1
|
Dong L, Jiang H, Kang Z, Guan M. Biomarkers for chemotherapy and drug resistance in the mismatch repair pathway. Clin Chim Acta 2023; 544:117338. [PMID: 37060988 DOI: 10.1016/j.cca.2023.117338] [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: 09/10/2022] [Revised: 04/09/2023] [Accepted: 04/10/2023] [Indexed: 04/17/2023]
Abstract
Drugs targeting DNA repair have developed rapidly in cancer therapy, and numerous inhibitors have already been utilized in preclinical and clinical stages. To optimize the selection of patients for treatment, it is essential to discover biomarkers to anticipate chemotherapy response. The DNA mismatch repair (MMR) pathway is closely correlated with cancer susceptibility and plays an important role in the occurrence and development of cancers. Here, we give a concise introduction of the MMR genes and focus on the potential biomarkers of chemotherapeutic response and resistance. It has been clarified that the status of MMR may affect the outcome of chemotherapy. However, the specific underlying mechanisms as well as contradictory results continue to raise considerable controversy and concern. In this review, we summarize the current literature to provide a general overview.
Collapse
Affiliation(s)
- Liu Dong
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, People's Republic of China
| | - Haoqin Jiang
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, People's Republic of China
| | - Zhihua Kang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, USA.
| | - Ming Guan
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
2
|
Shu J, Wang X, Yang X, Zhao G. ATM inhibitor KU60019 synergistically sensitizes lung cancer cells to topoisomerase II poisons by multiple mechanisms. Sci Rep 2023; 13:882. [PMID: 36650267 PMCID: PMC9845372 DOI: 10.1038/s41598-023-28185-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Type II topoisomerases (TOP2) poisons represent one class of the most successful and widely prescribed chemotherapeutics, which is frontline therapy for a myriad of systemic cancers and solid tumors, including lymphomas, leukemias, and lung cancer. Despite this, treatment with this class of drugs induces unwanted side effects (including cardiovascular morbidity and secondary malignancies). Additionally, the emergence of drug resistance also greatly compromises the clinical use of these drugs. To enhance therapeutic efficiency while lowering unwanted side effects, new insights into effective combination therapy are required. In this study we found that KU60019, a novel, and highly specific ATM kinase inhibitor interferes with the association of ATM with TOP2β and stabilizes TOP2β-DNA cleavage complex, thereby impairing the repair of TOP2 poison-induced DSBs and contributes to genome stability, leading to accelerated cell death. In H1299 as well as in A549 lung cancer cell lines, biologically, KU60019 combined with VP-16 (one of the TOP2 poisons) synergistically suppressed the growth of cells and survival and triggered a much higher apoptosis rate. In summary, we provide a proof-of-concept strategy that ATM inhibitors combined with TOP2 poison would synergistically suppresses lung cancer cell survival as well as reduce DNA damage responses, thus may lowering the possibility of cardiotoxicity and secondary malignancy linked to therapy.
Collapse
Affiliation(s)
- Jianfeng Shu
- HwaMei Hospital, University of Chinese Academy of Sciences, 41 Xibei Road, Ningbo, 315010, Zhejiang, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, 315000, Zhejiang, China
| | - Xiaofang Wang
- HwaMei Hospital, University of Chinese Academy of Sciences, 41 Xibei Road, Ningbo, 315010, Zhejiang, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, 315000, Zhejiang, China
| | - Xuejie Yang
- HwaMei Hospital, University of Chinese Academy of Sciences, 41 Xibei Road, Ningbo, 315010, Zhejiang, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, 315000, Zhejiang, China
| | - Guofang Zhao
- HwaMei Hospital, University of Chinese Academy of Sciences, 41 Xibei Road, Ningbo, 315010, Zhejiang, China.
| |
Collapse
|
3
|
Zhao H, Chen C, Song H, Qin R, Wang X, He Q, Li F, Zhao H, Li Y. DNA Topoisomerase II-α Regulated by miR-22-5p Promotes Hepatocellular Carcinoma Invasion and Migration through the Hippo Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4277254. [PMID: 36299605 PMCID: PMC9592219 DOI: 10.1155/2022/4277254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/27/2022] [Indexed: 10/10/2023]
Abstract
DNA topoisomerases (TOPs) are dysregulated in various types of cancer. However, how TOP II-alpha (TOP2A) contributes to hepatocellular carcinoma (HCC) progression remains elusive. Cohort analysis revealed that the increased expression of TOP2A was associated with poor clinical outcomes and TOP2A was significantly upregulated in HCC tissues and cell lines. In vitro, TOP2A expression level is related to cell invasion and migration, which may be due to the alteration of epithelial-mesenchymal transition by the TOP2A. Moreover, we used verteporfin (a Hippo inhibitor) to test how the Hippo pathway promotes the effect of TOP2A on the HCC phenotype and found that TOP2A induces tumor progression through the Hippo pathway. Finally, miR-22-5p inhibited tumor progression by sponging TOP2A.
Collapse
Affiliation(s)
- Haichao Zhao
- Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan 030032, China
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 300032, China
| | - Changzhou Chen
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 300032, China
| | - Huangqin Song
- Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan 030032, China
| | - Rongyi Qin
- Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan 030032, China
| | - Xiaoxiao Wang
- Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan 030032, China
| | - Qizu He
- Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan 030032, China
| | - Feng Li
- Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan 030032, China
| | - Haoliang Zhao
- Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan 030032, China
| | - Yanjun Li
- Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan 030032, China
| |
Collapse
|
4
|
Alsalhi A, Ayon NJ, Sikder S, Youan BBC. Self-Assembled Nanomicelles to Enhance Solubility and Anticancer Activity of Etoposide. Assay Drug Dev Technol 2021; 19:526-538. [PMID: 34813380 DOI: 10.1089/adt.2021.089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
It is hypothesized that etoposide/VP-16 nanomicellar formulation (VP-16 NMF) utilizing D-α-Tocopherol polyethylene glycol 1000 succinate (TPGS) can improve etoposide solubility and anticancer activity. The following four different concentrations of TPGS: 3, 6, 8, and 10 wt% were used to solubilize the drug. Among these four formulations, 10 wt% of TPGS loaded with VP-16 NMF dramatically enhanced etoposide apparent solubility by 26-folds compared with the native drug. The physicochemical properties of the optimized formulation were further analyzed by dynamic light scattering, X-ray powder diffraction, scanning electron microscopy, proton nuclear magnetic resonance (1HNMR) and Fourier transform infrared spectroscopy. Liquid chromatography tandem-mass spectrometry (LC-MS/MS) was used to assess solubility and intracellular uptake of the drug from the NMF. Cell viability assay ([3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H tetrazolium solution [MTS]) was performed on MCF-7 and MCF-10A cell lines to assess intracellular uptake and anticancer activity of etoposide. The MTS assay results showed that the VP-16 NMF platform provides a higher anticancer activity than the native VP-16 on the MCF-7 cells line as it integrates a dual anticancer activity of VP-16 and TPGS. LC-MS/MS data showed a threefold increase in cellular uptake of VP-16 NMF in MCF-7 cell line compared with the native etoposide. These data suggest that an optimal TPGS concentration can improve VP-16 bioavailability and efficacy with potential benefits for chemotherapy.
Collapse
Affiliation(s)
- Abdullah Alsalhi
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Navid J Ayon
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Sadia Sikder
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Bi-Botti C Youan
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri, USA
| |
Collapse
|
5
|
Ma Y, North BJ, Shu J. Regulation of topoisomerase II stability and activity by ubiquitination and SUMOylation: clinical implications for cancer chemotherapy. Mol Biol Rep 2021; 48:6589-6601. [PMID: 34476738 DOI: 10.1007/s11033-021-06665-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 08/17/2021] [Indexed: 12/09/2022]
Abstract
DNA topoisomerases II (TOP2) are peculiar enzymes (TOP2α and TOP2β) that modulate the conformation of DNA by momentarily breaking double-stranded DNA to allow another strand to pass through, and then rejoins the DNA phosphodiester backbone. TOP2α and TOP2β play vital roles in nearly all events involving DNA metabolism, including DNA transcription, replication, repair, and chromatin remodeling. Beyond these vital functions, TOP2 enzymes are therapeutic targets for various anticancer drugs, termed TOP2 poisons, such as teniposide, etoposide, and doxorubicin. These drugs exert their antitumor activity by inhibiting the activity of TOP2-DNA cleavage complexes (TOP2ccs) containing DNA double-strand breaks (DSBs), subsequently leading to the degradation of TOP2 by the 26S proteasome, thereby exposing the DSBs and eliciting a DNA damage response. Failure of the DSBs to be appropriately repaired leads to genomic instability. Due to this mechanism, patients treated with TOP2-based drugs have a high incidence of secondary malignancies and cardiotoxicity. While the cytotoxicity associated with TOP2 poisons appears to be TOP2α-dependent, the DNA sequence rearrangements and formation of DSBs appear to be mediated primarily through TOP2β inhibition, likely due to the differential degradation patterns of TOP2α and TOP2β. Research over the past few decades has shown that under various conditions, the ubiquitin-proteasome system (UPS) and the SUMOylation pathway are primarily responsible for regulating the stability and activity of TOP2 and are therefore critical regulators of the therapeutic effect of TOP2-targeting drugs. In this review, we summarize the current progress on the regulation of TOP2α and TOP2β by ubiquitination and SUMOylation. By fully elucidating the basic biology of these essential and complex molecular mechanisms, better strategies may be developed to improve the therapeutic efficacy of TOP2 poisons and minimize the risks of therapy-related secondary malignancy.
Collapse
Affiliation(s)
- Ying Ma
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310029, China
- Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Brian J North
- Biomedical Sciences Department, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE, 68178, USA.
| | - Jianfeng Shu
- HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, China.
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, 315020, China.
| |
Collapse
|
6
|
Kanne J, Hussong M, Isensee J, Muñoz-López Á, Wolffgramm J, Heß F, Grimm C, Bessonov S, Meder L, Wang J, Reinhardt HC, Odenthal M, Hucho T, Büttner R, Summerer D, Schweiger MR. Pericentromeric Satellite III transcripts induce etoposide resistance. Cell Death Dis 2021; 12:530. [PMID: 34031359 PMCID: PMC8144429 DOI: 10.1038/s41419-021-03810-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 03/04/2021] [Accepted: 05/10/2021] [Indexed: 02/05/2023]
Abstract
Non-coding RNA from pericentromeric satellite repeats are involved in stress-dependent splicing processes, maintenance of heterochromatin, and are required to protect genome stability. Here we show that the long non-coding satellite III RNA (SatIII) generates resistance against the topoisomerase IIa (TOP2A) inhibitor etoposide in lung cancer. Because heat shock conditions (HS) protect cells against the toxicity of etoposide, and SatIII is significantly induced under HS, we hypothesized that the protective effect could be traced back to SatIII. Using genome methylation profiles of patient-derived xenograft mouse models we show that the epigenetic modification of the SatIII DNA locus and the resulting SatIII expression predict chemotherapy resistance. In response to stress, SatIII recruits TOP2A to nuclear stress bodies, which protects TOP2A from a complex formation with etoposide and results in decreased DNA damage after treatment. We show that BRD4 inhibitors reduce the expression of SatIII, restoring etoposide sensitivity.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Cell Cycle Proteins/antagonists & inhibitors
- Centromere/genetics
- Centromere/metabolism
- DNA Methylation/physiology
- DNA Topoisomerases, Type II/drug effects
- DNA Topoisomerases, Type II/genetics
- DNA Topoisomerases, Type II/metabolism
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Etoposide/therapeutic use
- Gene Expression Regulation, Neoplastic/drug effects
- HEK293 Cells
- HeLa Cells
- Humans
- Male
- Mice, Inbred NOD
- Mice, SCID
- Poly-ADP-Ribose Binding Proteins/drug effects
- Poly-ADP-Ribose Binding Proteins/genetics
- Poly-ADP-Ribose Binding Proteins/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/physiology
- Transcription Factors/antagonists & inhibitors
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Julian Kanne
- Institute for Translational Epigenetics, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Michelle Hussong
- Institute for Translational Epigenetics, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Jörg Isensee
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, University Hospital Cologne, Faculty of Medicine, University Cologne, Cologne, Germany
| | - Álvaro Muñoz-López
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Jan Wolffgramm
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Felix Heß
- Institute for Translational Epigenetics, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
- Rheinische Fachhochschule Cologne, Cologne, Germany
| | - Christina Grimm
- Institute for Translational Epigenetics, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Sergey Bessonov
- Institute for Translational Epigenetics, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
- Department I of Internal Medicine, University Hospital Cologne, Medical Faculty, Cologne, Germany
| | - Lydia Meder
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Department I of Internal Medicine, University Hospital Cologne, Medical Faculty, Cologne, Germany
| | - Jie Wang
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Institute of Pathology, University Hospital of Cologne, Medical Faculty, Cologne, Germany
| | - H Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, German Cancer Consortium (DKTK partner site Essen), Essen, Germany
| | - Margarete Odenthal
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Institute of Pathology, University Hospital of Cologne, Medical Faculty, Cologne, Germany
- Center of Integrated Oncology Cologne-Bonn, Medical Faculty, University of Cologne, Cologne, Germany
| | - Tim Hucho
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, University Hospital Cologne, Faculty of Medicine, University Cologne, Cologne, Germany
| | - Reinhard Büttner
- Institute of Pathology, University Hospital of Cologne, Medical Faculty, Cologne, Germany
| | - Daniel Summerer
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Michal R Schweiger
- Institute for Translational Epigenetics, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
7
|
Chen P, Kuang P, Wang L, Li W, Chen B, Liu Y, Wang H, Zhao S, Ye L, Yu F, He Y, Zhou C. Mechanisms of drugs-resistance in small cell lung cancer: DNA-related, RNA-related, apoptosis-related, drug accumulation and metabolism procedure. Transl Lung Cancer Res 2020; 9:768-786. [PMID: 32676338 PMCID: PMC7354133 DOI: 10.21037/tlcr-19-547] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Small-cell lung cancer (SCLC), the highest malignant cancer amongst different types of lung cancer, has the feature of lower differentiation, rapid growth, and poor survival rate. Despite the dramatically initial sensitivity of SCLC to various types of treatment methods, including chemotherapy, radiotherapy and immunotherapy, the emergence of drugs-resistance is still a grandly clinical challenge. Therefore, in order to improve the prognosis and develop new therapeutic approaches, having a better understanding of the complex mechanisms of resistance in SCLC is of great clinical significance. This review summarized recent advances in understanding of multiple mechanisms which are involved in the resistance during SCLC treatment, including DNA-related process, RNA-related process, apoptosis-related mechanism, and the process of drug accumulation and metabolism.
Collapse
Affiliation(s)
- Peixin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Department of Medical School, Tongji University, Shanghai, China
| | - Peng Kuang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Department of Medical Oncology, The First Affiliated Hospital Of Nanchang University, Nanchang, China
| | - Lei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Wei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Bin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yu Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Department of Medical School, Tongji University, Shanghai, China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Department of Medical School, Tongji University, Shanghai, China
| | - Sha Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Lingyun Ye
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Feng Yu
- Department of Medical Oncology, The First Affiliated Hospital Of Nanchang University, Nanchang, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Van Den Borg R, Leonetti A, Tiseo M, Giovannetti E, Peters GJ. Novel targeted strategies to overcome resistance in small-cell lung cancer: focus on PARP inhibitors and rovalpituzumab tesirine. Expert Rev Anticancer Ther 2019; 19:461-471. [PMID: 31148500 DOI: 10.1080/14737140.2019.1624530] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Small-cell lung cancer (SCLC) is a highly aggressive neuroendocrine tumour, and its outcome is strongly conditioned by the rapid onset of resistance to conventional chemotherapeutics. First-line treatment with a combination of platinum agents and topoisomerase inhibitors has been the standard of care for over 30 years, with disappointing clinical outcome caused by early-acquired chemoresistance. In this disheartening scenario, novel treatment strategies are being implemented in order to either revert or bypass resistance mechanisms. Areas covered: The general mechanism of action of the standard frontline treatment regimens for SCLC, as well as the known resistance mechanisms to these drugs, is reviewed. Moreover, we focus on the current preclinical and clinical evidence on the potential role of PARP inhibitors and rovalpituzumab tesirine (Rova-T) to tackle chemoresistance in SCLC. Expert opinion: Preliminary evidence supports PARP inhibitors and Rova-T as two promising approaches to either revert or bypass chemoresistance in SCLC, respectively. The identification of potential predictive biomarkers of response to these innovative treatments (SLFN11 and DLL3) has shortened the gap between SCLC and personalized targeted therapy. Further large-scale clinical studies are urgently needed for a better designation of PARP inhibitors and Rova-T in the therapeutic algorithm of SCLC patients.
Collapse
Affiliation(s)
- Robin Van Den Borg
- a Laboratory Medical Oncology , Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam , Amsterdam , Netherlands
| | - Alessandro Leonetti
- a Laboratory Medical Oncology , Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam , Amsterdam , Netherlands.,b Medical Oncology Unit , University Hospital of Parma , Parma , Italy
| | - Marcello Tiseo
- b Medical Oncology Unit , University Hospital of Parma , Parma , Italy.,c Department of Medicine and Surgery , University of Parma , Parma , Italy
| | - Elisa Giovannetti
- a Laboratory Medical Oncology , Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam , Amsterdam , Netherlands.,d Cancer Pharmacology Lab , AIRC Start-Up Unit , Pisa , Italy
| | - Godefridus J Peters
- a Laboratory Medical Oncology , Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam , Amsterdam , Netherlands
| |
Collapse
|
9
|
Yang W, Zhao X, Han Y, Duan L, Lu X, Wang X, Zhang Y, Zhou W, Liu J, Zhang H, Zhao Q, Hong L, Fan D. Identification of hub genes and therapeutic drugs in esophageal squamous cell carcinoma based on integrated bioinformatics strategy. Cancer Cell Int 2019; 19:142. [PMID: 31139019 PMCID: PMC6530124 DOI: 10.1186/s12935-019-0854-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/10/2019] [Indexed: 12/13/2022] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is one of leading malignant cancers of gastrointestinal tract worldwide. Until now, the involved mechanisms during the development of ESCC are largely unknown. This study aims to explore the driven-genes and biological pathways in ESCC. Methods mRNA expression datasets of GSE29001, GSE20347, GSE100942, and GSE38129, containing 63 pairs of ESCC and non-tumor tissues data, were integrated and deeply analyzed. The bioinformatics approaches include identification of differentially expressed genes (DEGs) and hub genes, gene ontology (GO) terms analysis and biological pathway enrichment analysis, construction and analysis of protein-protein interaction (PPI) network, and miRNA-gene network construction. Subsequently, GEPIA2 database and qPCR assay were utilized to validate the expression of hub genes. DGIdb database was performed to search the candidate drugs for ESCC. Results Finally, 120 upregulated and 26 downregulated DEGs were identified. The functional enrichment of DEGs in ESCC were mainly correlated with cell cycle, DNA replication, deleted in colorectal cancer (DCC) mediated attractive signaling pathway, and Netrin-1 signaling pathway. The PPI network was constructed using STRING software with 146 nodes and 2392 edges. The most significant three modules in PPI were filtered and analyzed. Totally ten genes were selected and considered as the hub genes and nuclear division cycle 80 (NDC80) was closely related to the survival of ESCC patients. DGIdb database predicted 33 small molecules as the possible drugs for treating ESCC. Conclusions In summary, the data may provide new insights into ESCC pathogenesis and treatments. The candidate drugs may improve the efficiency of personalized therapy in future.
Collapse
Affiliation(s)
- Wanli Yang
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Xinhui Zhao
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Yu Han
- 2Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lili Duan
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Xin Lu
- 3The School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoqian Wang
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Yujie Zhang
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Wei Zhou
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Jinqiang Liu
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Hongwei Zhang
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Qingchuan Zhao
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Liu Hong
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Daiming Fan
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
10
|
Chai Y, Tan F, Ye S, Liu F, Fan Q. Identification of core genes and prediction of miRNAs associated with osteoporosis using a bioinformatics approach. Oncol Lett 2018; 17:468-481. [PMID: 30655789 DOI: 10.3892/ol.2018.9508] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/23/2018] [Indexed: 12/24/2022] Open
Abstract
Osteoporosis (OP) is an age-related disease, and osteoporotic fracture is one of the major causes of disability and mortality in elderly patients (>70 years old). As the pathogenesis and molecular mechanism of OP remain unclear, the identification of disease biomarkers is important for guiding research and providing therapeutic targets. In the present study, core genes and microRNAs (miRNAs) associated with OP were identified. Differentially expressed genes (DEGs) between human mesenchymal stem cell specimens from normal osseous tissues and OP tissues were detected using the GEO2R tool of the Gene Expression Omnibus database and Morpheus. Network topological parameters were determined using NetworkAnalyzer. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed using the Database for Annotation, Visualization and Integrated Discovery, and ClueGO. Cytoscape with the Search Tool for the Retrieval of Interacting Genes and Molecular Complex Detection plug-in was used to visualize protein-protein interactions (PPIs). Additionally, miRNA-gene regulatory modules were predicted using CyTargetLinker in order to guide future research. In total, 915 DEGs were identified, including 774 upregulated and 141 downregulated genes. Enriched GO terms and pathways were determined, including 'nervous system development', 'regulation of molecular function', 'glutamatergic synapse pathway' and 'pathways in cancer'. The node degrees of DEGs followed power-law distributions. A PPI network with 541 nodes and 1,431 edges was obtained. Overall, 3 important modules were identified from the PPI network. The following 10 genes were identified as core genes based on high degrees of connectivity: Albumin, PH domain leucine-rich repeat-containing protein phosphatase 2 (PHLPP2), DNA topoisomerase 2-α, kininogen 1 (KNG1), interleukin 2 (IL2), leucine-rich repeats and guanylate kinase domain containing, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit γ (PIK3CG), leptin, transferrin and RNA polymerase II subunit A (POLR2A). Additionally, 15 miRNA-target interactions were obtained using CyTargetLinker. Overall, 7 miRNAs co-regulated IL2, 3 regulated PHLPP2, 3 regulated KNG1, 1 regulated PIK3CG and 1 modulated POLR2A. These results indicate potential biomarkers in the pathogenesis of OP and therapeutic targets.
Collapse
Affiliation(s)
- Yi Chai
- Department of Formulaology of Traditional Chinese Medicine, School of Basic Medical Science, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, P.R. China
| | - Feng Tan
- Department of Formulaology of Traditional Chinese Medicine, School of Basic Medical Science, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, P.R. China
| | - Sumin Ye
- Department of Formulaology of Traditional Chinese Medicine, School of Basic Medical Science, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, P.R. China
| | - Feixiang Liu
- Department of Formulaology of Traditional Chinese Medicine, School of Basic Medical Science, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, P.R. China
| | - Qiaoling Fan
- Department of Formulaology of Traditional Chinese Medicine, School of Basic Medical Science, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, P.R. China
| |
Collapse
|
11
|
Chen F, Shen C, Wang X, Wang H, Liu Y, Yu C, Lv J, He J, Wen Z. Identification of genes and pathways in nasopharyngeal carcinoma by bioinformatics analysis. Oncotarget 2017; 8:63738-63749. [PMID: 28969025 PMCID: PMC5609957 DOI: 10.18632/oncotarget.19478] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 06/30/2017] [Indexed: 01/04/2023] Open
Abstract
Nasopharyngeal carcinoma is a metastatic malignant tumor originating from nasopharyngeal epithelium. Lacking or nonspecific symptoms of patients with early stage nasopharyngeal carcinoma have significantly reduced the accuracy of diagnosing and predicting nasopharyngeal carcinoma development. This study aimed to identify gene signatures of nasopharyngeal carcinoma and uncover potential mechanisms. Two gene expression profiles (GSE12452 and GSE13597) containing 56 nasopharyngeal carcinoma samples and 13 normal control samples were analyzed to identify the differentially expressed genes. In total, 179 up-regulated genes and 238 down-regulated genes were identified. Functional and pathway enrichment analysis showed that up-regulated genes were significantly involved in cell cycle, oocyte meiosis, DNA replication and p53 signaling pathway, while down-regulated genes were enriched in Huntington's disease,metabolic pathways. Subsequently, the top 10 hub genes, TOP2A (topoisomerase (DNA) II alpha), CDK1 (cyclin-dependent kinase 1), CCNB1 (cyclin B1), PCNA (proliferating cell nuclear antigen), MAD2L1 (mitotic arrest deficient 2 like 1), BUB1 (budding uninhibited by benzimidazoles 1 homolog), CCNB2 (cyclin B2), AURKA (aurora kinase A), CCNA2 (cyclin A2), CDC6 (cell division cycle 6 homolog), were identified from protein-protein interaction network. Furthermore, Module analysis revealed that the ten hub genes except TOP2A were belonged to module 1, indicating the upregulation of these hub genes associated molecular pathways in nasopharyngeal carcinoma might activate nasopharyngeal carcinoma pathogenesis. In conclusion, this study indicated that the identified differentially expressed genes and hub genes enrich our understanding of the molecular mechanisms of nasopharyngeal carcinoma, which could eventually translate into additional biomarkers to facilitate the early diagnosis and therapeutic approaches.
Collapse
Affiliation(s)
- Fang Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Congxiang Shen
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoqi Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huigang Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yanhui Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Chaosheng Yu
- Department of Otorhinolaryngology-Head and Neck Surgery, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Jieyu Lv
- Department of Otorhinolaryngology-Head and Neck Surgery, Jiangmen Central Hospital, Jiangmen, China
| | - Jingjing He
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Zhong Wen
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Resistance to anticancer drugs permanently alters electrophoretic mobility of cancer cell lines. Electrophoresis 2017; 38:1201-1205. [DOI: 10.1002/elps.201600494] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/24/2017] [Accepted: 01/25/2017] [Indexed: 12/13/2022]
|
13
|
Onguru O, Yalcin S, Rosemblit C, Zhang PJ, Kilic S, Gunduz U. APOBEC3B expression in drug resistant MCF-7 breast cancer cell lines. Biomed Pharmacother 2016; 79:87-92. [PMID: 27044816 DOI: 10.1016/j.biopha.2016.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 02/03/2016] [Accepted: 02/06/2016] [Indexed: 10/22/2022] Open
Abstract
APOBEC3B belongs to a protein family of cytidine deaminases that can insert mutations in DNA and RNA as a result of their ability to deaminate cytidine to uridine. It has been shown that APOBEC3B-catalysed deamination provides a chronic source of DNA damage in breast cancers. We investigated APOBEC3B expression in four drug resistant breast cancer cell lines (Doxorubicin, Etoposide, Paclitaxel and Docetaxel resistant MCF-7 cell lines) using a novel RNA in situ hybridization technology (RNAscope) and compared expression levels with drug sensitive MCF-7 cell line. After RNAscope staining, slides were scanned and saved as digital images using Aperio scanner and software. Quantitative scoring utilizing the number of punctate dots present within each cell boundary was performed for the parameters including positive cell percentage and signal intensity per positive cell. In Doxorubicin and Etoposide resistant MCF-7 cell lines, APOBEC3B expression was approximately five-fold increased (23% and 24% respectively) with higher signal intensity (1.92 and 1.44 signal/cell, respectively) compared to drug sensitive MCF-7 cell line (5%, 1.00 signal/cell) with statistical significance. The increase of APOBEC3B expression in Docataxel resitant and Paclitaxel resistant MCF-7 cell lines was not very high. In conclusion, APOBEC3B expression was increased in some population of tumor cells of drug resistant cell lines. At least for some drugs, APOBEC3B expression may be related to drug resistance, subjecting to some tumor cells to frequent mutation.
Collapse
Affiliation(s)
- Onder Onguru
- Department of Pathology, GATA, Ankara, Turkey; Department of Pathology and Laboratory Medicine, University of Pennsylvania Medical Center, Philadelphia, PA, United States.
| | - Serap Yalcin
- Department of Food Engineering, Ahi Evran University, Kırsehir, Turkey
| | - Cinthia Rosemblit
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Paul J Zhang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Medical Center, Philadelphia, PA, United States
| | - Selim Kilic
- Department of Public Health, GATA, Ankara, Turkey
| | - Ufuk Gunduz
- Department of Biology, Middle East Technical University, Ankara, Turkey
| |
Collapse
|
14
|
Ponnusamy L, Mahalingaiah PKS, Singh KP. Chronic Oxidative Stress Increases Resistance to Doxorubicin-Induced Cytotoxicity in Renal Carcinoma Cells Potentially Through Epigenetic Mechanism. Mol Pharmacol 2015; 89:27-41. [DOI: 10.1124/mol.115.100206] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/29/2015] [Indexed: 12/11/2022] Open
|
15
|
Alpsoy A, Gündüz U. Protein kinase D2 silencing reduced motility of doxorubicin-resistant MCF7 cells. Tumour Biol 2015; 36:4417-26. [PMID: 25874490 DOI: 10.1007/s13277-015-3081-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/08/2015] [Indexed: 11/26/2022] Open
Abstract
Success of chemotherapy is generally impaired by multidrug resistance, intrinsic resistance, or acquired resistance to functionally and structurally irrelevant drugs. Multidrug resistance emerges via distinct mechanisms: increased drug export, decreased drug internalization, dysfunctional apoptotic machinery, increased DNA damage repair, altered cell cycle regulation, and increased drug detoxification. Several reports demonstrated that multidrug resistance is a multifaceted problem such that multidrug resistance correlates with increased aggressiveness and metastatic potential. Here, we tested the involvement of protein kinase D2, a serine/threonine kinase that was previously implicated in proliferation, drug resistance, and motility in doxorubicin-resistant MCF7 (MCF7/DOX) cell line, which served as an in vitro model for drug resistance and invasiveness. We showed that basal level activity of protein kinase D2 (PKD2) was higher in MCF7/DOX cells than parental MCF7 cells. To elucidate the roles of PKD2 MCF7/DOX, PKD2 expression was reduced via small interfering RNA (siRNA)-mediated knockdown. Results showed that acquired resistance of MCF7/DOX to doxorubicin was not affected by PKD2 silencing, while motility of MCF7/DOX cells was reduced. The results implied that PKD2 silencing might inhibit migration of MCF7/DOX cells without affecting chemoresistance significantly.
Collapse
Affiliation(s)
- Aktan Alpsoy
- Department of Biological Sciences, Middle East Technical University, 06800, Ankara, Turkey
| | | |
Collapse
|
16
|
Camargo MS, Oliveira MT, Santoni MM, Resende FA, Oliveira-Höhne AP, Espanha LG, Nogueira CH, Cuesta-Rubio O, Vilegas W, Varanda EA. Effects of nemorosone, isolated from the plant Clusia rosea, on the cell cycle and gene expression in MCF-7 BUS breast cancer cell lines. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:153-157. [PMID: 25636884 DOI: 10.1016/j.phymed.2014.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 09/30/2014] [Accepted: 11/15/2014] [Indexed: 06/04/2023]
Abstract
BACKGROUND Breast cancer is the cause of considerable morbidity and mortality in women. While estrogen receptor antagonists have been widely used in breast cancer treatment, patients have increasingly shown resistance to these agents and the identification of novel targeted therapies is therefore required. Nemorosone is the major constituent of the floral resin from Clusia rosea and belongs to the class of polycyclic polyisoprenylated benzophenones of the acylphloroglucinol group. The cytotoxicity of nemorosone in human cancer cell lines has been reported in recent years and has been related to estrogen receptors in breast cancer cells. METHODS Changes induced by nemorosone in the cell cycle and gene expression of the MCF-7 BUS (estrogen-dependent) breast cancer cell line were analyzed using flow cytometry and the RT(2) Profiler PCR array, respectively. RESULTS In comparison to breast cancer cells without treatment, nemorosone induced discrete cell cycle arrest in the G1 phase and significant depletion in the G2 phase. Moreover, the compound altered the expression of 19 genes related to different pathways, especially the cell cycle, apoptosis and hormone receptors. CONCLUSION These promising results justify further studies to clarify mechanisms of action of nemorosone, in view of evaluate the possible use of this benzophenone as adjuvant in the treatment of breast cancer.
Collapse
Affiliation(s)
- M S Camargo
- UNESP - Univ. Estadual Paulista, Faculdade de Ciências Farmacêuticas de Araraquara, Departamento de Ciências Biológicas, CEP 14801-902, Araraquara, São Paulo, Brazil
| | - M T Oliveira
- UEL - Univ. Estadual Londrina, Londrina, Paraná, Brazil
| | - M M Santoni
- UNESP - Univ. Estadual Paulista, Faculdade de Ciências Farmacêuticas de Araraquara, Departamento de Ciências Biológicas, CEP 14801-902, Araraquara, São Paulo, Brazil
| | - F A Resende
- UNESP - Univ. Estadual Paulista, Faculdade de Ciências Farmacêuticas de Araraquara, Departamento de Ciências Biológicas, CEP 14801-902, Araraquara, São Paulo, Brazil.
| | - A P Oliveira-Höhne
- UNESP - Univ. Estadual Paulista, Faculdade de Ciências Farmacêuticas de Araraquara, Departamento de Ciências Biológicas, CEP 14801-902, Araraquara, São Paulo, Brazil
| | - L G Espanha
- UNESP - Univ. Estadual Paulista, Faculdade de Ciências Farmacêuticas de Araraquara, Departamento de Ciências Biológicas, CEP 14801-902, Araraquara, São Paulo, Brazil
| | - C H Nogueira
- UNESP - Univ. Estadual Paulista, Faculdade de Ciências Farmacêuticas de Araraquara, Departamento de Ciências Biológicas, CEP 14801-902, Araraquara, São Paulo, Brazil
| | - O Cuesta-Rubio
- Instituto de Farmacia y Alimentos (IFAL), Universidad de La Habana, La Habana, Cuba
| | - W Vilegas
- UNESP - Univ. Estadual Paulista, Instituto de Química, Campus Araraquara, c.p. 355, CEP 14800-900, Araraquara, São Paulo, Brazil
| | - E A Varanda
- UNESP - Univ. Estadual Paulista, Faculdade de Ciências Farmacêuticas de Araraquara, Departamento de Ciências Biológicas, CEP 14801-902, Araraquara, São Paulo, Brazil
| |
Collapse
|
17
|
Fruhwirth GO, Diocou S, Blower PJ, Ng T, Mullen G. A whole-body dual-modality radionuclide optical strategy for preclinical imaging of metastasis and heterogeneous treatment response in different microenvironments. J Nucl Med 2014; 55:686-94. [PMID: 24604910 PMCID: PMC6205625 DOI: 10.2967/jnumed.113.127480] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
UNLABELLED Imaging spontaneous cancer cell metastasis or heterogeneous tumor responses to drug treatment in vivo is difficult to achieve. The goal was to develop a new highly sensitive and reliable preclinical longitudinal in vivo imaging model for this purpose, thereby facilitating discovery and validation of anticancer therapies or molecular imaging agents. METHODS The strategy is based on breast cancer cells stably expressing the human sodium iodide symporter (NIS) fused to a red fluorescent protein, thereby permitting radionuclide and fluorescence imaging. Using whole-body nano-SPECT/CT with (99m)TcO4(-), we followed primary tumor growth and spontaneous metastasis in the presence or absence of etoposide treatment. NIS imaging was used to classify organs as small as individual lymph nodes (LNs) to be positive or negative for metastasis, and results were confirmed by confocal fluorescence microscopy. Etoposide treatment efficacy was proven by ex vivo anticaspase 3 staining and fluorescence microscopy. RESULTS In this preclinical model, we found that the NIS imaging strategy outperformed state-of-the-art (18)F-FDG imaging in its ability to detect small tumors (18.5-fold-better tumor-to-blood ratio) and metastases (LN, 3.6-fold) because of improved contrast in organs close to metastatic sites (12- and 8.5-fold-lower standardized uptake value in the heart and kidney, respectively). We applied the model to assess the treatment response to the neoadjuvant etoposide and found a consistent and reliable improvement in spontaneous metastasis detection. Importantly, we also found that tumor cells in different microenvironments responded in a heterogeneous manner to etoposide treatment, which could be determined only by the NIS-based strategy and not by (18)F-FDG imaging. CONCLUSION We developed a new strategy for preclinical longitudinal in vivo cancer cell tracking with greater sensitivity and reliability than (18)F-FDG PET and applied it to track spontaneous and distant metastasis in the presence or absence of genotoxic stress therapy. Importantly, the model provides sufficient sensitivity and dynamic range to permit the reliable assessment of heterogeneous treatment responses in various microenvironments.
Collapse
Affiliation(s)
- Gilbert O. Fruhwirth
- Comprehensive Cancer Imaging Centre, King's College London (KCL) & UCL
- The Richard Dimbleby Department of Cancer Research, KCL, London SE1 1UL
- Division of Imaging Sciences and Biomedical Engineering, St.Thomas’ Hospital, London SE1 7EH
| | - Seckou Diocou
- Comprehensive Cancer Imaging Centre, King's College London (KCL) & UCL
- Division of Imaging Sciences and Biomedical Engineering, St.Thomas’ Hospital, London SE1 7EH
| | - Philip J. Blower
- Comprehensive Cancer Imaging Centre, King's College London (KCL) & UCL
- Division of Imaging Sciences and Biomedical Engineering, St.Thomas’ Hospital, London SE1 7EH
| | - Tony Ng
- Comprehensive Cancer Imaging Centre, King's College London (KCL) & UCL
- The Richard Dimbleby Department of Cancer Research, KCL, London SE1 1UL
| | - Greg Mullen
- Division of Imaging Sciences and Biomedical Engineering, St.Thomas’ Hospital, London SE1 7EH
| |
Collapse
|
18
|
Alpsoy A, Yasa S, Gündüz U. Etoposide resistance in MCF-7 breast cancer cell line is marked by multiple mechanisms. Biomed Pharmacother 2014; 68:351-5. [DOI: 10.1016/j.biopha.2013.09.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 09/24/2013] [Indexed: 12/25/2022] Open
|
19
|
TOP2A overexpression as a poor prognostic factor in patients with nasopharyngeal carcinoma. Tumour Biol 2013; 35:179-87. [PMID: 23897556 DOI: 10.1007/s13277-013-1022-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 07/12/2013] [Indexed: 01/18/2023] Open
Abstract
Despite the advances in diagnostic imaging and treatment modalities, the risk stratification and final outcomes in patients with nasopharyngeal carcinomas (NPC) still remain suboptimal. Through data mining from published transcriptomic database, topoisomerase IIα (TOP2A) was first identified as a differentially upregulated gene in NPC tissues, which implicates cell division via selective cleavage, rearrangement, and re-ligation of DNA strands. Given the roles of TOP2A in prognostication and in the frontline therapeutic regimen of common carcinomas, such as breast cancer, we explored TOP2A immunoexpression status and its associations with clinicopathological variables and survival in a well-defined cohort of NPC patients. TOP2A immunohistochemistry was retrospectively performed and analyzed using H-score method for biopsy specimens from 124 NPC patients who received standard treatment without distant metastasis at initial diagnosis. Those cases with H-score larger than the median value were construed as featuring TOP2A overexpression. The findings were correlated with the clinicopathological variables, disease-specific survival (DSS) and distant metastasis-free survival (DMFS). TOP2A overexpression was significantly associated with American Joint of Cancer Committee (AJCC) stages III-IV (p = 0.019) and univariately predictive of adverse outcomes for DSS (p = 0.0078) and DMFS (p = 0.0003). In the multivariate comparison, TOP2A overexpression remained prognostically independent to portend worse DSS (p = 0.047, hazard ratio = 1.732) and DMFS (p = 0.003, hazard ratio = 2.569), together with advanced AJCC stages III-IV. TOP2A expression is upregulated in a subset of NPCs and its increased immunoexpression significantly correlated with advanced stages and tumor aggressiveness, justifying the potentiality of TOP2A as a prognostic biomarker and a novel therapeutic target of NPC.
Collapse
|