1
|
Kar A, Degtyareva NP, Doetsch PW. Human NTHL1 expression and subcellular distribution determines cisplatin sensitivity in human lung epithelial and non-small cell lung cancer cells. NAR Cancer 2024; 6:zcae006. [PMID: 38384388 PMCID: PMC10880605 DOI: 10.1093/narcan/zcae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/11/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024] Open
Abstract
Base excision repair is critical for maintaining genomic stability and for preventing malignant transformation. NTHL1 is a bifunctional DNA glycosylase/AP lyase that initiates repair of oxidatively damaged pyrimidines. Our recent work established that transient over-expression of NTHL1 leads to acquisition of several hallmarks of cancer in non-tumorigenic immortalized cells likely through interaction with nucleotide excision repair protein XPG. Here, we investigate how NTHL1 expression levels impact cellular sensitivity to cisplatin in non-tumorigenic immortalized cells and five non-small cell lung carcinomas cell lines. The cell line with lowest expression of NTHL1 (H522) shows the highest resistance to cisplatin indicating that decrease in NTHL1 levels may modulate resistance to crosslinking agents in NSCLC tumors. In a complementation study, overexpression of NTHL1 in H522 cell line sensitized it to cisplatin. Using NTHL1 N-terminal deletion mutants defective in nuclear localization we show that cisplatin treatment can alter NTHL1 subcellular localization possibly leading to altered protein-protein interactions and affecting cisplatin sensitivity. Experiments presented in this study reveal a previously unknown link between NTHL1 expression levels and cisplatin sensitivity of NSCLC tumor cells. These findings provide an opportunity to understand how altered NTHL1 expression levels and subcellular distribution can impact cisplatin sensitivity in NSCLC tumor cells.
Collapse
Affiliation(s)
- Anirban Kar
- Mutagenesis & DNA Repair Regulation Group, National Institute of Environmental Health Sciences, 111 TW Alexander Dr, Durham, NC 27709, USA
| | - Natalya P Degtyareva
- Mutagenesis & DNA Repair Regulation Group, National Institute of Environmental Health Sciences, 111 TW Alexander Dr, Durham, NC 27709, USA
| | - Paul W Doetsch
- Mutagenesis & DNA Repair Regulation Group, National Institute of Environmental Health Sciences, 111 TW Alexander Dr, Durham, NC 27709, USA
| |
Collapse
|
2
|
Neary B, Lin S, Qiu P. Methylation of CpG Sites as Biomarkers Predictive of Drug-Specific
Patient Survival in Cancer. Cancer Inform 2022; 21:11769351221131124. [PMID: 36340286 PMCID: PMC9634212 DOI: 10.1177/11769351221131124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 09/18/2022] [Indexed: 11/06/2022] Open
Abstract
Background: Though the development of targeted cancer drugs continues to accelerate,
doctors still lack reliable methods for predicting patient response to
standard-of-care therapies for most cancers. DNA methylation has been
implicated in tumor drug response and is a promising source of predictive
biomarkers of drug efficacy, yet the relationship between drug efficacy and
DNA methylation remains largely unexplored. Method: In this analysis, we performed log-rank survival analyses on patients grouped
by cancer and drug exposure to find CpG sites where binary methylation
status is associated with differential survival in patients treated with a
specific drug but not in patients with the same cancer who were not exposed
to that drug. We also clustered these drug-specific CpG sites based on
co-methylation among patients to identify broader methylation patterns that
may be related to drug efficacy, which we investigated for transcription
factor binding site enrichment using gene set enrichment analysis. Results: We identified CpG sites that were drug-specific predictors of survival in 38
cancer-drug patient groups across 15 cancers and 20 drugs. These included 11
CpG sites with similar drug-specific survival effects in multiple cancers.
We also identified 76 clusters of CpG sites with stronger associations with
patient drug response, many of which contained CpG sites in gene promoters
containing transcription factor binding sites. Conclusion: These findings are promising biomarkers of drug response for a variety of
drugs and contribute to our understanding of drug-methylation interactions
in cancer. Investigation and validation of these results could lead to the
development of targeted co-therapies aimed at manipulating methylation in
order to improve efficacy of commonly used therapies and could improve
patient survival and quality of life by furthering the effort toward drug
response prediction.
Collapse
Affiliation(s)
- Bridget Neary
- School of Biological Sciences, Georgia
Institute of Technology, Atlanta, GA, USA
| | - Shuting Lin
- School of Biological Sciences, Georgia
Institute of Technology, Atlanta, GA, USA
| | - Peng Qiu
- Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA,Peng Qiu, Department of Biomedical
Engineering, Georgia Institute of Technology and Emory University, 950 Atlantic
Dr. NW, Atlanta, GA 30332 USA.
| |
Collapse
|
3
|
Lin S, Zhou J, Xiao Y, Neary B, Teng Y, Qiu P. Integrative analysis of TCGA data identifies miRNAs as drug-specific survival biomarkers. Sci Rep 2022; 12:6785. [PMID: 35474090 PMCID: PMC9042876 DOI: 10.1038/s41598-022-10662-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 04/11/2022] [Indexed: 01/03/2023] Open
Abstract
Biomarkers predictive of drug-specific outcomes are important tools for personalized medicine. In this study, we present an integrative analysis to identify miRNAs that are predictive of drug-specific survival outcome in cancer. Using the clinical data from TCGA, we defined subsets of cancer patients who suffered from the same cancer and received the same drug treatment, which we call cancer-drug groups. We then used the miRNA expression data in TCGA to evaluate each miRNA’s ability to predict the survival outcome of patients in each cancer-drug group. As a result, the identified miRNAs are predictive of survival outcomes in a cancer-specific and drug-specific manner. Notably, most of the drug-specific miRNA survival markers and their target genes showed consistency in terms of correlations in their expression and their correlations with survival. Some of the identified miRNAs were supported by published literature in contexts of various cancers. We explored several additional breast cancer datasets that provided miRNA expression and survival data, and showed that our drug-specific miRNA survival markers for breast cancer were able to effectively stratify the prognosis of patients in those additional datasets. Together, this analysis revealed drug-specific miRNA markers for cancer survival, which can be promising tools toward personalized medicine.
Collapse
Affiliation(s)
- Shuting Lin
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, USA
| | - Jie Zhou
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, USA
| | - Yiqiong Xiao
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, USA
| | - Bridget Neary
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, USA
| | - Peng Qiu
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, USA.
| |
Collapse
|
4
|
Neary B, Zhou J, Qiu P. Identifying gene expression patterns associated with drug-specific survival in cancer patients. Sci Rep 2021; 11:5004. [PMID: 33654134 PMCID: PMC7925648 DOI: 10.1038/s41598-021-84211-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/11/2021] [Indexed: 11/13/2022] Open
Abstract
The ability to predict the efficacy of cancer treatments is a longstanding goal of precision medicine that requires improved understanding of molecular interactions with drugs and the discovery of biomarkers of drug response. Identifying genes whose expression influences drug sensitivity can help address both of these needs, elucidating the molecular pathways involved in drug efficacy and providing potential ways to predict new patients’ response to available therapies. In this study, we integrated cancer type, drug treatment, and survival data with RNA-seq gene expression data from The Cancer Genome Atlas to identify genes and gene sets whose expression levels in patient tumor biopsies are associated with drug-specific patient survival using a log-rank test comparing survival of patients with low vs. high expression for each gene. This analysis was successful in identifying thousands of such gene–drug relationships across 20 drugs in 14 cancers, several of which have been previously implicated in the respective drug’s efficacy. We then clustered significant genes based on their expression patterns across patients and defined gene sets that are more robust predictors of patient outcome, many of which were significantly enriched for target genes of one or more transcription factors, indicating several upstream regulatory mechanisms that may be involved in drug efficacy. We identified a large number of genes and gene sets that were potentially useful as transcript-level biomarkers for predicting drug-specific patient survival outcome. Our gene sets were robust predictors of drug-specific survival and our results included both novel and previously reported findings, suggesting that the drug-specific survival marker genes reported herein warrant further investigation for insights into drug mechanisms and for validation as biomarkers to aid cancer therapy decisions.
Collapse
Affiliation(s)
- Bridget Neary
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jie Zhou
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Peng Qiu
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
5
|
Lainetti PF, Leis-Filho AF, Kobayashi PE, de Camargo LS, Laufer-Amorim R, Fonseca-Alves CE, Souza FF. Proteomics Approach of Rapamycin Anti-Tumoral Effect on Primary and Metastatic Canine Mammary Tumor Cells In Vitro. Molecules 2021; 26:molecules26051213. [PMID: 33668689 PMCID: PMC7956669 DOI: 10.3390/molecules26051213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/05/2022] Open
Abstract
Rapamycin is an antifungal drug with antitumor activity and acts inhibiting the mTOR complex. Due to drug antitumor potential, the aim of this study was to evaluate its effect on a preclinical model of primary mammary gland tumors and their metastases from female dogs. Four cell lines from our cell bank, two from primary canine mammary tumors (UNESP-CM1, UNESP-CM60) and two metastases (UNESP-MM1, and UNESP-MM4) were cultured in vitro and investigated for rapamycin IC50. Then, cell lines were treated with rapamycin IC50 dose and mRNA and protein were extracted in treated and non-treated cells to perform AKT, mTOR, PTEN and 4EBP1 gene expression and global proteomics by mass spectrometry. MTT assay demonstrated rapamycin IC50 dose for all different tumor cells between 2 and 10 μM. RT-qPCR from cultured cells, control versus treated group and primary tumor cells versus metastatic tumor cells, did not shown statistical differences. In proteomics were found 273 proteins in all groups, and after data normalization 49 and 92 proteins were used for statistical analysis for comparisons between control versus rapamycin treatment groups, and metastasis versus primary tumor versus metastasis rapamycin versus primary tumor rapamycin, respectively. Considering the two statistical analysis, four proteins, phosphoglycerate mutase, malate dehydrogenase, l-lactate dehydrogenase and nucleolin were found in decreased abundance in the rapamycin group and they are related with cellular metabolic processes and enhanced tumor malignant behavior. Two proteins, dihydrolipoamide dehydrogenase and superoxide dismutase, also related with metabolic processes, were found in higher abundance in rapamycin group and are associated with apoptosis. The results suggested that rapamycin was able to inhibit cell growth of mammary gland tumor and metastatic tumors cells in vitro, however, concentrations needed to reach the IC50 were higher when compared to other studies.
Collapse
Affiliation(s)
- Patrícia F. Lainetti
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University—UNESP, Botucatu 18618-681, Brazil; (P.F.L.); (L.S.d.C.); (C.E.F.-A.)
| | - Antonio F. Leis-Filho
- Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University—UNESP, Botucatu 18618-681, Brazil; (A.F.L.-F.); (P.E.K.); (R.L.-A.)
| | - Priscila E. Kobayashi
- Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University—UNESP, Botucatu 18618-681, Brazil; (A.F.L.-F.); (P.E.K.); (R.L.-A.)
| | - Laíza S. de Camargo
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University—UNESP, Botucatu 18618-681, Brazil; (P.F.L.); (L.S.d.C.); (C.E.F.-A.)
| | - Renee Laufer-Amorim
- Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University—UNESP, Botucatu 18618-681, Brazil; (A.F.L.-F.); (P.E.K.); (R.L.-A.)
| | - Carlos E. Fonseca-Alves
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University—UNESP, Botucatu 18618-681, Brazil; (P.F.L.); (L.S.d.C.); (C.E.F.-A.)
- Institute of Health Sciences, Universidade Paulista—UNIP, Bauru 17048-290, Brazil
| | - Fabiana F. Souza
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University—UNESP, Botucatu 18618-681, Brazil; (P.F.L.); (L.S.d.C.); (C.E.F.-A.)
- Correspondence: ; Tel.: +55-14-38802237
| |
Collapse
|
6
|
Bhattacharyya R, Ha MJ, Liu Q, Akbani R, Liang H, Baladandayuthapani V. Personalized Network Modeling of the Pan-Cancer Patient and Cell Line Interactome. JCO Clin Cancer Inform 2020; 4:399-411. [PMID: 32374631 PMCID: PMC7265783 DOI: 10.1200/cci.19.00140] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2020] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Personalized network inference on diverse clinical and in vitro model systems across cancer types can be used to delineate specific regulatory mechanisms, uncover drug targets and pathways, and develop individualized predictive models in cancer. METHODS We developed TransPRECISE (personalized cancer-specific integrated network estimation model), a multiscale Bayesian network modeling framework, to analyze the pan-cancer patient and cell line interactome to identify differential and conserved intrapathway activities, to globally assess cell lines as representative models for patients, and to develop drug sensitivity prediction models. We assessed pan-cancer pathway activities for a large cohort of patient samples (> 7,700) from the Cancer Proteome Atlas across ≥ 30 tumor types, a set of 640 cancer cell lines from the MD Anderson Cell Lines Project spanning 16 lineages, and ≥ 250 cell lines' response to > 400 drugs. RESULTS TransPRECISE captured differential and conserved proteomic network topologies and pathway circuitry between multiple patient and cell line lineages: ovarian and kidney cancers shared high levels of connectivity in the hormone receptor and receptor tyrosine kinase pathways, respectively, between the two model systems. Our tumor stratification approach found distinct clinical subtypes of the patients represented by different sets of cell lines: patients with head and neck tumors were classified into two different subtypes that are represented by head and neck and esophagus cell lines and had different prognostic patterns (456 v 654 days of median overall survival; P = .02). High predictive accuracy was observed for drug sensitivities in cell lines across multiple drugs (median area under the receiver operating characteristic curve > 0.8) using Bayesian additive regression tree models with TransPRECISE pathway scores. CONCLUSION Our study provides a generalizable analytic framework to assess the translational potential of preclinical model systems and to guide pathway-based personalized medical decision making, integrating genomic and molecular data across model systems.
Collapse
Affiliation(s)
| | - Min Jin Ha
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Qingzhi Liu
- Department of Biostatistics, University of Michigan, Ann Arbor, MI
| | - Rehan Akbani
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
7
|
Bertsimas D, Zhuo YD. Novel Target Discovery of Existing Therapies: Path to Personalized Cancer Therapy. ACTA ACUST UNITED AC 2020. [DOI: 10.1287/ijoo.2019.0019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Dimitris Bertsimas
- Sloan School of Management and Operations Research Center, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Ying Daisy Zhuo
- Sloan School of Management and Operations Research Center, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| |
Collapse
|
8
|
Kalari KR, Sinnwell JP, Thompson KJ, Tang X, Carlson EE, Yu J, Vedell PT, Ingle JN, Weinshilboum RM, Boughey JC, Wang L, Goetz MP, Suman V. PANOPLY: Omics-Guided Drug Prioritization Method Tailored to an Individual Patient. JCO Clin Cancer Inform 2019; 2:1-11. [PMID: 30652605 DOI: 10.1200/cci.18.00012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE The majority of patients with cancer receive treatments that are minimally informed by omics data. We propose a precision medicine computational framework, PANOPLY (Precision Cancer Genomic Report: Single Sample Inventory), to identify and prioritize drug targets and cancer therapy regimens. MATERIALS AND METHODS The PANOPLY approach integrates clinical data with germline and somatic features obtained from multiomics platforms and applies machine learning and network analysis approaches in the context of the individual patient and matched controls. The PANOPLY workflow uses the following four steps: selection of matched controls to the patient of interest; identification of patient-specific genomic events; identification of suitable drugs using the driver-gene network and random forest analyses; and provision of an integrated multiomics case report of the patient with prioritization of anticancer drugs. RESULTS The PANOPLY workflow can be executed on a stand-alone virtual machine and is also available for download as an R package. We applied the method to an institutional breast cancer neoadjuvant chemotherapy study that collected clinical and genomic data as well as patient-derived xenografts to investigate the prioritization offered by PANOPLY. In a chemotherapy-resistant patient-derived xenograft model, we found that that the prioritized drug, olaparib, was more effective than placebo in treating the tumor ( P < .05). We also applied PANOPLY to in-house and publicly accessible multiomics tumor data sets with therapeutic response or survival data available. CONCLUSION PANOPLY shows promise as a means to prioritize drugs on the basis of clinical and multiomics data for an individual patient with cancer. Additional studies are needed to confirm this approach.
Collapse
Affiliation(s)
| | | | | | | | | | - Jia Yu
- All authors: Mayo Clinic, Rochester, MN
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Shibahara I, Hanihara M, Watanabe T, Dan M, Sato S, Kuroda H, Inamura A, Inukai M, Hara A, Yasui Y, Kumabe T. Tumor microenvironment after biodegradable BCNU wafer implantation: special consideration of immune system. J Neurooncol 2018; 137:417-427. [DOI: 10.1007/s11060-017-2733-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 12/24/2017] [Indexed: 02/07/2023]
|
10
|
Spainhour JCG, Lim J, Qiu P. GDISC: a web portal for integrative analysis of gene-drug interaction for survival in cancer. Bioinformatics 2018; 33:1426-1428. [PMID: 28453687 DOI: 10.1093/bioinformatics/btw830] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 12/27/2016] [Indexed: 11/14/2022] Open
Abstract
Summary Survival analysis has been applied to The Cancer Genome Atlas (TCGA) data. Although drug exposure records are available in TCGA, existing survival analyses typically did not consider drug exposure, partly due to naming inconsistencies in the data. We have spent extensive effort to standardize the drug exposure data, which enabled us to perform survival analysis on drug-stratified subpopulations of cancer patients. Using this strategy, we integrated gene copy number data, drug exposure data and patient survival data to infer gene-drug interactions that impact survival. The collection of all analyzed gene-drug interactions in 32 cancer types are organized and presented in a searchable web-portal called gene-drug Interaction for survival in cancer (GDISC). GDISC allows biologists and clinicians to interactively explore the gene-drug interactions identified in the context of TCGA, and discover interactions associated to their favorite cancer, drug and/or gene of interest. In addition, GDISC provides the standardized drug exposure data, which is a valuable resource for developing new methods for drug-specific analysis. Availability and Implementation GDISC is available at https://gdisc.bme.gatech.edu/. Contact peng.qiu@bme.gatech.edu.
Collapse
Affiliation(s)
| | - Juho Lim
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Peng Qiu
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| |
Collapse
|