1
|
Kim HS, Kim Y, Lee HS. Clinicopathologic Characteristics of Trop Family Proteins (Trop-2 and EpCAM) in Gastric Carcinoma. J Gastric Cancer 2024; 24:391-405. [PMID: 39375055 PMCID: PMC11471318 DOI: 10.5230/jgc.2024.24.e32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 10/09/2024] Open
Abstract
PURPOSE Trop family proteins, including epithelial cell adhesion molecule (EpCAM) and Trop-2, have garnered attention as potential therapeutic and diagnostic targets for various malignancies. This study aimed to elucidate the clinicopathological significance of these proteins in gastric carcinoma (GC) and to reinforce their potential as biomarkers for patient stratification in targeted therapies. MATERIALS AND METHODS Immunohistochemical (IHC) analyses of EpCAM and Trop-2 were performed on GC and precancerous lesions, following rigorous orthogonal validation of the antibodies to ensure specificity and sensitivity. RESULTS Strong membranous staining (3+) for Trop-2 was observed in 49.3% of the GC cases, whereas EpCAM was strongly expressed in almost all cases (93.2%), indicating its widespread expression in GC. A high Trop-2 expression level, characterized by an elevated H-score, was significantly associated with intestinal type by Lauren classification, gastric mucin type, presence of lymph node metastasis, human epidermal growth factor receptor 2-positivity, and Epstein-Barr virus (EBV)-positivity. Patients with a high Trop-2 expression level exhibited poorer survival outcomes on univariate and multivariate analyses. High EpCAM expression levels were prevalent in differentiated histologic type, microsatellite instability-high, and EBV-negative cancer, and were correlated with high densities of CD3 and CD8 T cells and elevated combined positive score for programmed death-ligand 1. CONCLUSIONS These results highlight the differential expression of Trop-2 and EpCAM and their prognostic implications in GC. The use of meticulously validated antibodies ensured the reliability of our IHC data, thereby offering a robust foundation for future therapeutic strategies targeting Trop family members in GC.
Collapse
Affiliation(s)
- Hye Sung Kim
- Department of Pathology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Younghoon Kim
- Department of Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hye Seung Lee
- Department of Pathology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Pathology, Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
2
|
Ooki A, Osumi H, Yoshino K, Yamaguchi K. Potent therapeutic strategy in gastric cancer with microsatellite instability-high and/or deficient mismatch repair. Gastric Cancer 2024; 27:907-931. [PMID: 38922524 PMCID: PMC11335850 DOI: 10.1007/s10120-024-01523-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024]
Abstract
Gastric cancer (GC) is a common malignancy that presents challenges in patient care worldwide. The mismatch repair (MMR) system is a highly conserved DNA repair mechanism that protects genome integrity during replication. Deficient MMR (dMMR) results in an increased accumulation of genetic errors in microsatellite sequences, leading to the development of a microsatellite instability-high (MSI-H) phenotype. Most MSI-H/dMMR GCs arise sporadically, mainly due to MutL homolog 1 (MLH1) epigenetic silencing. Unlike microsatellite-stable (MSS)/proficient MMR (pMMR) GCs, MSI-H/dMMR GCs are relatively rare and represent a distinct subtype with genomic instability, a high somatic mutational burden, favorable immunogenicity, different responses to treatment, and prognosis. dMMR/MSI-H status is a robust predictive biomarker for treatment with immune checkpoint inhibitors (ICIs) due to high neoantigen load, prominent tumor-infiltrating lymphocytes, and programmed cell death ligand 1 (PD-L1) overexpression. However, a subset of MSI-H/dMMR GC patients does not benefit from immunotherapy, highlighting the need for further research into predictive biomarkers and resistance mechanisms. This review provides a comprehensive overview of the clinical, molecular, immunogenic, and therapeutic aspects of MSI-H/dMMR GC, with a focus on the impact of ICIs in immunotherapy and their potential as neoadjuvant therapies. Understanding the complexity and diversity of the molecular and immunological profiles of MSI-H/dMMR GC will drive the development of more effective therapeutic strategies and molecular targets for future precision medicine.
Collapse
Affiliation(s)
- Akira Ooki
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan.
| | - Hiroki Osumi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Koichiro Yoshino
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| |
Collapse
|
3
|
Sharma NK, Dwivedi P, Bhushan R, Maurya PK, Kumar A, Dakal TC. Engineering circular RNA for molecular and metabolic reprogramming. Funct Integr Genomics 2024; 24:117. [PMID: 38918231 DOI: 10.1007/s10142-024-01394-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
The role of messenger RNA (mRNA) in biological systems is extremely versatile. However, it's extremely short half-life poses a fundamental restriction on its application. Moreover, the translation efficiency of mRNA is also limited. On the contrary, circular RNAs, also known as circRNAs, are a common and stable form of RNA found in eukaryotic cells. These molecules are synthesized via back-splicing. Both synthetic circRNAs and certain endogenous circRNAs have the potential to encode proteins, hence suggesting the potential of circRNA as a gene expression machinery. Herein, we aim to summarize all engineering aspects that allow exogenous circular RNA (circRNA) to prolong the time that proteins are expressed from full-length RNA signals. This review presents a systematic engineering approach that have been devised to efficiently assemble circRNAs and evaluate several aspects that have an impact on protein production derived from. We have also reviewed how optimization of the key components of circRNAs, including the topology of vector, 5' and 3' untranslated sections, entrance site of the internal ribosome, and engineered aptamers could be efficiently impacting the translation machinery for molecular and metabolic reprogramming. Collectively, molecular and metabolic reprogramming present a novel way of regulating distinctive cellular features, for instance growth traits to neoplastic cells, and offer new possibilities for therapeutic inventions.
Collapse
Affiliation(s)
- Narendra Kumar Sharma
- Department of Bioscience and Biotechnology, Banasthali Vidyapith (Deemed University), P.O. Banasthali Vidyapith Distt. Tonk, Rajasthan, 304 022, India.
| | - Pragya Dwivedi
- Department of Bioscience and Biotechnology, Banasthali Vidyapith (Deemed University), P.O. Banasthali Vidyapith Distt. Tonk, Rajasthan, 304 022, India
| | - Ravi Bhushan
- Department of Zoology, M.S. College, Motihari, Bihar, India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendergarh, 123031, Haryana, India
| | - Abhishek Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, Karnataka, India
- Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Tikam Chand Dakal
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan, 313001, India.
| |
Collapse
|
4
|
Xie K, Li C, Wang M, Fu S, Cai Y. miR-135a-5p overexpression in peripheral blood-derived exosomes mediates vascular injury in type 2 diabetes patients. Front Endocrinol (Lausanne) 2023; 14:1035029. [PMID: 38027164 PMCID: PMC10657216 DOI: 10.3389/fendo.2023.1035029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/03/2023] [Indexed: 12/01/2023] Open
Abstract
Objective Diabetes pathology relies on exosomes (Exos). This study investigated how peripheral blood Exo-containing microRNAs (miRNAs) cause vascular injury in type 2 diabetes (T2D). Methods We removed DEmiRNA from T2D chip data from the GEO database. We isolated Exo from 15 peripheral blood samples from T2D patients and 15 healthy controls and measured Exo DEmiRNA levels. We employed the intersection of Geneards and mirWALK database queries to find T2D peripheral blood mRNA-related chip target genes. Next, we created a STRING database candidate target gene interaction network map. Next, we performed GO and KEGG enrichment analysis on T2D-related potential target genes using the ClusterProfiler R package. Finally, we selected T2D vascular damage core genes and signaling pathways using GSEA and PPI analysis. Finally, we used HEK293 cells for luciferase assays, co-cultured T2D peripheral blood-derived Exo with HVSMC, and detected HVSMC movement alterations. Results We found 12 T2D-related DEmiRNAs in GEO. T2D patient-derived peripheral blood Exo exhibited significantly up-regulated miR-135a-3p by qRT-PCR. Next, we projected miR-135a-3p's downstream target mRNA and screened 715 DEmRNAs to create a regulatory network diagram. DEmRNAs regulated biological enzyme activity and vascular endothelial cells according to GO function and KEGG pathway analysis. ErbB signaling pathway differences stood out. PPI network study demonstrated that DEmRNA ATM genes regulate the ErbB signaling pathway. The luciferase experiment validated miR-135a-3p and ATM target-binding. Co-culture of T2D patient-derived peripheral blood Exo with HVSMC cells increases HVSMC migration, ErbB2, Bcl-2, and VEGF production, and decreases BAX and ATM. However, miR-135a-3p can reverse the production of the aforesaid functional proteins and impair HVSMC cell movement. Conclusion T2D patient-derived peripheral blood Exo carrying miR-135a-3p enter HVSMC, possibly targeting and inhibiting ATM, activating the ErbB signaling pathway, promoting abnormal HVSMC proliferation and migration, and aggravating vascular damage.
Collapse
Affiliation(s)
| | | | | | | | - Ying Cai
- National Clinical Research Center for Geriatric Disorders, Department of Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
| |
Collapse
|
5
|
Lavacchi D, Fancelli S, Buttitta E, Vannini G, Guidolin A, Winchler C, Caliman E, Vannini A, Giommoni E, Brugia M, Cianchi F, Pillozzi S, Roviello G, Antonuzzo L. Perioperative Tailored Treatments for Gastric Cancer: Times Are Changing. Int J Mol Sci 2023; 24:4877. [PMID: 36902306 PMCID: PMC10003389 DOI: 10.3390/ijms24054877] [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: 01/26/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Resectable gastric or gastroesophageal (G/GEJ) cancer is a heterogeneous disease with no defined molecularly based treatment strategy. Unfortunately, nearly half of patients experience disease recurrence despite standard treatments (neoadjuvant and/or adjuvant chemotherapy/chemoradiotherapy and surgery). In this review, we summarize the evidence of potential tailored approaches in perioperative treatment of G/GEJ cancer, with a special focus on patients with human epidermal growth factor receptor-2(HER2)-positive and microsatellite instability-high (MSI-H) tumors. In patients with resectable MSI-H G/GEJ adenocarcinoma, the ongoing INFINITY trial introduces the concept of non-operative management for patients with complete clinical-pathological-molecular response, and this could be a novel and potential practice changing strategy. Other pathways involving vascular endothelial growth factor receptor (VEGFR), fibroblast growth factor receptor (FGFR), claudin18 isoform 2 (CLDN18.2), and DNA damage repair proteins are also described, with limited evidence until now. Although tailored therapy appears to be a promising strategy for resectable G/GEJ cancer, there are several methodological issues to address: inadequate sample size for pivotal trials, underestimation of subgroup effects, and choice of primary endpoint (tumor-centered vs. patient-centered endpoints). A better optimization of G/GEJ cancer treatment allows maximizing patient outcomes. In the perioperative phase, although caution is mandatory, times are changing and tailored strategies could introduce new treatment concepts. Overall, MSI-H G/GEJ cancer patients possess the characteristics to be the subgroup that could receive the most benefit from a tailored approach.
Collapse
Affiliation(s)
- Daniele Lavacchi
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Sara Fancelli
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Eleonora Buttitta
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Gianmarco Vannini
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Alessia Guidolin
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Costanza Winchler
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Enrico Caliman
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Agnese Vannini
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Elisa Giommoni
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Marco Brugia
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Fabio Cianchi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Unit of Digestive Surgery, Careggi University Hospital, 50134 Florence, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | | | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| |
Collapse
|
6
|
He AT, Liu J, Li F, Yang BB. Targeting circular RNAs as a therapeutic approach: current strategies and challenges. Signal Transduct Target Ther 2021; 6:185. [PMID: 34016945 PMCID: PMC8137869 DOI: 10.1038/s41392-021-00569-5] [Citation(s) in RCA: 265] [Impact Index Per Article: 88.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/28/2021] [Accepted: 03/16/2021] [Indexed: 02/04/2023] Open
Abstract
Significant progress has been made in circular RNA (circRNA) research in recent years. Increasing evidence suggests that circRNAs play important roles in many cellular processes, and their dysregulation is implicated in the pathogenesis of various diseases. CircRNAs are highly stable and usually expressed in a tissue- or cell type-specific manner. Therefore, they are currently being explored as potential therapeutic targets. Gain-of-function and loss-of-function approaches are typically performed using circRNA expression plasmids and RNA interference-based strategies, respectively. These strategies have limitations that can be mitigated using nanoparticle and exosome delivery systems. Furthermore, recent developments show that the cre-lox system can be used to knockdown circRNAs in a cell-specific manner. While still in the early stages of development, the CRISPR/Cas13 system has shown promise in knocking down circRNAs with high specificity and efficiency. In this review, we describe circRNA properties and functions and highlight their significance in disease. We summarize strategies that can be used to overexpress or knockdown circRNAs as a therapeutic approach. Lastly, we discuss major challenges and propose future directions for the development of circRNA-based therapeutics.
Collapse
Affiliation(s)
- Alina T. He
- grid.17063.330000 0001 2157 2938Sunnybrook Research Institute, Toronto, ON Canada
| | - Jinglei Liu
- Department of Bioinformatics, ATCGene Inc, Guangzhou, China
| | - Feiya Li
- grid.17063.330000 0001 2157 2938Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
| | - Burton B. Yang
- grid.17063.330000 0001 2157 2938Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
| |
Collapse
|
7
|
Kasi A, Al-Jumayli M, Park R, Baranda J, Sun W. Update on the Role of Poly (ADP-Ribose) Polymerase Inhibitors in the DNA Repair-Deficient Pancreatic Cancers: A Narrative Review. J Pancreat Cancer 2020; 6:107-115. [PMID: 33376937 PMCID: PMC7757687 DOI: 10.1089/pancan.2020.0010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose: Pancreatic ductal adenocarcinoma (PDAC) is the most common cancer found in the pancreas. It has a dismal prognosis and current therapeutic options, including surgical resection, provide only a temporary or limited response due to the development of treatment resistance. Methods: A narrative review of studies investigating poly (ADP-ribose) polymerase (PARP) pathway inhibitors in metastatic PDAC to highlight recent advances. Results: Mutations in BRCA genes confer a higher risk of PDAC, while germ line mutations are found in 4-7% of individuals harboring pancreatic cancer. Although solid tumors with defective DNA damage repair defect (DDR) genes such as BRCA show heightened sensitivity to platinum agents, tumors can exploit the PARP pathway as salvage pathways. Therefore, blocking this pathway will trigger cell death in vulnerable tumor cells with BRCA/DNA repair deficiency. Several drugs with inhibitory activity on the PARP pathway have been approved for breast and ovarian tumors harboring germ line or somatic BRCA mutations. Based on these results, the phase III POLO study showed a significant improvement in progression-free survival compared with placebo in BRCA mutant pancreatic tumors and highlighted the importance of germ line testing in everyone diagnosed with pancreatic cancer. In addition, expansion of the PARP inhibitor indication beyond BRCA mutations to other genes involved in DDR such as ATM and PALB2 merits attention. Conclusion: PARP inhibitors represent a safe and efficacious treatment for a subset of PDAC patients with BRCA mutations. Ongoing trials are evaluating PARP inhibitors in PDAC patients with non-BRCA DDR gene deficiencies as well as PARP inhibitors in combination with other agents, notably immune checkpoint inhibitors to expand the group of patients that derive benefit from this treatment.
Collapse
Affiliation(s)
- Anup Kasi
- Division of Medical Oncology, Department of Medicine, Kansas University Cancer Center, Kansas City, Kansas, USA
| | - Mohammed Al-Jumayli
- Division of Medical Oncology, Department of Medicine, Kansas University Cancer Center, Kansas City, Kansas, USA
| | - Robin Park
- Department of Medicine, MetroWest Medical Center/Tufts University School of Medicine, Framingham, Massachusetts, USA
| | - Joaquina Baranda
- Division of Medical Oncology, Department of Medicine, Kansas University Cancer Center, Kansas City, Kansas, USA
| | - Weijing Sun
- Division of Medical Oncology, Department of Medicine, Kansas University Cancer Center, Kansas City, Kansas, USA
| |
Collapse
|
8
|
Li Y, Shi P, Jiang D. Polymorphism rs1801516 (G > A) in the ATM gene is not associated with overall cancer risk: an updated meta-analysis. J Int Med Res 2020; 48:300060520937618. [PMID: 32674635 PMCID: PMC7370572 DOI: 10.1177/0300060520937618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/06/2020] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE The ataxia telangiectasia mutated (ATM) gene contains a functional single nucleotide polymorphism (SNP) rs1801516 (G > A) that may be associated with cancer risk. This meta-analysis aimed to interrogate the relationship between rs1801516 and cancer occurrence and disease etiology. METHODS We retrieved and identified the available case-control studies that met the inclusion criteria from the PubMed, Web of Science, and Embase databases. Odds ratio (OR) and 95% confidence intervals (CIs) were used to measure the association between rs1801516 and cancer risk. Additionally, we performed sensitivity, subgroup, and publication bias analyses. RESULTS After inclusion criteria were met, the meta-analysis included 29 studies, with 9,453 cancer patients (cases) and 14,646 controls. No association was found between rs1801516 and cancer risk (pooled OR = 0.911; 95% CI, 0.740-1.123). Concordantly, no association was found between rs1801516 and cancer risk after subgroup analysis by source of controls, cancer type, or ethnicity, which confirmed the finding of the dominant model that this SNP is not involved in the occurrence of cancer. CONCLUSIONS Through this meta-analysis, we found no association between rs1801516 and cancer occurrence as a risk factor. These data provide useful information for future case-control studies on cancer etiology.
Collapse
Affiliation(s)
- Yueting Li
- Department of Breast Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning Province, P.R. China
| | - Pengxu Shi
- Department of Bone Surgery, People’s Hospital of Liaoning Province, Shenyang, Liaoning Province, P.R. China
| | - Daqing Jiang
- Department of Breast Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning Province, P.R. China
| |
Collapse
|
9
|
Ji X, Mukherjee S, Landi MT, Bosse Y, Joubert P, Zhu D, Gorlov I, Xiao X, Han Y, Gorlova O, Hung RJ, Brhane Y, Carreras-Torres R, Christiani DC, Caporaso N, Johansson M, Liu G, Bojesen SE, Le Marchand L, Albanes D, Bickeböller H, Aldrich MC, Bush WS, Tardon A, Rennert G, Chen C, Byun J, Dragnev KH, Field JK, Kiemeney LF, Lazarus P, Zienolddiny S, Lam S, Schabath MB, Andrew AS, Bertazzi PA, Pesatori AC, Diao N, Su L, Song L, Zhang R, Leighl N, Johansen JS, Mellemgaard A, Saliba W, Haiman C, Wilkens L, Fernandez-Somoano A, Fernandez-Tardon G, Heijden EHFMVD, Kim JH, Davies MPA, Marcus MW, Brunnström H, Manjer J, Melander O, Muller DC, Overvad K, Trichopoulou A, Tumino R, Goodman GE, Cox A, Taylor F, Woll P, Wichmann E, Muley T, Risch A, Rosenberger A, Grankvist K, Johansson M, Shepherd F, Tsao MS, Arnold SM, Haura EB, Bolca C, Holcatova I, Janout V, Kontic M, Lissowska J, Mukeria A, Ognjanovic S, Orlowski TM, Scelo G, Swiatkowska B, Zaridze D, Bakke P, Skaug V, Butler LM, Offit K, Srinivasan P, Bandlamudi C, Hellmann MD, Solit DB, Robson ME, Rudin CM, Stadler ZK, Taylor BS, Berger MF, Houlston R, McLaughlin J, Stevens V, Nickle DC, Obeidat M, Timens W, Artigas MS, Shete S, Brenner H, Chanock S, Brennan P, McKay JD, Amos CI. Protein-altering germline mutations implicate novel genes related to lung cancer development. Nat Commun 2020; 11:2220. [PMID: 32393777 PMCID: PMC7214407 DOI: 10.1038/s41467-020-15905-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 03/25/2020] [Indexed: 01/24/2023] Open
Abstract
Few germline mutations are known to affect lung cancer risk. We performed analyses of rare variants from 39,146 individuals of European ancestry and investigated gene expression levels in 7,773 samples. We find a large-effect association with an ATM L2307F (rs56009889) mutation in adenocarcinoma for discovery (adjusted Odds Ratio = 8.82, P = 1.18 × 10-15) and replication (adjusted OR = 2.93, P = 2.22 × 10-3) that is more pronounced in females (adjusted OR = 6.81 and 3.19 and for discovery and replication). We observe an excess loss of heterozygosity in lung tumors among ATM L2307F allele carriers. L2307F is more frequent (4%) among Ashkenazi Jewish populations. We also observe an association in discovery (adjusted OR = 2.61, P = 7.98 × 10-22) and replication datasets (adjusted OR = 1.55, P = 0.06) with a loss-of-function mutation, Q4X (rs150665432) of an uncharacterized gene, KIAA0930. Our findings implicate germline genetic variants in ATM with lung cancer susceptibility and suggest KIAA0930 as a novel candidate gene for lung cancer risk.
Collapse
Affiliation(s)
- Xuemei Ji
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
| | - Semanti Mukherjee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yohan Bosse
- Institut universitaire de cardiologie et de pneumologie de Québec, Department of Molecular Medicine, Laval University, Québec, Canada
| | - Philippe Joubert
- Institut universitaire de cardiologie et de pneumologie de Québec, Department of Molecular Medicine, Laval University, Québec, Canada
| | - Dakai Zhu
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Ivan Gorlov
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Xiangjun Xiao
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Younghun Han
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Olga Gorlova
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System and University of Toronto, Toronto, Canada
| | - Yonathan Brhane
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System and University of Toronto, Toronto, Canada
| | | | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital/Harvard, Boston, MA, USA
| | - Neil Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mattias Johansson
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Geoffrey Liu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System and University of Toronto, Toronto, Canada
| | - Stig E Bojesen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Demetrios Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Heike Bickeböller
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| | - Melinda C Aldrich
- Department of Thoracic Surgery, Division of Epidemiology, Vanderbilt University Medical Center, Göttingen, Germany
| | - William S Bush
- Department of Epidemiology and Biostatistics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Adonina Tardon
- IUOPA. University of Oviedo and CIBERESP, Faculty of Medicine, Campus del Cristo s/n, Oviedo, Spain
| | - Gad Rennert
- Clalit National Cancer Control Center at Carmel Medical Center and Technion Faculty of Medicine, Haifa, Israel
| | - Chu Chen
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jinyoung Byun
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Konstantin H Dragnev
- The Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - John K Field
- Roy Castle lung Cancer Research Programme, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Lambertus Fa Kiemeney
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, WA, USA
| | | | - Stephen Lam
- British Columbia Cancer Agency, Vancouver, Canada
| | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Angeline S Andrew
- Department of Epidemiology, Geisel School of Medicine, Hanover, NH, USA
| | - Pier A Bertazzi
- Department of Preventive Medicine, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Angela C Pesatori
- Department of Preventive Medicine, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Nancy Diao
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Li Su
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lei Song
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ruyang Zhang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Natasha Leighl
- University Health Network- The Princess Margaret Cancer Centre, Toronto, CA, USA
| | - Jakob S Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Anders Mellemgaard
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Walid Saliba
- Clalit National Cancer Control Center at Carmel Medical Center and Technion Faculty of Medicine, Haifa, Israel
| | - Christopher Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Lynne Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Ana Fernandez-Somoano
- IUOPA. University of Oviedo and CIBERESP, Faculty of Medicine, Campus del Cristo s/n, Oviedo, Spain
| | | | | | - Jin Hee Kim
- Department of Integrative Bioscience & Biotechnology, Sejong University, Gwangjin-gu, Seoul, Republic of Korea
| | - Michael P A Davies
- Roy Castle lung Cancer Research Programme, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Michael W Marcus
- Roy Castle lung Cancer Research Programme, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | | | - Jonas Manjer
- Faculty of Medicine, Lund University, Lund, Sweden
| | | | - David C Muller
- School of Public Health, St Mary's Campus, Imperial College London, London, UK
| | - Kim Overvad
- Faculty of Medicine, Lund University, Lund, Sweden
| | | | - Rosario Tumino
- Cancer Registry and Histopathology Department, "Civic - M.P. Arezzo" Hospital, Asp Ragusa, Italy
| | - Gary E Goodman
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Swedish Medical Group, Seattle, WA, USA
| | - Angela Cox
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Fiona Taylor
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Penella Woll
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Erich Wichmann
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Muley
- Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), Heidelberg, Germany
| | - Angela Risch
- University of Salzburg and Cancer Cluster Salzburg, Salzburg, Austria
| | - Albert Rosenberger
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| | - Kjell Grankvist
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | | | | | | | - Susanne M Arnold
- University of Kentucky, Markey Cancer Center, Lexington, KY, USA
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ciprian Bolca
- Institute of Pneumology "Marius Nasta", Bucharest, Romania
| | - Ivana Holcatova
- Charles University, 1st Faculty of Medicine, Prague, Czech Republic
| | - Vladimir Janout
- Faculty of Health Sciences, Palacky University, Olomouc, Czech Republic
| | - Milica Kontic
- Clinical Center of Serbia, Clinic for Pulmonology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Prevention, M. Sklodowska-Curie Institute - Oncology Center, Warsaw, Poland
| | - Anush Mukeria
- Department of Epidemiology and Prevention, Russian N.N.Blokhin Cancer Research Centre, Moscow, Russian Federation
| | - Simona Ognjanovic
- International Organization for Cancer Prevention and Research, Belgrade, Serbia
| | - Tadeusz M Orlowski
- Department of Surgery, National Tuberculosis and Lung Diseases Research Institute, Warsaw, Poland
| | - Ghislaine Scelo
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Beata Swiatkowska
- Nofer Institute of Occupational Medicine, Department of Environmental Epidemiology, Lodz, Poland
| | - David Zaridze
- Department of Epidemiology and Prevention, Russian N.N.Blokhin Cancer Research Centre, Moscow, Russian Federation
| | - Per Bakke
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Vidar Skaug
- National Institute of Occupational Health, Oslo, Norway
| | | | - Kenneth Offit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Preethi Srinivasan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Chaitanya Bandlamudi
- Marie-Josée and Henry R. KravisCenter for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Matthew D Hellmann
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David B Solit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. KravisCenter for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Mark E Robson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Barry S Taylor
- Marie-Josée and Henry R. KravisCenter for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Michael F Berger
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
- Marie-Josée and Henry R. KravisCenter for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | | | | | | | - David C Nickle
- Merck Research Laboratories, Genetics and Pharmacogenomics, Boston, MA, USA
| | - Ma'en Obeidat
- The University of British Columbia Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, GRIAC research institute, Groningen, The Netherlands
| | - María Soler Artigas
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, LE1 7RH, UK
- National Institute for Health Research (NIHR) Leicester Respiratory Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| | - Sanjay Shete
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Paul Brennan
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - James D McKay
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Christopher I Amos
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, 7200 Cambridge St., 7th Floor, Houston, TX, 77030, USA.
| |
Collapse
|
10
|
Qu X, Zhao L, Zhang R, Wei Q, Wang M. Differential microRNA expression profiles associated with microsatellite status reveal possible epigenetic regulation of microsatellite instability in gastric adenocarcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:484. [PMID: 32395528 PMCID: PMC7210178 DOI: 10.21037/atm.2020.03.54] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background Although microsatellite instability (MSI) is a powerful predictive biomarker for the efficacy of immunotherapy, the mechanism of MSI in sporadic gastrointestinal cancer is not fully understood. However, epigenetics, particularly microRNAs, has been suggested as one of the main regulators that contribute to the MSI formation. Methods We used microRNA expression data of 386 gastric adenocarcinoma samples from The Cancer Genome Atlas (TCGA) database to identify differential microRNA expression profiles by different MSI status. We also obtained putative common target genes of the top differential microRNAs with miRanda online tools, and we analyzed these data by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway enrichment (KEGG). Results We found that 56 and 67 gastric adenocarcinoma samples were positive for low and high MSI, respectively, and that a high MSI status was associated with age, sex and subregion (P=0.049, 0.014 and 0.007, respectively). In the 67 samples with a high MSI status, expression levels of 14 microRNAs were upregulated but five microRNAs were downregulated as assessed by the fold change (FC), compared with that of the 56 samples with a low MSI status (P<0.05, |FC| >2). Further analysis suggested that the expression of miR-210-3p, miR-582-3p, miR-30a-3p and miR-105-5p predicted a high MSI status (P=4.93×10−10, 5.63×10−10, 3.23×10−9 and 7.64×10−4, respectively). Regulation of the transcription pathways ranked the top of lists from both GO and KEGG analyses, and these microRNAs might regulate DNA damage-repair genes that were also associated with a high MSI status. Conclusions MiR-30a-3p and miR-105-5p are potential biomarkers for the MSI-H gastric adenocarcinoma, possibly by altering expression of DNA damage-repair genes.
Collapse
Affiliation(s)
- Xiaofei Qu
- Cancer institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Liqin Zhao
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Ruoxin Zhang
- Cancer institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Epidemiology and Biostatistics, Fudan University School of Public Health, Shanghai 200032, China
| | - Qingyi Wei
- Cancer institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Mengyun Wang
- Cancer institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
11
|
Tsai CY, Lin TA, Huang SC, Hsu JT, Yeh CN, Chen TC, Chiu CT, Chen JS, Yeh TS. Is Adjuvant Chemotherapy Necessary for Patients with Deficient Mismatch Repair Gastric Cancer?-Autophagy Inhibition Matches the Mismatched. Oncologist 2020; 25:e1021-e1030. [PMID: 32058649 DOI: 10.1634/theoncologist.2019-0419] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/20/2019] [Indexed: 12/15/2022] Open
Abstract
PURPOSE The use of microsatellite instability (MSI) and mismatch repair (MMR) as predictive biomarkers for fluorouracil-based adjuvant chemotherapy in colorectal cancer has been a paradigm shift. However, whether this applies to gastric cancer is questionable. Furthermore, we herein investigated whether and how autophagy plays a role in MSI-relevant chemoresistance. MATERIALS AND METHODS A total of 929 patients with deficient MMR (dMMR) and proficient MMR (pMMR) gastric cancers who underwent curative-intent gastrectomy were enrolled. We compared clinicopathological variables and survival among dMMR and pMMR cohorts and tested the responses of MSI-high and microsatellite stable (MSS) gastric cancer cell lines to 5-fluorouracil (5-FU) with or without chloroquine, an autophagy inhibitor. RESULTS We identified an 8.9% prevalence of dMMR cases (83 out of 929) in our cohort. This was associated with old age, tumor site at the distal stomach, an intestinal phenotype, fewer nodal metastasis, and early pathological stages. MMR was an independent prognostic factor after multivariate adjustment. Overall survival (OS) of dMMR patients was better than that of the pMMR patients but was only applicable to stage III patients. There was no difference in OS between dMMR patients treated with or without adjuvant chemotherapy, although the latter showed more medical morbidities. The MSI-high gastric cancer cell lines, versus the MSS counterparts, displayed increased resistance to 5-FU and increased autophagy. Interestingly, autophagy inhibition abrogated the chemoresistance. CONCLUSION Our data show that fluorouracil-based adjuvant chemotherapy does not work for dMMR cases, if not worse. Autophagy inhibition and/or immune checkpoint inhibition might be promising alternative strategies for gastric cancer treatment. IMPLICATIONS FOR PRACTICE The use of microsatellite instability (MSI) and mismatch repair (MMR) as predictive biomarkers for adjuvant chemotherapy in colorectal cancer has caused a paradigm shift in cancer therapy, although its implications in gastric cancer are still questionable. The data obtained in the current study indicate that MSI-MMR is an independent prognostic factor for gastric cancer. Standard fluorouracil-based adjuvant chemotherapy did not work for deficient MMR cases, and was likely worse. Instead, strategies like autophagy inhibition and/or immune checkpoint inhibition should be taken into consideration in the future.
Collapse
Affiliation(s)
- Chun-Yi Tsai
- Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Tien-An Lin
- Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Shih-Chiang Huang
- Department of Pathology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Jun-Te Hsu
- Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chun-Nan Yeh
- Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Tse-Ching Chen
- Department of Pathology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Cheng-Tang Chiu
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Jen-Shi Chen
- Medical Oncology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ta-Sen Yeh
- Department of Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| |
Collapse
|
12
|
Ding X, Cheng J, Pang Q, Wei X, Zhang X, Wang P, Yuan Z, Qian D. BIBR1532, a Selective Telomerase Inhibitor, Enhances Radiosensitivity of Non-Small Cell Lung Cancer Through Increasing Telomere Dysfunction and ATM/CHK1 Inhibition. Int J Radiat Oncol Biol Phys 2019; 105:861-874. [PMID: 31419512 DOI: 10.1016/j.ijrobp.2019.08.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 07/30/2019] [Accepted: 08/06/2019] [Indexed: 01/06/2023]
Abstract
PURPOSE Telomerase is reactivated in non-small cell lung cancer (NSCLC), and it increases cell resistance to irradiation through protecting damaged telomeres and enhancing DNA damage repair. We investigated the radiosensitizing effect of BIBR1532, a highly selective telomerase inhibitor, and its corresponding mechanism in NSCLC. METHODS AND MATERIALS Cell proliferation, telomerase activity, and telomere dysfunction-induced foci were measured with CCK-8 assay, real-time fluorescent quantitative polymerase chain reaction, and immunofluorescence. The effect of BIBR1532 on the response of NSCLC cells to radiation was analyzed using clonogenic survival and xenograft tumor assays. Cell death and cell senescence induced by BIBR1532 or ionizing radiation (IR), or both, were detected with western blotting, flow cytometry, and senescence-association β-galactosidase staining assay. RESULTS We observed dose-dependent direct cytotoxicity of BIBR1532 at relatively high concentrations in NSCLC cells. Low concentrations of BIBR1532 did not appear toxic to NSCLC cells; however, they substantially increased the therapeutic efficacy of IR in vitro by enhancing IR-induced apoptosis, senescence, and mitotic catastrophe. Moreover, in a mouse xenograft model, BIBR1532 treatment synergized with IR at nontoxic dose levels promoted the antitumor efficacy of IR without toxicity to hematologic and internal organs. Mechanistically, lower concentrations of BIBR1532 effectively inhibited telomerase activity and increased IR-induced telomere dysfunction, resulting in disruption of chromosomal stability and inhibition of the ATM/CHK1 (ataxia-telangiectasia-mutated/Checkpoint kinase 1) pathway, which impaired DNA damage repair. CONCLUSIONS Our findings demonstrate that disturbances in telomerase function by nontoxic dose levels of BIBR1532 effectively enhance the radiosensitivity of NSCLC cells. This finding provides a rationale for the clinical assessment of BIBR1532 as a radiosensitizer.
Collapse
Affiliation(s)
- Xiaofeng Ding
- Department of Radiation Oncology and Cyberknife Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jingjing Cheng
- Department of Radiation Oncology and Cyberknife Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China; Department of Radiation Oncology, First Affiliated Hospital of USTC(University of Science and Technology of China), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Qingsong Pang
- Department of Radiation Oncology and Cyberknife Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiaoying Wei
- Department of Radiation Oncology and Cyberknife Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China; Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Ximei Zhang
- Department of Radiation Oncology and Cyberknife Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ping Wang
- Department of Radiation Oncology and Cyberknife Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhiyong Yuan
- Department of Radiation Oncology and Cyberknife Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| | - Dong Qian
- Department of Radiation Oncology and Cyberknife Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China; Department of Radiation Oncology, First Affiliated Hospital of USTC(University of Science and Technology of China), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
13
|
Sokolova O, Naumann M. Crosstalk Between DNA Damage and Inflammation in the Multiple Steps of Gastric Carcinogenesis. Curr Top Microbiol Immunol 2019; 421:107-137. [PMID: 31123887 DOI: 10.1007/978-3-030-15138-6_5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Over the last years, intensive investigations in molecular biology and cell physiology extended tremendously the knowledge about the association of inflammation and cancer. In frame of this paradigm, the human pathogen Helicobacter pylori triggers gastritis and gastric ulcer disease, and contributes to the development of gastric cancer. Mechanisms, by which the bacteria-induced inflammation in gastric mucosa leads to intestinal metaplasia and carcinoma, are represented in this review. An altered cell-signaling response and increased production of free radicals by epithelial and immune cells account for the accumulation of DNA damage in gastric mucosa, if infection stays untreated. Host genetics and environmental factors, especially diet, can accelerate the process, which offers the opportunity of intervention based on a balanced nutrition. It is supposed that inflammation might influence stem- or progenitor cells in gastric tissue predisposing for metaplasia or tumor relapse. Herein, DNA is strongly mutated and labile, which restricts therapy options. Thus, the understanding of the mechanisms that underlie gastric carcinogenesis will be of preeminent importance for the development of strategies for screening and early detection. As most gastric cancer patients face late-stage disease with a poor overall survival, the development of multi-targeted therapeutic intervention strategies is a major challenge for the future.
Collapse
Affiliation(s)
- Olga Sokolova
- Institute of Experimental Internal Medicine, Otto von Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| |
Collapse
|
14
|
Wu Q, Li P, Wu M, Liu Q. Deregulation of Circular RNAs in Cancer From the Perspectives of Aberrant Biogenesis, Transport and Removal. Front Genet 2019; 10:16. [PMID: 30774645 PMCID: PMC6367250 DOI: 10.3389/fgene.2019.00016] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/14/2019] [Indexed: 12/28/2022] Open
Abstract
CircRNAs (circular RNAs) are a class of RNAs generated from circularization with multiple novel functions. Recent studies have revealed the aberrant expression and aberrant functions of circRNAs in various tumors; thus, circRNAs have been recognized as promising cancer biomarkers. However, the underlying mechanisms behind their aberrant expression and functions remain unclear. In this review, we discuss at length the cancer-specific deregulation of circRNAs and the potential underlying aberrant events in circRNA biogenesis, localization and removal in cancer cells.
Collapse
Affiliation(s)
- Qiongqiong Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Peiyao Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Minghua Wu
- Key Laboratory of Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Qiang Liu
- Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
15
|
Zhao ZL, Xia L, Zhao C, Yao J. ATM rs189037 (G > A) polymorphism increased the risk of cancer: an updated meta-analysis. BMC MEDICAL GENETICS 2019; 20:28. [PMID: 30709340 PMCID: PMC6359756 DOI: 10.1186/s12881-019-0760-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 01/25/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Rs189037 (G > A) is a functional single nucleotide polymorphism (SNP) in the Ataxia-telangiectasia mutated (ATM) gene that may be associated with the risk of cancer. We performed a meta-analysis to determine whether rs189037 polymorphism influences the occurrence of cancer and examined the relationship between this SNP and the etiology of cancer. METHODS Case-control studies were retrieved from literature databases in accordance with established inclusion criteria. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to evaluate the strength of the association between rs189037 and cancer. Subgroup analysis and sensitivity analysis also were performed. RESULTS After inclusion criteria were met, fifteen studies-comprising 8660 patients with cancer (cases) and 9259 controls-were included in this meta-analysis. Summary results indicated that an association was found between rs189037 and cancer risk. In the dominant model, the pooled OR using a random effects model was 1.207 (95% CI, 1.090-1.337; P < 0.001). The A allele of rs189037 increased the risk of lung cancer, breast cancer, and oral cancer. Results of subgroup analysis by ethnicity indicated that the SNP was associated with the risk of cancer among East Asian and Latino, but not Caucasian. CONCLUSIONS Results of this meta-analysis suggest that rs189037 is associated with the occurrence of lung cancer, breast cancer, and oral cancer as the risk factor. These data provide possible avenues for future case-control studies related to cancer.
Collapse
Affiliation(s)
- Zhi-liang Zhao
- Hospital Office, Chengdu First People’s Hospital, Chengdu, 610000 Sichuan Province People’s Republic of China
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA USA
| | - Lu Xia
- Department of Rehabilitation, Chengdu First People’s Hospital, Chengdu, 610000 Sichuan Province People’s Republic of China
| | - Cong Zhao
- Department of Gastroenterology, Chengdu First People’s Hospital, Chengdu, 610000 Sichuan Province People’s Republic of China
| | - Jun Yao
- School of Forensic Medicine, China Medical University, No. 77 Puhe Road, Shenbei New District, Shenyang, 110122 China
| |
Collapse
|
16
|
Han M, Ma L, Qu Y, Tang Y. Decreased expression of the ATM gene linked to poor prognosis for gastric cancer of different nationalities in Xinjiang. Pathol Res Pract 2017. [DOI: 10.1016/j.prp.2017.05.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Ward TA, McHugh PJ, Durant ST. Small molecule inhibitors uncover synthetic genetic interactions of human flap endonuclease 1 (FEN1) with DNA damage response genes. PLoS One 2017. [PMID: 28628639 PMCID: PMC5476263 DOI: 10.1371/journal.pone.0179278] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Flap endonuclease 1 (FEN1) is a structure selective endonuclease required for proficient DNA replication and the repair of DNA damage. Cellularly active inhibitors of this enzyme have previously been shown to induce a DNA damage response and, ultimately, cell death. High-throughput screens of human cancer cell-lines identify colorectal and gastric cell-lines with microsatellite instability (MSI) as enriched for cellular sensitivity to N-hydroxyurea series inhibitors of FEN1, but not the PARP inhibitor olaparib or other inhibitors of the DNA damage response. This sensitivity is due to a synthetic lethal interaction between FEN1 and MRE11A, which is often mutated in MSI cancers through instabilities at a poly(T) microsatellite repeat. Disruption of ATM is similarly synthetic lethal with FEN1 inhibition, suggesting that disruption of FEN1 function leads to the accumulation of DNA double-strand breaks. These are likely a result of the accumulation of aberrant replication forks, that accumulate as a consequence of a failure in Okazaki fragment maturation, as inhibition of FEN1 is toxic in cells disrupted for the Fanconi anemia pathway and post-replication repair. Furthermore, RAD51 foci accumulate as a consequence of FEN1 inhibition and the toxicity of FEN1 inhibitors increases in cells disrupted for the homologous recombination pathway, suggesting a role for homologous recombination in the resolution of damage induced by FEN1 inhibition. Finally, FEN1 appears to be required for the repair of damage induced by olaparib and cisplatin within the Fanconi anemia pathway, and may play a role in the repair of damage associated with its own disruption.
Collapse
Affiliation(s)
- Thomas A. Ward
- AstraZeneca, Innovative Medicines and Early Development Biotech Unit, Oncology Bioscience, Alderley Park, Macclesfield, Cheshire, United Kingdom
- Department of Oncology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- * E-mail: (TAW); (STD)
| | - Peter J. McHugh
- Department of Oncology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Stephen T. Durant
- AstraZeneca, Innovative Medicines and Early Development Biotech Unit, Oncology Bioscience, Alderley Park, Macclesfield, Cheshire, United Kingdom
- AstraZeneca, Innovative Medicines and Early Development Biotech Unit, Oncology Bioscience, Little Chesterford, Cambridge, United Kingdom
- * E-mail: (TAW); (STD)
| |
Collapse
|
18
|
Wang C, Jette N, Moussienko D, Bebb DG, Lees-Miller SP. ATM-Deficient Colorectal Cancer Cells Are Sensitive to the PARP Inhibitor Olaparib. Transl Oncol 2017; 10:190-196. [PMID: 28182994 PMCID: PMC5299208 DOI: 10.1016/j.tranon.2017.01.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 01/18/2017] [Accepted: 01/18/2017] [Indexed: 02/06/2023] Open
Abstract
The ataxia telangiectasia mutated (ATM) protein kinase plays a central role in the cellular response to DNA damage. Loss or inactivation of both copies of the ATM gene (ATM) leads to ataxia telangiectasia, a devastating childhood condition characterized by neurodegeneration, immune deficiencies, and cancer predisposition. ATM is also absent in approximately 40% of mantle cell lymphomas (MCLs), and we previously showed that MCL cell lines with loss of ATM are sensitive to poly-ADP ribose polymerase (PARP) inhibitors. Next-generation sequencing of patient tumors has revealed that ATM is altered in many human cancers including colorectal, lung, prostate, and breast. Here, we show that the colorectal cancer cell line SK-CO-1 lacks detectable ATM protein expression and is sensitive to the PARP inhibitor olaparib. Similarly, HCT116 colorectal cancer cells with shRNA depletion of ATM are sensitive to olaparib, and depletion of p53 enhances this sensitivity. Moreover, HCT116 cells are sensitive to olaparib in combination with the ATM inhibitor KU55933, and sensitivity is enhanced by deletion of p53. Together our studies suggest that PARP inhibitors may have potential for treating colorectal cancer with ATM dysfunction and/or colorectal cancer with mutation of p53 when combined with an ATM kinase inhibitor.
Collapse
Affiliation(s)
- Chen Wang
- Department of Biochemistry & Molecular Biology, Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Nicholas Jette
- Department of Biochemistry & Molecular Biology, Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Daniel Moussienko
- Department of Biochemistry & Molecular Biology, Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - D Gwyn Bebb
- Department on Oncology, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Susan P Lees-Miller
- Department of Biochemistry & Molecular Biology, Robson DNA Science Centre, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada; Department on Oncology, Tom Baker Cancer Centre, Calgary, Alberta, Canada.
| |
Collapse
|
19
|
Targeted Therapies for Advanced Oesophagogastric Cancer: Recent Progress and Future Directions. Drugs 2016; 76:13-26. [PMID: 26620367 DOI: 10.1007/s40265-015-0510-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The genomic landscape of oesophagogastric (OG) cancer is highly complex. The recent elucidation of some of the pathways involved has suggested a number of novel targets for therapy. This therapy is urgently required as with conventional chemotherapy regimens patients with advanced OG cancer still have a median overall survival of under a year. This review outlines the rationale for the current treatment of OG cancer with chemotherapy and describes both previously conducted and ongoing clinical trials of novel agents in this area. The targets and associated treatments discussed include HER-2, EGFR, VEGF, c-Met, FGFR-2, PI3K, mTOR andIGF-1. To date only two targeted treatments, trastuzumab and ramucirumab, have become part of the treatment paradigm for OG cancer, partly due to difficulties in defining predictive biomarkers in this disease. However, there are a number of promising drugs in the pipeline and this article seeks to describe these and other potential novel approaches including targeting DNA repair deficiencies and the immune system.
Collapse
|
20
|
Young K, Starling N, Cunningham D. Targeting deficient DNA damage repair in gastric cancer. Expert Opin Pharmacother 2016; 17:1757-66. [PMID: 27488684 DOI: 10.1080/14656566.2016.1217992] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Over recent years our understanding of DNA damage repair has evolved leading to an expansion of therapies attempting to exploit DNA damage repair deficiencies across multiple solid tumours. Gastric cancer has been identified as a tumour where a subgroup of patients demonstrates deficiencies in the homologous recombination pathway providing a potential novel treatment approach for this poor prognosis disease. AREA COVERED This review provides an overview of DNA damage repair and how this has been targeted to date in other tumour types exploiting the concept of synthetic lethality. This is followed by a discussion of how deficiencies in homologous recombination may be identified across tumour types and on recent progress in targeting DNA repair deficiencies in gastric cancer. EXPERT OPINION Gastric cancer remains a difficult malignancy to treat and the possibility of targeting deficient DNA repair in a subgroup of patients is an exciting prospect. Future combinations with immunotherapy and radiotherapy are appealing and appear to have a sound biological rationale. However, much work remains to be done to understand the significance of the genetic and epigenetic alterations involved, to elucidate the optimum predictive signatures or biomarkers and to consider means of overcoming treatment resistance.
Collapse
Affiliation(s)
- Kate Young
- a Department of Medicine , The Royal Marsden NHS Foundation Trust, GI and Lymphoma Unit , Sutton , UK
| | - Naureen Starling
- a Department of Medicine , The Royal Marsden NHS Foundation Trust, GI and Lymphoma Unit , Sutton , UK
| | - David Cunningham
- a Department of Medicine , The Royal Marsden NHS Foundation Trust, GI and Lymphoma Unit , Sutton , UK
| |
Collapse
|
21
|
Suh KJ, Ryu HS, Lee KH, Kim H, Min A, Kim TY, Yang Y, Moon HG, Han SW, Oh DY, Han W, Park IA, Noh DY, Im SA. Loss of ataxia-telangiectasia-mutated protein expression correlates with poor prognosis but benefits from anthracycline-containing adjuvant chemotherapy in breast cancer. Breast Cancer Res Treat 2016; 158:233-41. [DOI: 10.1007/s10549-016-3869-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 06/14/2016] [Indexed: 10/21/2022]
|
22
|
Mahon S. Management of Individuals With a Mutation in the Ataxia Telangiectasia Mutated Gene. Oncol Nurs Forum 2016; 43:114-7. [DOI: 10.1188/16.onf.114-117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
23
|
Zhang S, Huo X, Li Z, Li X, Tang W, Li C, Guo M, Du X, Chen Z. Microsatellite instability detected in tumor-related genes in C57BL/6J mice with thymic lymphoma induced by N-methyl-N-nitrosourea. Mutat Res 2015; 782:7-16. [PMID: 26498208 DOI: 10.1016/j.mrfmmm.2015.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 09/19/2015] [Accepted: 10/07/2015] [Indexed: 11/19/2022]
Abstract
Microsatellite instability (MSI) has been observed within tumors and found to be closely associated with the degree of malignancy and prognosis in tumors. However, whether MSI in tumor-related genes can be induced by a chemical and whether a connection exists between MSI and tumors remain unclear. In the present study, we detected MSI in the tissues of N-methyl-N-nitrosourea (MNU) treated mice by targeting to 5, 29, 30 microsatellite loci in 3 mismatch repair (MMR) genes, 1 DNA repair gene, and 5 tumor suppressor (TS) genes, respectively. Among 26 mice survived in the MNU-group, 18 (69%) mice presented thymic lymphomas. Moreover, 61% (11/18) of the tumors metastasized to the other organs, including the liver, spleen, and kidney. We examined 104 tissues from MNU-treated mice using the 64 loci, and found 8 MSI events involved 4 loci in 4 tissues types. The MSI incidence in MMR, DNA repair, and TS genes was 67% (2/3), 0% (0/1) and 40% (2/5), respectively. MSI occurrence in tumor and non-tumor tissues was 5.6% (1/18) and 0% (0/8) and that in metastasis and non-metastasis tissues was 7.1% (1/14) and 9.4% (6/64), showing no significant difference. MSI loci in intronic regions of Atm, Msh6 and p21 and MSI in the 3'UTR of Pms2 were detected in MNU-treated mice. Specifically, we found a loss of heterozygosity in intron of Atm (ATM-8) in one metastasis mouse. Four similar events occurred in p21 gene intron (P21-1) of another non-metastasis mouse. Another MSI was a heterozygous mutation existed in an Msh6 allele (MSH6-2) in metastasis mouse. We also found a homozygous 2-bp insertion in the 3'UTR of Pms2 in two non-metastasis mice. These results imply that MNU can induce MSI in MMR and TS genes in C57BL/6J mice. MSI frequency does not seem to be associated with tumorigenesis or metastasis.
Collapse
Affiliation(s)
- Shuangyue Zhang
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Xueyun Huo
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Zhenkun Li
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Xiaohong Li
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Wang Tang
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Changlong Li
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Meng Guo
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China
| | - Xiaoyan Du
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China.
| | - Zhenwen Chen
- School of Basic Medical Science, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing 100069, China.
| |
Collapse
|
24
|
Burtness B. Complexity in the Gastric Cancer Genome and a Biomarker-Driven Trial of Poly (ADP-Ribose) Polymerase Inhibition in Gastric Cancer. J Clin Oncol 2015; 33:3845-6. [PMID: 26282638 DOI: 10.1200/jco.2015.62.8487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
25
|
Huo X, Zhang S, Li Z, Gao J, Wang C, Li C, Guo M, Du X, Chen Z. Analysis of the relationship between microsatellite instability and thymic lymphoma induced by N-methyl-N-nitrosourea in C57BL/6J mice. Mutat Res 2014; 771:21-8. [PMID: 25771976 DOI: 10.1016/j.mrfmmm.2014.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 11/04/2014] [Accepted: 11/21/2014] [Indexed: 01/21/2023]
Abstract
Microsatellite instability (MSI) has been found to be closely associated with many types of human tumors and often shows strong correlations with specific tumor features. However, the relationship between MSI and tumors are still unclear. The aim of the present study is to explore the relationships between MSI, tumor formation under the mutagenic effects of N-methyl-N-nitrosourea (MNU). Mice were administered with either MNU (90 mg/kg) or PBS and DMSO (control) at the beginning of the 1st week of the experiment. Of the 31 mice that survived the entire experimental time course, 19 (61.3%) mice developed thymic lymphomas. In addition, 52.6% (10/19) of the tumors had metastasized to the liver. We detected MSI in MNU-treated mice using a panel of 42 mutation-sensitive loci. Nineteen loci (45.2%) in six organs showed 70 MSI events. Locus D8Mit14 showed enhanced MSI compared with the other examined loci. MSI frequency in thymus was higher than in other organs. Interestingly, there was no significant difference observed between the metastatic and non-metastatic livers. The MSI frequency (4.6%, 23/(42×12)) in the MNU-treated thymus that had never developed tumor was significantly higher than this in the thymus that had developed lymphoma (0.5%, 4/(42×19)) (p<0.0001). These results indicate that, although thymic tumorigenesis is associated with MSI, it occurs with higher frequency in these that have not developed tumors upon the MNU-treatment. Our study provides additional insights into the relationship between MSI occurrence and tumorigenesis.
Collapse
Affiliation(s)
- Xueyun Huo
- Department of Laboratory Animal Science, School of Basic Medical Science, Capital Medical University, Beijing 100069, China
| | - Shuangyue Zhang
- Department of Laboratory Animal Science, School of Basic Medical Science, Capital Medical University, Beijing 100069, China
| | - Zhenkun Li
- Department of Laboratory Animal Science, School of Basic Medical Science, Capital Medical University, Beijing 100069, China
| | - Juan Gao
- Department of Laboratory Animal Science, School of Basic Medical Science, Capital Medical University, Beijing 100069, China
| | - Chao Wang
- Department of Laboratory Animal Science, School of Basic Medical Science, Capital Medical University, Beijing 100069, China
| | - Changlong Li
- Department of Laboratory Animal Science, School of Basic Medical Science, Capital Medical University, Beijing 100069, China
| | - Meng Guo
- Department of Laboratory Animal Science, School of Basic Medical Science, Capital Medical University, Beijing 100069, China
| | - Xiaoyan Du
- Department of Laboratory Animal Science, School of Basic Medical Science, Capital Medical University, Beijing 100069, China.
| | - Zhenwen Chen
- Department of Laboratory Animal Science, School of Basic Medical Science, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
26
|
Mahe E, Akhter A, Le A, Street L, Pournaziri P, Kosari F, Shabani-Rad MT, Stewart D, Mansoor A. PARP1 expression in mantle cell lymphoma: the utility of PARP1 immunohistochemistry and its relationship with markers of DNA damage. Hematol Oncol 2014; 33:159-65. [PMID: 25143154 DOI: 10.1002/hon.2160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 07/08/2014] [Accepted: 07/10/2014] [Indexed: 11/05/2022]
Abstract
Mantle cell lymphoma (MCL) is an aggressive disease with poor overall survival, attributable in part to frequent defects of the DNA repair genes. In such malignancies, additional inhibition of the ubiquitous DNA damage repair protein, poly-ADP ribose polymerase-1 (PARP1) has shown enhanced cytotoxicity (so-called synthetic lethality). We studied PARP1 expression in a series of clinical cases of MCL, with the secondary aim to ascertain the relationship between PARP1 expression and DNA repair gene expression (namely ATM and p53) by immunohistochemical methods. We also examined the relationship between PARP1 expression and the well-established prognostic biomarker Ki-67, in addition to correlating PARP1 expression with the overall survival. From amongst our series of 79 unselected cases of MCL, we detected PARP1 expression in all but two cases with variable intensity. We also noted correlations between PARP1 expression and ATM and p53 expression. As described in previous studies, we identified a significant survival difference on the basis of Ki-67 and p53 expression. When digital H-score analysis of PARP1 expression was performed, there was a distinct survival advantage noted in patients with lower levels of expression. When our biomarker data were assessed by Cox regression, furthermore, the dominant effects of p53 and PARP1 expression were highlighted. Our data support the need for further research into the potential utility of PARP1 as a biomarker in MCL and for the potential direction of future PARP1 inhibitor-targeted therapy studies.
Collapse
Affiliation(s)
- Etienne Mahe
- Department of Pathology & Laboratory Medicine, University of Calgary/Calgary Laboratory Services, Calgary, AB, Canada
| | - Ariz Akhter
- Department of Pathology & Laboratory Medicine, University of Calgary/Calgary Laboratory Services, Calgary, AB, Canada
| | - Anne Le
- Department of Pathology & Laboratory Medicine, University of Calgary/Calgary Laboratory Services, Calgary, AB, Canada
| | - Lelsey Street
- Division of Hematology, Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Payam Pournaziri
- Department of Pathology & Laboratory Medicine, University of Calgary/Calgary Laboratory Services, Calgary, AB, Canada
| | - Farid Kosari
- Department of Pathology & Laboratory Medicine, University of Calgary/Calgary Laboratory Services, Calgary, AB, Canada
| | - Meer-Taher Shabani-Rad
- Department of Pathology & Laboratory Medicine, University of Calgary/Calgary Laboratory Services, Calgary, AB, Canada
| | - Douglas Stewart
- Division of Hematology, Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Adnan Mansoor
- Department of Pathology & Laboratory Medicine, University of Calgary/Calgary Laboratory Services, Calgary, AB, Canada
| |
Collapse
|
27
|
Kubota E, Williamson CT, Ye R, Elegbede A, Peterson L, Lees-Miller SP, Bebb DG. Low ATM protein expression and depletion of p53 correlates with olaparib sensitivity in gastric cancer cell lines. Cell Cycle 2014; 13:2129-37. [PMID: 24841718 DOI: 10.4161/cc.29212] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Small-molecule inhibitors of poly (ADP-ribose) polymerase (PARP) have shown considerable promise in the treatment of homologous recombination (HR)-defective tumors, such as BRCA1- and BRCA2-deficient breast and ovarian cancers. We previously reported that mantle cell lymphoma cells with deficiency in ataxia telangiectasia mutated (ATM) are sensitive to PARP-1 inhibitors in vitro and in vivo. Here, we report that PARP inhibitors can potentially target ATM deficiency arising in a solid malignancy. We show that ATM protein expression varies between gastric cancer cell lines, with NUGC4 having significantly reduced protein levels. Significant correlation was found between ATM protein expression and sensitivity to the PARP inhibitor olaparib, with NUGC4 being the most sensitive. Moreover, reducing ATM kinase activity using a small-molecule inhibitor (KU55933) or shRNA-mediated depletion of ATM protein enhanced olaparib sensitivity in gastric cancer cell lines with depletion or inactivation of p53. Our results demonstrate that ATM is a potential predictive biomarker for PARP-1 inhibitor activity in gastric cancer harboring disruption of p53, and that combined inhibition of ATM and PARP-1 is a rational strategy for expanding the utility of PARP-1 inhibitors to gastric cancer with p53 disruption.
Collapse
Affiliation(s)
- Eiji Kubota
- Department of Biochemistry & Molecular Biology; University of Calgary; Calgary, Alberta, Canada; Southern Alberta Cancer Research Institute; University of Calgary; Calgary, Alberta, Canada
| | - Christopher T Williamson
- Department of Biochemistry & Molecular Biology; University of Calgary; Calgary, Alberta, Canada; Southern Alberta Cancer Research Institute; University of Calgary; Calgary, Alberta, Canada
| | - Ruiqiong Ye
- Department of Biochemistry & Molecular Biology; University of Calgary; Calgary, Alberta, Canada; Southern Alberta Cancer Research Institute; University of Calgary; Calgary, Alberta, Canada
| | - Anifat Elegbede
- Southern Alberta Cancer Research Institute; University of Calgary; Calgary, Alberta, Canada; Tom Baker Cancer Center; Calgary, Alberta, Canada
| | - Lars Peterson
- Southern Alberta Cancer Research Institute; University of Calgary; Calgary, Alberta, Canada; Tom Baker Cancer Center; Calgary, Alberta, Canada
| | - Susan P Lees-Miller
- Department of Biochemistry & Molecular Biology; University of Calgary; Calgary, Alberta, Canada; Southern Alberta Cancer Research Institute; University of Calgary; Calgary, Alberta, Canada
| | - D Gwyn Bebb
- Southern Alberta Cancer Research Institute; University of Calgary; Calgary, Alberta, Canada; Tom Baker Cancer Center; Calgary, Alberta, Canada
| |
Collapse
|