1
|
Carconi C, Bosi C, Scartozzi M, Cergnul M, Cinausero M, Faloppi L, Garajova I, Lonardi S, Pecora I, Pisanu L, Spadi R, Spallanzani A, Peretti U, Macchini M, Orsi G, Reni M. A pilot study of chlorambucil in pre-treated metastatic pancreatic adenocarcinoma patients bearing germline BRCA or other DNA damage repair system variants. Pancreatology 2024:S1424-3903(24)00736-1. [PMID: 39277480 DOI: 10.1016/j.pan.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/27/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
BACKGORUND Pancreatic adenocarcinoma remains a malignancy with a grim prognosis and scarce personalized treatment options. Pathogenic variants of DNA damage repair (DDR) genes are emerging as molecular targets, as they confer a higher sensitivity to DNA-damaging agents. This study aimed at assessing the activity of chlorambucil as salvage therapy in metastatic pancreatic cancer patients bearing a germline pathogenetic variant or variant of uncertain significance on a DDR-related gene. METHODS Platinum-pretreated metastatic pancreatic cancer patients harbouring a germline variant on a DDR gene received chlorambucil at a daily oral dose of 6 mg/m2 for 42 every 56 days for the first cycle and for 14 every 28 days for the following cycles, until disease progression or unacceptable toxicity. The primary endpoint was 6-month progression-free survival rate (PFS-6). Median progression-free survival (PFS) and overall survival (OS) were secondarily described. RESULTS Twenty patients were enrolled between December 2020 and September 2022. PFS-6 was 5%, median PFS and OS were 1.6 months and 3.0 months, respectively. Grade-3 adverse events were observed in 25% of patients, while no Grade-4 toxicity was reported. CONCLUSIONS Single agent chlorambucil did not show sufficient signal of activity to warrant its further investigation in metastatic pancreatic cancer patients bearing a DDR-related germline alteration.
Collapse
Affiliation(s)
- Catia Carconi
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Carlo Bosi
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Mario Scartozzi
- Medical Oncology, University and University Hospital, Cagliari, Italy
| | - Massimiliano Cergnul
- Medical Oncology, Ospedale Civile di Legnano - ASST Ovest Milanese, Legnano, Italy
| | - Marika Cinausero
- Department of Oncology, Azienda Sanitaria Universitaria Friuli Centrale Santa Maria Della Misericordia, Udine, Italy
| | - Luca Faloppi
- Medical Oncology Unit, Ospedali Santa Maria Della Pietà e Bartolomeo Eustachio - AST di Macerata, Camerino, San Severino Marche, Italy
| | - Ingrid Garajova
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Sara Lonardi
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Irene Pecora
- Department of Medical Oncology, Azienda Toscana Sud-Est, Misericordia Hospital, Grosseto, Italy
| | | | - Rosella Spadi
- Department of Oncology, Medical Oncology, 1, Città Della Salute e Della Scienza, Turin, Italy
| | - Andrea Spallanzani
- Division of Oncology, Department of Oncology and Hematology, University Hospital Modena, Modena, Italy
| | - Umberto Peretti
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Marina Macchini
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Giulia Orsi
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Michele Reni
- Department of Medical Oncology, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
2
|
Yang J, Ouedraogo SY, Wang J, Li Z, Feng X, Ye Z, Zheng S, Li N, Zhan X. Clinically relevant stratification of lung squamous carcinoma patients based on ubiquitinated proteasome genes for 3P medical approach. EPMA J 2024; 15:67-97. [PMID: 38463626 PMCID: PMC10923771 DOI: 10.1007/s13167-024-00352-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 01/30/2024] [Indexed: 03/12/2024]
Abstract
Relevance The proteasome is a crucial mechanism that regulates protein fate and eliminates misfolded proteins, playing a significant role in cellular processes. In the context of lung cancer, the proteasome's regulatory function is closely associated with the disease's pathophysiology, revealing multiple connections within the cell. Therefore, studying proteasome inhibitors as a means to identify potential pathways in carcinogenesis and metastatic progression is crucial in in-depth insight into its molecular mechanism and discovery of new therapeutic target to improve its therapy, and establishing effective biomarkers for patient stratification, predictive diagnosis, prognostic assessment, and personalized treatment for lung squamous carcinoma in the framework of predictive, preventive, and personalized medicine (PPPM; 3P medicine). Methods This study identified differentially expressed proteasome genes (DEPGs) in lung squamous carcinoma (LUSC) and developed a gene signature validated through Kaplan-Meier analysis and ROC curves. The study used WGCNA analysis to identify proteasome co-expression gene modules and their interactions with the immune system. NMF analysis delineated distinct LUSC subtypes based on proteasome gene expression patterns, while ssGSEA analysis quantified immune gene-set abundance and classified immune subtypes within LUSC samples. Furthermore, the study examined correlations between clinicopathological attributes, immune checkpoints, immune scores, immune cell composition, and mutation status across different risk score groups, NMF clusters, and immunity clusters. Results This study utilized DEPGs to develop an eleven-proteasome gene-signature prognostic model for LUSC, which divided samples into high-risk and low-risk groups with significant overall survival differences. NMF analysis identified six distinct LUSC clusters associated with overall survival. Additionally, ssGSEA analysis classified LUSC samples into four immune subtypes based on the abundance of immune cell infiltration with clinical relevance. A total of 145 DEGs were identified between high-risk and low-risk score groups, which had significant biological effects. Moreover, PSMD11 was found to promote LUSC progression by depending on the ubiquitin-proteasome system for degradation. Conclusions Ubiquitinated proteasome genes were effective in developing a prognostic model for LUSC patients. The study emphasized the critical role of proteasomes in LUSC processes, such as drug sensitivity, immune microenvironment, and mutation status. These data will contribute to the clinically relevant stratification of LUSC patients for personalized 3P medical approach. Further, we also recommend the application of the ubiquitinated proteasome system in multi-level diagnostics including multi-omics, liquid biopsy, prediction and targeted prevention of chronic inflammation and metastatic disease, and mitochondrial health-related biomarkers, for LUSC 3PM practice. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00352-w.
Collapse
Affiliation(s)
- Jingru Yang
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People's Republic of China
| | - Serge Yannick Ouedraogo
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People's Republic of China
| | - Jingjing Wang
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University, 440 Jiyan Road, Jinan, Shandong 250117 People's Republic of China
| | - Zhijun Li
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People's Republic of China
| | - Xiaoxia Feng
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People's Republic of China
| | - Zhen Ye
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People's Republic of China
- School of Basic Medicine, Shandong First Medical University, 6699 Qingdao Road, Jinan, Shandong 250117 People's Republic of China
| | - Shu Zheng
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People's Republic of China
| | - Na Li
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People's Republic of China
| | - Xianquan Zhan
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People's Republic of China
| |
Collapse
|
3
|
Rudd SG. Targeting pan-essential pathways in cancer with cytotoxic chemotherapy: challenges and opportunities. Cancer Chemother Pharmacol 2023; 92:241-251. [PMID: 37452860 PMCID: PMC10435635 DOI: 10.1007/s00280-023-04562-3] [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: 02/06/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023]
Abstract
Cytotoxic chemotherapy remains a key modality in cancer treatment. These therapies, successfully used for decades, continue to transform the lives of cancer patients daily. With the high attrition rate of current oncology drug development, combined with the knowledge that most new therapies do not displace standard-of-care treatments and that many healthcare systems cannot afford these new therapies; cytotoxic chemotherapies will remain an important component of cancer therapy for many years to come. The clinical value of these therapies is often under-appreciated within the pre-clinical cancer research community, where this diverse class of agents are often grouped together as non-specific cellular poisons killing tumor cells based solely upon proliferation rate; however, this is inaccurate. This review article seeks to reaffirm the importance of focusing research efforts upon improving our basic understanding of how these drugs work, discussing their ability to target pan-essential pathways in cancer cells, the relationship of this to the chemotherapeutic window, and highlighting basic science approaches that can be employed towards refining their use.
Collapse
Affiliation(s)
- Sean G Rudd
- Science For Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
4
|
Longo MA, Roy S, Chen Y, Tomaszowski KH, Arvai AS, Pepper JT, Boisvert RA, Kunnimalaiyaan S, Keshvani C, Schild D, Bacolla A, Williams GJ, Tainer JA, Schlacher K. RAD51C-XRCC3 structure and cancer patient mutations define DNA replication roles. Nat Commun 2023; 14:4445. [PMID: 37488098 PMCID: PMC10366140 DOI: 10.1038/s41467-023-40096-1] [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: 04/12/2023] [Accepted: 07/07/2023] [Indexed: 07/26/2023] Open
Abstract
RAD51C is an enigmatic predisposition gene for breast, ovarian, and prostate cancer. Currently, missing structural and related functional understanding limits patient mutation interpretation to homology-directed repair (HDR) function analysis. Here we report the RAD51C-XRCC3 (CX3) X-ray co-crystal structure with bound ATP analog and define separable RAD51C replication stability roles informed by its three-dimensional structure, assembly, and unappreciated polymerization motif. Mapping of cancer patient mutations as a functional guide confirms ATP-binding matching RAD51 recombinase, yet highlights distinct CX3 interfaces. Analyses of CRISPR/Cas9-edited human cells with RAD51C mutations combined with single-molecule, single-cell and biophysics measurements uncover discrete CX3 regions for DNA replication fork protection, restart and reversal, accomplished by separable functions in DNA binding and implied 5' RAD51 filament capping. Collective findings establish CX3 as a cancer-relevant replication stress response complex, show how HDR-proficient variants could contribute to tumor development, and identify regions to aid functional testing and classification of cancer mutations.
Collapse
Affiliation(s)
- Michael A Longo
- Department of Molecular & Cellular Oncology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Sunetra Roy
- Department of Cancer Biology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Yue Chen
- Department of Cancer Biology, UT MD Anderson Cancer Center, Houston, TX, USA
| | | | - Andrew S Arvai
- The Department of Integrative Structural & Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Jordan T Pepper
- Department of Biochemistry and Molecular Biology, Robson DNA Science Centre, Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Rebecca A Boisvert
- Department of Cancer Biology, UT MD Anderson Cancer Center, Houston, TX, USA
| | | | - Caezanne Keshvani
- Department of Cancer Biology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - David Schild
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Albino Bacolla
- Department of Molecular & Cellular Oncology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Gareth J Williams
- Department of Biochemistry and Molecular Biology, Robson DNA Science Centre, Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - John A Tainer
- Department of Molecular & Cellular Oncology, UT MD Anderson Cancer Center, Houston, TX, USA.
- Department of Cancer Biology, UT MD Anderson Cancer Center, Houston, TX, USA.
| | - Katharina Schlacher
- Department of Cancer Biology, UT MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
5
|
Fay EM, Newton A, Berney M, El‐Sagheer AH, Brown T, McGouran JF. Two-Step Validation Approach for Tools To Study the DNA Repair Enzyme SNM1A. Chembiochem 2023; 24:e202200756. [PMID: 36917742 PMCID: PMC10962688 DOI: 10.1002/cbic.202200756] [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: 12/16/2022] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 03/16/2023]
Abstract
We report a two-step validation approach to evaluate the suitability of metal-binding groups for targeting DNA damage-repair metalloenzymes using model enzyme SNM1A. A fragment-based screening approach was first used to identify metal-binding fragments suitable for targeting the enzyme. Effective fragments were then incorporated into oligonucleotides using the copper-catalysed azide-alkyne cycloaddition reaction. These modified oligonucleotides were recognised by SNM1A at >1000-fold lower concentrations than their fragment counterparts. The exonuclease SNM1A is a key enzyme involved in the repair of interstrand crosslinks, a highly cytotoxic form of DNA damage. However, SNM1A and other enzymes of this class are poorly understood, as there is a lack of tools available to facilitate their study. Our novel approach of incorporating functional fragments into oligonucleotides is broadly applicable to generating modified oligonucleotide structures with high affinity for DNA damage-repair enzymes.
Collapse
Affiliation(s)
- Ellen M. Fay
- School of Chemistry and Trinity Biomedical Sciences InstituteTrinity College DublinThe University of DublinDublin 2D02 R590Ireland
| | - Ailish Newton
- School of Chemistry and Trinity Biomedical Sciences InstituteTrinity College DublinThe University of DublinDublin 2D02 R590Ireland
| | - Mark Berney
- School of Chemistry and Trinity Biomedical Sciences InstituteTrinity College DublinThe University of DublinDublin 2D02 R590Ireland
| | - Afaf H. El‐Sagheer
- Department of ChemistryUniversity of OxfordMansfield RoadOX1 3TAOxfordUK
| | - Tom Brown
- Department of ChemistryUniversity of OxfordMansfield RoadOX1 3TAOxfordUK
| | - Joanna F. McGouran
- School of Chemistry and Trinity Biomedical Sciences InstituteTrinity College DublinThe University of DublinDublin 2D02 R590Ireland
| |
Collapse
|
6
|
Xie T, Dickson KA, Yee C, Ma Y, Ford CE, Bowden NA, Marsh DJ. Targeting Homologous Recombination Deficiency in Ovarian Cancer with PARP Inhibitors: Synthetic Lethal Strategies That Impact Overall Survival. Cancers (Basel) 2022; 14:4621. [PMID: 36230543 PMCID: PMC9563432 DOI: 10.3390/cancers14194621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/09/2022] [Accepted: 09/21/2022] [Indexed: 11/23/2022] Open
Abstract
The advent of molecular targeted therapies has made a significant impact on survival of women with ovarian cancer who have defects in homologous recombination repair (HRR). High-grade serous ovarian cancer (HGSOC) is the most common histological subtype of ovarian cancer, with over 50% displaying defective HRR. Poly ADP ribose polymerases (PARPs) are a family of enzymes that catalyse the transfer of ADP-ribose to target proteins, functioning in fundamental cellular processes including transcription, chromatin remodelling and DNA repair. In cells with deficient HRR, PARP inhibitors (PARPis) cause synthetic lethality leading to cell death. Despite the major advances that PARPis have heralded for women with ovarian cancer, questions and challenges remain, including: can the benefits of PARPis be brought to a wider range of women with ovarian cancer; can other drugs in clinical use function in a similar way or with greater efficacy than currently clinically approved PARPis; what can we learn from long-term responders to PARPis; can PARPis sensitise ovarian cancer cells to immunotherapy; and can synthetic lethal strategies be employed more broadly to develop new therapies for women with ovarian cancer. We examine these, and other, questions with focus on improving outcomes for women with ovarian cancer.
Collapse
Affiliation(s)
- Tao Xie
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Kristie-Ann Dickson
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Christine Yee
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Yue Ma
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Caroline E. Ford
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Nikola A. Bowden
- Centre for Drug Repurposing and Medicines Research, University of Newcastle, Newcastle, NSW 2289, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW 2289, Australia
- Hunter Medical Research Institute, Newcastle, NSW 2289, Australia
| | - Deborah J. Marsh
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
7
|
van der Wiel AMA, Schuitmaker L, Cong Y, Theys J, Van Hoeck A, Vens C, Lambin P, Yaromina A, Dubois LJ. Homologous Recombination Deficiency Scar: Mutations and Beyond-Implications for Precision Oncology. Cancers (Basel) 2022; 14:cancers14174157. [PMID: 36077694 PMCID: PMC9454578 DOI: 10.3390/cancers14174157] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 02/05/2023] Open
Abstract
Homologous recombination deficiency (HRD) is a prevalent in approximately 17% of tumors and is associated with enhanced sensitivity to anticancer therapies inducing double-strand DNA breaks. Accurate detection of HRD would therefore allow improved patient selection and outcome of conventional and targeted anticancer therapies. However, current clinical assessment of HRD mainly relies on determining germline BRCA1/2 mutational status and is insufficient for adequate patient stratification as mechanisms of HRD occurrence extend beyond functional BRCA1/2 loss. HRD, regardless of BRCA1/2 status, is associated with specific forms of genomic and mutational signatures termed HRD scar. Detection of this HRD scar might therefore be a more reliable biomarker for HRD. This review discusses and compares different methods of assessing HRD and HRD scar, their advances into the clinic, and their potential implications for precision oncology.
Collapse
Affiliation(s)
- Alexander M. A. van der Wiel
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Lesley Schuitmaker
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ying Cong
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Jan Theys
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Arne Van Hoeck
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Conchita Vens
- Institute of Cancer Science, University of Glasgow, Glasgow G61 1BD, Scotland, UK
- Department of Radiation Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Philippe Lambin
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ludwig J. Dubois
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
- Correspondence:
| |
Collapse
|
8
|
Antitumor effects of pyrrole-imidazole polyamide modified with alkylating agent on prostate cancer cells. Biochem Biophys Res Commun 2022; 623:9-16. [PMID: 35868070 DOI: 10.1016/j.bbrc.2022.07.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 11/21/2022]
Abstract
Androgens and androgen receptor (AR) have a central role in prostate cancer progression by regulating its downstream signaling. Although androgen depletion therapy (ADT) is the primary treatment for most prostate cancers, they acquires resistance to ADT and become castration resistant prostate cancers (CRPC). AR complex formation with multiple transcription factors is important for enhancer activity and transcriptional regulation, which can contribute to cancer progression and resistance to ADT. We previously demonstrated that OCT1 collaborates with AR in prostate cancer, and that a pyrrole-imidazole (PI) polyamide (PIP) targeting OCT1 inhibits cell and castration-resistant tumor growth (Obinata D et al. Oncogene 2016). PIP can bind to DNA non-covalently without a drug delivery system unlike most DNA targeted therapeutics. In the present study, we developed a PIP modified with a DNA alkylating agent, chlorambucil (ChB) (OCT1-PIP-ChB). Then its effect on the growth of prostate cancer LNCaP, 22Rv1, and PC3 cells, pancreatic cancer BxPC3 cells, and colon cancer HCT116 cells, as well as non-cancerous MCF-10A epithelial cells, were analyzed. It was shown that the IC50s of OCT1-PIP-ChB for 22Rv1 and LNCaP were markedly lower compared to other cells, including non-cancerous MCF-10A cells. Comprehensive gene expression analysis of CRPC model 22Rv1 cells treated with IC50 concentrations of OCT1-PIP-ChB revealed that the gene group involved in DNA double-strand break repair was the most enriched among gene sets repressed by OCT1-PIP-ChB treatment. Importantly, in vivo study using 22Rv1 xenografts, we showed that OCT1-PIP-ChB significantly reduced tumor growth compared to the control group without showing obvious adverse effects. Thus, the PIP combined with ChB can exert a significant inhibitory effect on prostate cancer cell proliferation and castration-resistant tumor growth, suggesting a potential role as a therapeutic agent.
Collapse
|
9
|
Wang N, Gu Y, Li L, Chi J, Liu X, Xiong Y, Zhong C. Development and Validation of a Prognostic Classifier Based on Lipid Metabolism-Related Genes for Breast Cancer. J Inflamm Res 2022; 15:3477-3499. [PMID: 35726216 PMCID: PMC9206459 DOI: 10.2147/jir.s357144] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 06/07/2022] [Indexed: 11/23/2022] Open
Abstract
Background The changes of lipid metabolism have been implicated in the development of many tumors, but its role in breast invasive carcinoma (BRCA) remains to be fully established. Here, we attempted to ascertain the prognostic value of lipid metabolism-related genes in BRCA. Methods We obtained RNA expression data and clinical information for BRCA and normal samples from public databases and downloaded a lipid metabolism-related gene set. Ingenuity Pathway Analysis (IPA) was applied to identify the potential pathways and functions of Differentially Expressed Genes (DEGs) related to lipid metabolism. Subsequently, univariate and multivariate Cox regression analyses were utilized to construct the prognostic gene signature. Functional enrichment analysis of prognostic genes was achieved by the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Kaplan-Meier analysis, Receiver Operating Characteristic (ROC) curves, clinical follow-up results were employed to assess the prognostic potency. Potential compounds targeting prognostic genes were screened by Connectivity Map (CMap) database and a prognostic gene-drug interaction network was constructed using Comparative Toxicogenomics Database (CTD). Furthermore, we separately validated the selected marker genes in BRCA samples and human breast cancer cell lines (MCF-7, MDA-MB-231). Results IPA and functional enrichment analysis demonstrated that the 162 lipid metabolism-related DEGs we obtained were involved in many lipid metabolism and BRCA pathological signatures. The prognostic classifier we constructed comprising SDC1 and SORBS1 can serve as an independent prognostic marker for BRCA. CMap filtered 37 potential compounds against prognostic genes, of which 16 compounds could target both two prognostic genes were identified by CTD. The functions of the two prognostic genes in breast cancer cells were verified by cell function experiments. Conclusion Within this study, we identified a novel prognostic classifier based on two lipid metabolism-related genes: SDC1 and SORBS1. This result highlighted a new perspective on the metabolic exploration of BRCA.
Collapse
Affiliation(s)
- Nan Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yuanting Gu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Lin Li
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Jiangrui Chi
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Xinwei Liu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Youyi Xiong
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Chaochao Zhong
- Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
10
|
Kim Y, Ahn I, Cho HN, Gwon H, Kang HJ, Seo H, Choi H, Kim KP, Jun TJ, Kim YH. RIDAB: Electronic medical record-integrated real world data platform for predicting and summarizing interactions in biomedical research from heterogeneous data resources. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 221:106866. [PMID: 35594580 DOI: 10.1016/j.cmpb.2022.106866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 04/27/2022] [Accepted: 05/07/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND OBJECTIVE With the advent of bioinformatics, biological databases have been constructed to computerize data. Biological systems can be described as interactions and relationships between elements constituting the systems, and they are organized in various biomedical open databases. These open databases have been used in approaches to predict functional interactions such as protein-protein interactions (PPI), drug-drug interactions (DDI) and disease-disease relationships (DDR). However, just combining interaction data has limited effectiveness in predicting the complex relationships occurring in a whole context. Each contributing source contains information on each element in a specific field of knowledge but there is a lack of inter-disciplinary insight in combining them. METHODS In this study, we propose the RWD Integrated platform for Discovering Associations in Biomedical research (RIDAB) to predict interactions between biomedical entities. RIDAB is established as a graph network to construct a platform that predicts the interactions of target entities. Biomedical open database is combined with EMRs each representing a biomedical network and a real-world data. To integrate databases from different domains to build the platform, mapping of the vocabularies was required. In addition, the appropriate structure of the network and the graph embedding method to be used were needed to be selected to fit the tasks. RESULTS The feasibility of the platform was evaluated using node similarity and link prediction for drug repositioning task, a commonly used task for biomedical network. In addition, we compared the US Food and Drug Administration (FDA)-approved repositioned drugs with the predicted result. By integrating EMR database with biomedical networks, the platform showed increased f1 score in predicting repositioned drugs, from 45.62% to 57.26%, compared to platforms based on biomedical networks alone. CONCLUSIONS This study demonstrates that the elements of biomedical research findings can be reflected by integrating EMR data with open-source biomedical networks. In addition, showed the feasibility of using the established platform to represent the integration of biomedical networks and reflected the relationship between real world networks.
Collapse
Affiliation(s)
- Yunha Kim
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea.
| | - Imjin Ahn
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea.
| | - Ha Na Cho
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea.
| | - Hansle Gwon
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea.
| | - Hee Jun Kang
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea.
| | - Hyeram Seo
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea.
| | - Heejung Choi
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea.
| | - Kyu-Pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea.
| | - Tae Joon Jun
- Big Data Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea.
| | - Young-Hak Kim
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea.
| |
Collapse
|
11
|
Devico Marciano N, Kroening G, Dayyani F, Zell JA, Lee FC, Cho M, Valerin JG. BRCA-Mutated Pancreatic Cancer: From Discovery to Novel Treatment Paradigms. Cancers (Basel) 2022; 14:cancers14102453. [PMID: 35626055 PMCID: PMC9140002 DOI: 10.3390/cancers14102453] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/02/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Approximately 10–20% of pancreatic cancer patients will have a mutation in their DNA, passed on in families, that contributes to the development of their pancreatic cancer. These mutations are important in that they effect the biology of the disease as well as contribute to sensitivity to specific treatments. We describe the critical role that these genes play in various cellular processes in the body that contribute to their role in cancer development and normal cellular function. In this review, we aim to describe the role of certain genes (BRCA1 and BRCA2) in the development of pancreatic cancer and the current and future research efforts underway to treat this subtype of disease. Abstract The discovery of BRCA1 and BRCA2 in the 1990s revolutionized the way we research and treat breast, ovarian, and pancreatic cancers. In the case of pancreatic cancers, germline mutations occur in about 10–20% of patients, with mutations in BRCA1 and BRCA2 being the most common. BRCA genes are critical in DNA repair pathways, particularly in homologous recombination, which has a serious impact on genomic stability and can contribute to cancerous cell proliferation. However, BRCA1 also plays a fundamental role in cell cycle checkpoint control, ubiquitination, control of gene expression, and chromatin remodeling, while BRCA2 also plays a role in transcription and immune system response. Therefore, mutations in these genes lead to multiple defects in cells that may be utilized when treating cancer. BRCA mutations seem to confer a prognostic benefit with an improved overall survival due to differing underlying biology. These mutations also appear to be a predictive marker, with patients showing increased sensitivity to certain treatments, such as platinum chemotherapy and PARP inhibitors. Olaparib is currently indicated for maintenance therapy in metastatic PDAC after induction with platinum-based chemotherapy. Resistance has been found to these therapies, and with a 10.8% five-year OS, novel therapies are desperately needed.
Collapse
|
12
|
Groelly FJ, Porru M, Zimmer J, Benainous H, De Visser Y, Kosova AA, Di Vito S, Serra V, Ryan A, Leonetti C, Bruna A, Biroccio A, Tarsounas M. Anti-tumoural activity of the G-quadruplex ligand pyridostatin against BRCA1/2-deficient tumours. EMBO Mol Med 2022; 14:e14501. [PMID: 35107878 PMCID: PMC8899905 DOI: 10.15252/emmm.202114501] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 12/20/2021] [Accepted: 01/07/2022] [Indexed: 12/28/2022] Open
Abstract
The cells with compromised BRCA1 or BRCA2 (BRCA1/2) function accumulate stalled replication forks, which leads to replication‐associated DNA damage and genomic instability, a signature of BRCA1/2‐mutated tumours. Targeted therapies against BRCA1/2‐mutated tumours exploit this vulnerability by introducing additional DNA lesions. Because homologous recombination (HR) repair is abrogated in the absence of BRCA1 or BRCA2, these lesions are specifically lethal to tumour cells, but not to the healthy tissue. Ligands that bind and stabilise G‐quadruplexes (G4s) have recently emerged as a class of compounds that selectively eliminate the cells and tumours lacking BRCA1 or BRCA2. Pyridostatin is a small molecule that binds G4s and is specifically toxic to BRCA1/2‐deficient cells in vitro. However, its in vivo potential has not yet been evaluated. Here, we demonstrate that pyridostatin exhibits a high specific activity against BRCA1/2‐deficient tumours, including patient‐derived xenograft tumours that have acquired PARP inhibitor (PARPi) resistance. Mechanistically, we demonstrate that pyridostatin disrupts replication leading to DNA double‐stranded breaks (DSBs) that can be repaired in the absence of BRCA1/2 by canonical non‐homologous end joining (C‐NHEJ). Consistent with this, chemical inhibitors of DNA‐PKcs, a core component of C‐NHEJ kinase activity, act synergistically with pyridostatin in eliminating BRCA1/2‐deficient cells and tumours. Furthermore, we demonstrate that pyridostatin triggers cGAS/STING‐dependent innate immune responses when BRCA1 or BRCA2 is abrogated. Paclitaxel, a drug routinely used in cancer chemotherapy, potentiates the in vivo toxicity of pyridostatin. Overall, our results demonstrate that pyridostatin is a compound suitable for further therapeutic development, alone or in combination with paclitaxel and DNA‐PKcs inhibitors, for the benefit of cancer patients carrying BRCA1/2 mutations.
Collapse
Affiliation(s)
- Florian J Groelly
- Genome Stability and Tumourigenesis Group, The MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Manuela Porru
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Jutta Zimmer
- Genome Stability and Tumourigenesis Group, The MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Hugo Benainous
- Genome Stability and Tumourigenesis Group, The MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Yanti De Visser
- Genome Stability and Tumourigenesis Group, The MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Anastasiya A Kosova
- Genome Stability and Tumourigenesis Group, The MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Serena Di Vito
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy.,Department of Ecological and Biological Sciences (DEB), University of Tuscia, Viterbo, Italy
| | - Violeta Serra
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Anderson Ryan
- Lung Cancer Translational Science Research Group, The MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Carlo Leonetti
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alejandra Bruna
- Molecular Pathology Division, Centre for Cancer Evolution, The Institute of Cancer Research, London, UK
| | - Annamaria Biroccio
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Madalena Tarsounas
- Genome Stability and Tumourigenesis Group, The MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| |
Collapse
|
13
|
Macchini M, Centonze F, Peretti U, Orsi G, Militello AM, Valente MM, Cascinu S, Reni M. Treatment opportunities and future perspectives for pancreatic cancer patients with germline BRCA1-2 pathogenic variants. Cancer Treat Rev 2021; 100:102262. [PMID: 34418781 DOI: 10.1016/j.ctrv.2021.102262] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/17/2021] [Accepted: 07/19/2021] [Indexed: 01/07/2023]
Abstract
Personalized treatments and predictive biomarkers of pancreatic cancer (PDAC) are still lacking. Recently germline mutations in BRCA 1 and 2 genes, leading to homologous repair deficiency, have emerged as new targets for more specific and effective therapies, exploiting the increased susceptibility to platinum salts and PARP inhibitors. In addition to BRCA, pathogenic variants in PALB2 and in other genes involved in the DNA damage response pathway (DDR) represent potential targets, as well as their respective somatic alterations. This enlarged molecularly-selected population sharing the BRCAness phenotype, is expected to show a higher sensibility to a number of DNA damaging agents and DDR inhibitors. However, the possibility of new therapeutic opportunities for DDR defective PDAC patients has to face the lack of solid evidence about the proper type and timing of targeted-treatments, the potential combination strategies and most importantly, the lack of informations on the functional impact of each specific pathogenic variant on the DDR pathway. This review summarizes the current and near-future options for the clinical management of PDAC patients harboring a DDR deficiency, analyzing the state of the art of the indications of platinum salts and other cytotoxic agents in the advanced and early stage PDAC, the development of PARP inhibitors and the rational for new combinations with immunotherapy and cycle checkpoint inhibitors, as well as the strategy to overcome the development of resistance over treatments.
Collapse
Affiliation(s)
- Marina Macchini
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy; Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Federico Centonze
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy; Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Umberto Peretti
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy; Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Orsi
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy; Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Anna Maria Militello
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy; Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Maria Maddalena Valente
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy; Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Cascinu
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy; Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy
| | - Michele Reni
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy; Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
14
|
Cheun YK, Groehler AS, Schärer OD. New Synthetic Analogs of Nitrogen Mustard DNA Interstrand Cross-Links and Their Use to Study Lesion Bypass by DNA Polymerases. Chem Res Toxicol 2021; 34:1790-1799. [PMID: 34133118 PMCID: PMC11246215 DOI: 10.1021/acs.chemrestox.1c00123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Nitrogen mustards are a widely used class of antitumor agents that exert their cytotoxic effects through the formation of DNA interstrand cross-links (ICLs). Despite being among the first antitumor agents used, the biological responses to NM ICLs remain only partially understood. We have previously reported the generation of NM ICL mimics by incorporation of ICL precursors into DNA using solid-phase synthesis at defined positions, followed by a double reductive amination reaction. However, the structure of these mimics deviated from the native NM ICLs. Using further development of our approach, we report a new class of NM ICL mimics that only differ from their native counterpart by substitution of dG with 7-deaza-dG at the ICL. Importantly, this approach allows for the synthesis of diverse NM ICLs, illustrated here with a mimic of the adduct formed by chlorambucil. We used the newly generated ICLs in reactions with replicative and translesion synthesis DNA polymerase to demonstrate their stability and utility for functional studies. These new NM ICLs will allow for the further characterization of the biological responses to this important class of antitumor agents.
Collapse
Affiliation(s)
- Young K Cheun
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| | - Arnold S Groehler
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| | - Orlando D Schärer
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| |
Collapse
|
15
|
Liu X, Qiu R, Xu M, Meng M, Zhao S, Ji J, Yang Y. KMT2C is a potential biomarker of prognosis and chemotherapy sensitivity in breast cancer. Breast Cancer Res Treat 2021; 189:347-361. [PMID: 34240274 DOI: 10.1007/s10549-021-06325-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/05/2021] [Indexed: 12/27/2022]
Abstract
PURPOSE Epigenetic regulation plays critical roles in cancer progression, and high-frequency mutations or expression variations in epigenetic regulators have been frequently observed in tumorigenesis, serving as biomarkers and targets for cancer therapy. Here, we aimed to explore the function of epigenetic regulators in breast cancer. METHODS The mutational landscape of epigenetic regulators in breast cancer samples was investigated based on datasets from the Cancer Genome Atlas. The Kaplan-Meier method was used for survival analysis. RNA sequencing (RNA-seq) in MCF-7 cells transfected with control siRNA or KMT2C siRNA was performed. Quantitative reverse transcription-PCR and chromatin immunoprecipitation were used to validate the RNA-seq results. RESULTS Among the 450 epigenetic regulators, KMT2C was frequently mutated in breast cancer samples. The tumor mutational burden (TMB) was elevated in breast cancer samples with KMT2C mutations or low KMT2C mRNA levels compared to their counterparts with wild-type KMT2C or high KMT2C mRNA levels. Somatic mutation and low expression of KMT2C were independently correlated with the poor overall survival (OS) and disease-free survival (DFS) of the breast cancer samples, respectively. RNA-seq analysis combined with chromatin immunoprecipitation and qRT-PCR assays revealed that the depletion of KMT2C remarkably affected the expression of DNA damage repair-related genes. More importantly, the low expression of KMT2C was related to breast cancer cell sensitivity to chemotherapy and longer OS of breast cancer patients who underwent chemotherapy. CONCLUSION We conclude that KMT2C could serve as a potential biomarker of prognosis and chemotherapy sensitivity by affecting the DNA damage repair-related genes of breast cancer.
Collapse
Affiliation(s)
- Xinhua Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, 311121, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Rongfang Qiu
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, School of Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, Zhejiang, China.,Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Min Xu
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, School of Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, Zhejiang, China.,Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Miaomiao Meng
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, School of Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, Zhejiang, China.,Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Siyu Zhao
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, School of Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, Zhejiang, China.,Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Jiansong Ji
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, School of Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, Zhejiang, China. .,Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| | - Yang Yang
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, School of Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, Zhejiang, China. .,Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China. .,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
16
|
Buzon B, Grainger RA, Rzadki C, Huang SYM, Junop M. Identification of Bioactive SNM1A Inhibitors. ACS OMEGA 2021; 6:9352-9361. [PMID: 33869915 PMCID: PMC8047731 DOI: 10.1021/acsomega.0c03528] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 03/02/2021] [Indexed: 06/12/2023]
Abstract
SNM1A is a nuclease required to repair DNA interstrand cross-links (ICLs) caused by some anticancer compounds, including cisplatin. Unlike other nucleases involved in ICL repair, SNM1A is not needed to restore other forms of DNA damage. As such, SNM1A is an attractive target for selectively increasing the efficacy of ICL-based chemotherapy. Using a fluorescence-based exonuclease assay, we screened a bioactive library of compounds for inhibition of SNM1A. Of the 52 compounds initially identified as hits, 22 compounds showed dose-response inhibition of SNM1A. An orthogonal gel-based assay further confirmed nine small molecules as SNM1A nuclease activity inhibitors with IC50 values in the mid-nanomolar to low micromolar range. Finally, three compounds showed no toxicity at concentrations able to significantly potentiate the cytotoxicity of cisplatin. These compounds represent potential leads for further optimization to sensitize cells toward chemotherapeutic agents inducing ICL damage.
Collapse
Affiliation(s)
- Beverlee Buzon
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department
of Biochemistry, Western University, London, Ontario N6A 5C1, Canada
| | - Ryan A. Grainger
- Department
of Biochemistry, Western University, London, Ontario N6A 5C1, Canada
| | - Cameron Rzadki
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Simon York Ming Huang
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Murray Junop
- Department
of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4L8, Canada
- Department
of Biochemistry, Western University, London, Ontario N6A 5C1, Canada
| |
Collapse
|
17
|
Carraro C, Helbing T, Francke A, Zuravka I, Sosic A, De Franco M, Gandin V, Gatto B, Göttlich DR. Appended Aromatic Moieties in Flexible Bis-3-chloropiperidines Confer Tropism against Pancreatic Cancer Cells. ChemMedChem 2021; 16:860-868. [PMID: 33200541 PMCID: PMC7984046 DOI: 10.1002/cmdc.202000814] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Indexed: 12/24/2022]
Abstract
Nitrogen mustards (NMs) are an old but still largely diffused class of anticancer drugs. However, spreading mechanisms of resistance undermine their efficacy and therapeutic applicability. To expand their antitumour value, we developed bis-3-chloropiperidines (B-CePs), a new class of mustard-based alkylating agent, and we recently reported the striking selectivity for BxPC-3 pancreatic tumour cells of B-CePs bearing aromatic moieties embedded in the linker. In this study, we demonstrate that such tropism is shared by bis-3-chloropiperidines bearing appended aromatic groups in flexible linkers, whereas esters substituted by aliphatic groups or by efficient DNA-interacting groups are potent but nonselective cytotoxic agents. Besides, we describe how the critical balance between water stability and DNA reactivity can affect the properties of bis-3-chloropiperidines. Together, these findings support the exploitation of B-CePs as potential antitumour clinical candidates.
Collapse
Affiliation(s)
- Caterina Carraro
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Francesco Marzolo 535131PadovaItaly
| | - Tim Helbing
- Institute of Organic ChemistryJustus Liebig University GiessenHeinrich-Buff-Ring 1735392GiessenGermany
| | - Alexander Francke
- Institute of Organic ChemistryJustus Liebig University GiessenHeinrich-Buff-Ring 1735392GiessenGermany
| | - Ivonne Zuravka
- Institute of Organic ChemistryJustus Liebig University GiessenHeinrich-Buff-Ring 1735392GiessenGermany
| | - Alice Sosic
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Francesco Marzolo 535131PadovaItaly
| | - Michele De Franco
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Francesco Marzolo 535131PadovaItaly
| | - Valentina Gandin
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Francesco Marzolo 535131PadovaItaly
| | - Barbara Gatto
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Francesco Marzolo 535131PadovaItaly
| | - D. Richard Göttlich
- Institute of Organic ChemistryJustus Liebig University GiessenHeinrich-Buff-Ring 1735392GiessenGermany
| |
Collapse
|
18
|
Helbing T, Carraro C, Francke A, Sosic A, De Franco M, Gandin V, Göttlich R, Gatto B. Aromatic Linkers Unleash the Antiproliferative Potential of 3-Chloropiperidines Against Pancreatic Cancer Cells. ChemMedChem 2020; 15:2040-2051. [PMID: 32744774 PMCID: PMC7692949 DOI: 10.1002/cmdc.202000457] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Indexed: 12/27/2022]
Abstract
In this study, we describe the synthesis and biological evaluation of a set of bis-3-chloropiperidines (B-CePs) containing rigid aromatic linker structures. A modification of the synthetic strategy also enabled the synthesis of a pilot tris-3-chloropiperidine (Tri-CeP) bearing three reactive meta-chloropiperidine moieties on the aromatic scaffold. A structure-reactivity relationship analysis of B-CePs suggests that the arrangement of the reactive units affects the DNA alkylating activity, while also revealing correlations between the electron density of the aromatic system and the reactivity with biologically relevant nucleophiles, both on isolated DNA and in cancer cells. Interestingly, all aromatic 3-chloropiperidines exhibited a marked cytotoxicity and tropism for 2D and 3D cultures of pancreatic cancer cells. Therefore, the new aromatic 3-chloropiperidines appear to be promising contenders for further development of mustard-based anticancer agents aimed at pancreatic cancers.
Collapse
Affiliation(s)
- Tim Helbing
- Institute of Organic ChemistryJustus Liebig University GiessenHeinrich-Buff-Ring 1735392GiessenGermany
| | - Caterina Carraro
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Francesco Marzolo 535131PadovaItaly
| | - Alexander Francke
- Institute of Organic ChemistryJustus Liebig University GiessenHeinrich-Buff-Ring 1735392GiessenGermany
| | - Alice Sosic
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Francesco Marzolo 535131PadovaItaly
| | - Michele De Franco
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Francesco Marzolo 535131PadovaItaly
| | - Valentina Gandin
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Francesco Marzolo 535131PadovaItaly
| | - Richard Göttlich
- Institute of Organic ChemistryJustus Liebig University GiessenHeinrich-Buff-Ring 1735392GiessenGermany
| | - Barbara Gatto
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaVia Francesco Marzolo 535131PadovaItaly
| |
Collapse
|
19
|
Rieckhoff J, Meyer F, Classen S, Zielinski A, Riepen B, Wikman H, Petersen C, Rothkamm K, Borgmann K, Parplys AC. Exploiting Chromosomal Instability of PTEN-Deficient Triple-Negative Breast Cancer Cell Lines for the Sensitization against PARP1 Inhibition in a Replication-Dependent Manner. Cancers (Basel) 2020; 12:cancers12102809. [PMID: 33003585 PMCID: PMC7601067 DOI: 10.3390/cancers12102809] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/11/2020] [Accepted: 09/25/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary The poor prognosis of patients with TNBC have fostered a major effort to identify more patients who would benefit from targeted therapies. Here we recognize PTEN as a potential CIN-causing gene in TNBC and consider PTEN-deficient TNBC for the treatment with PARP1 inhibitors due to the protective role of PTEN during DNA replication. Abstract Chromosomal instability (CIN) is an emerging hallmark of cancer and its role in therapeutic responses has been increasingly attracting the attention of the research community. To target the vulnerability of tumors with high CIN, it is important to identify the genes and mechanisms involved in the maintenance of CIN. In our work, we recognize the tumor suppressor gene Phosphatase and Tensin homolog (PTEN) as a potential gene causing CIN in triple-negative breast cancer (TNBC) and show that TNBC with low expression levels of PTEN can be sensitized for the treatment with poly-(ADP-ribose)-polymerase 1 (PARP1) inhibitors, independent of Breast Cancer (BRCA) mutations or a BRCA-like phenotype. In silico analysis of mRNA expression data from 200 TNBC patients revealed low expression of PTEN in tumors with a high CIN70 score. Western blot analysis of TNBC cell lines confirm lower protein expression of PTEN compared to non TNBC cell lines. Further, PTEN-deficient cell lines showed cellular sensitivity towards PARP1 inhibition treatment. DNA fiber assays and examination of chromatin bound protein fractions indicate a protective role of PTEN at stalled replication forks. In this study, we recognize PTEN as a potential CIN-causing gene in TNBC and identify its important role in the replication processes.
Collapse
Affiliation(s)
- Johanna Rieckhoff
- Laboratory of Radiobiology & Experimental Radio Oncology, Centre of Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.R.); (F.M.); (S.C.); (A.Z.); (B.R.); (K.R.); (K.B.)
| | - Felix Meyer
- Laboratory of Radiobiology & Experimental Radio Oncology, Centre of Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.R.); (F.M.); (S.C.); (A.Z.); (B.R.); (K.R.); (K.B.)
| | - Sandra Classen
- Laboratory of Radiobiology & Experimental Radio Oncology, Centre of Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.R.); (F.M.); (S.C.); (A.Z.); (B.R.); (K.R.); (K.B.)
| | - Alexandra Zielinski
- Laboratory of Radiobiology & Experimental Radio Oncology, Centre of Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.R.); (F.M.); (S.C.); (A.Z.); (B.R.); (K.R.); (K.B.)
| | - Britta Riepen
- Laboratory of Radiobiology & Experimental Radio Oncology, Centre of Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.R.); (F.M.); (S.C.); (A.Z.); (B.R.); (K.R.); (K.B.)
| | - Harriet Wikman
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center, Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Cordula Petersen
- Department of Radiotherapy and Radio Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Kai Rothkamm
- Laboratory of Radiobiology & Experimental Radio Oncology, Centre of Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.R.); (F.M.); (S.C.); (A.Z.); (B.R.); (K.R.); (K.B.)
| | - Kerstin Borgmann
- Laboratory of Radiobiology & Experimental Radio Oncology, Centre of Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.R.); (F.M.); (S.C.); (A.Z.); (B.R.); (K.R.); (K.B.)
| | - Ann Christin Parplys
- Laboratory of Radiobiology & Experimental Radio Oncology, Centre of Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (J.R.); (F.M.); (S.C.); (A.Z.); (B.R.); (K.R.); (K.B.)
- Correspondence:
| |
Collapse
|
20
|
Chen X, Peng F, Ji Y, Xiang H, Wang X, Liu T, Wang H, Han Y, Wang C, Zhang Y, Kong X, Lang JY. Brca2 deficiency drives gastrointestinal tumor formation and is selectively inhibited by mitomycin C. Cell Death Dis 2020; 11:812. [PMID: 32980867 PMCID: PMC7519908 DOI: 10.1038/s41419-020-03013-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/13/2020] [Accepted: 09/15/2020] [Indexed: 12/24/2022]
Abstract
BRCA2 is crucial for repairing DNA double-strand breaks with high fidelity, and loss of BRCA2 increases the risks of developing breast and ovarian cancers. Herein, we show that BRCA2 is inactively mutated in 10% of gastric and 7% of colorectal adenocarcinomas, and that this inactivation is significantly correlated with microsatellite instability. Villin-driven Brca2 depletion promotes mouse gastrointestinal tumor formation when genome instability is increased. Whole-genome screening data showed that these BRCA2 monoallelic and biallelic mutant tumors were selectively inhibited by mitomycin C. Mechanistically, mitomycin C provoked double-strand breaks in cancer cells that often recruit wild-type BRCA2 for repair; the failure to repair double-strand breaks caused cell-cycle arrest at the S phase and p53-mediated cell apoptosis of BRCA2 monoallelic and biallelic mutant tumor cells. Our study unveils the role of BRCA2 loss in the development of gastrointestinal tumors and provides a potential therapeutic strategy to eliminate BRCA2 monoallelic and biallelic mutant tumors through mitomycin C.
Collapse
Affiliation(s)
- Xiaomin Chen
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Fangfei Peng
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yan Ji
- Bioinformatics Core, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Honggang Xiang
- Department of General Surgery, Pudong New Area People's Hospital affiliated to Shanghai University of Medicine & Health Science, 201299, Shanghai, China
| | - Xiang Wang
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Tingting Liu
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Heng Wang
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yumin Han
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Changxu Wang
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yongfeng Zhang
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xiangyin Kong
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Jing-Yu Lang
- CAS_Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China.
| |
Collapse
|
21
|
Liu L, Zhang Z, Zhang G, Wang T, Ma Y, Guo W. Down-regulation of PADI2 prevents proliferation and epithelial-mesenchymal transition in ovarian cancer through inhibiting JAK2/STAT3 pathway in vitro and in vivo, alone or in combination with Olaparib. J Transl Med 2020; 18:357. [PMID: 32951601 PMCID: PMC7504643 DOI: 10.1186/s12967-020-02528-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022] Open
Abstract
Background Epithelial ovarian cancer (EOC) is the most lethal disease among female genital malignant tumors. Peptidylarginine deiminase type II(PADI II) has been shown to enhance a variety of cancers carcinogenesis, including ovarian cancer. The purpose of this study was to investigate the biological role of PADI2 in ovarian cancer (OC) and the relative mechanism. Methods Gene Expression Profiling Interactive Analysis (GEPIA) (https://gepia.pku.cn/) and ONCOMINE (https://www.oncomine.org/) were used to analyze PADI2 Gene Expression data. The survival curve for the PADI2 gene was generated by using the online Kaplan–Meier mapping site (https://www.kmplot.com/). We conducted MTT assay, cloning formation assay and EdU cell proliferation assay to detect the cell activity of PADI2 knockdown A2780 and SKOV3 ovarian cancer cells treated with Olaparib. Cell migration and invasion were observed by would healing and transwell assay. The pathway changes after the treatment of PADI2 were detected by transcriptome sequencing and western blot. The role of PADI2 combined with Olaparib treatment in vivo was studied in nude mouse model bearing ovarian cancer tumor. Results We investigated the role of PADI2 on EOC in vitro and in vivo. PADI2 was upregulated in ovarian cancer samples and high PADI2 expression was correlated with poor outcome. Downregulating PADI2 suppressed colony formation, proliferation, migration and invasion of A2780 and SKOV3 cells. Furthermore, downregulating PADI2 and Olaparib combination treatment attenuated the viability, migration and invasion of A2780 and SKOV3 cells. We identified differentially expressed genes in A2780-shPADI2 and SKOV3-shPADI2 cell by transcriptome sequencing analysis and verified that downregulating PADI2 and Olaparib combination treatment suppresses EMT and JAK2/STAT3 signaling pathway in A2780 and SKOV3 cells in vitro and in vivo. Conclusions Downregulation of PADI2 and Olaparib combination treatment attenuated the proliferation, migration and invasion of A2780 and SKOV3 cells by inhibiting the EMT through JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Lidong Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, Shandong, People's Republic of China.,Medical Research Center, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, Shandong, People's Republic of China.,Obstetrics Department, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, People's Republic of China
| | - Zhiwei Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, Shandong, People's Republic of China
| | - Guoxiang Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, Shandong, People's Republic of China
| | - Ting Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, Shandong, People's Republic of China
| | - Yingchun Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, Shandong, People's Republic of China
| | - Wei Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, Shandong, People's Republic of China.
| |
Collapse
|
22
|
Shi B, Xu FF, Xiang CP, Jia R, Yan CH, Ma SQ, Wang N, Wang AJ, Fan P. Effect of sodium butyrate on ABC transporters in lung cancer A549 and colorectal cancer HCT116 cells. Oncol Lett 2020; 20:148. [PMID: 32934716 PMCID: PMC7471751 DOI: 10.3892/ol.2020.12011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/22/2020] [Indexed: 12/18/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors and DNA alkylators are effective components of combination chemotherapy. The aim of the present study was to investigate the possible mechanism of their synergism by detecting the effect of HDAC inhibitors on the expression levels of drug transporters that export DNA alkylators. It was demonstrated that the HDAC inhibitor sodium butyrate (NaB) induced the differential expression of multidrug resistant ATP-binding cassette (ABC) transporters in lung cancer and colorectal cancer cells. Specifically, NaB increased the mRNA expression levels of ABC subfamily B member 1 (ABCB1), ABCC10 and ABCC12, and protein expression levels of multidrug resistance-1 (MDR1), multidrug resistance-associated protein 7 (MRP7) and MRP9. Moreover, NaB decreased the expression levels of ABCC1, ABCC2 and ABCC3 mRNAs, as well as those of MRP1, MRP2 and MRP3 proteins. The molecular mechanism underlying this process was subsequently investigated. NaB decreased the expression of HDAC4, but not HDAC1, HDAC2 or HDAC3. In addition, NaB promoted histone H3 acetylation and methylation at lysine 9, as well as MDR1 acetylation, suggesting that acetylation and methylation may be involved in NaB-mediated ABC transporter expression. Thus, the present results indicated that the synergism of the HDAC inhibitors with the DNA alkylating agents may due to the inhibitory effect of MRPs by HDAC inhibitors. The findings also suggested the possibility of antagonistic effects following the combined treatment of HDAC inhibitors with MDR1 ligands.
Collapse
Affiliation(s)
- Bin Shi
- Department of Anorectal Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230000, P.R. China
| | - Fang-Fang Xu
- Department of Anorectal Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230000, P.R. China
| | - Cai-Ping Xiang
- Department of Anorectal Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230000, P.R. China
| | - Ru Jia
- Department of Anorectal Surgery, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230000, P.R. China
| | - Chun-Hong Yan
- Department of Anorectal Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230000, P.R. China
| | - Se-Qing Ma
- Department of Anorectal Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230000, P.R. China
| | - Ning Wang
- Department of Anorectal Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230000, P.R. China
| | - An-Jiao Wang
- Department of Anorectal Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230000, P.R. China
| | - Ping Fan
- Department of Anorectal Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230000, P.R. China
| |
Collapse
|
23
|
Tarsounas M, Sung P. The antitumorigenic roles of BRCA1-BARD1 in DNA repair and replication. Nat Rev Mol Cell Biol 2020; 21:284-299. [PMID: 32094664 PMCID: PMC7204409 DOI: 10.1038/s41580-020-0218-z] [Citation(s) in RCA: 187] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2020] [Indexed: 11/09/2022]
Abstract
The tumour suppressor breast cancer type 1 susceptibility protein (BRCA1) promotes DNA double-strand break (DSB) repair by homologous recombination and protects DNA replication forks from attrition. BRCA1 partners with BRCA1-associated RING domain protein 1 (BARD1) and other tumour suppressor proteins to mediate the initial nucleolytic resection of DNA lesions and the recruitment and regulation of the recombinase RAD51. The discovery of the opposing functions of BRCA1 and the p53-binding protein 1 (53BP1)-associated complex in DNA resection sheds light on how BRCA1 influences the choice of homologous recombination over non-homologous end joining and potentially other mutagenic pathways of DSB repair. Understanding the functional crosstalk between BRCA1-BARD1 and its cofactors and antagonists will illuminate the molecular basis of cancers that arise from a deficiency or misregulation of chromosome damage repair and replication fork maintenance. Such knowledge will also be valuable for understanding acquired tumour resistance to poly(ADP-ribose) polymerase (PARP) inhibitors and other therapeutics and for the development of new treatments. In this Review, we discuss recent advances in elucidating the mechanisms by which BRCA1-BARD1 functions in DNA repair, replication fork maintenance and tumour suppression, and its therapeutic relevance.
Collapse
Affiliation(s)
- Madalena Tarsounas
- Genome Stability and Tumourigenesis Group, Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK.
| | - Patrick Sung
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA.
| |
Collapse
|
24
|
Chang R, Chen L, Su G, Du L, Qin Y, Xu J, Tan H, Zhou C, Cao Q, Yuan G, Kijlstra A, Yang P. Identification of Ribosomal Protein S4, Y-Linked 1 as a cyclosporin A plus corticosteroid resistance gene. J Autoimmun 2020; 112:102465. [PMID: 32331925 DOI: 10.1016/j.jaut.2020.102465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/07/2020] [Accepted: 04/10/2020] [Indexed: 02/06/2023]
Abstract
Combination of corticosteroids (CS) with cyclosporin A (CsA) is widely used in the treatment of autoimmune diseases, autoinflammatory diseases and transplantation rejection. However, some patients fail to respond or develop resistance to the combination regimen. In Vogt-Koyanagi-Harada (VKH) disease model, we performed RNA sequencing (RNA-seq) based transcriptomics, isobaric tags for relative and absolute quantification (iTRAQ) based proteomics and assays in vitro to screen and validate potential resistant molecules. We found that a total of 1697 differentially expressed genes (DEGs) and 21 differentially expressed proteins (DEPs) in CD4+ T cells between CsA & CS-resistant and -sensitive VKH patients. Ribosomal Protein S4, Y-Linked 1 (RPS4Y1) was verified to regulate the resistance of CD4+ T cells from male VKH patients to CsA & CS. Importantly, we showed that chlorambucil (CLB) could reverse the resistance by RPS4Y1 suppression. Taken together, we identify RPS4Y1 as an important CsA & CS resistance gene in VKH disease. Researchers should consider validating the resistant effect of RPS4Y1 in other autoimmune diseases or organ transplantation.
Collapse
Affiliation(s)
- Rui Chang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, PR China
| | - Lin Chen
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, PR China
| | - Guannan Su
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, PR China
| | - Liping Du
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Province Eye Hospital, Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Zhengzhou, PR China
| | - Yang Qin
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, PR China
| | - Jing Xu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, PR China
| | - Handan Tan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, PR China
| | - Chunjiang Zhou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, PR China
| | - Qingfeng Cao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, PR China
| | - Gangxiang Yuan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, PR China
| | - Aize Kijlstra
- University Eye Clinic Maastricht, Maastricht, the Netherlands
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, PR China.
| |
Collapse
|
25
|
Zhong Y, Cao L, Ma H, Wang Q, Wei P, Yang J, Mo Y, Cao L, Shuai C, Peng S. Lin28A Regulates Stem-like Properties of Ovarian Cancer Cells by Enriching RAN and HSBP1 mRNA and Up-regulating its Protein Expression. Int J Biol Sci 2020; 16:1941-1953. [PMID: 32398961 PMCID: PMC7211169 DOI: 10.7150/ijbs.43504] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 03/15/2020] [Indexed: 12/18/2022] Open
Abstract
Ovarian cancer (OC) is one of the malignant tumors that seriously threaten women's health, with the highest mortality rate in gynecological malignancies. The prognosis of patients with advanced OC is still poor, and the 5-year survival rate is only 20-30%. Therefore, how to improve the early diagnosis rate and therapeutic effect are urgent for patients with OC. In this research, we found that Lin28A can promote the expression of stem cell marker molecules CD133, CD44, OCT4 and Nanog. We later confirmed that Lin28A can enrich the mRNA of ras-related nuclear protein (RAN) and heat shock factor binding protein 1 (HSBP1) through RIP assay, and that Lin28A can regulate their protein expression. We also identified that RAN and HSBP1 are highly expressed in OC tissues, and that they are significantly positively correlated with the expression of Lin28A and negatively correlated with the survival prognosis of OC patients. After stable knockdown of RAN or HSBP1 in OC cells with high expression of Lin28A, the expression of the stem cell marker molecules such as OCT4, CD44 and Nanog are reduced. And after knocking down of RAN or HSBP1 in Lin28A highly expressed OC cells, the survival and invasion of OC cells and tumor size of OC xenograft in nude mice were markedly inhibited and apoptosis was increased. Our data also showed that knock down of RAN or HSBP1 can inhibit the invasion ability of OC cells by decreasing the expression of N-cadherin, Vimentin and promoting the expression of E-cadherin. Meanwhile, knockdown of RAN or HSBP1 induced cell apoptosis by inhibiting the expression of PARP. Our results indicated that Lin28A could regulate the biological behaviors in OC cells through RAN/HSBP1. These findings suggest that Lin28A/RAN/HSBP1 can be used as a marker for diagnosis and prognosis of OC patients, and RAN/HSBP1 may be a potential new target for gene therapy of OC.
Collapse
Affiliation(s)
- Yancheng Zhong
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine; School of basic Medical Science, Central South University, Changsha, Hunan 410013, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, 410078, China.,Hunan Key Laboratory of Non-resolving Inflammation and Cancer, Disease Genome Research Center, the Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Lanqin Cao
- The department of gynecology of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haotian Ma
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine; School of basic Medical Science, Central South University, Changsha, Hunan 410013, China
| | - Qian Wang
- The department of gynecology of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Pingpin Wei
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine; School of basic Medical Science, Central South University, Changsha, Hunan 410013, China
| | - Juan Yang
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine; School of basic Medical Science, Central South University, Changsha, Hunan 410013, China
| | - Yuqing Mo
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine; School of basic Medical Science, Central South University, Changsha, Hunan 410013, China
| | - Lihua Cao
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine; School of basic Medical Science, Central South University, Changsha, Hunan 410013, China
| | - Cijun Shuai
- Jiangxi University of Science and Technology, Ganzhou, 341000, China; State Key Laboratory of High Performance Complex Manufacturing, Central South University, Changsha, 410083, China
| | - Shuping Peng
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine; School of basic Medical Science, Central South University, Changsha, Hunan 410013, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, 410078, China.,Hunan Key Laboratory of Non-resolving Inflammation and Cancer, Disease Genome Research Center, the Third Xiangya Hospital, Central South University, Changsha, 410013, China
| |
Collapse
|
26
|
Tacconi EMC, Badie S, De Gregoriis G, Reisländer T, Lai X, Porru M, Folio C, Moore J, Kopp A, Baguña Torres J, Sneddon D, Green M, Dedic S, Lee JW, Batra AS, Rueda OM, Bruna A, Leonetti C, Caldas C, Cornelissen B, Brino L, Ryan A, Biroccio A, Tarsounas M. Chlorambucil targets BRCA1/2-deficient tumours and counteracts PARP inhibitor resistance. EMBO Mol Med 2019; 11:e9982. [PMID: 31273933 PMCID: PMC6609913 DOI: 10.15252/emmm.201809982] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 01/03/2023] Open
Abstract
Due to compromised homologous recombination (HR) repair, BRCA1- and BRCA2-mutated tumours accumulate DNA damage and genomic rearrangements conducive of tumour progression. To identify drugs that target specifically BRCA2-deficient cells, we screened a chemical library containing compounds in clinical use. The top hit was chlorambucil, a bifunctional alkylating agent used for the treatment of chronic lymphocytic leukaemia (CLL). We establish that chlorambucil is specifically toxic to BRCA1/2-deficient cells, including olaparib-resistant and cisplatin-resistant ones, suggesting the potential clinical use of chlorambucil against disease which has become resistant to these drugs. Additionally, chlorambucil eradicates BRCA2-deficient xenografts and inhibits growth of olaparib-resistant patient-derived tumour xenografts (PDTXs). We demonstrate that chlorambucil inflicts replication-associated DNA double-strand breaks (DSBs), similarly to cisplatin, and we identify ATR, FANCD2 and the SNM1A nuclease as determinants of sensitivity to both drugs. Importantly, chlorambucil is substantially less toxic to normal cells and tissues in vitro and in vivo relative to cisplatin. Because chlorambucil and cisplatin are equally effective inhibitors of BRCA2-compromised tumours, our results indicate that chlorambucil has a higher therapeutic index than cisplatin in targeting BRCA-deficient tumours.
Collapse
MESH Headings
- Animals
- BRCA1 Protein/deficiency
- BRCA2 Protein/deficiency
- Cell Line, Tumor
- Chlorambucil/pharmacology
- Cricetinae
- Drug Delivery Systems
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Male
- Mice
- Mice, SCID
- Peroxisome Proliferator-Activated Receptors/antagonists & inhibitors
- Peroxisome Proliferator-Activated Receptors/metabolism
- Phthalazines/pharmacology
- Piperazines/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Eliana MC Tacconi
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Sophie Badie
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Giuliana De Gregoriis
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Timo Reisländer
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Xianning Lai
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Manuela Porru
- Area of Translational ResearchIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Cecilia Folio
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - John Moore
- Lung Cancer Translational Science Research GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Arnaud Kopp
- Institut de Génétique et de Biologie Cellulaire et Moléculaire (IGBMC)Inserm U1258, CNRS (UMR 7104)Université de StrasbourgIllkirchFrance
| | - Júlia Baguña Torres
- Radiopharmaceuticals and Molecular Imaging GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Deborah Sneddon
- Radiopharmaceuticals and Molecular Imaging GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Marcus Green
- Lung Cancer Translational Science Research GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Simon Dedic
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Jonathan W Lee
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Ankita Sati Batra
- Department of OncologyCancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Oscar M Rueda
- Department of OncologyCancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Alejandra Bruna
- Department of OncologyCancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Carlo Leonetti
- Area of Translational ResearchIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Carlos Caldas
- Department of OncologyCancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Bart Cornelissen
- Radiopharmaceuticals and Molecular Imaging GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Laurent Brino
- Institut de Génétique et de Biologie Cellulaire et Moléculaire (IGBMC)Inserm U1258, CNRS (UMR 7104)Université de StrasbourgIllkirchFrance
| | - Anderson Ryan
- Lung Cancer Translational Science Research GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Annamaria Biroccio
- Area of Translational ResearchIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Madalena Tarsounas
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| |
Collapse
|