were a primary effect of ERCC1 deficiency, and caused selective exquisite sensitivity to small-molecule NAMPT inhibitors, both in vitro - ERCC1-deficient cells being approximately 1,000 times more sensitive than ERCC1-WT cells - and in vivo. Using transmission electronic microscopy and functional metabolic studies, we found that ERCC1-deficient cells harbor mitochondrial defects. We propose a model where NAD+ acts as a regulator of ERCC1-deficient NSCLC cell fitness. These findings open therapeutic opportunities that exploit a yet-undescribed nuclear-mitochondrial synthetic lethal relationship in NSCLC models, and highlight the potential for targeting DNA repair/metabolic crosstalks for cancer therapy.
Collapse
Affiliation(s)
- Mehdi Touat
- Inserm U981, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Inserm U1127, CNRS UMR 7225, Sorbonne Universités, UPMC Université Paris 06 UMRS1127, Institut du Cerveau et de la Moelle Epiniere, ICM, Paris, France
| | - Tony Sourisseau
- Inserm U981, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Nicolas Dorvault
- Inserm U981, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Inserm U981, ATIP-Avenir Team, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Roman M. Chabanon
- Inserm U981, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Inserm U981, ATIP-Avenir Team, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Marlène Garrido
- Inserm U981, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Inserm U981, ATIP-Avenir Team, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Daphné Morel
- Inserm U981, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Inserm U981, ATIP-Avenir Team, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Dragomir B. Krastev
- The CRUK Gene Function Laboratory and Breast Cancer Now Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Ludovic Bigot
- Inserm U981, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Julien Adam
- Inserm U981, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Département de Biologie et Pathologies Médicales, and
| | - Jessica R. Frankum
- The CRUK Gene Function Laboratory and Breast Cancer Now Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Sylvère Durand
- Metabolomics Platform, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Clement Pontoizeau
- Centre de Référence des Maladies Héréditaires du Métabolisme, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
- Service de Biochimie Métabolomique et Protéomique, Hôpital Necker-Enfants Malades, Assistance Publique–Hôpitaux de Paris, Paris, France
- Inserm U1163, Institut Imagine, Equipe “Génétique des Maladies Mitochondriales” and Paris Descartes University, Paris, France
| | - Sylvie Souquère
- CNRS UMR-9196, Functional Organization of the Cell, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Mei-Shiue Kuo
- Inserm U981, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | | | - Faraz Mardakheh
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Alain Sarasin
- CNRS UMR-8200, Laboratory of Genetic Stability and Oncogenesis, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Ken A. Olaussen
- Inserm U981, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Faculté de médecine Paris-Sud XI, Kremlin-Bicêtre
| | - Luc Friboulet
- Inserm U981, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Frédéric Bouillaud
- Inserm U1016, CNRS UMR 8104, Institut Cochin, Université Paris-Descartes-Paris 5, Paris, France
| | - Gérard Pierron
- CNRS UMR-9196, Functional Organization of the Cell, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Alan Ashworth
- UCSF Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, USA
| | - Anne Lombès
- Inserm U1016, CNRS UMR 8104, Institut Cochin, Université Paris-Descartes-Paris 5, Paris, France
| | - Christopher J. Lord
- The CRUK Gene Function Laboratory and Breast Cancer Now Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Jean-Charles Soria
- Inserm U981, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Faculté de médecine Paris-Sud XI, Kremlin-Bicêtre
| | - Sophie Postel-Vinay
- Inserm U981, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Inserm U981, ATIP-Avenir Team, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- The CRUK Gene Function Laboratory and Breast Cancer Now Research Centre, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
30
|
ERCC1 as a prognostic factor for survival in patients with advanced urothelial cancer treated with platinum based chemotherapy: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2017; 120:120-126. [PMID: 29198325 DOI: 10.1016/j.critrevonc.2017.10.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 10/12/2017] [Accepted: 10/29/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The predictive role of excision repair cross-complementing group 1 (ERCC1) as a predictive factor in patients with advanced urothelial cancer (AUC) treated with platinum-based treatment is not well defined. Here, we evaluate the role of ERCC1 in patients with AUC treated with platinum-based treatment. METHODS We performed comprehensive, systematic computerized search to identify relevant studies through Medline, Embase, Cochrane Controlled Trials Register (CCTR) databases and abstracts from American Society of Clinical Oncology (ASCO) and ASCO Genitourinary Cancers Symposium, European Society For Medical Oncology (ESMO) and European Association of Urology (EAU) meeting up to July 2015. A systematic review and meta-analysis were performed. RESULTS We included a total of 1475 patients from 13 studies. We found that ERCC1 positivity was significantly associated with worse progression-free survival (pooled HR: 1.54, 95% CI: 1.13-2.11, p=0.006). There was no significant association with overall survival (pooled HR1.63, 95% CI: 0.93-2.88, p=0.09) and disease-free survival (pooled HR: 1.092, 95% CI: 0.63-1.90, p=0.75). CONCLUSION ERCC1 positivity might be a prognostic indicator for poorer survival outcomes among patients with AUC. ERCC1 positivity was trending to poorer OS but was statistically worse for PFS. Further large prospective studies are warranted as ERCC1 could be used as a predictive marker to direct treatment of patients with AUC.
Collapse
|
31
|
Wang X, Zhu X, Zhang H, Fan X, Xue X, Chen Y, Ding C, Zhao J, Wu G. ERCC1_202 Is A Prognostic Biomarker in Advanced Stage Non-Small Cell Lung Cancer Patients Treated with Platinum-Based Chemotherapy. J Cancer 2017; 8:2846-2853. [PMID: 28928874 PMCID: PMC5604217 DOI: 10.7150/jca.19897] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/25/2017] [Indexed: 12/12/2022] Open
Abstract
Purpose: To develop a qPCR method to examine the 202 isoform of excision repair cross-complementation group 1 (ERCC1_202) and to evaluate its clinical utility as a predictive biomarker for platinum-based chemotherapy in non-small cell lung cancer (NSCLC). Methods: The relative complementary DNA (cDNA) quantification for ERCC1_202 was conducted using a fluorescence-based, real-time detection method and β-actin was used as a reference gene. Results: A strong correlation was observed between ERCC1_202 mRNA and ERCC1 mRNA levels in NSCLC cells (P < 0.001). 28 patients completed this research. Our results implied that as ERCC1_202 levels increased, the risk of progression (HR = 4.296, P = 0.011) and death (HR = 6.503, P = 0.001) increased. At multivariate analysis, high expression of ERCC1_202 was shown to be an independent predictive factor for time to progression (P = 0.047), and progression-free survival (P = 0.014). However, the high expression of ERCC1_202 was not an independent predictive factor for response (P = 0.324). Conclusions: This study suggests that the efficacy of platinum-based chemotherapy can be improved when customized according to the expression of ERCC1_202.
Collapse
Affiliation(s)
- Xiyong Wang
- Medical school of Southeast University, Nanjing 210009, China.,Department of Respiratory Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Xiaoli Zhu
- Medical school of Southeast University, Nanjing 210009, China.,Department of Respiratory Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Hongming Zhang
- Medical school of Southeast University, Nanjing 210009, China.,Department of Respiratory Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Xiaobo Fan
- Medical school of Southeast University, Nanjing 210009, China
| | - Xiulei Xue
- Medical school of Southeast University, Nanjing 210009, China
| | - Yan Chen
- Medical school of Southeast University, Nanjing 210009, China
| | - Chenbo Ding
- Medical school of Southeast University, Nanjing 210009, China
| | - Jianwen Zhao
- Medical school of Southeast University, Nanjing 210009, China
| | - Guoqiu Wu
- Medical school of Southeast University, Nanjing 210009, China.,Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| |
Collapse
|
32
|
Postel-Vinay S, Soria JC. ERCC1 as Predictor of Platinum Benefit in Non–Small-Cell Lung Cancer. J Clin Oncol 2017; 35:384-386. [DOI: 10.1200/jco.2016.70.5053] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Sophie Postel-Vinay
- Sophie Postel-Vinay and Jean-Charles Soria, Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Paris-Saclay University, and INSERM, UMR981, Villejuif, France
| | - Jean-Charles Soria
- Sophie Postel-Vinay and Jean-Charles Soria, Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Paris-Saclay University, and INSERM, UMR981, Villejuif, France
| |
Collapse
|
33
|
Lee SM, Falzon M, Blackhall F, Spicer J, Nicolson M, Chaudhuri A, Middleton G, Ahmed S, Hicks J, Crosse B, Napier M, Singer JM, Ferry D, Lewanski C, Forster M, Rolls SA, Capitanio A, Rudd R, Iles N, Ngai Y, Gandy M, Lillywhite R, Hackshaw A. Randomized Prospective Biomarker Trial of ERCC1 for Comparing Platinum and Nonplatinum Therapy in Advanced Non-Small-Cell Lung Cancer: ERCC1 Trial (ET). J Clin Oncol 2017; 35:402-411. [PMID: 27893326 DOI: 10.1200/jco.2016.68.1841] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Purpose Retrospective studies indicate that expression of excision repair cross complementing group 1 (ERCC1) protein is associated with platinum resistance and survival in non-small-cell lung cancer (NSCLC). We conducted the first randomized trial, to our knowledge, to evaluate ERCC1 prospectively and to assess the superiority of nonplatinum therapy over platinum doublet therapy for ERCC1-positive NSCLC as well as noninferiority for ERCC1-negative NSCLC. Patients and Methods This trial had a marker-by-treatment interaction phase III design, with ERCC1 (8F1 antibody) status as a randomization stratification factor. Chemonaïve patients with NSCLC (stage IIIB and IV) were eligible. Patients with squamous histology were randomly assigned to cisplatin and gemcitabine or paclitaxel and gemcitabine; nonsquamous patients received cisplatin and pemetrexed or paclitaxel and pemetrexed. Primary end point was overall survival (OS). We also evaluated an antibody specific for XPF (clone 3F2). The target hazard ratio (HR) for patients with ERCC1-positive NSCLC was ≤ 0.78. Results Of patients, 648 were recruited (177 squamous, 471 nonsquamous). ERCC1-positive rates were 54.5% and 76.7% in nonsquamous and squamous patients, respectively, and the corresponding XPF-positive rates were 70.5% and 68.5%. Accrual stopped early in 2012 for squamous patients because OS for nonplatinum therapy was inferior to platinum therapy (median OS, 7.6 months [paclitaxel and gemcitabine] v 10.7 months [cisplatin and gemcitabine]; HR, 1.46; P = .02). Accrual for nonsquamous patients halted in 2013. Median OS was 8.0 (paclitaxel and pemetrexed) versus 9.6 (cisplatin and pemetrexed) months for ERCC1-positive patients (HR, 1.11; 95% CI, 0.85 to 1.44), and 10.3 (paclitaxel and pemetrexed) versus 11.6 (cisplatin and pemetrexed) months for ERCC1-negative patients (HR, 0.99; 95% CI, 0.73 to 1.33; interaction P = .64). OS HR was 1.09 (95% CI, 0.83 to 1.44) for XPF-positive patients, and 1.39 (95% CI, 0.90 to 2.15) for XPF-negative patients (interaction P = .35). Neither ERCC1 nor XPF were prognostic: among nonsquamous patients, OS HRs for positive versus negative were ERCC1, 1.11 ( P = .32), and XPF, 1.08 ( P = .55). Conclusion Superior outcomes were observed for patients with squamous histology who received platinum therapy compared with nonplatinum chemotherapy; however, selecting chemotherapy by using commercially available ERCC1 or XPF antibodies did not confer any extra survival benefit.
Collapse
Affiliation(s)
- Siow Ming Lee
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Mary Falzon
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Fiona Blackhall
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - James Spicer
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Marianne Nicolson
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Abhro Chaudhuri
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Gary Middleton
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Samreen Ahmed
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Jonathan Hicks
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Barbara Crosse
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Mark Napier
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Julian M Singer
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - David Ferry
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Conrad Lewanski
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Martin Forster
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Sally-Ann Rolls
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Arrigo Capitanio
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Robin Rudd
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Natasha Iles
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Yenting Ngai
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Michael Gandy
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Rachel Lillywhite
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| | - Allan Hackshaw
- Siow Ming Lee, Mary Falzon, Martin Forster, Arrigo Capitanio, Robin Rudd, Natasha Iles, Yenting Ngai, Michael Gandy, Rachel Lillywhite, and Allan Hackshaw, University College London, University College London Hospitals; James Spicer, Guy's and St Thomas's NHS Foundation Trust; Conrad Lewanski, Charing Cross Hospital, London; Fiona Blackhall, The Christie NHS Foundation Trust, Manchester; Marianne Nicolson, Aberdeen Royal Infirmary, Aberdeen; Abhro Chaudhuri, Lincoln County Hospital, Lincoln; Gary Middleton, University of Birmingham, Birmingham; Samreen Ahmed, Leicester Royal Infirmary, Leicester; Jonathan Hicks, New Victoria Hospital, Kingston Upon Thames; Barbara Crosse, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield; Mark Napier, North Devon District Hospital, Barnstaple; Julian M. Singer, Princess Alexandra Hospital NHS Foundation Trust, Harlow; David Ferry, New Cross Hospital, Wolverhampton; and Sally-Ann Rolls, Withybush General Hospital, Haverfordwest, United Kingdom
| |
Collapse
|
34
|
Li MX, Bi XY, Zhao H, Huang Z, Han Y, Zhao DB, Zhao JJ, Cai JQ. Excision Repair Cross-complementation Group 1 is a Prognostic Biomarker in Patients with Colorectal Cancer Receiving Chemotherapy. Chin Med J (Engl) 2017; 129:586-93. [PMID: 26904994 PMCID: PMC4804441 DOI: 10.4103/0366-6999.176993] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: Conflicting results about the association between expression level of excision repair cross-complementation group 1 (ERCC1) and clinical outcome in patients with colorectal cancer (CRC) receiving chemotherapy have been reported. Thus, we searched the available articles and performed the meta-analysis to elucidate the prognostic role of ERCC1 expression in patients with CRC. Methods: A thorough literature search using PubMed (Medline), Embase, Cochrane Library, Web of Science databases, and Chinese Science Citation Database was conducted to obtain the relevant studies. Pooled hazard ratios (HRs) or odds ratios (ORs) with 95% confidence intervals (CIs) were calculated to estimate the results. Results: A total of 11 studies were finally enrolled in this meta-analysis. Compared with patients with lower ERCC1 expression, patients with higher ERCC1 expression tended to have unfavorable overall survival (OS) (HR = 2.325, 95% CI: 1.720–3.143, P < 0.001), progression-free survival (PFS) (HR = 1.917, 95% CI: 1.366–2.691, P < 0.001) and poor response to chemotherapy (OR = 0.491, 95% CI: 0.243–0.990, P = 0.047). Subgroup analyses by treatment setting, ethnicity, HR extraction, detection methods, survival analysis, and study design demonstrated that our results were robust. Conclusions: ERCC1 expression may be taken as an effective prognostic factor predicting the response to chemotherapy, OS, and PFS. Further studies with better study design and longer follow-up are warranted in order to gain a deeper understanding of ERCC1's prognostic value.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jian-Qiang Cai
- Department of Abdominal Surgical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
35
|
López-Valverde G, Garcia-Martin E, Fernández-Mateos J, Cruz-González F, Larrosa-Povés JM, Polo-Llorens V, Pablo-Júlvez LE, González-Sarmiento R. Study of association between pre-senile cataracts and rs11615 of ERCC1, rs13181 of ERCC2, and rs25487 of XRCC1 polymorphisms in a Spanish population. Ophthalmic Genet 2016; 38:314-319. [DOI: 10.1080/13816810.2016.1217548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Gloria López-Valverde
- Department of Ophthalmology, University Hospital of Salamanca, Salamanca, Spain
- Department of Ophthalmology, Royo Villanova Hospital, Zaragoza, Spain
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain
| | - Elena Garcia-Martin
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain
- Department of Ophthalmology, Instituto de Investigaciones Sanitarias de Aragón (IIS Aragon), Zaragoza, Spain
| | - Javier Fernández-Mateos
- Molecular Medicine Unit, Department of Medicine, Institute of Molecular and Cellular Biology of Cancer and Institute of Biomedical Research of Salamanca, University of Salamanca-University Hospital of Salamanca CSIC, Salamanca, Spain
| | | | - José M. Larrosa-Povés
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain
- Department of Ophthalmology, Instituto de Investigaciones Sanitarias de Aragón (IIS Aragon), Zaragoza, Spain
| | - Vicente Polo-Llorens
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain
- Department of Ophthalmology, Instituto de Investigaciones Sanitarias de Aragón (IIS Aragon), Zaragoza, Spain
| | - Luis E. Pablo-Júlvez
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain
- Department of Ophthalmology, Instituto de Investigaciones Sanitarias de Aragón (IIS Aragon), Zaragoza, Spain
| | - Rogelio González-Sarmiento
- Molecular Medicine Unit, Department of Medicine, Institute of Molecular and Cellular Biology of Cancer and Institute of Biomedical Research of Salamanca, University of Salamanca-University Hospital of Salamanca CSIC, Salamanca, Spain
| |
Collapse
|
36
|
Taniguchi K, Yamachika S, He F, Karin M. p62/SQSTM1-Dr. Jekyll and Mr. Hyde that prevents oxidative stress but promotes liver cancer. FEBS Lett 2016; 590:2375-97. [PMID: 27404485 DOI: 10.1002/1873-3468.12301] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/08/2016] [Accepted: 07/09/2016] [Indexed: 12/17/2022]
Abstract
p62/SQSTM1 is a multifunctional signaling hub and autophagy adaptor with many binding partners, which allow it to activate mTORC1-dependent nutrient sensing, NF-κB-mediated inflammatory responses, and the NRF2-activated antioxidant defense. p62 recognizes polyubiquitin chains via its C-terminal domain and binds to LC3 via its LIR motif, thereby promoting the autophagic degradation of ubiquitinated cargos. p62 accumulates in many human liver diseases, including nonalcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC), where it is a component of Mallory-Denk bodies and intracellular hyaline bodies. Chronic p62 elevation contributes to HCC development by preventing oncogene-induced senescence and death of cancer-initiating cells and enhancing their proliferation. In this review, we discuss p62-mediated signaling pathways and their roles in liver pathophysiology, especially NASH and HCC.
Collapse
Affiliation(s)
- Koji Taniguchi
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA, USA.,Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Shinichiro Yamachika
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA, USA
| | - Feng He
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
37
|
Lack of Correlation between Aberrant p16, RAR-β2, TIMP3, ERCC1, and BRCA1 Protein Expression and Promoter Methylation in Squamous Cell Carcinoma Accompanying Candida albicans-Induced Inflammation. PLoS One 2016; 11:e0159090. [PMID: 27410681 PMCID: PMC4943641 DOI: 10.1371/journal.pone.0159090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/27/2016] [Indexed: 02/06/2023] Open
Abstract
Hyperplastic candidiasis is characterized by thickening of the mucosal epithelia with Candida albicans infection with occasional progression to squamous cell carcinoma (SCC). C. albicans is a critical factor in tumor development; however, the oncogenic mechanism is unclear. We have previously produced an animal model for hyperplastic candidiasis in the rat forestomach. In the present study, we investigate whether impaired DNA methylation and associated protein expression of tumor suppressor and DNA repair genes are involved in the SCC carcinogenesis process using this hyperplastic candidiasis model. Promoter methylation and protein expression were analyzed by methylation specific PCR and immunohistochemical staining, respectively, of 5 areas in the forestomachs of alloxan-induced diabetic rats with hyperplastic candidiasis: normal squamous epithelia, squamous hyperplasia, squamous hyperplasia adjacent to SCC, squamous hyperplasia transitioning to SCC, and SCC. We observed nuclear p16 overexpression despite increases in p16 gene promoter methylation during the carcinogenic process. TIMP3 and RAR-β2 promoter methylation progressed until the precancerous stage but disappeared upon malignant transformation. In comparison, TIMP3 protein expression was suppressed during carcinogenesis and RAR-β2 expression was attenuated in the cytoplasm but enhanced in nuclei. ERCC1 and BRCA1 promoters were not methylated at any stage; however, their protein expression disappeared beginning at hyperplasia and nuclear protein re-expression in SCC was observed only for ERCC1. These results suggest that aberrant p16, RAR-β2, TIMP3, ERCC1, and BRCA1 expression might occur that is inconsistent with the respective gene promoter methylation status, and that this overexpression might serve to promote the inflammatory carcinogenesis caused by C. albicans infection.
Collapse
|
38
|
Shen Y, Lee YH, Panneerselvam J, Zhang J, Loo LWM, Fei P. Mutated Fanconi anemia pathway in non-Fanconi anemia cancers. Oncotarget 2016; 6:20396-403. [PMID: 26015400 PMCID: PMC4653013 DOI: 10.18632/oncotarget.4056] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 04/22/2015] [Indexed: 01/01/2023] Open
Abstract
An extremely high cancer incidence and the hypersensitivity to DNA crosslinking agents associated with Fanconi Anemia (FA) have marked it to be a unique genetic model system to study human cancer etiology and treatment, which has emerged an intense area of investigation in cancer research. However, there is limited information about the relationship between the mutated FA pathway and the cancer development or/and treatment in patients without FA. Here we analyzed the mutation rates of the seventeen FA genes in 68 DNA sequence datasets. We found that the FA pathway is frequently mutated across a variety of human cancers, with a rate mostly in the range of 15 to 35 % in human lung, brain, bladder, ovarian, breast cancers, or others. Furthermore, we found a statistically significant correlation (p < 0.05) between the mutated FA pathway and the development of human bladder cancer that we only further analyzed. Together, our study demonstrates a previously unknown fact that the mutated FA pathway frequently occurs during the development of non-FA human cancers, holding profound implications directly in advancing our understanding of human tumorigenesis as well as tumor sensitivity/resistance to crosslinking drug-relevant chemotherapy.
Collapse
Affiliation(s)
- Yihang Shen
- Program of Cancer Biology, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Yuan-Hao Lee
- Program of Cancer Biology, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Jayabal Panneerselvam
- Program of Cancer Biology, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Jun Zhang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Lenora W M Loo
- Program of Epidemiology, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Peiwen Fei
- Program of Cancer Biology, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| |
Collapse
|
39
|
Tian J, Han S. [Role of RRM1 in the Treatment and Prognosis of Advanced Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2015; 18:381-6. [PMID: 26104896 PMCID: PMC5999903 DOI: 10.3779/j.issn.1009-3419.2015.06.09] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lung cancer is one of the most common and highest mortality rates malignant tumors, further, 75%-80% is non-small cell lung cancer (NSCLC). For the majority of patients, lost the chance of radical operation or radiotherapy, chemotherapy is the main treatment. However, because of the diversities of tumor behavior and drug-resistant, the chemotherapy of advanced NSCLC is not optimistic. In recent years, with the application of molecular markers for individual chemotherapy, these patients have achieved prolong life and improved life quality. Individualized chemotherapy based on molecular markers to select the appropriate drug is the problem that needs to be solved. The paper gives a brief review on the role of ribonucleotide reductase subunit 1 (RRM1) in the treatment and prognosis of advanced NSCLC. Individualized chemotherapy by RRM1 can't become commonplace for advanced NSCLC and needs further research.
Collapse
Affiliation(s)
- Jiawei Tian
- Medical College, Southeast University, Nanjing 210009, China
| | - Shuhua Han
- Department of Respiration,
Zhongda Hospital Affiliated to Southeast University, Nanjing 210009, China
| |
Collapse
|
40
|
Massari F, Santoni M, Ciccarese C, Brunelli M, Conti A, Santini D, Montironi R, Cascinu S, Tortora G. Emerging concepts on drug resistance in bladder cancer: Implications for future strategies. Crit Rev Oncol Hematol 2015; 96:81-90. [PMID: 26022449 DOI: 10.1016/j.critrevonc.2015.05.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 02/16/2015] [Accepted: 05/05/2015] [Indexed: 02/07/2023] Open
Abstract
The combination chemotherapies with methotrexate plus vinblastine, doxorubicin and cisplatin (MVAC or CMV regimens) or gemcitabine plus cisplatin represent the standard as first-line therapy for patients with metastatic urothelial cancer. In Europe, vinflunine is an option for second-line therapy for patients progressed during first-line or perioperative platinum-containing regimen. Alternative regimens containing taxanes and/or gemcitabine may be valuated case by case. Furthermore, carboplatin should be considered in patients unfit for cisplatin both in the first and second-line setting. Based on these findings, a better comprehension of the mechanisms underlying the development of drug resistance in patients with bladder cancer will represent a major step forward in optimizing patients' outcome. This article reviews the current knowledge of the mechanisms and emerging strategies to overcome resistance in patients with advanced urothelial cancer.
Collapse
Affiliation(s)
- Francesco Massari
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Matteo Santoni
- Medical Oncology, AOU Ospedali Riuniti, Polytechnic University of the Marche Region, Ancona, Italy.
| | - Chiara Ciccarese
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Matteo Brunelli
- Department of Pathology and Diagnostic, A.O.U.I., University of Verona, Verona, Italy
| | - Alessandro Conti
- Department of Clinic and Specialistic Sciences-Urology, Polytechnic University of the Marche Region, Ancona, Italy
| | - Daniele Santini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, AOU Ospedali Riuniti, Ancona, Italy
| | - Stefano Cascinu
- Medical Oncology, AOU Ospedali Riuniti, Polytechnic University of the Marche Region, Ancona, Italy
| | - Giampaolo Tortora
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| |
Collapse
|
41
|
Prognostic value of ERCC1, RRM1, and TS proteins in patients with resected non-small cell lung cancer. Cancer Chemother Pharmacol 2015; 75:861-7. [DOI: 10.1007/s00280-015-2714-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 02/23/2015] [Indexed: 12/19/2022]
|
42
|
Cui S, Jiang L. [Current translational research status of ERCC1 expression of
non-small cell lung cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2014; 17:428-32. [PMID: 24854562 PMCID: PMC6000451 DOI: 10.3779/j.issn.1009-3419.2014.05.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Shaohua Cui
- Shanghai Chest Hospital, Shanghai JiaoTong University, Shanghai, 200030, China
| | - Liyan Jiang
- Shanghai Chest Hospital, Shanghai JiaoTong University, Shanghai, 200030, China
| |
Collapse
|