1
|
Sokolov P, Nifontova G, Samokhvalov P, Karaulov A, Sukhanova A, Nabiev I. Nontoxic Fluorescent Nanoprobes for Multiplexed Detection and 3D Imaging of Tumor Markers in Breast Cancer. Pharmaceutics 2023; 15:pharmaceutics15030946. [PMID: 36986807 PMCID: PMC10052755 DOI: 10.3390/pharmaceutics15030946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Multiplexed fluorescent immunohistochemical analysis of breast cancer (BC) markers and high-resolution 3D immunofluorescence imaging of the tumor and its microenvironment not only facilitate making the disease prognosis and selecting effective anticancer therapy (including photodynamic therapy), but also provides information on signaling and metabolic mechanisms of carcinogenesis and helps in the search for new therapeutic targets and drugs. The characteristics of imaging nanoprobe efficiency, such as sensitivity, target affinity, depth of tissue penetration, and photostability, are determined by the properties of their components, fluorophores and capture molecules, and by the method of their conjugation. Regarding individual nanoprobe components, fluorescent nanocrystals (NCs) are widely used for optical imaging in vitro and in vivo, and single-domain antibodies (sdAbs) are well established as highly specific capture molecules in diagnostic and therapeutic applications. Moreover, the technologies of obtaining functionally active sdAb–NC conjugates with the highest possible avidity, with all sdAb molecules bound to the NC in a strictly oriented manner, provide 3D-imaging nanoprobes with strong comparative advantages. This review is aimed at highlighting the importance of an integrated approach to BC diagnosis, including the detection of biomarkers of the tumor and its microenvironment, as well as the need for their quantitative profiling and imaging of their mutual location, using advanced approaches to 3D detection in thick tissue sections. The existing approaches to 3D imaging of tumors and their microenvironment using fluorescent NCs are described, and the main comparative advantages and disadvantages of nontoxic fluorescent sdAb–NC conjugates as nanoprobes for multiplexed detection and 3D imaging of BC markers are discussed.
Collapse
Affiliation(s)
- Pavel Sokolov
- Laboratory of Nano-Bioengineering, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115522 Moscow, Russia
| | - Galina Nifontova
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Université de Reims Champagne-Ardenne, 51100 Reims, France
| | - Pavel Samokhvalov
- Laboratory of Nano-Bioengineering, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115522 Moscow, Russia
| | - Alexander Karaulov
- Department of Clinical Immunology and Allergology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia
| | - Alyona Sukhanova
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Université de Reims Champagne-Ardenne, 51100 Reims, France
| | - Igor Nabiev
- Laboratory of Nano-Bioengineering, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115522 Moscow, Russia
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Université de Reims Champagne-Ardenne, 51100 Reims, France
- Department of Clinical Immunology and Allergology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia
- Correspondence:
| |
Collapse
|
2
|
Ng CCY, Lim S, Lim AH, Md Nasir ND, Zhang J, Rajasegaran V, Lee JY, Kok JST, Thike AA, Lim JX, Weng R, Yee S, Choudhury Y, Chan JY, Tan PH, Tan MH, Teh BT. A comprehensive next generation sequencing tissue assay for Asian-prevalent cancers—Analytical validation and performance evaluation with clinical samples. Front Mol Biosci 2022; 9:963243. [PMID: 36213130 PMCID: PMC9532579 DOI: 10.3389/fmolb.2022.963243] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction: A well-validated diagnostic assay with curated biomarkers complements clinicopathological factors to facilitate early diagnosis and ensure timely treatment delivery. This study focuses on an Asian-centric cancer diagnostic assay designed and thoroughly validated against commercially available standard references and a cohort of over 200 clinical specimens spanning 12 diverse Asian-centric cancer types. Methods: The assay uses hybrid-capture probes capable of profiling DNA aberrations from 572 cancer-related genes and 91 RNA fusion partners. The panel can detect clinically-tractable biomarkers such as microsatellite instability (MSI) and tumor mutation burden (TMB). Results: Analytical evaluation demonstrated 100% specificity and 99.9% sensitivity within a ≥5% VAF limit of detection (LoD) for SNV/Indels. RNA-based fusion features an LoD of ≥5 copies per nanogram input when evaluated against commercial references. Excellent linearity and concordance were observed when benchmarking against orthogonal methods in identifying MSI status, TMB scores and RNA fusions. Actionable genetic alterations were identified in 65% of the clinical samples. Conclusion: These results demonstrate a molecular diagnostic assay that accurately detects genomic alterations and complex biomarkers. The data also supports an excellent performance of this assay for making critical diagnoses and well-informed therapeutic decisions in Asian prevalent cancers.
Collapse
Affiliation(s)
- Cedric Chuan-Young Ng
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Sandy Lim
- Diagnostics Development Hub (DxD Hub), A National Platform Hosted by A*STAR, Singapore, Singapore
| | - Abner Herbert Lim
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Nur Diyana Md Nasir
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Jingxian Zhang
- Diagnostics Development Hub (DxD Hub), A National Platform Hosted by A*STAR, Singapore, Singapore
| | - Vikneswari Rajasegaran
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Jing Yi Lee
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Jessica Sook Ting Kok
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Aye Aye Thike
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | | | - Ruifen Weng
- Diagnostics Development Hub (DxD Hub), A National Platform Hosted by A*STAR, Singapore, Singapore
| | - Sidney Yee
- Diagnostics Development Hub (DxD Hub), A National Platform Hosted by A*STAR, Singapore, Singapore
| | | | - Jason Yongsheng Chan
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Puay Hoon Tan
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Min-Han Tan
- Lucence Diagnostics Pte Ltd, Singapore, Singapore
- *Correspondence: Bin Tean Teh, ; Min-Han Tan,
| | - Bin Tean Teh
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
- *Correspondence: Bin Tean Teh, ; Min-Han Tan,
| |
Collapse
|
3
|
Spanjaard A, Shah R, de Groot D, Buoninfante OA, Morris B, Lieftink C, Pritchard C, Zürcher LM, Ormel S, Catsman JJI, de Korte-Grimmerink R, Siteur B, Proost N, Boadum T, van de Ven M, Song JY, Kreft M, van den Berk PCM, Beijersbergen RL, Jacobs H. Division of labor within the DNA damage tolerance system reveals non-epistatic and clinically actionable targets for precision cancer medicine. Nucleic Acids Res 2022; 50:7420-7435. [PMID: 35819193 PMCID: PMC9303390 DOI: 10.1093/nar/gkac545] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/02/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Crosslink repair depends on the Fanconi anemia pathway and translesion synthesis polymerases that replicate over unhooked crosslinks. Translesion synthesis is regulated via ubiquitination of PCNA, and independently via translesion synthesis polymerase REV1. The division of labor between PCNA-ubiquitination and REV1 in interstrand crosslink repair is unclear. Inhibition of either of these pathways has been proposed as a strategy to increase cytotoxicity of platinating agents in cancer treatment. Here, we defined the importance of PCNA-ubiquitination and REV1 for DNA in mammalian ICL repair. In mice, loss of PCNA-ubiquitination, but not REV1, resulted in germ cell defects and hypersensitivity to cisplatin. Loss of PCNA-ubiquitination, but not REV1 sensitized mammalian cancer cell lines to cisplatin. We identify polymerase Kappa as essential in tolerating DNA damage-induced lesions, in particular cisplatin lesions. Polk-deficient tumors were controlled by cisplatin treatment and it significantly delayed tumor outgrowth and increased overall survival of tumor bearing mice. Our results indicate that PCNA-ubiquitination and REV1 play distinct roles in DNA damage tolerance. Moreover, our results highlight POLK as a critical TLS polymerase in tolerating multiple genotoxic lesions, including cisplatin lesions. The relative frequent loss of Polk in cancers indicates an exploitable vulnerability for precision cancer medicine.
Collapse
Affiliation(s)
- Aldo Spanjaard
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Ronak Shah
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Daniël de Groot
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Olimpia Alessandra Buoninfante
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Ben Morris
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Cor Lieftink
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Colin Pritchard
- Intervention unit of the Mouse Clinic for Cancer and Aging research (MCCA), The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Lisa M Zürcher
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Shirley Ormel
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Joyce J I Catsman
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Renske de Korte-Grimmerink
- Intervention unit of the Mouse Clinic for Cancer and Aging research (MCCA), The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Bjørn Siteur
- Intervention unit of the Mouse Clinic for Cancer and Aging research (MCCA), The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Natalie Proost
- Intervention unit of the Mouse Clinic for Cancer and Aging research (MCCA), The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Terry Boadum
- NKI Animal facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Marieke van de Ven
- Intervention unit of the Mouse Clinic for Cancer and Aging research (MCCA), The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Ji-Ying Song
- Division of Experimental Animal Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Maaike Kreft
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Paul C M van den Berk
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Roderick L Beijersbergen
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Heinz Jacobs
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
4
|
Jeong HY, Yoo J, Kim H, Kim TM. Identification of potential candidate genes for lip and oral cavity cancer using network analysis. Genomics Inform 2021; 19:e40. [PMID: 35172473 PMCID: PMC8752981 DOI: 10.5808/gi.21047] [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: 08/19/2021] [Accepted: 09/28/2021] [Indexed: 11/21/2022] Open
Abstract
Mutation signatures represent unique sequence footprints of somatic mutations resulting from specific DNA mutagenic and repair processes. However, their causal associations and the potential utility for genome research remain largely unknown. In this study, we performed PanCancer-scale correlative analyses to identify the genomic features associated with tumor mutation burdens (TMB) and individual mutation signatures. We observed that TMB was correlated with tumor purity, ploidy, and the level of aneuploidy, as well as with the expression of cell proliferation-related genes representing genomic covariates in evaluating TMB. Correlative analyses of mutation signature levels with genes belonging to specific DNA damage-repair processes revealed that deficiencies of NHEJ1 and ALKBH3 may contribute to mutations in the settings of APOBEC cytidine deaminase activation and DNA mismatch repair deficiency, respectively. We further employed a strategy to identify feature-driven, de novo mutation signatures and demonstrated that mutation signatures can be reconstructed using known causal features. Using the strategy, we further identified tumor hypoxia-related mutation signatures similar to the APOBEC-related mutation signatures, suggesting that APOBEC activity mediates hypoxia-related mutational consequences in cancer genomes. Our study advances the mechanistic insights into the TMB and signature-based DNA mutagenic and repair processes in cancer genomes. We also propose that feature-driven mutation signature analysis can further extend the categories of cancer-relevant mutation signatures and their causal relationships.
Collapse
Affiliation(s)
- Hye Young Jeong
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Jinseon Yoo
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Hyunwoo Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Tae-Min Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
5
|
Errazquin R, Sieiro E, Moreno P, Ramirez MJ, Lorz C, Peral J, Ortiz J, Casado JA, Roman-Rodriguez FJ, Hanenberg H, Río P, Surralles J, Segrelles C, Garcia-Escudero R. Generating New FANCA-Deficient HNSCC Cell Lines by Genomic Editing Recapitulates the Cellular Phenotypes of Fanconi Anemia. Genes (Basel) 2021; 12:548. [PMID: 33918752 PMCID: PMC8069753 DOI: 10.3390/genes12040548] [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: 11/28/2020] [Revised: 03/30/2021] [Accepted: 04/07/2021] [Indexed: 01/22/2023] Open
Abstract
Fanconi anemia (FA) patients have an exacerbated risk of head and neck squamous cell carcinoma (HNSCC). Treatment is challenging as FA patients display enhanced toxicity to standard treatments, including radio/chemotherapy. Therefore, better therapies as well as new disease models are urgently needed. We have used CRISPR/Cas9 editing tools in order to interrupt the human FANCA gene by the generation of insertions/deletions (indels) in exon 4 in two cancer cell lines from sporadic HNSCC having no mutation in FA-genes: CAL27 and CAL33 cells. Our approach allowed efficient editing, subsequent purification of single-cell clones, and Sanger sequencing validation at the edited locus. Clones having frameshift indels in homozygosis did not express FANCA protein and were selected for further analysis. When compared with parental CAL27 and CAL33, FANCA-mutant cell clones displayed a FA-phenotype as they (i) are highly sensitive to DNA interstrand crosslink (ICL) agents such as mitomycin C (MMC) or cisplatin, (ii) do not monoubiquitinate FANCD2 upon MMC treatment and therefore (iii) do not form FANCD2 nuclear foci, and (iv) they display increased chromosome fragility and G2 arrest after diepoxybutane (DEB) treatment. These FANCA-mutant clones display similar growth rates as their parental cells. Interestingly, mutant cells acquire phenotypes associated with more aggressive disease, such as increased migration in wound healing assays. Therefore, CAL27 and CAL33 cells with FANCA mutations are phenocopies of FA-HNSCC cells.
Collapse
Affiliation(s)
- Ricardo Errazquin
- Biomedical Research Institute I+12, University Hospital 12 de Octubre, 28041 Madrid, Spain; (R.E.); (C.L.); (C.S.)
- Molecular Oncology Unit, CIEMAT, 28040 Madrid, Spain; (E.S.); (P.M.); (J.P.); (J.O.)
| | - Esther Sieiro
- Molecular Oncology Unit, CIEMAT, 28040 Madrid, Spain; (E.S.); (P.M.); (J.P.); (J.O.)
| | - Pilar Moreno
- Molecular Oncology Unit, CIEMAT, 28040 Madrid, Spain; (E.S.); (P.M.); (J.P.); (J.O.)
| | - María José Ramirez
- Join Research Unit on Genomic Medicine UAB-Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (M.J.R.); (J.S.)
- Centro de Investigación Biomédica en Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (J.A.C.); (F.J.R.-R.); (P.R.)
| | - Corina Lorz
- Biomedical Research Institute I+12, University Hospital 12 de Octubre, 28041 Madrid, Spain; (R.E.); (C.L.); (C.S.)
- Molecular Oncology Unit, CIEMAT, 28040 Madrid, Spain; (E.S.); (P.M.); (J.P.); (J.O.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Jorge Peral
- Molecular Oncology Unit, CIEMAT, 28040 Madrid, Spain; (E.S.); (P.M.); (J.P.); (J.O.)
| | - Jessica Ortiz
- Molecular Oncology Unit, CIEMAT, 28040 Madrid, Spain; (E.S.); (P.M.); (J.P.); (J.O.)
| | - José Antonio Casado
- Centro de Investigación Biomédica en Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (J.A.C.); (F.J.R.-R.); (P.R.)
- Hematopoietic Innovative Therapies Division, CIEMAT, 28040 Madrid, Spain
- Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - Francisco J. Roman-Rodriguez
- Centro de Investigación Biomédica en Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (J.A.C.); (F.J.R.-R.); (P.R.)
- Hematopoietic Innovative Therapies Division, CIEMAT, 28040 Madrid, Spain
- Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - Helmut Hanenberg
- University Children’s Hospital Essen, University of Duisburg-Essen, 47057 Essen, Germany;
- Department of Otorhinolaryngology & Head/Neck Surgery, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Paula Río
- Centro de Investigación Biomédica en Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (J.A.C.); (F.J.R.-R.); (P.R.)
- Hematopoietic Innovative Therapies Division, CIEMAT, 28040 Madrid, Spain
- Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - Jordi Surralles
- Join Research Unit on Genomic Medicine UAB-Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (M.J.R.); (J.S.)
- Centro de Investigación Biomédica en Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (J.A.C.); (F.J.R.-R.); (P.R.)
| | - Carmen Segrelles
- Biomedical Research Institute I+12, University Hospital 12 de Octubre, 28041 Madrid, Spain; (R.E.); (C.L.); (C.S.)
- Molecular Oncology Unit, CIEMAT, 28040 Madrid, Spain; (E.S.); (P.M.); (J.P.); (J.O.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Ramon Garcia-Escudero
- Biomedical Research Institute I+12, University Hospital 12 de Octubre, 28041 Madrid, Spain; (R.E.); (C.L.); (C.S.)
- Molecular Oncology Unit, CIEMAT, 28040 Madrid, Spain; (E.S.); (P.M.); (J.P.); (J.O.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
6
|
de Kort WWB, Spelier S, Devriese LA, van Es RJJ, Willems SM. Predictive Value of EGFR-PI3K-AKT-mTOR-Pathway Inhibitor Biomarkers for Head and Neck Squamous Cell Carcinoma: A Systematic Review. Mol Diagn Ther 2021; 25:123-136. [PMID: 33686517 PMCID: PMC7956931 DOI: 10.1007/s40291-021-00518-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Understanding molecular pathogenesis of head and neck squamous cell carcinomas (HNSCC) has considerably improved in the last decades. As a result, novel therapeutic strategies have evolved, amongst which are epidermal growth factor receptor (EGFR)-targeted therapies. With the exception of cetuximab, targeted therapies for HNSCC have not yet been introduced into clinical practice. One important aspect of new treatment regimes in clinical practice is presence of robust biomarkers predictive for therapy response. METHODS We performed a systematic search in PubMed, Embase and the Cochrane library. Articles were included if they investigated a biomarker for targeted therapy in the EGFR-PI3K-AKT-mTOR-pathway. RESULTS Of 83 included articles, 52 were preclinical and 33 were clinical studies (two studies contained both a preclinical and a clinical part). We classified EGFR pathway inhibitor types and investigated the type of biomarker (biomarker on epigenetic, DNA, mRNA or protein level). CONCLUSION Several EGFR-PI3K-AKT-mTOR-pathway inhibitor biomarkers have been researched for HNSCC but few of the investigated biomarkers have been adequately confirmed in clinical trials. A more systematic approach is needed to discover proper biomarkers as stratifying patients is essential to prevent unnecessary costs and side effects.
Collapse
Affiliation(s)
- W. W. B. de Kort
- Department of Pathology, University Medical Center Utrecht, PO Box 885500, 3508 GA Utrecht, The Netherlands
| | - S. Spelier
- Department of Pathology, University Medical Center Utrecht, PO Box 885500, 3508 GA Utrecht, The Netherlands
| | - L. A. Devriese
- Department of Medical Oncology, University Medical Center Utrecht, PO Box 885500, 3508 GA Utrecht, The Netherlands
| | - R. J. J. van Es
- Department of Oral and Maxillofacial Surgery, University Medical Center Utrecht, PO Box 885500, 3508 GA Utrecht, The Netherlands
- Department of Head and Neck Surgical Oncology, Utrecht Cancer Center, University Medical Center Utrecht, PO Box 885500, 3508 GA Utrecht, The Netherlands
| | - S. M. Willems
- Department of Pathology, University Medical Center Utrecht, PO Box 885500, 3508 GA Utrecht, The Netherlands
- Department of Pathology, University Medical Center Groningen, PO Box 30001, 9700 RB Groningen, The Netherlands
| |
Collapse
|
7
|
Shilkin ES, Boldinova EO, Stolyarenko AD, Goncharova RI, Chuprov-Netochin RN, Smal MP, Makarova AV. Translesion DNA Synthesis and Reinitiation of DNA Synthesis in Chemotherapy Resistance. BIOCHEMISTRY (MOSCOW) 2021; 85:869-882. [PMID: 33045948 DOI: 10.1134/s0006297920080039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Many chemotherapy drugs block tumor cell division by damaging DNA. DNA polymerases eta (Pol η), iota (Pol ι), kappa (Pol κ), REV1 of the Y-family and zeta (Pol ζ) of the B-family efficiently incorporate nucleotides opposite a number of DNA lesions during translesion DNA synthesis. Primase-polymerase PrimPol and the Pol α-primase complex reinitiate DNA synthesis downstream of the damaged sites using their DNA primase activity. These enzymes can decrease the efficacy of chemotherapy drugs, contribute to the survival of tumor cells and to the progression of malignant diseases. DNA polymerases are promising targets for increasing the effectiveness of chemotherapy, and mutations and polymorphisms in some DNA polymerases can serve as additional prognostic markers in a number of oncological disorders.
Collapse
Affiliation(s)
- E S Shilkin
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| | - E O Boldinova
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| | - A D Stolyarenko
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| | - R I Goncharova
- Institute of Genetics and Cytology, National Academy of Sciences of Belarus, Minsk, 220072, Republic of Belarus
| | - R N Chuprov-Netochin
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia
| | - M P Smal
- Institute of Genetics and Cytology, National Academy of Sciences of Belarus, Minsk, 220072, Republic of Belarus.
| | - A V Makarova
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia.
| |
Collapse
|
8
|
Wang D, Zhou X, Yin J, Zhou Y. Lnc-PICSAR contributes to cisplatin resistance by miR-485-5p/REV3L axis in cutaneous squamous cell carcinoma. Open Life Sci 2020; 15:488-500. [PMID: 33817237 PMCID: PMC7874590 DOI: 10.1515/biol-2020-0049] [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: 12/26/2019] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/22/2022] Open
Abstract
Background Dysregulation of long noncoding RNAs (lncRNAs) is associated with drug resistance in multiple cancers. We explored the roles of lncRNA p38 inhibited cutaneous squamous cell carcinoma-associated lincRNA (PICSAR) in cisplatin (DDP) resistance of cutaneous squamous cell carcinoma (CSCC). Methods Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to measure the expression of lnc-PICSAR, miR-485-5p and reversionless 3-like (REV3L) mRNA. The cell counting kit-8 (CCK-8) assay was conducted to evaluate DDP resistance and cell viability. The transwell assay was performed to determine cell migration and invasion. Western blot assay and immunohistochemistry (IHC) staining assay were carried out to measure protein levels. The dual-luciferase reporter assay was used to investigate the association between miR-485-5p and lnc-PICSAR or REV3L. Murine xenograft model was constructed to explore the function of lnc-PICSAR in vivo. The morphology of exosomes was analyzed by transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). Results Lnc-PICSAR was elevated in DDP-resistant CSCC cells. Lnc-PICSAR silencing suppressed cell viability, DDP resistance, migration and invasion in DDP-resistant CSCC cells. MiR-485-5p acted as a target of lnc-PICSAR, and miR-485-5p inhibition reversed the impacts of lnc-PICSAR silencing on DDP resistance and cell progression in DDP-resistant CSCC cells. Lnc-PICSAR promoted REV3L expression via sponging miR-485-5p. Moreover, REV3L overexpression overturned the effects of lnc-PICSAR on cell progression and DDP resistance. Lnc-PICSAR knockdown suppressed DDP resistance in vivo. In addition, lnc-PICSAR was increased in the exosomes derived from CSCC patients’ serum and CSCC cells. Conclusion Lnc-PICSAR enhanced DDP resistance via miR-485-5p/REV3L axis in DDP-resistant CSCC cells. Besides, exosome-mediated lnc-PICSAR might be involved in the regulation of drug resistance in CSCC.
Collapse
Affiliation(s)
- Dan Wang
- Plastic Surgery Center, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Xiaoqiang Zhou
- Department of Dermatology, Medical College of Shaoguan University, Shaoguan, 512026, China
| | - Jing Yin
- Department of Interventional Vascular, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Yang Zhou
- Department of Dermatology, Yancheng First People's Hospital, Jiangsu Province, No.166 yulong west road, Yancheng, Jiangsu, 224001, China
| |
Collapse
|
9
|
Xu W, Hua Y, Deng F, Wang D, Wu Y, Zhang W, Tang J. MiR-145 in cancer therapy resistance and sensitivity: A comprehensive review. Cancer Sci 2020; 111:3122-3131. [PMID: 32506767 PMCID: PMC7469794 DOI: 10.1111/cas.14517] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/22/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022] Open
Abstract
MircoRNA (miRNA) are a group of small, non–coding, regulatory RNA with an average length of approximately 22 nucleotides, which mostly modulate gene expression post–transcriptionally through complementary binding to the 3ʹ‐untranslated region (3ʹ‐UTR) of multiple target genes. Emerging evidence has shown that miRNA are frequently dysregulated in a variety of human malignancies. Among them, microRNA‐145 (miR‐145) has been increasingly identified as a critical suppressor of carcinogenesis and therapeutic resistance. Resistance to tumor therapy is a challenge in cancer treatment due to the daunting range of resistance mechanisms. We reviewed the status quo of recent advancements in the knowledge of the functional role of miR‐145 in therapeutic resistance and the tumor microenvironment. It may serve as an innovative biomarker for therapeutic response and cancer prognosis.
Collapse
Affiliation(s)
- Wenxiu Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuting Hua
- Department of Gastroenterology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Fei Deng
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dandan Wang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Wu
- The Jiangsu Province Research Institute for Clinical Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Zhang
- The Jiangsu Province Research Institute for Clinical Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jinhai Tang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Zhou L, Mudianto T, Ma X, Riley R, Uppaluri R. Targeting EZH2 Enhances Antigen Presentation, Antitumor Immunity, and Circumvents Anti-PD-1 Resistance in Head and Neck Cancer. Clin Cancer Res 2019; 26:290-300. [PMID: 31562203 DOI: 10.1158/1078-0432.ccr-19-1351] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/12/2019] [Accepted: 09/23/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE Anti-programmed death-1 (PD-1) receptor-based therapeutics improve survival in patients with recurrent head and neck squamous cell carcinoma (HNSCC), but many do not benefit due to a low response rate. Herein, we identified EZH2 as a therapeutic target that enhanced tumor cell antigen presentation and subsequently sensitized resistant tumors to anti-PD-1 therapy. EXPERIMENTAL DESIGN EZH2 regulation of antigen presentation was defined using EZH2 inhibitors (GSK126 and EPZ6438) in human and mouse HNSCC cell lines. Mechanistic dissection of EZH2 in regulation of antigen presentation was investigated using flow cytometry, qRT-PCR, ELISA, and chromatin-immunoprecipitation assays. EZH2-deficient cell lines were generated using CRISPR-CAS9. GSK126 and anti-PD-1-blocking antibody were used in testing combinatorial therapy in vivo. RESULTS EZH2 expression was negatively correlated with antigen-processing machinery pathway components in HNSCC data sets in The Cancer Genome Atlas. EZH2 inhibition resulted in significant upregulation of MHC class I expression in human and mouse human papillomavirus-negative HNSCC lines in vitro and in mouse models in vivo. Enhanced antigen presentation on the tumor cells by EZH2 inhibitors or CRISPR-mediated EZH2 deficiency increased antigen-specific CD8+ T-cell proliferation, IFNγ production, and tumor cell cytotoxicity. Mechanistically, EZH2 inhibition reduced the histone H3K27me3 modification on the β-2-microglobulin promoter. Finally, in an anti-PD-1-resistant model of HNSCC, tumor growth was suppressed with combination therapy. CONCLUSIONS Our results demonstrated that targeting EZH2 enhanced antigen presentation and was able to circumvent anti-PD-1 resistance. Thus, combining EZH2 targeting with anti-PD-1 may increase therapeutic susceptibility in HNSCC.
Collapse
Affiliation(s)
- Liye Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tenny Mudianto
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Xiaojing Ma
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Rachel Riley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ravindra Uppaluri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Surgery/Otolaryngology, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
11
|
Van Hoeck A, Tjoonk NH, van Boxtel R, Cuppen E. Portrait of a cancer: mutational signature analyses for cancer diagnostics. BMC Cancer 2019; 19:457. [PMID: 31092228 PMCID: PMC6521503 DOI: 10.1186/s12885-019-5677-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 05/03/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND In the past decade, systematic and comprehensive analyses of cancer genomes have identified cancer driver genes and revealed unprecedented insight into the molecular mechanisms underlying the initiation and progression of cancer. These studies illustrate that although every cancer has a unique genetic make-up, there are only a limited number of mechanisms that shape the mutational landscapes of cancer genomes, as reflected by characteristic computationally-derived mutational signatures. Importantly, the molecular mechanisms underlying specific signatures can now be dissected and coupled to treatment strategies. Systematic characterization of mutational signatures in a cancer patient's genome may thus be a promising new tool for molecular tumor diagnosis and classification. RESULTS In this review, we describe the status of mutational signature analysis in cancer genomes and discuss the opportunities and relevance, as well as future challenges, for further implementation of mutational signatures in clinical tumor diagnostics and therapy guidance. CONCLUSIONS Scientific studies have illustrated the potential of mutational signature analysis in cancer research. As such, we believe that the implementation of mutational signature analysis within the diagnostic workflow will improve cancer diagnosis in the future.
Collapse
Affiliation(s)
- Arne Van Hoeck
- Center for Molecular Medicine and Oncode Institute, University Medical Centre Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - Niels H. Tjoonk
- Center for Molecular Medicine and Oncode Institute, University Medical Centre Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology and Oncode Institute, Heidelberglaan 25, 3584CS Utrecht, The Netherlands
| | - Ruben van Boxtel
- Princess Máxima Center for Pediatric Oncology and Oncode Institute, Heidelberglaan 25, 3584CS Utrecht, The Netherlands
| | - Edwin Cuppen
- Center for Molecular Medicine and Oncode Institute, University Medical Centre Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
- Hartwig Medical Foundation, Science Park 408, 1098XH Amsterdam, The Netherlands
| |
Collapse
|
12
|
Chen Q, Hou J, Wu Z, Zhao J, Ma D. miR-145 Regulates the sensitivity of esophageal squamous cell carcinoma cells to 5-FU via targeting REV3L. Pathol Res Pract 2019; 215:152427. [PMID: 31072625 DOI: 10.1016/j.prp.2019.04.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 03/28/2019] [Accepted: 04/26/2019] [Indexed: 12/24/2022]
Abstract
Aberrant expression of miR-145 was associated with chemotherapy in multitype cancers. However, the underlying role and molecular mechanism of miR-145 in the sensitivity of esophageal squamous cell carcinoma (ESCC) to 5-FU remained largely unknown. Cell viability was determined by Cell Counting Kit-8 (CCK-8) assay. Gene expression levels were detected by real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR). Protein expression levels were evaluated by Western blot. TargetScan was used for the prediction of binding sites for miRNA in mRNAs. The interaction between mRNA 3' UTR and miRNA was verified by dual luciferase reporter assay. The results showed that miR-145 was downregulated in ESCC tumor tissues and cells, while REV3L was upregulated in ESCC tumor tissues. Overexpression of miR-145 decreased REV3L mRNA and protein level in ESCC cell line KYSE150, while decreased miR-145 increased REV3L mRNA and protein level in esophageal epithelium cell line (HEEC). In addition, the luciferase activity of ESCC cells was decreased after the treatment of miR-145 mimic and mRNA 3'UTR-WT. Overexpressed miR-145 significantly inhibited cell viability and elevated cell apoptosis rate upon 5-FU treatment. Additionally, transfection of miR-145 mimic further altered expression of key genes involved in cell apoptosis (Bcl-2, Bax, Caspase3) in ESCC cells treated with 5-FU. miR-145 might be a therapeutic target for the treatment of ESCC.
Collapse
Affiliation(s)
- Qing Chen
- Department of Oncology, Jingjiang Peoples' Hospital, Jingjiang, 214500, China
| | - Juan Hou
- Department of Oncology, Jingjiang Peoples' Hospital, Jingjiang, 214500, China
| | - Zhiwei Wu
- Department of Oncology, Jingjiang Peoples' Hospital, Jingjiang, 214500, China
| | - Jie Zhao
- Department of Oncology, Jingjiang Peoples' Hospital, Jingjiang, 214500, China
| | - De Ma
- Department of Oncology, Jingjiang Peoples' Hospital, Jingjiang, 214500, China.
| |
Collapse
|
13
|
Englinger B, Pirker C, Heffeter P, Terenzi A, Kowol CR, Keppler BK, Berger W. Metal Drugs and the Anticancer Immune Response. Chem Rev 2018; 119:1519-1624. [DOI: 10.1021/acs.chemrev.8b00396] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Bernhard Englinger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Alessio Terenzi
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Christian R. Kowol
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Bernhard K. Keppler
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| |
Collapse
|
14
|
Arivazhagan R, Lee J, Bayarsaikhan D, Kwak P, Son M, Byun K, Salekdeh GH, Lee B. MicroRNA-340 inhibits the proliferation and promotes the apoptosis of colon cancer cells by modulating REV3L. Oncotarget 2017; 9:5155-5168. [PMID: 29435169 PMCID: PMC5797040 DOI: 10.18632/oncotarget.23703] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 12/05/2017] [Indexed: 11/25/2022] Open
Abstract
DNA Directed Polymerase Zeta Catalytic Subunit (REV3L) has recently emerged as an important oncogene. Although the expressions of REV3L are similar in normal and cancer cells, several mutations in REV3L have been shown to play important roles in cancer. These mutations cause proteins misfolding and mislocalization, which in turn alters their interactions and biological functions. miRNAs play important regulatory roles during the progression and metastasis of several human cancers. This study was undertaken to determine how changes in the location and interactions of REV3L regulate colon cancer progression. REV3L protein mislocalization confirmed from the immunostaining results and the known interactions of REV3L was found to be broken as seen from the PLA assay results. The mislocalized REV3L might interact with new proteins partners in the cytoplasm which in turn may play role in regulating colon cancer progression. hsa-miR-340 (miR-340), a microRNA down-regulated in colon cancer, was used to bind to and downregulate REV3L, and found to control the proliferation and induce the apoptosis of colon cancer cells (HCT-116 and DLD-1) via the MAPK pathway. Furthermore, this down-regulation of REV3L also diminished colon cancer cell migration, and down-regulated MMP-2 and MMP-9. Combined treatment of colon cancer cells with miR-340 and 5-FU enhanced the inhibitory effects of 5-FU. In addition, in vivo experiments conducted on nude mice revealed tumor sizes were smaller in a HCT-116-miR-340 injected group than in a HCT-116-pCMV injected group. Our findings suggest mutations in REV3L causes protein mislocalization to the cytoplasm, breaking its interaction and is believed to form new protein interactions in cytoplasm contributing to colon cancer progression. Accordingly, microRNA-340 appears to be a good candidate for colon cancer therapy.
Collapse
Affiliation(s)
- Roshini Arivazhagan
- Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Jaesuk Lee
- Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Delger Bayarsaikhan
- Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Peter Kwak
- Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Myeongjoo Son
- Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea.,Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon, Republic of Korea
| | - Kyunghee Byun
- Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea.,Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon, Republic of Korea
| | - Ghasem Hosseini Salekdeh
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Molecular Sciences, Macquarie University Sydney, New South Wales, Australia
| | - Bonghee Lee
- Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea.,Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon, Republic of Korea
| |
Collapse
|
15
|
Hadjadj D, Kim SJ, Denecker T, Driss LB, Cadoret JC, Maric C, Baldacci G, Fauchereau F. A hypothesis-driven approach identifies CDK4 and CDK6 inhibitors as candidate drugs for treatments of adrenocortical carcinomas. Aging (Albany NY) 2017; 9:2695-2716. [PMID: 29283884 PMCID: PMC5764399 DOI: 10.18632/aging.101356] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 12/17/2017] [Indexed: 12/17/2022]
Abstract
High proliferation rate and high mutation density are both indicators of poor prognosis in adrenocortical carcinomas. We performed a hypothesis-driven association study between clinical features in adrenocortical carcinomas and the expression levels of 136 genes involved in DNA metabolism and G1/S phase transition. In 79 samples downloaded from The Cancer Genome Atlas portal, high Cyclin Dependent Kinase 6 (CDK6) mRNA levels gave the most significant association with shorter time to relapse and poorer survival of patients. A hierarchical clustering approach assembled most tumors with high levels of CDK6 mRNA into one group. These tumors tend to cumulate mutations activating the Wnt/β-catenin pathway and show reduced MIR506 expression. Actually, the level of MIR506 RNA is inversely correlated with the levels of both CDK6 and CTNNB1 (encoding β-catenin). Together these results indicate that high CDK6 expression is found in aggressive tumors with activated Wnt/β-catenin pathway. Thus we tested the impact of Food and Drug Administration-approved CDK4 and CDK6 inhibitors, namely palbociclib and ribociclib, on SW-13 and NCI-H295R cells. While both drugs reduced viability and induced senescence in SW-13 cells, only palbociclib was effective on the retinoblastoma protein (pRB)-negative NCI-H295R cells, by inducing apoptosis. In NCI-H295R cells, palbociclib induced an increase of the active form of Glycogen Synthase Kinase 3β (GSK3β) responsible for the reduced amount of active β-catenin, and altered the amount of AXIN2 mRNA. Taken together, these data underline the impact of CDK4 and CDK6 inhibitors in treating adrenocortical carcinomas.
Collapse
Affiliation(s)
- Djihad Hadjadj
- Pathologies de la Réplication de l'ADN, Université Paris-Diderot – Paris 7, Sorbonne Paris Cité, CNRS UMR7592, Institut Jacques-Monod, 75205 Paris Cedex 13, France
| | - Su-Jung Kim
- Pathologies de la Réplication de l'ADN, Université Paris-Diderot – Paris 7, Sorbonne Paris Cité, CNRS UMR7592, Institut Jacques-Monod, 75205 Paris Cedex 13, France
| | - Thomas Denecker
- Pathologies de la Réplication de l'ADN, Université Paris-Diderot – Paris 7, Sorbonne Paris Cité, CNRS UMR7592, Institut Jacques-Monod, 75205 Paris Cedex 13, France
| | - Laura Ben Driss
- Pathologies de la Réplication de l'ADN, Université Paris-Diderot – Paris 7, Sorbonne Paris Cité, CNRS UMR7592, Institut Jacques-Monod, 75205 Paris Cedex 13, France
| | - Jean-Charles Cadoret
- Pathologies de la Réplication de l'ADN, Université Paris-Diderot – Paris 7, Sorbonne Paris Cité, CNRS UMR7592, Institut Jacques-Monod, 75205 Paris Cedex 13, France
| | - Chrystelle Maric
- Pathologies de la Réplication de l'ADN, Université Paris-Diderot – Paris 7, Sorbonne Paris Cité, CNRS UMR7592, Institut Jacques-Monod, 75205 Paris Cedex 13, France
| | - Giuseppe Baldacci
- Pathologies de la Réplication de l'ADN, Université Paris-Diderot – Paris 7, Sorbonne Paris Cité, CNRS UMR7592, Institut Jacques-Monod, 75205 Paris Cedex 13, France
| | - Fabien Fauchereau
- Pathologies de la Réplication de l'ADN, Université Paris-Diderot – Paris 7, Sorbonne Paris Cité, CNRS UMR7592, Institut Jacques-Monod, 75205 Paris Cedex 13, France
- ePôle de Génoinformatique, Université Paris-Diderot – Paris 7, Sorbonne Paris Cité, CNRS UMR7592, Institut Jacques-Monod, 75205 Paris Cedex 13, France
| |
Collapse
|
16
|
Liu D, Abbosh P, Keliher D, Reardon B, Miao D, Mouw K, Weiner-Taylor A, Wankowicz S, Han G, Teo MY, Cipolla C, Kim J, Iyer G, Al-Ahmadie H, Dulaimi E, Chen DYT, Alpaugh RK, Hoffman-Censits J, Garraway LA, Getz G, Carter SL, Bellmunt J, Plimack ER, Rosenberg JE, Van Allen EM. Mutational patterns in chemotherapy resistant muscle-invasive bladder cancer. Nat Commun 2017; 8:2193. [PMID: 29259186 PMCID: PMC5736752 DOI: 10.1038/s41467-017-02320-7] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 11/21/2017] [Indexed: 12/20/2022] Open
Abstract
Despite continued widespread use, the genomic effects of cisplatin-based chemotherapy and implications for subsequent treatment are incompletely characterized. Here, we analyze whole exome sequencing of matched pre- and post-neoadjuvant cisplatin-based chemotherapy primary bladder tumor samples from 30 muscle-invasive bladder cancer patients. We observe no overall increase in tumor mutational burden post-chemotherapy, though a significant proportion of subclonal mutations are unique to the matched pre- or post-treatment tumor, suggesting chemotherapy-induced and/or spatial heterogeneity. We subsequently identify and validate a novel mutational signature in post-treatment tumors consistent with known characteristics of cisplatin damage and repair. We find that post-treatment tumor heterogeneity predicts worse overall survival, and further observe alterations in cell-cycle and immune checkpoint regulation genes in post-treatment tumors. These results provide insight into the clinical and genomic dynamics of tumor evolution with cisplatin-based chemotherapy, suggest mechanisms of clinical resistance, and inform development of clinically relevant biomarkers and trials of combination therapies. The impact of cisplatin-based chemotherapy on tumor genomes is complex. Here, the authors study matched pre- and post-chemotherapy primary samples in muscle-invasive bladder cancer, finding a cisplatin-based mutational signature, and highlighting the impact of intratumor heterogeneity on survival.
Collapse
Affiliation(s)
- David Liu
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Philip Abbosh
- Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Daniel Keliher
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Brendan Reardon
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Diana Miao
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Kent Mouw
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | | | - Stephanie Wankowicz
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Garam Han
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Min Yuen Teo
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | - Jaegil Kim
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Gopa Iyer
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | - Essel Dulaimi
- Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | | | | | | | - Levi A Garraway
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Gad Getz
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Scott L Carter
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Joaquim Bellmunt
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | | | | | - Eliezer M Van Allen
- Dana-Farber Cancer Institute, Boston, MA, 02215, USA. .,Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA.
| |
Collapse
|
17
|
Brandsma I, Fleuren ED, Williamson CT, Lord CJ. Directing the use of DDR kinase inhibitors in cancer treatment. Expert Opin Investig Drugs 2017; 26:1341-1355. [PMID: 28984489 PMCID: PMC6157710 DOI: 10.1080/13543784.2017.1389895] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Defects in the DNA damage response (DDR) drive the development of cancer by fostering DNA mutation but also provide cancer-specific vulnerabilities that can be exploited therapeutically. The recent approval of three different PARP inhibitors for the treatment of ovarian cancer provides the impetus for further developing targeted inhibitors of many of the kinases involved in the DDR, including inhibitors of ATR, ATM, CHEK1, CHEK2, DNAPK and WEE1. Areas covered: We summarise the current stage of development of these novel DDR kinase inhibitors, and describe which predictive biomarkers might be exploited to direct their clinical use. Expert opinion: Novel DDR inhibitors present promising candidates in cancer treatment and have the potential to elicit synthetic lethal effects. In order to fully exploit their potential and maximize their utility, identifying highly penetrant predictive biomarkers of single agent and combinatorial DDR inhibitor sensitivity are critical. Identifying the optimal drug combination regimens that could used with DDR inhibitors is also a key objective.
Collapse
Affiliation(s)
- Inger Brandsma
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Emmy D.G. Fleuren
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Chris T. Williamson
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Christopher J. Lord
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| |
Collapse
|
18
|
Wu P, Wu H, Tang Y, Luo S, Fang X, Xie C, He J, Zhao S, Wang X, Xu J, Chen X, Li D, Yang H, Wang J. Whole-exome sequencing reveals novel mutations and epigenetic regulation in hypopharyngeal carcinoma. Oncotarget 2017; 8:85326-85340. [PMID: 29156722 PMCID: PMC5689612 DOI: 10.18632/oncotarget.19674] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 05/23/2017] [Indexed: 02/01/2023] Open
Abstract
Hypopharyngeal cancer (HPC) frequently presents at an advanced stage, resulting in poor prognosis. Although combined surgical therapy and chemoradiotherapy have improved the survival for patients with HPC over the past 3 decades, the mortality rate in late-stage diagnosis of HPC is unsatisfactory. In this study, we performed whole-exome sequencing (WES) of 23 hypopharyngeal tumor and paired adjacent normal tissue to identify novel candidate driver genes associated with hypopharyngeal carcinoma. We identified several copy number variants (CNVs) and 15 somatic mutation genes that were associated with hypopharyngeal carcinoma. Mutations in nine new genes (PRB4, NSD1, REC8, ZNF772, ZNF69, EI24, CYFIP2, NEFH, KRTAP4-5) were also indentified. PRB4 and NSD1 expression were significantly upregulated in hypopharyngeal carcinoma, which was confirmed in an independent cohort using IHC. There was a positive relationship between PRB4 and NSD1. Downregulation of PRB4 by siRNA could inhibit cell growth, colony formation and cell invasion. Notably, we here demonstrate that NSD1 could bind to the promoter regions of PRB4 and activate promoter activity by reducing the binding of H3K27me2 and increasing the binding of H3K36me2 on PRB4 promoter. In summary, we pinpoint the predominant mutations in hypopharyngeal carcinoma by WES, highlighting the substantial genetic alterations contributing to hypopharyngeal carcinoma tumorigenesis. We also indentify a novel epigenetically regulatory between PRB4 and NSD1 that contribute to hypopharyngeal carcinoma tumorigenesis. They may become potential prognostic biomarkers and therapeutic target for hypopharyngeal carcinoma treatment.
Collapse
Affiliation(s)
- Ping Wu
- Department of Otorhinolaryngology Head & Neck Surgery, Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Honglong Wu
- Binhai Genomics Institute, BGI-Tianjin, BGI-Shenzhen, Tianjin 300308, China.,Tianjin Translational Genomics Center, BGI-Tianjin, BGI-Shenzhen, Tianjin 300308, China.,Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yaoyun Tang
- Department of Otorhinolaryngology Head & Neck Surgery, Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Shi Luo
- Department of Otorhinolaryngology Head & Neck Surgery, Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Xing Fang
- Department of Otorhinolaryngology Head & Neck Surgery, Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Chubo Xie
- Department of Otorhinolaryngology Head & Neck Surgery, Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Jian He
- Department of Otorhinolaryngology Head & Neck Surgery, Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Suping Zhao
- Department of Otorhinolaryngology Head & Neck Surgery, Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Xiaofeng Wang
- Binhai Genomics Institute, BGI-Tianjin, BGI-Shenzhen, Tianjin 300308, China.,Tianjin Translational Genomics Center, BGI-Tianjin, BGI-Shenzhen, Tianjin 300308, China
| | - Jiajia Xu
- Binhai Genomics Institute, BGI-Tianjin, BGI-Shenzhen, Tianjin 300308, China.,Tianjin Translational Genomics Center, BGI-Tianjin, BGI-Shenzhen, Tianjin 300308, China
| | - Xi Chen
- Binhai Genomics Institute, BGI-Tianjin, BGI-Shenzhen, Tianjin 300308, China.,Tianjin Translational Genomics Center, BGI-Tianjin, BGI-Shenzhen, Tianjin 300308, China
| | - Dongfang Li
- Binhai Genomics Institute, BGI-Tianjin, BGI-Shenzhen, Tianjin 300308, China.,Tianjin Translational Genomics Center, BGI-Tianjin, BGI-Shenzhen, Tianjin 300308, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen 518083, China.,James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen 518083, China.,James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| |
Collapse
|
19
|
Perisanidis C. Prevalence of EGFR Tyrosine Kinase Domain Mutations in Head and Neck Squamous Cell Carcinoma: Cohort Study and Systematic Review. ACTA ACUST UNITED AC 2017; 31:23-34. [PMID: 28064216 DOI: 10.21873/invivo.11020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/06/2016] [Accepted: 12/08/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Mutations in the epidermal growth factor receptor (EGFR) tyrosine kinase domain (TKD) are associated with response and resistance to targeted therapy. The EGFR mutation status in patients with advanced oral and oropharyngeal squamous cell carcinoma (OOSCC) was evaluated. A systematic literature review was undertaken to summarize current evidence and estimate the overall prevalence of EGFR TKD mutations in patients with head and neck squamous cell carcinoma (HNSCC). MATERIALS AND METHODS Genomic DNA was extracted from formalin-fixed, paraffin-embedded tumor samples of 113 patients with OOSCC. Pyrosequencing was performed to investigate mutations in EGFR exons 18 to 21. Medline databases were searched for relevant studies. Studies reporting mutations in the EGFR TKD in HNSCC were eligible for inclusion in the systematic review. RESULTS No mutations in the EGFR TKD were observed in 113 samples of OOSCC. A total of 53 eligible studies were included in the systematic review. In total, from the review, 117 patients harboring a total of 159 EGFR TKD mutations were reported among 4122 patients with HNSCC. The overall prevalence of EGFR TKD mutations in HNSCC was 2.8%. CONCLUSION Large-scale studies are warranted to provide further evidence regarding the mutation status of EGFR in patients with HNSCC.
Collapse
Affiliation(s)
- Christos Perisanidis
- Department of Cranio-, Maxillofacial and Oral Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
20
|
Hypermutation signature reveals a slippage and realignment model of translesion synthesis by Rev3 polymerase in cisplatin-treated yeast. Proc Natl Acad Sci U S A 2017; 114:2663-2668. [PMID: 28223526 DOI: 10.1073/pnas.1618555114] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Gene-gene or gene-drug interactions are typically quantified using fitness as a readout because the data are continuous and easily measured in high throughput. However, to what extent fitness captures the range of other phenotypes that show synergistic effects is usually unknown. Using Saccharomyces cerevisiae and focusing on a matrix of DNA repair mutants and genotoxic drugs, we quantify 76 gene-drug interactions based on both mutation rate and fitness and find that these parameters are not connected. Independent of fitness defects, we identified six cases of synthetic hypermutation, where the combined effect of the drug and mutant on mutation rate was greater than predicted. One example occurred when yeast lacking RAD1 were exposed to cisplatin, and we characterized this interaction using whole-genome sequencing. Our sequencing results indicate mutagenesis by cisplatin in rad1Δ cells appeared to depend almost entirely on interstrand cross-links at GpCpN motifs. Interestingly, our data suggest that the following base on the template strand dictates the addition of the mutated base. This result differs from cisplatin mutation signatures in XPF-deficient Caenorhabditis elegans and supports a model in which translesion synthesis polymerases perform a slippage and realignment extension across from the damaged base. Accordingly, DNA polymerase ζ activity was essential for mutagenesis in cisplatin-treated rad1Δ cells. Together these data reveal the potential to gain new mechanistic insights from nonfitness measures of gene-drug interactions and extend the use of mutation accumulation and whole-genome sequencing analysis to define DNA repair mechanisms.
Collapse
|
21
|
Szikriszt B, Póti Á, Pipek O, Krzystanek M, Kanu N, Molnár J, Ribli D, Szeltner Z, Tusnády GE, Csabai I, Szallasi Z, Swanton C, Szüts D. A comprehensive survey of the mutagenic impact of common cancer cytotoxics. Genome Biol 2016; 17:99. [PMID: 27161042 PMCID: PMC4862131 DOI: 10.1186/s13059-016-0963-7] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 04/22/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Genomic mutations caused by cytotoxic agents used in cancer chemotherapy may cause secondary malignancies as well as contribute to the evolution of treatment-resistant tumour cells. The stable diploid genome of the chicken DT40 lymphoblast cell line, an established DNA repair model system, is well suited to accurately assay genomic mutations. RESULTS We use whole genome sequencing of multiple DT40 clones to determine the mutagenic effect of eight common cytotoxics used for the treatment of millions of patients worldwide. We determine the spontaneous mutagenesis rate at 2.3 × 10(-10) per base per cell division and find that cisplatin, cyclophosphamide and etoposide induce extra base substitutions with distinct spectra. After four cycles of exposure, cisplatin induces 0.8 mutations per Mb, equivalent to the median mutational burden in common leukaemias. Cisplatin-induced mutations, including short insertions and deletions, are mainly located at sites of putative intrastrand crosslinks. We find two of the newly defined cisplatin-specific mutation types as causes of the reversion of BRCA2 mutations in emerging cisplatin-resistant tumours or cell clones. Gemcitabine, 5-fluorouracil, hydroxyurea, doxorubicin and paclitaxel have no measurable mutagenic effect. The cisplatin-induced mutation spectrum shows good correlation with cancer mutation signatures attributed to smoking and other sources of guanine-directed base damage. CONCLUSION This study provides support for the use of cell line mutagenesis assays to validate or predict the mutagenic effect of environmental and iatrogenic exposures. Our results suggest genetic reversion due to cisplatin-induced mutations as a distinct mechanism for developing resistance.
Collapse
Affiliation(s)
- Bernadett Szikriszt
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, 1117, Budapest, Hungary
| | - Ádám Póti
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, 1117, Budapest, Hungary
| | - Orsolya Pipek
- Department of Physics of Complex Systems, Eötvös Loránd University, 1117, Budapest, Hungary
| | - Marcin Krzystanek
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, 2800, Lyngby, Denmark
| | - Nnennaya Kanu
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK
| | - János Molnár
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, 1117, Budapest, Hungary
| | - Dezső Ribli
- Department of Physics of Complex Systems, Eötvös Loránd University, 1117, Budapest, Hungary
| | - Zoltán Szeltner
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, 1117, Budapest, Hungary
| | - Gábor E Tusnády
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, 1117, Budapest, Hungary
| | - István Csabai
- Department of Physics of Complex Systems, Eötvös Loránd University, 1117, Budapest, Hungary
| | - Zoltan Szallasi
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, 2800, Lyngby, Denmark.
- Computational Health Informatics Program (CHIP), Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, 02215, USA.
- MTA-SE-NAP, Brain Metastasis Research Group, 2nd Department of Pathology, Semmelweis University, 1091, Budapest, Hungary.
| | - Charles Swanton
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK.
- Francis Crick Institute, 44 Lincoln's Inn Fields, London, WCA2 3PX, UK.
| | - Dávid Szüts
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, 1117, Budapest, Hungary.
| |
Collapse
|