1
|
Lomov NA, Viushkov VS, Rubtsov MA. Mechanisms of Secondary Leukemia Development Caused by Treatment with DNA Topoisomerase Inhibitors. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:892-911. [PMID: 37751862 DOI: 10.1134/s0006297923070040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/14/2023] [Accepted: 04/20/2023] [Indexed: 09/28/2023]
Abstract
Leukemia is a blood cancer originating in the blood and bone marrow. Therapy-related leukemia is associated with prior chemotherapy. Although cancer therapy with DNA topoisomerase II inhibitors is one of the most effective cancer treatments, its side effects include development of secondary leukemia characterized by the chromosomal rearrangements affecting AML1 or MLL genes. Recurrent chromosomal translocations in the therapy-related leukemia differ from chromosomal rearrangements associated with other neoplasias. Here, we reviewed the factors that drive chromosomal translocations induced by cancer treatment with DNA topoisomerase II inhibitors, such as mobility of ends of double-strand DNA breaks formed before the translocation and gain of function of fusion proteins generated as a result of translocation.
Collapse
Affiliation(s)
- Nikolai A Lomov
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
| | - Vladimir S Viushkov
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Mikhail A Rubtsov
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
- Department of Biochemistry, Center for Industrial Technologies and Entrepreneurship Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
| |
Collapse
|
2
|
Zhang J, Gao Y, Zhang Z, Zhao J, Jia W, Xia C, Wang F, Liu T. Multi-therapies Based on PARP Inhibition: Potential Therapeutic Approaches for Cancer Treatment. J Med Chem 2022; 65:16099-16127. [PMID: 36512711 DOI: 10.1021/acs.jmedchem.2c01352] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The nuclear enzymes called poly(ADP-ribose)polymerases (PARPs) are known to catalyze the process of PARylation, which plays a vital role in various cellular functions. They have become important targets for the discovery of novel antitumor drugs since their inhibition can induce significant lethality in tumor cells. Therefore, researchers all over the world have been focusing on developing novel and potent PARP inhibitors for cancer therapy. Studies have shown that PARP inhibitors and other antitumor agents, such as EZH2 and EGFR inhibitors, play a synergistic role in cancer cells. The combined inhibition of PARP and the targets with synergistic effects may provide a rational strategy to improve the effectiveness of current anticancer regimens. In this Perspective, we sum up the recent advance of PARP-targeted agents, including single-target inhibitors/degraders and dual-target inhibitors/degraders, discuss the fundamental theory of developing these dual-target agents, and give insight into the corresponding structure-activity relationships of these agents.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Yuqi Gao
- College of Radiology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, China.,Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong 250117, China
| | - Zipeng Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong 250117, China
| | - Jinbo Zhao
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, China.,Department of Chemistry and Biology, Jilin Provincial Key Laboratory of Carbon Fiber Development and Application, Changchun University of Technology, Changchun, Jilin 130012, China
| | - Wenshuang Jia
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong 250117, China
| | - Chengcai Xia
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Fugang Wang
- Department of Pharmacology, School of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Tingting Liu
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| |
Collapse
|
3
|
Gupta P, Majumdar AG, Patro BS. Non-enzymatic function of WRN RECQL helicase regulates removal of topoisomerase-I-DNA covalent complexes and triggers NF-κB signaling in cancer. Aging Cell 2022; 21:e13625. [PMID: 35582959 PMCID: PMC9197415 DOI: 10.1111/acel.13625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/25/2022] [Accepted: 04/20/2022] [Indexed: 11/26/2022] Open
Abstract
Mutation in Werner (WRN) RECQL helicase is associated with premature aging syndrome (Werner syndrome, WS) and predisposition to multiple cancers. In patients with solid cancers, deficiency of the WRN RECQL helicase is paradoxically associated with enhanced overall survival in response to treatment with TOP1 inhibitors, which stabilize pathological TOP1-DNA-covalent-complexes (TOP1cc) on the genome. However, the underlying mechanism of WRN in development of chemoresistance to TOP1 inhibitors is not yet explored. Our whole-genome transcriptomic analysis for ~25,000 genes showed robust activation of NF-κB-dependent prosurvival genes in response to TOP1cc. CRISPR-Cas9 knockout, shRNA silencing, and under-expression of WRN confer high-sensitivity of multiple cancers to TOP1 inhibitor. We demonstrated that WRN orchestrates TOP1cc repair through proteasome-dependent and proteasome-independent process, unleashing robust ssDNA generation. This in turn ensues signal transduction for CHK1 mediated NF-κB-activation through IκBα-degradation and nuclear localization of p65 protein. Intriguingly, our site-directed mutagenesis and rescue experiments revealed that neither RECQL-helicase nor DNA-exonuclease enzyme activity of WRN (WRNE84A , WRNK577M , and WRNE84A-K577M ) were required for TOP1cc removal, ssDNA generation and signaling for NF-κB activation. In correlation with patient data and above results, the TOP1 inhibitor-based targeted therapy showed that WRN-deficient melanoma tumors were highly sensitive to TOP1 inhibition in preclinical in vivo mouse model. Collectively, our findings identify hitherto unknown non-enzymatic role of WRN RECQL helicase in pathological mechanisms underlying TOP1cc processing and subsequent NF-κB-activation, offering a potential targeted therapy for WRN-deficient cancer patients.
Collapse
Affiliation(s)
- Pooja Gupta
- Bio‐Organic DivisionBhabha Atomic Research CentreTrombayMumbaiIndia
- Homi Bhabha National InstituteAnushaktinagarMumbaiIndia
| | - Ananda Guha Majumdar
- Bio‐Organic DivisionBhabha Atomic Research CentreTrombayMumbaiIndia
- Homi Bhabha National InstituteAnushaktinagarMumbaiIndia
| | - Birija Sankar Patro
- Bio‐Organic DivisionBhabha Atomic Research CentreTrombayMumbaiIndia
- Homi Bhabha National InstituteAnushaktinagarMumbaiIndia
| |
Collapse
|
4
|
The PARP1 Inhibitor Niraparib Represses DNA Damage Repair and Synergizes with Temozolomide for Antimyeloma Effects. JOURNAL OF ONCOLOGY 2022; 2022:2800488. [PMID: 35422863 PMCID: PMC9005285 DOI: 10.1155/2022/2800488] [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: 01/17/2022] [Revised: 02/15/2022] [Accepted: 03/09/2022] [Indexed: 11/18/2022]
Abstract
Purpose Poly(ADP-ribose) polymerase 1 (PARP1) is necessary for single-strand break (SSB) repair by sensing DNA breaks and facilitating DNA repair through poly ADP-ribosylation of several DNA-binding and repair proteins. Inhibition of PARP1 results in collapsed DNA replication fork and double-strand breaks (DSBs). Accumulation of DSBs goes beyond the capacity of DNA repair response, ultimately resulting in cell death. This work is aimed at assessing the synergistic effects of the DNA-damaging agent temozolomide (TMZ) and the PARP inhibitor niraparib (Nira) in human multiple myeloma (MM) cells. Materials and Methods MM RPMI8226 and NCI-H929 cells were administered TMZ and/or Nira for 48 hours. CCK-8 was utilized for cell viability assessment. Cell proliferation and apoptosis were detected flow-cytometrically. Immunofluorescence was performed for detecting γH2A.X expression. Soft-agar colony formation assay was applied to evaluate the antiproliferative effect. The amounts of related proteins were obtained by immunoblot. The combination index was calculated with the CompuSyn software. A human plasmacytoma xenograft model was established to assess the anti-MM effects in vivo. The anti-MM activities of TMZ and/or Nira were evaluated by H&E staining, IHC, and the TUNEL assay. Results The results demonstrated that cotreatment with TMZ and Nira promoted DNA damage, cell cycle arrest, and apoptotic death in cultured cells but also reduced MM xenograft growth in nude mice, yielding highly synergistic effects. Immunoblot revealed that TMZ and Nira cotreatment markedly increased the expression of p-ATM, p-CHK2, RAD51, and γH2A.X, indicating the suppression of DNA damage response (DDR) and elevated DSB accumulation. Conclusion Inhibition of PARP1 sensitizes genotoxic agents and represents an important therapeutic approach for MM. These findings provide preliminary evidence for combining PARP1 inhibitors with TMZ for MM treatment.
Collapse
|
5
|
Fucoxanthin, a Marine-Derived Carotenoid from Brown Seaweeds and Microalgae: A Promising Bioactive Compound for Cancer Therapy. Int J Mol Sci 2020; 21:ijms21239273. [PMID: 33291743 PMCID: PMC7730715 DOI: 10.3390/ijms21239273] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022] Open
Abstract
Fucoxanthin is a well-known carotenoid of the xanthophyll family, mainly produced by marine organisms such as the macroalgae of the fucus genus or microalgae such as Phaeodactylum tricornutum. Fucoxanthin has antioxidant and anti-inflammatory properties but also several anticancer effects. Fucoxanthin induces cell growth arrest, apoptosis, and/or autophagy in several cancer cell lines as well as in animal models of cancer. Fucoxanthin treatment leads to the inhibition of metastasis-related migration, invasion, epithelial–mesenchymal transition, and angiogenesis. Fucoxanthin also affects the DNA repair pathways, which could be involved in the resistance phenotype of tumor cells. Moreover, combined treatments of fucoxanthin, or its metabolite fucoxanthinol, with usual anticancer treatments can support conventional therapeutic strategies by reducing drug resistance. This review focuses on the current knowledge of fucoxanthin with its potential anticancer properties, showing that fucoxanthin could be a promising compound for cancer therapy by acting on most of the classical hallmarks of tumor cells.
Collapse
|
6
|
Induction of Redox-Mediated Cell Death in ER-Positive and ER-Negative Breast Cancer Cells by a Copper(II)-Phenolate Complex: An In Vitro and In Silico Study. Molecules 2020; 25:molecules25194504. [PMID: 33019623 PMCID: PMC7583785 DOI: 10.3390/molecules25194504] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/25/2022] Open
Abstract
This research was aimed at finding the cytotoxic potential of the mixed ligand copper(II) complex [Cu(tdp)(phen)](ClO4)—where H(tdp) is the tetradentate ligand 2-[(2-(2-hydroxyethylamino)-ethylimino)methyl]phenol, and phen is 1,10-phenanthroline—to two genotypically different breast cancer cells, MCF-7 (p53+ and ER+) and MDA-MB-231 (p53- and ER-). The complex has been already shown to be cytotoxic to ME180 cervical carcinoma cells. The special focus in this study was the induction of cell death by apoptosis and necrosis, and its link with ROS. The treatment brought about nuclear fragmentation, phosphatidylserine externalization, disruption of mitochondrial trans-membrane potential, DNA damage, cell cycle arrest at sub-G1 phase, and increase of ROS generation, followed by apoptotic death of cells during early hours and a late onset of necrosis in the cells surviving the apoptosis. The efficacy of the complex against genotypically different breast cancer cells is attributed to a strong association through p53-mitochondrial redox—cell cycle junction. The ADMET properties and docking of the complex at the active site of Top1 are desirable attributes of a lead molecule for development into a therapeutic. Thus, it is shown that the copper(II)–phenolate complex[Cu(tdp)(phen)]+ offers potential to be developed into a therapeutic for breast cancers in general and ER-negative ones in particular.
Collapse
|
7
|
Oster S, Aqeilan RI. Programmed DNA Damage and Physiological DSBs: Mapping, Biological Significance and Perturbations in Disease States. Cells 2020; 9:cells9081870. [PMID: 32785139 PMCID: PMC7463922 DOI: 10.3390/cells9081870] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/02/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022] Open
Abstract
DNA double strand breaks (DSBs) are known to be the most toxic and threatening of the various types of breaks that may occur to the DNA. However, growing evidence continuously sheds light on the regulatory roles of programmed DSBs. Emerging studies demonstrate the roles of DSBs in processes such as T and B cell development, meiosis, transcription and replication. A significant recent progress in the last few years has contributed to our advanced knowledge regarding the functions of DSBs is the development of many next generation sequencing (NGS) methods, which have considerably advanced our capabilities. Other studies have focused on the implications of programmed DSBs on chromosomal aberrations and tumorigenesis. This review aims to summarize what is known about DNA damage in its physiological context. In addition, we will examine the advancements of the past several years, which have made an impact on the study of genome landscape and its organization.
Collapse
Affiliation(s)
- Sara Oster
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Hebrew University-Hadassah Medical School, Jerusalem 9112001, Israel;
| | - Rami I. Aqeilan
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Hebrew University-Hadassah Medical School, Jerusalem 9112001, Israel;
- Department of Cancer Biology and Genetics, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
- Correspondence:
| |
Collapse
|
8
|
Liu Z, Yu M, Fei B, Sun J, Wang D. Identification Of Natural Compound Derivative For Inhibition Of XLF And Overcoming Chemoresistance In Colorectal Cancer Cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3823-3834. [PMID: 31806933 PMCID: PMC6847993 DOI: 10.2147/dddt.s215967] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/27/2019] [Indexed: 12/14/2022]
Abstract
Purpose A previous study has identified that XRCC4-like factor (XLF) is a potential target to overcome resistance to 5-fluorouracil (5-Fu) and oxaliplatin (OXA) in colorectal cancer (CRC). The purpose of this study is to develop potent XLF inhibitors to chemoresistance in CRC. Methods Virtual screening was adopted to identify novel XLF-binding compounds by initially testing 6800 molecules in Chemical Entities of Biological Interest library. Hit compounds were further validated by Western blot assay. Cell sensitivity to 5-Fu and OXA was measured using sulforhodamine B assay. The effect of XLF inhibitor on DNA repair efficiency was evaluated by comet assay, fluorescent-based nonhomologous end joining (NHEJ) and homologous recombination (HR) reporter assays. DNA-binding activity of NHEJ key factors was examined by chromatin fractionation assay. Results We identified G3, a novel and potent XLF inhibitor (IC50 0.47±0.02 µM). G3 induced XLF protein degradation in CRC cells. Significantly, G3 improved cell sensitivity to 5-Fu and OXA in chemoresistant CRC cell lines. Mechanistically, G3 depleted XLF expression, severely compromised NHEJ efficiency by up to 65% and inhibited NHEJ key factor assembly on DNA. G3 also inhibited HR efficiency in a time-dependent manner. Conclusion These results suggest that G3 overcomes 5-Fu and OXA resistance in CRC cells by inhibiting XLF expression. Thus, XLF is a promising target and its inhibitor G3 is a potential candidate for treatment of chemoresistant CRC patients.
Collapse
Affiliation(s)
- Zhuo Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Miao Yu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Bingyuan Fei
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Jing Sun
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, USA
| | - Dongxin Wang
- Department of Anesthesiology, Jilin Cancer Hospital, Jilin, People's Republic of China
| |
Collapse
|
9
|
Volasertib preclinical activity in high-risk hepatoblastoma. Oncotarget 2019; 10:6403-6417. [PMID: 31741706 PMCID: PMC6849653 DOI: 10.18632/oncotarget.27237] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/12/2019] [Indexed: 12/17/2022] Open
Abstract
Relapsed and metastatic hepatoblastoma represents an unmet clinical need with limited chemotherapy treatment options. In a chemical screen, we identified volasertib as an agent with in vitro activity, inhibiting hepatoblastoma cell growth while sparing normal hepatocytes. Volasertib targets PLK1 and prevents the progression of mitosis, resulting in eventual cell death. PLK1 is overexpressed in hepatoblastoma biopsies relative to normal liver tissue. As a potential therapeutic strategy, we tested the combination of volasertib and the relapse-related hepatoblastoma chemotherapeutic irinotecan. We found both in vitro and in vivo efficacy of this combination, which may merit further preclinical investigation and exploration for a clinical trial concept.
Collapse
|
10
|
Byun MR, Kim CH, Lee HS, Choi JW, Lee SK. Repurposing of ginseng extract as topoisomerase I inhibitor based on the comparative analysis of gene expression patterns. PHYTOCHEMISTRY 2019; 164:223-227. [PMID: 31181353 DOI: 10.1016/j.phytochem.2019.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 03/25/2019] [Accepted: 04/10/2019] [Indexed: 06/09/2023]
Abstract
Repositioning of plant extracts and chemical drugs can accelerate drug development. However, its success rate may depend on what the clue is for the repositioning. Recently, repositioning based on correction of unwarranted gene expression pattern has suggested the possibility of new drug development. Here, we designed a similar method for the repositioning of nutraceutical ginseng (Panax ginseng C.A.Mey.), which is one of the most validated natural therapeutic products for various diseases. We analyzed ginseng-induced gene expression profiles using the connectivity map algorithm, which is a database that connects diseases, chemical drugs, and gene expression. Ginseng was predicted to show the same effects as those of topoisomerase I inhibitors. In a subsequent in vitro assay, ginseng extract unwound coiled or supercoiled DNA, an effect comparable to that of the topoisomerase I inhibitor, camptothecin. Furthermore, ginseng extract induced synthetic lethality with suppression of the Werner syndrome gene. The collected data implicate ginseng as a candidate antitumor agent owing to its topoisomerase I inhibitory activity and further validate the usefulness of differentially expressed gene similarity-based repurposing of other natural products.
Collapse
Affiliation(s)
- Mi Ran Byun
- College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Cheol Hyun Kim
- College of Korean Medicine, Wonkwang University, Iksan, Cheonbuk, 54538, Republic of Korea
| | - Ho Sub Lee
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, 460, Iksan-daero, Iksan, Jeonbuk, 54538, Republic of Korea; College of Korean Medicine, Wonkwang University, Iksan, Cheonbuk, 54538, Republic of Korea.
| | - Jin Woo Choi
- College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Sang Kwan Lee
- College of Korean Medicine, Wonkwang University, Iksan, Cheonbuk, 54538, Republic of Korea.
| |
Collapse
|
11
|
Chung SY, Huang WC, Chen ZS, Chao TC, Su Y. Elucidation of the mechanism underlying CD44v6-induced transformation of IEC-6 normal intestinal epithelial cells. J Cell Physiol 2019; 235:194-209. [PMID: 31219187 DOI: 10.1002/jcp.28959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 05/24/2019] [Indexed: 02/06/2023]
Abstract
The transformation abilities of CD44s and CD44v6 in normal intestinal epithelial cells have not yet been reported. Herein, we established both CD44s and CD44v6 overexpressing stable clones from rat IEC-6 cells and demonstrated that the CD44v6 clones had higher saturation density and anchorage independence. Additionally, CD44v6 clones were more resistant to oxaliplatin and irinotecan which might be attributed to a significantly increased B-cell lymphoma 2 level and a reduced DNA damage response in these cells. Moreover, c-Met and vascular endothelial growth factor receptor 2 signalings were involved in modulating the saturation density in CD44v6 clones. Interestingly, higher activation of both AKT and extracellular-signal-regulated kinase (ERK) were detected in CD44v6 clones which might account in part for the cell density-independent nuclear localization of Yes-associated protein (YAP). To no surprise, increases of both saturation density and anchorage independence in CD44v6 clones were markedly diminished by PI3K, AKT, MEK, and ERK inhibitors as well as YAP knockdown. By contrast, overexpression of a constitutively active YAP robustly increased the aforementioned phenotypes in IEC-6 cells. Collectively, our results suggest that upregulation of CD44v6, but not CD44s, induces the transformation of normal intestinal epithelial cells possibly via activating the c-Met/AKT/YAP pathway which might also explain the important role of CD44v6 in the initiation of various carcinomas.
Collapse
Affiliation(s)
- Shin-Yi Chung
- Institute of Biopharmaceutical Sciences, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Wen-Chen Huang
- Institute of Biopharmaceutical Sciences, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Zong-Siang Chen
- Institute of Biopharmaceutical Sciences, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Ta-Chung Chao
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC.,Faculty of Medicine, School of Medicine, National Yang-Min University, Taipei, Taiwan, ROC
| | - Yeu Su
- Institute of Biopharmaceutical Sciences, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan, ROC
| |
Collapse
|
12
|
Liu C, Mu X, Wang X, Zhang C, Zhang L, Yu B, Sun G. Ponatinib Inhibits Proliferation and Induces Apoptosis of Liver Cancer Cells, but Its Efficacy Is Compromised by Its Activation on PDK1/Akt/mTOR Signaling. Molecules 2019; 24:molecules24071363. [PMID: 30959969 PMCID: PMC6480565 DOI: 10.3390/molecules24071363] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/30/2019] [Accepted: 04/04/2019] [Indexed: 12/14/2022] Open
Abstract
Ponatinib is a multi-target protein tyrosine kinase inhibitor, and its effects on hepatocellular carcinoma cells have not been previously explored. In the present study, we investigated its effects on hepatocellular carcinoma cell growth and the underlying mechanisms. Toward SK-Hep-1 and SNU-423 cells, ponatinib induces apoptosis by upregulation of cleaved caspase-3 and -7 and promotes cell cycle arrest in the G1 phase by inhibiting CDK4/6/CyclinD1 complex and phosphorylation of retinoblastoma protein. It inhibits the growth-stimulating mitogen-activated protein (MAP) kinase pathway, the phosphorylation of Src on both negative and positive regulation sites, and Jak2 and Stat3 phosphorylation. Surprisingly, it also activates the PDK1, the protein kinase B (Akt), and the mechanistic target of rapamycin (mTOR) signaling pathway. Blocking mTOR signaling strongly sensitizes cells to inhibition by ponatinib and makes ponatinib a much more potent inhibitor of hepatocellular carcinoma cell proliferation. These findings demonstrate that ponatinib exerts both positive and negative effects on hepatocellular cell proliferation, and eliminating its growth-stimulating effects by drug combination or potentially by chemical medication can significantly improve its efficacy as an anti-cancer drug.
Collapse
Affiliation(s)
- Chang Liu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Xiuli Mu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Xuan Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Chan Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Lina Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Baofeng Yu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Gongqin Sun
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA.
| |
Collapse
|
13
|
Munera López J, Ganuza A, Bogado SS, Muñoz D, Ruiz DM, Sullivan WJ, Vanagas L, Angel SO. Evaluation of ATM Kinase Inhibitor KU-55933 as Potential Anti- Toxoplasma gondii Agent. Front Cell Infect Microbiol 2019; 9:26. [PMID: 30815397 PMCID: PMC6381018 DOI: 10.3389/fcimb.2019.00026] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/25/2019] [Indexed: 01/01/2023] Open
Abstract
Toxoplasma gondii is an apicomplexan protozoan parasite with a complex life cycle composed of multiple stages that infect mammals and birds. Tachyzoites rapidly replicate within host cells to produce acute infection during which the parasite disseminates to tissues and organs. Highly replicative cells are subject to Double Strand Breaks (DSBs) by replication fork collapse and ATM, a member of the phosphatidylinositol 3-kinase (PI3K) family, is a key factor that initiates DNA repair and activates cell cycle checkpoints. Here we demonstrate that the treatment of intracellular tachyzoites with the PI3K inhibitor caffeine or ATM kinase-inhibitor KU-55933 affects parasite replication rate in a dose-dependent manner. KU-55933 affects intracellular tachyzoite growth and induces G1-phase arrest. Addition of KU-55933 to extracellular tachyzoites also leads to a significant reduction of tachyzoite replication upon infection of host cells. ATM kinase phosphorylates H2A.X (γH2AX) to promote DSB damage repair. The level of γH2AX increases in tachyzoites treated with camptothecin (CPT), a drug that generates fork collapse, but this increase was not observed when co-administered with KU-55933. These findings support that KU-55933 is affecting the Toxoplasma ATM-like kinase (TgATM). The combination of KU-55933 and other DNA damaging agents such as methyl methane sulfonate (MMS) and CPT produce a synergic effect, suggesting that TgATM kinase inhibition sensitizes the parasite to damaged DNA. By contrast, hydroxyurea (HU) did not further inhibit tachyzoite replication when combined with KU-55933.
Collapse
Affiliation(s)
- Jonathan Munera López
- Laboratorio de Parasitología Molecular, IIB-INTECH, Consejo Nacional de Investigaciones Científicas (CONICET)-Universidad Nacional General San Martin (UNSAM), Chascomús, Argentina
| | - Agustina Ganuza
- Laboratorio de Parasitología Molecular, IIB-INTECH, Consejo Nacional de Investigaciones Científicas (CONICET)-Universidad Nacional General San Martin (UNSAM), Chascomús, Argentina
| | - Silvina S Bogado
- Laboratorio de Parasitología Molecular, IIB-INTECH, Consejo Nacional de Investigaciones Científicas (CONICET)-Universidad Nacional General San Martin (UNSAM), Chascomús, Argentina
| | - Daniela Muñoz
- Laboratorio de Parasitología Molecular, IIB-INTECH, Consejo Nacional de Investigaciones Científicas (CONICET)-Universidad Nacional General San Martin (UNSAM), Chascomús, Argentina
| | - Diego M Ruiz
- Laboratorio de Parasitología Molecular, IIB-INTECH, Consejo Nacional de Investigaciones Científicas (CONICET)-Universidad Nacional General San Martin (UNSAM), Chascomús, Argentina
| | - William J Sullivan
- Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States.,Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Laura Vanagas
- Laboratorio de Parasitología Molecular, IIB-INTECH, Consejo Nacional de Investigaciones Científicas (CONICET)-Universidad Nacional General San Martin (UNSAM), Chascomús, Argentina
| | - Sergio O Angel
- Laboratorio de Parasitología Molecular, IIB-INTECH, Consejo Nacional de Investigaciones Científicas (CONICET)-Universidad Nacional General San Martin (UNSAM), Chascomús, Argentina
| |
Collapse
|
14
|
Wu J, Xiao S, Yuan M, Li Q, Xiao G, Wu W, Ouyang Y, Huang L, Yao C. PARP inhibitor re‑sensitizes Adriamycin resistant leukemia cells through DNA damage and apoptosis. Mol Med Rep 2018; 19:75-84. [PMID: 30431088 PMCID: PMC6297734 DOI: 10.3892/mmr.2018.9628] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 09/10/2018] [Indexed: 01/09/2023] Open
Abstract
Resistance to Adriamycin (ADR) is an increasing problem in the treatment of leukemia and the development of novel therapeutic strategies is becoming increasingly important. Olaparib is a poly (adenosine diphosphate-ribose) polymerase (PARP) 1 inhibitor, which has promising antitumor activity in patients with metastatic breast cancer and germline BRCA mutations. Previously published studies have indicated that Olaparib is able to overcome drug resistance in cancer; however, its underlying mechanism of action is yet to be elucidated. The aim of the present study was to explore the mechanism underlying re-sensitization. Annexin V-propidium iodide staining indicated that the percentage of apoptotic ADR resistant cells was markedly increased and the cell cycle was blocked at the G2/M-phase following treatment with ADR combined with Olaparib, when compared with the control group. The alkaline comet assay demonstrated that ADR combined with Olaparib significantly upregulated the induction of the DNA damage response in ADR-resistant cells. Western blot analysis revealed that the protein expression of γ-H2A histone family member X, cleaved PARP, caspase 3 and cleaved caspase 3 was markedly enhanced, while the cell cycle-associated protein cyclin B1 was downregulated in K562/ADR cells following treatment with a combination of ADR and Olaparib. Similar synergistic cytotoxicity was observed in blood mononuclear cells, which were isolated from patients with chemotherapy-resistant leukemia. As Olaparib is available for clinical use, the results of the present study provide a rationale for the development of Olaparib combinational therapies for cases of ADR resistant leukemia.
Collapse
Affiliation(s)
- Jie Wu
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Sheng Xiao
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Miaomiao Yuan
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Qianyuan Li
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Guangfen Xiao
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Wei Wu
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Yuexian Ouyang
- Center for Medical Experiments, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Lihua Huang
- Center for Medical Experiments, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Chenjiao Yao
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
15
|
Zhou C, Zhu Y, Lu B, Zhao W, Zhao X. Survivin expression modulates the sensitivity of A549 lung cancer cells resistance to vincristine. Oncol Lett 2018; 16:5466-5472. [PMID: 30250619 PMCID: PMC6144891 DOI: 10.3892/ol.2018.9277] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 12/12/2017] [Indexed: 01/12/2023] Open
Abstract
Lung cancer is the leading cause of cancer-associated mortalities worldwide. Chemotherapeutic drug vincristine is widely used to treat lung cancer; however, the acquisition of drug resistance is the major limitation of chemotherapy, and it is thus important to determine the mechanism underlying vincristine resistance in lung cancer. Survivin has been reported to be associated with the development of drug resistance and be involved in the progression of non-small cell lung cancer. In the present study, a vincristine-resistant lung cancer cell line, A549/VCR, was used to investigate the possible involvement of survivin in the acquisition of vincristine resistance. Western blot analysis demonstrated that survivin protein expression level was markedly higher in A549/VCR cells compared with in control A549 cells, whereas p53 expression level was lower in A549/VCR cells compared with in A549 cells. Thus, wild-type p53 was overexpressed in A549/VCR cells and it reversed vincristine resistance of A549/VCR cells via the inhibition of survivin expression. Furthermore, survivin was knocked down by small interfering RNA technology and the effects on viability and apoptosis of resistant cells were investigated. MTT, Annexin V-fluorescein isothiocyanate/propidium iodide and caspase-3 activity assays indicated that survivin silencing significantly inhibited cell viability and enhanced apoptosis induced by vincristine treatment in A549/VCR cells compared with non-silenced A549/VCR cells. These results suggested that survivin expression regulated by p53 may serve an important role in drug resistance in A549/VCR cells and may be a potential target for enhancing vincristine sensitivity in A549 lung cancer cells. Additionally, the present study revealed that A549/VCR cells exhibited cross resistance to methotrexate (MTX) and survivin silencing re-sensitized A549/VCR cells to MTX, indicating the crucial role of survivin in regulating A549 cells sensitivity to anticancer drugs. The results of the present study are significant for determining the underlying mechanism of vincristine resistance in lung cancer.
Collapse
Affiliation(s)
- Chengwei Zhou
- Thoracic Department, Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| | - Yonggang Zhu
- Thoracic Department, Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| | - Bin Lu
- Thoracic Department, Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| | - Weijun Zhao
- Thoracic Department, Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| | - Xiaodong Zhao
- Thoracic Department, Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| |
Collapse
|
16
|
DNA Damage-Response Pathway Heterogeneity of Human Lung Cancer A549 and H1299 Cells Determines Sensitivity to 8-Chloro-Adenosine. Int J Mol Sci 2018; 19:ijms19061587. [PMID: 29843366 PMCID: PMC6032248 DOI: 10.3390/ijms19061587] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/10/2018] [Accepted: 05/21/2018] [Indexed: 01/29/2023] Open
Abstract
Human lung cancer H1299 (p53-null) cells often display enhanced susceptibility to chemotherapeutics comparing to A549 (p53-wt) cells. However, little is known regarding to the association of DNA damage-response (DDR) pathway heterogeneity with drug sensitivity in these two cells. We investigated the DDR pathway differences between A549 and H1299 cells exposed to 8-chloro-adenosine (8-Cl-Ado), a potential anticancer drug that can induce DNA double-strand breaks (DSBs), and found that the hypersensitivity of H1299 cells to 8-Cl-Ado is associated with its DSB overaccumulation. The major causes of excessive DSBs in H1299 cells are as follows: First, defect of p53-p21 signal and phosphorylation of SMC1 increase S phase cells, where replication of DNA containing single-strand DNA break (SSB) produces more DSBs in H1299 cells. Second, p53 defect and no available induction of DNA repair protein p53R2 impair DNA repair activity in H1299 cells more severely than A549 cells. Third, cleavage of PARP-1 inhibits topoisomerase I and/or topoisomerase I-like activity of PARP-1, aggravates DNA DSBs and DNA repair mechanism impairment in H1299 cells. Together, DDR pathway heterogeneity of cancer cells is linked to cancer susceptibility to DNA damage-based chemotherapeutics, which may provide aid in design of chemotherapy strategy to improve treatment outcomes.
Collapse
|
17
|
Romanova LY, Mushinski F, Kovalchuk AL. Transcriptional activation of p21 Waf1 contributes to suppression of HR by p53 in response to replication arrest induced by camptothecin. Oncotarget 2018; 9:25427-25440. [PMID: 29875999 PMCID: PMC5986631 DOI: 10.18632/oncotarget.25172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/21/2018] [Indexed: 11/25/2022] Open
Abstract
The inhibitory effect of p53 on homologous recombination (HR) is exerted through sequestration of replication protein A (RPA). Release of the p53/RPA complex in response to replication stress is crucially dependent on the phosphorylation status of both proteins and is required for efficient DNA repair by HR. Phosphorylation of RPA within its RPA2 subunit by cyclin-dependent kinases (CDK) is an early event in the replication stress response. Here we investigated the role of transcriptional activation of the p53 downstream target, p21Waf1, on RPA2 phosphorylation, the stability of the p53/RPA complex and HR in cells undergoing replication arrest induced by camptothecin (CPT). We show that in CPT-treated cells, activation of p53 and p21Waf1 impedes RPA2 phosphorylation, while their depletion by siRNA stimulates it. The p53/RPA complex is more stable in wild-type cells than in cells depleted of p21Waf1. We used nocodazole-synchronized cells treated with CPT at the entrance to S phase to assess rates of HR. Regardless of their p53 or p21Waf1 status, the cells proceed through S phase at a similar rate and enter G2. While HR is low in wild-type cells and high in p53-depleted cells, only partial inhibition of HR is observed in the p21Waf1-depleted cells. This correlates with the extent of RPA sequestration by p53. Thus, in CPT-treated cells, p53-induced transcriptional activation of p21Waf1 regulates RPA2 phosphorylation, the stability of the p53/RPA complex and HR.
Collapse
Affiliation(s)
- Larisa Y Romanova
- The Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.,The Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, Maryland, USA
| | - Frederick Mushinski
- The Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Alexander L Kovalchuk
- The Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, Maryland, USA
| |
Collapse
|
18
|
Xie J, Gurler Main H, Sacks JD, Muralidhar GG, Barbolina MV. Regulation of DNA damage repair and lipid uptake by CX 3CR1 in epithelial ovarian carcinoma. Oncogenesis 2018; 7:37. [PMID: 29712888 PMCID: PMC5928120 DOI: 10.1038/s41389-018-0046-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/17/2018] [Accepted: 03/26/2018] [Indexed: 12/25/2022] Open
Abstract
Failure of currently used cytotoxic chemotherapy is one of the main reasons behind high mortality from metastatic high grade serous ovarian carcinoma. We found that high expression of a receptor for fractalkine (CX3CR1) significantly correlated with shorter survival of patients with serous ovarian carcinoma treated with cytotoxic DNA damage chemotherapies, and reduction of CX3CR1 expression resulted in sensitization to several DNA damaging modalities, including x-ray radiation and cisplatin. Here, we show that CX3CR1 plays a role in double-strand DNA break response and repair by regulating expression of RAD50 by a MYC-dependent mechanism. We demonstrate that downregulation of CX3CR1 alone and in a combination with irradiation affects peritoneal metastasis in an organ-specific manner; we show that CX3CR1 regulates lipid uptake which could control omental metastasis. This study identifies CX3CR1 as a novel potential target for sensitization of ovarian carcinoma to DNA damage therapies and reduction of peritoneal carcinomatosis.
Collapse
Affiliation(s)
- Jia Xie
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Hilal Gurler Main
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Joelle D Sacks
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Goda G Muralidhar
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Maria V Barbolina
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
19
|
Warmoes M, Lam SW, van der Groep P, Jaspers JE, Smolders YHCM, de Boer L, Pham TV, Piersma SR, Rottenberg S, Boven E, Jonkers J, van Diest PJ, Jimenez CR. Secretome proteomics reveals candidate non-invasive biomarkers of BRCA1 deficiency in breast cancer. Oncotarget 2018; 7:63537-63548. [PMID: 27566577 PMCID: PMC5325383 DOI: 10.18632/oncotarget.11535] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/13/2016] [Indexed: 11/25/2022] Open
Abstract
Breast cancer arising in female BRCA1 mutation carriers is characterized by an aggressive phenotype and early age of onset. We performed tandem mass spectrometry-based proteomics of secretomes and exosome-like extracellular vesicles from BRCA1-deficient and BRCA1-proficient murine breast tumor models to identify extracellular protein biomarkers, which can be used as an adjunct to current diagnostic modalities in patients with BRCA1-deficient breast cancer. We identified 2,107 proteins, of which 215 were highly enriched in the BRCA1-deficient secretome. We demonstrated that BRCA1-deficient secretome proteins could cluster most human BRCA1- and BRCA2-related breast carcinomas at the transcriptome level. Topoisomerase I (TOP1) and P-cadherin (CDH3) expression was investigated by immunohistochemistry on tissue microarrays of a large panel of 253 human breast carcinomas with and without BRCA1/2 mutations. We showed that expression of TOP1 and CDH3 was significantly increased in human BRCA1-related breast carcinomas relative to sporadic cases (p = 0.002 and p < 0.001, respectively). Multiple logistic regression showed that TOP1 (adjusted odds ratio [OR] 3.75; 95% confidence interval [95% CI], 1.85 - 7.71, p < 0.001) as well as CDH3 positivity (adjusted OR 2.45; 95% CI, 1.08 - 5.49, p = 0.032) were associated with BRCA1/2-related breast carcinomas after adjustment for triple-negative phenotype and age. In conclusion, proteome profiling of secretome using murine breast tumor models is a powerful strategy to identify non-invasive candidate biomarkers of BRCA1-deficient breast cancer. We demonstrate that TOP1 and CDH3 are closely associated to BRCA1-deficient breast cancer. These data merit further investigation for early detection of tumors arising in BRCA1 mutation carriers.
Collapse
Affiliation(s)
- Marc Warmoes
- Oncoproteomics Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Siu W Lam
- Oncoproteomics Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Petra van der Groep
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Internal Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Janneke E Jaspers
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Yvonne H C M Smolders
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Leon de Boer
- Oncoproteomics Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Thang V Pham
- Oncoproteomics Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Sander R Piersma
- Oncoproteomics Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Sven Rottenberg
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Switzerland
| | - Epie Boven
- Oncoproteomics Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Jos Jonkers
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Connie R Jimenez
- Oncoproteomics Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Liu S, Liu B. Overexpression of Nitrogen Permease Regulator Like-2 (NPRL2) Enhances Sensitivity to Irinotecan (CPT-11) in Colon Cancer Cells by Activating the DNA Damage Checkpoint Pathway. Med Sci Monit 2018. [PMID: 29519997 PMCID: PMC5857147 DOI: 10.12659/msm.909186] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Background Colorectal cancer (CRC) is the third most common cancer worldwide, making it is a serious threat to human health. It is imperative to develop new therapeutics to improve the CRC treatment efficiency. The aim of this study was to investigate the role of NPRL2 in improving sensitivity to CPT-11 in colon cancer cells. Material/Methods NPRL2 overexpression was established by transfecting the recombinant lentivirus-encoding NPRL2 gene into HCT116 colon cancer cells. Cell proliferation was identified using Cell Counting Kit-8 (CCK8) assay. Cell cycle and apoptosis were examined by flow cytometry. An immunofluorescence staining assay was conducted to examine the expression of γ-H2AX. Wound-healing and Transwell assays were utilized to show cell migration and invasion capability. The expression of apoptosis-related proteins (cleaved caspase-3, caspase-9, cleaved PARP, BAX, and Bcl-2), invasion-related proteins (MMP2, MMP9, p-PI3K, and p-AKT), and DNA damage checkpoint pathway proteins (p-ATM, p-Chk2, Cdc25C, Cdc2, and Cyclin B1) were quantified by Western blotting. Results A CCK8 assay revealed that the overexpression of NPRL2 improved the sensitivity of CPT-11 in HCT116 cells (P<0.05). Functionally, NPRL2 overexpression elevated the sensitivity of CPT-11 by preventing colon cancer cell proliferation, cell movement, and invasion, and promoting cell apoptosis and G2/M cell cycle arrest. Mechanistically, NPRL2 overexpression enhanced CPT-11 sensitivity by activating the DNA damage checkpoint pathway. Conclusions NPRL2 overexpression enhances sensitivity to CPT-11 treatment in colon cancer cells, and it may serve as a molecular therapeutic agent to treat patients with CRC.
Collapse
Affiliation(s)
- Shasha Liu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Bingrong Liu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| |
Collapse
|
21
|
Kjeldsen E, Nielsen CJF, Roy A, Tesauro C, Jakobsen AK, Stougaard M, Knudsen BR. Characterization of Camptothecin-induced Genomic Changes in the Camptothecin-resistant T-ALL-derived Cell Line CPT-K5. Cancer Genomics Proteomics 2018; 15:91-114. [PMID: 29496689 PMCID: PMC5892604 DOI: 10.21873/cgp.20068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 12/27/2022] Open
Abstract
Acquisition of resistance to topoisomerase I (TOP1)-targeting camptothecin (CPT) derivatives is a major clinical problem. Little is known about the underlying chromosomal and genomic mechanisms. We characterized the CPT-K5 cell line expressing mutant CPT-resistant TOP1 and its parental T-cell derived acute lymphoblastic leukemia CPT-sensitive RPMI-8402 cell line by karyotyping and molecular genetic methods, including subtractive oligo-based array comparative genomic hybridization (soaCGH) analysis. Karyotyping revealed that CPT-K5 cells had acquired additional structural aberrations and a reduced modal chromosomal number compared to RPMI-8402. soaCGH analysis identified vast copy number alterations and >200 unbalanced DNA breakpoints distributed unevenly across the chromosomal complement in CPT-K5. In addition, the short tandem repeat alleles were found to be highly different between CPT-K5 and its parental cell line. We identified copy number alterations affecting genes important for maintaining genome integrity and reducing CPT-induced DNA damage. We show for the first time that short tandem repeats are targets for TOP1 cleavage, that can be differentially stimulated by CPT.
Collapse
Affiliation(s)
- Eigil Kjeldsen
- Cancer Cytogenetics Section, HemoDiagnostic Laboratory, Aarhus University Hospital, Aarhus, Denmark
| | - Christine J F Nielsen
- Department of Molecular Biology and Genetics, C.F. Møllers Allé, Aarhus University, Aarhus, Denmark
| | - Amit Roy
- Department of Molecular Biology and Genetics, C.F. Møllers Allé, Aarhus University, Aarhus, Denmark
| | - Cinzia Tesauro
- Department of Molecular Biology and Genetics, C.F. Møllers Allé, Aarhus University, Aarhus, Denmark
| | | | - Magnus Stougaard
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Birgitta R Knudsen
- Department of Molecular Biology and Genetics, C.F. Møllers Allé, Aarhus University, Aarhus, Denmark
| |
Collapse
|
22
|
Abstract
Topoisomerase I (Top1) resolves torsional stress that accumulates during transcription, replication and chromatin remodeling by introducing a transient single-strand break in DNA. The cleavage activity of Top1 has opposing roles, either promoting or destabilizing genome integrity depending on the context. Resolution of transcription-associated negative supercoils, for example, prevents pairing of the nascent RNA with the DNA template (R-loops) as well as DNA secondary structure formation. Reduced Top1 levels thus enhance CAG repeat contraction, somatic hypermutation, and class switch recombination. Actively transcribed ribosomal DNA is also destabilized in the absence of Top1, reflecting the importance of Top1 in ensuring efficient transcription. In terms of promoting genome instability, an aborted Top1 catalytic cycle stimulates deletions at short tandem repeats and the enzyme's transesterification activity supports illegitimate recombination. Finally, Top1 incision at ribonucleotides embedded in DNA generates deletions in tandem repeats, and induces gross chromosomal rearrangements and mitotic recombination.
Collapse
Affiliation(s)
- Jang-Eun Cho
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, CARL 384, Durham, NC, 27710, USA
| | - Sue Jinks-Robertson
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, CARL 384, Durham, NC, 27710, USA.
| |
Collapse
|
23
|
Shamanna RA, Lu H, Croteau DL, Arora A, Agarwal D, Ball G, Aleskandarany MA, Ellis IO, Pommier Y, Madhusudan S, Bohr VA. Camptothecin targets WRN protein: mechanism and relevance in clinical breast cancer. Oncotarget 2017; 7:13269-84. [PMID: 26959889 PMCID: PMC4924640 DOI: 10.18632/oncotarget.7906] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/09/2016] [Indexed: 12/22/2022] Open
Abstract
Werner syndrome protein (WRN) is a RecQ helicase that participates in DNA repair, genome stability and cellular senescence. The five human RecQ helicases, RECQL1, Bloom, WRN, RECQL4 and RECQL5 play critical roles in DNA repair and cell survival after treatment with the anticancer drug camptothecin (CPT). CPT derivatives are widely used in cancer chemotherapy to inhibit topoisomerase I and generate DNA double-strand breaks during replication. Here we studied the effects of CPT on the stability and expression dynamics of human RecQ helicases. In the cells treated with CPT, we observed distinct effects on WRN compared to other human RecQ helicases. CPT altered the cellular localization of WRN and induced its degradation by a ubiquitin-mediated proteasome pathway. WRN knockdown cells as well as CPT treated cells became senescent and stained positive for senescence-associated β-galactosidase at a higher frequency compared to control cells. However, the senescent phenotype was attenuated by ectopic expression of WRN suggesting functional implication of WRN degradation in CPT treated cells. Approximately 5-23% of breast cancer tumors are known to respond to CPT-based chemotherapy. Interestingly, we found that the extent of CPT-induced WRN degradation correlates with increasing sensitivity of breast cancer cells to CPT. The abundance of WRN decreased in CPT-treated sensitive cells; however, WRN remained relatively stable in CPT-resistant breast cancer cells. In a large clinical cohort of breast cancer patients, we find that WRN and topoisomerase I expression correlate with an aggressive tumor phenotype and poor prognosis. Our novel observations suggest that WRN abundance along with CPT-induced degradation could be a promising strategy for personalizing CPT-based cancer chemotherapeutic regimens.
Collapse
Affiliation(s)
- Raghavendra A Shamanna
- Laboratory of Molecular Gerontology, Biomedical Research Center, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Huiming Lu
- Laboratory of Molecular Gerontology, Biomedical Research Center, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Deborah L Croteau
- Laboratory of Molecular Gerontology, Biomedical Research Center, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Arvind Arora
- Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Devika Agarwal
- School of Science and Technology, Nottingham Trent University, Clifton Campus, Nottingham, UK
| | - Graham Ball
- School of Science and Technology, Nottingham Trent University, Clifton Campus, Nottingham, UK
| | - Mohammed A Aleskandarany
- Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Ian O Ellis
- Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, National Cancer Institute, Bethesda, Maryland, USA
| | - Srinivasan Madhusudan
- Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, Biomedical Research Center, National Institute on Aging, NIH, Baltimore, Maryland, USA
| |
Collapse
|
24
|
Meisenberg C, Ashour ME, El-Shafie L, Liao C, Hodgson A, Pilborough A, Khurram SA, Downs JA, Ward SE, El-Khamisy SF. Epigenetic changes in histone acetylation underpin resistance to the topoisomerase I inhibitor irinotecan. Nucleic Acids Res 2017; 45:1159-1176. [PMID: 28180300 PMCID: PMC5388393 DOI: 10.1093/nar/gkw1026] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 10/08/2016] [Accepted: 10/18/2016] [Indexed: 02/06/2023] Open
Abstract
The topoisomerase I (TOP1) inhibitor irinotecan triggers cell death by trapping TOP1 on DNA, generating cytotoxic protein-linked DNA breaks (PDBs). Despite its wide application in a variety of solid tumors, the mechanisms of cancer cell resistance to irinotecan remains poorly understood. Here, we generated colorectal cancer (CRC) cell models for irinotecan resistance and report that resistance is neither due to downregulation of the main cellular target of irinotecan TOP1 nor upregulation of the key TOP1 PDB repair factor TDP1. Instead, the faster repair of PDBs underlies resistance, which is associated with perturbed histone H4K16 acetylation. Subsequent treatment of irinotecan-resistant, but not parental, CRC cells with histone deacetylase (HDAC) inhibitors can effectively overcome resistance. Immunohistochemical analyses of CRC tissues further corroborate the importance of histone H4K16 acetylation in CRC. Finally, the resistant clones exhibit cross-resistance with oxaliplatin but not with ionising radiation or 5-fluoruracil, suggesting that the latter two could be employed following loss of irinotecan response. These findings identify perturbed chromatin acetylation in irinotecan resistance and establish HDAC inhibitors as potential therapeutic means to overcome resistance.
Collapse
Affiliation(s)
- Cornelia Meisenberg
- Mammalian Genome Stability Group, Krebs and Sheffield Institute for Nucleic Acids, University of Sheffield, Western Bank, Sheffield, UK.,The Wellcome Trust DNA Repair Group, University of Sussex, Brighton, UK
| | - Mohamed E Ashour
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City for Science and Technology, Giza, Egypt.,Mammalian Genome Stability Group, Krebs and Sheffield Institute for Nucleic Acids, University of Sheffield, Western Bank, Sheffield, UK
| | - Lamia El-Shafie
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City for Science and Technology, Giza, Egypt
| | - Chunyan Liao
- Mammalian Genome Stability Group, Krebs and Sheffield Institute for Nucleic Acids, University of Sheffield, Western Bank, Sheffield, UK
| | - Adam Hodgson
- Mammalian Genome Stability Group, Krebs and Sheffield Institute for Nucleic Acids, University of Sheffield, Western Bank, Sheffield, UK
| | - Alice Pilborough
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, UK
| | - Syed A Khurram
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, UK
| | - Jessica A Downs
- Genome Damage and Stability Centre, University of Sussex, Brighton, UK
| | - Simon E Ward
- Sussex Drug Discovery Centre, School of Life Sciences, University of Sussex, Brighton BN1 9QJ, UK
| | - Sherif F El-Khamisy
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City for Science and Technology, Giza, Egypt.,Mammalian Genome Stability Group, Krebs and Sheffield Institute for Nucleic Acids, University of Sheffield, Western Bank, Sheffield, UK.,The Wellcome Trust DNA Repair Group, University of Sussex, Brighton, UK
| |
Collapse
|
25
|
Uchihashi T, Ota K, Yabuno Y, Ohno S, Fukushima K, Naito Y, Kogo M, Yabuta N, Nojima H. ELAS1 induces apoptotic death in adenocarcinoma DU145 and squamous-cell carcinoma SAS cancer cells, but not in normal KD cells. Oncotarget 2017; 8:85868-85882. [PMID: 29156763 PMCID: PMC5689653 DOI: 10.18632/oncotarget.20696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 05/03/2017] [Indexed: 12/31/2022] Open
Abstract
We previously reported that an ELAS1 peptide containing 29 amino acids induces apoptotic death in U2OS human osteosarcoma cells following DNA double-strand break insults. Here, we show that ELAS1 also caused apoptosis in prostate adenocarcinoma DU145 cells and tongue squamous-cell carcinoma SAS cells. ELAS1 appears to be safe because it induced apoptosis only in cancer cells, not in normal KD cells. Because the effect of ELAS1 is dependent on increased stability of p53 and enhanced phosphorylation of p53-S46, we exogenously expressed wild-type p53 protein to fully promote ELAS1-mediated induction of apoptosis in SAS cells. Interestingly, simultaneous expression of Myc-ELAS1 and FLAG-p53 mediated by an internal ribosome entry site efficiently induced apoptosis in SAS cells. Moreover, we prepared a recombinant adenovirus that simultaneously expressed Myc-ELAS1 and FLAG-p53. This adenovirus also killed SAS cells, as determined by a cell viability assay, in the presence of camptothecin, an inducer of DNA double-strand breaks. Moreover, nude mice harboring Myc-ELAS1-expressing SAS cells lived longer than mice harboring Myc-vector-expressing SAS cells, suggesting the usefulness of ELAS1 in vivo. Notably, Cy5-tagged ELAS1-t, which contained only ten amino acids, also efficiently induced apoptosis in both DU145 and SAS cells, suggesting the usefulness of ELAS1-t as a peptide. Taken together, our results suggest that ELAS1 is therapeutically useful as a peptide drug.
Collapse
Affiliation(s)
- Toshihiro Uchihashi
- First Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Kaori Ota
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Yusuke Yabuno
- First Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Shouichi Ohno
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Kohshiro Fukushima
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Yoko Naito
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Mikihiko Kogo
- First Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Norikazu Yabuta
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Hiroshi Nojima
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
26
|
Lehmann J, Seebode C, Smolorz S, Schubert S, Emmert S. XPF knockout via CRISPR/Cas9 reveals that ERCC1 is retained in the cytoplasm without its heterodimer partner XPF. Cell Mol Life Sci 2017; 74:2081-2094. [PMID: 28130555 PMCID: PMC11107539 DOI: 10.1007/s00018-017-2455-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/01/2016] [Accepted: 01/03/2017] [Indexed: 01/05/2023]
Abstract
The XPF/ERCC1 heterodimeric complex is essentially involved in nucleotide excision repair (NER), interstrand crosslink (ICL), and double-strand break repair. Defects in XPF lead to severe diseases like xeroderma pigmentosum (XP). Up until now, XP-F patient cells have been utilized for functional analyses. Due to the multiple roles of the XPF/ERCC1 complex, these patient cells retain at least one full-length allele and residual repair capabilities. Despite the essential function of the XPF/ERCC1 complex for the human organism, we successfully generated a viable immortalised human XPF knockout cell line with complete loss of XPF using the CRISPR/Cas9 technique in fetal lung fibroblasts (MRC5Vi cells). These cells showed a markedly increased sensitivity to UVC, cisplatin, and psoralen activated by UVA as well as reduced repair capabilities for NER and ICL repair as assessed by reporter gene assays. Using the newly generated knockout cells, we could show that human XPF is markedly involved in homologous recombination repair (HRR) but dispensable for non-homologous end-joining (NHEJ). Notably, ERCC1 was not detectable in the nucleus of the XPF knockout cells indicating the necessity of a functional XPF/ERCC1 heterodimer to allow ERCC1 to enter the nucleus. Overexpression of wild-type XPF could reverse this effect as well as the repair deficiencies.
Collapse
Affiliation(s)
- Janin Lehmann
- Clinic and Policlinic for Dermatology and Venereology, University Medical Centre Rostock, Strempelstrasse 13, 18057, Rostock, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Centre Goettingen, Robert-Koch-Strasse 40, 37075, Goettingen, Germany
| | - Christina Seebode
- Clinic and Policlinic for Dermatology and Venereology, University Medical Centre Rostock, Strempelstrasse 13, 18057, Rostock, Germany
| | - Sabine Smolorz
- Department of Dermatology, Venereology and Allergology, University Medical Centre Goettingen, Robert-Koch-Strasse 40, 37075, Goettingen, Germany
| | - Steffen Schubert
- Department of Dermatology, Venereology and Allergology, University Medical Centre Goettingen, Robert-Koch-Strasse 40, 37075, Goettingen, Germany
| | - Steffen Emmert
- Clinic and Policlinic for Dermatology and Venereology, University Medical Centre Rostock, Strempelstrasse 13, 18057, Rostock, Germany.
- Department of Dermatology, Venereology and Allergology, University Medical Centre Goettingen, Robert-Koch-Strasse 40, 37075, Goettingen, Germany.
| |
Collapse
|
27
|
Deneka AY, Haber L, Kopp MC, Gaponova AV, Nikonova AS, Golemis EA. Tumor-targeted SN38 inhibits growth of early stage non-small cell lung cancer (NSCLC) in a KRas/p53 transgenic mouse model. PLoS One 2017; 12:e0176747. [PMID: 28453558 PMCID: PMC5409145 DOI: 10.1371/journal.pone.0176747] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/14/2017] [Indexed: 11/19/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer death worldwide, with a 5-year survival of only ~16%. Potential strategies to address NSCLC mortality include improvements in early detection and prevention, and development of new therapies suitable for use in patients with early and late stage diagnoses. Controlling the growth of early stage tumors could yield significant clinical benefits for patients with comorbidities that make them poor candidates for surgery: however, many drugs that limit cancer growth are not useful in the setting of long-term use or in comorbid patients, because of associated toxicities. In this study, we explored the use of a recently described small molecule agent, STA-8666, as a potential agent for controlling early stage tumor growth. STA-8666 uses a cleavable linker to merge a tumor-targeting moiety that binds heat shock protein 90 (HSP90) with the cytotoxic chemical SN38, and has been shown to have high efficacy and low toxicity, associated with efficient tumor targeting, in preclinical studies using patient-derived and other xenograft models for pancreatic, bladder, and small cell lung cancer. Using a genetically engineered model of NSCLC arising from induced mutation of KRas and knockout of Trp53, we continuously dosed mice with STA-8666 from immediately after tumor induction for 15 weeks. STA-8666 significantly slowed the rate of tumor growth, and was well tolerated over this extended dosing period. STA-8666 induced DNA damage and apoptosis, and reduced proliferation and phosphorylation of the proliferation-associated protein ERK1/2, selectively in tumor tissue. In contrast, STA-8666 did not affect tumor features, such as degree of vimentin staining, associated with epithelial-mesenchymal transition (EMT), or downregulate tumor expression of HSP90. These data suggest STA-8666 and other similar targeted compounds may be useful additions to control the growth of early stage NSCLC in patient populations.
Collapse
Affiliation(s)
- Alexander Y. Deneka
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
- Department of Biochemistry, Kazan Federal University, Kazan, Russia
- * E-mail: (EG); (AD)
| | - Leora Haber
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Meghan C. Kopp
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
- Program in Molecular and Cell Biology and Genetics, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Anna V. Gaponova
- Laboratory of Genome Engineering, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Immanuel Kant Baltic Federal University, Konigsberg, Russia
| | - Anna S. Nikonova
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Erica A. Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
- * E-mail: (EG); (AD)
| |
Collapse
|
28
|
Apoptosis induced by temozolomide and nimustine in glioblastoma cells is supported by JNK/c-Jun-mediated induction of the BH3-only protein BIM. Oncotarget 2016; 6:33755-68. [PMID: 26418950 PMCID: PMC4741800 DOI: 10.18632/oncotarget.5274] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 09/03/2015] [Indexed: 12/16/2022] Open
Abstract
The outcome of cancer therapy strongly depends on the complex network of cell signaling pathways, including transcription factor activation following drug exposure. Here we assessed whether and how the MAP kinase (MAPK) cascade and its downstream target, the transcription factor AP-1, influence the sensitivity of malignant glioma cells to the anticancer drugs temozolomide (TMZ) and nimustine (ACNU). Both drugs induce apoptosis in glioma cells at late times following treatment. Activation of the MAPK cascade precedes apoptosis, as shown by phosphorylation of Jun kinase (JNK) and c-Jun, a main component of AP-1. Pharmacological inhibition and siRNA mediated knockdown of JNK and c-Jun reduced the level of apoptosis in LN-229 glioma cells treated with TMZ or ACNU. Analyzing the underlying molecular mechanism, we identified the pro-apoptotic gene BIM as a critical target of AP-1, which is upregulated following TMZ and ACNU. Importantly, shRNA mediated downregulation of BIM in the malignant glioma cell lines LN-229 and U87MG led to an attenuated cleavage of caspase-9 and, consequently, reduced the level of apoptosis following TMZ and ACNU treatment. Overall, we identified JNK/c-Jun activation and BIM induction as a late pro-apoptotic response of glioma cells treated with alkylating anticancer drugs.
Collapse
|
29
|
Gmeiner WH, Gearhart PJ, Pommier Y, Nakamura J. F10 cytotoxicity via topoisomerase I cleavage complex repair consistent with a unique mechanism for thymineless death. Future Oncol 2016; 12:2183-8. [PMID: 27333295 DOI: 10.2217/fon-2016-0127] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- William H Gmeiner
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Patricia J Gearhart
- Laboratory of Molecular Biology & Immunology, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892-4255, USA
| | - Jun Nakamura
- University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
30
|
Gaponova AV, Nikonova AS, Deneka A, Kopp MC, Kudinov AE, Skobeleva N, Khazak V, Ogawa LS, Cai KQ, Duncan KE, Duncan JS, Egleston BL, Proia DA, Boumber Y, Golemis EA. A Novel HSP90 Inhibitor-Drug Conjugate to SN38 Is Highly Effective in Small Cell Lung Cancer. Clin Cancer Res 2016; 22:5120-5129. [PMID: 27267850 DOI: 10.1158/1078-0432.ccr-15-3068] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 05/26/2016] [Indexed: 01/08/2023]
Abstract
PURPOSE Small cell lung cancer (SCLC) is a highly aggressive disease representing 12% to 13% of total lung cancers, with median survival of <2 years. No targeted therapies have proven effective in SCLC. Although most patients respond initially to cytotoxic chemotherapies, resistance rapidly emerges, response to second-line agents is limited, and dose-limiting toxicities (DLT) are a major issue. This study performs preclinical evaluation of a new compound, STA-8666, in SCLC. EXPERIMENTAL DESIGN To avoid DLT for useful cytotoxic agents, the recently developed drug STA-8666 combines a chemical moiety targeting active HSP90 (concentrated in tumors) fused via cleavable linker to SN38, the active metabolite of irinotecan. We compare potency and mechanism of action of STA-8666 and irinotecan in vitro and in vivo RESULTS: In two SCLC xenograft and patient-derived xenograft models, STA-8666 was tolerated without side effects up to 150 mg/kg. At this dose, STA-8666 controlled or eliminated established tumors whether used in a first-line setting or in tumors that had progressed following treatment on standard first- and second-line agents for SCLC. At 50 mg/kg, STA-8666 strongly enhanced the action of carboplatin. Pharmacokinetic profiling confirmed durable STA-8666 exposure in tumors compared with irinotecan. STA-8666 induced a more rapid, robust, and stable induction of cell-cycle arrest, expression of signaling proteins associated with DNA damage and cell-cycle checkpoints, and apoptosis in vitro and in vivo, in comparison with irinotecan. CONCLUSIONS Together, these results strongly support clinical development of STA-8666 for use in the first- or second-line setting for SCLC. Clin Cancer Res; 22(20); 5120-9. ©2016 AACR.
Collapse
Affiliation(s)
- Anna V Gaponova
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.,Department of Biochemistry, Kazan Federal University, Kazan 420008, Russia
| | - Anna S Nikonova
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Alexander Deneka
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.,Department of Biochemistry, Kazan Federal University, Kazan 420008, Russia
| | - Meghan C Kopp
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.,Program in Molecular and Cell Biology and Genetics, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Alexander E Kudinov
- University of New Mexico Comprehensive Cancer Center, University of New Mexico School of Medicine, Division of Hematology/Oncology, Department of Internal Medicine, Albuquerque, NM 87131
| | - Natalia Skobeleva
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | | - Kathy Q Cai
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Kelly E Duncan
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - James S Duncan
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Brian L Egleston
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | - Yanis Boumber
- University of New Mexico Comprehensive Cancer Center, University of New Mexico School of Medicine, Division of Hematology/Oncology, Department of Internal Medicine, Albuquerque, NM 87131
| | - Erica A Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| |
Collapse
|
31
|
Fenoy IM, Bogado SS, Contreras SM, Gottifredi V, Angel SO. The Knowns Unknowns: Exploring the Homologous Recombination Repair Pathway in Toxoplasma gondii. Front Microbiol 2016; 7:627. [PMID: 27199954 PMCID: PMC4853372 DOI: 10.3389/fmicb.2016.00627] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 04/18/2016] [Indexed: 12/17/2022] Open
Abstract
Toxoplasma gondii is an apicomplexan parasite of medical and veterinary importance which causes toxoplasmosis in humans. Great effort is currently being devoted toward the identification of novel drugs capable of targeting such illness. In this context, we believe that the thorough understanding of the life cycle of this model parasite will facilitate the identification of new druggable targets in T. gondii. It is important to exploit the available knowledge of pathways which could modulate the sensitivity of the parasite to DNA damaging agents. The homologous recombination repair (HRR) pathway may be of particular interest in this regard as its inactivation sensitizes other cellular models such as human cancer to targeted therapy. Herein we discuss the information available on T. gondii's HRR pathway from the perspective of its conservation with respect to yeast and humans. Special attention was devoted to BRCT domain-containing and end-resection associated proteins in T. gondii as in other experimental models such proteins have crucial roles in early/late steps or HRR and in the pathway choice for double strand break resolution. We conclude that T. gondii HRR pathway is a source of several lines of investigation that allow to to comprehend the extent of diversification of HRR in T. gondii. Such an effort will serve to determine if HRR could represent a potential targer for the treatment of toxoplasmosis.
Collapse
Affiliation(s)
- Ignacio M Fenoy
- Laboratorio de Parasitología Molecular, IIB-INTECH, CONICET-UNSAM Chascomús, Argentina
| | - Silvina S Bogado
- Laboratorio de Parasitología Molecular, IIB-INTECH, CONICET-UNSAM Chascomús, Argentina
| | - Susana M Contreras
- Laboratorio de Parasitología Molecular, IIB-INTECH, CONICET-UNSAM Chascomús, Argentina
| | - Vanesa Gottifredi
- Cell Cycle Genomic Instability Laboratory, Fundación Instituto Leloir, IIBBA-CONICET Chascomús, Argentina
| | - Sergio O Angel
- Laboratorio de Parasitología Molecular, IIB-INTECH, CONICET-UNSAM Chascomús, Argentina
| |
Collapse
|
32
|
Patel AG, Flatten KS, Peterson KL, Beito TG, Schneider PA, Perkins AL, Harki DA, Kaufmann SH. Immunodetection of human topoisomerase I-DNA covalent complexes. Nucleic Acids Res 2016; 44:2816-26. [PMID: 26917015 PMCID: PMC4824114 DOI: 10.1093/nar/gkw109] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/15/2016] [Indexed: 12/29/2022] Open
Abstract
A number of established and investigational anticancer drugs slow the religation step of DNA topoisomerase I (topo I). These agents induce cytotoxicity by stabilizing topo I-DNA covalent complexes, which in turn interact with advancing replication forks or transcription complexes to generate lethal lesions. Despite the importance of topo I-DNA covalent complexes, it has been difficult to detect these lesions within intact cells and tumors. Here, we report development of a monoclonal antibody that specifically recognizes covalent topo I-DNA complexes, but not free topo I or DNA, by immunoblotting, immunofluorescence or flow cytometry. Utilizing this antibody, we demonstrate readily detectable topo I-DNA covalent complexes after treatment with camptothecins, indenoisoquinolines and cisplatin but not nucleoside analogues. Topotecan-induced topo I-DNA complexes peak at 15-30 min after drug addition and then decrease, whereas indotecan-induced complexes persist for at least 4 h. Interestingly, simultaneous staining for covalent topo I-DNA complexes, phospho-H2AX and Rad51 suggests that topotecan-induced DNA double-strand breaks occur at sites distinct from stabilized topo I-DNA covalent complexes. These studies not only provide new insight into the action of topo I-directed agents, but also illustrate a strategy that can be applied to study additional topoisomerases and their inhibitors in vitro and in vivo.
Collapse
Affiliation(s)
- Anand G Patel
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Karen S Flatten
- Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Kevin L Peterson
- Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Thomas G Beito
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Paula A Schneider
- Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Angela L Perkins
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel A Harki
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Scott H Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
33
|
Shao J, Xu Z, Peng X, Chen M, Zhu Y, Xu L, Zhu H, Yang B, Luo P, He Q. Gefitinib Synergizes with Irinotecan to Suppress Hepatocellular Carcinoma via Antagonizing Rad51-Mediated DNA-Repair. PLoS One 2016; 11:e0146968. [PMID: 26752698 PMCID: PMC4709237 DOI: 10.1371/journal.pone.0146968] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/23/2015] [Indexed: 11/24/2022] Open
Abstract
Chemotherapy is the only choice for most of the advanced hepatocellular carcinoma (HCC) patients, while few agents were available, making it an urgent need to develop new chemotherapy strategies. A phase II clinical trial suggested that the efficacy of irinotecan in HCC was limited due to dose-dependent toxicities. Here, we found that gefitinib exhibited synergistic activity in combination with SN-38, an active metabolite of irinotecan, in HCC cell lines. And the enhanced apoptosis induced by gefitinib plus SN-38 was a result from caspase pathway activation. Mechanistically, gefitinib dramatically promoted the ubiquitin–proteasome-dependent degradation of Rad51 protein, suppressed the DNA repair, gave rise to more DNA damages, and ultimately resulted in the synergism of these two agents. In addition, the increased antitumor efficacy of gefitinib combined with irinotecan was further validated in a HepG2 xenograft mice model. Taken together, our data demonstrated for the first time that the combination of irinotecan and gefitinib showed potential benefit in HCC, which suggests that Rad51 is a promising target and provides a rationale for clinical trials investigating the efficacy of the combination of topoisomerase I inhibitors and gefitinib in HCC.
Collapse
Affiliation(s)
- Jinjin Shao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhifei Xu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xueming Peng
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Min Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuanrun Zhu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Li Xu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hong Zhu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Peihua Luo
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- * E-mail: (PL); (QH)
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- * E-mail: (PL); (QH)
| |
Collapse
|
34
|
Tabanor K, Lee P, Kiptoo P, Choi IY, Sherry EB, Eagle CS, Williams TD, Siahaan TJ. Brain Delivery of Drug and MRI Contrast Agent: Detection and Quantitative Determination of Brain Deposition of CPT-Glu Using LC-MS/MS and Gd-DTPA Using Magnetic Resonance Imaging. Mol Pharm 2016; 13:379-90. [PMID: 26705088 DOI: 10.1021/acs.molpharmaceut.5b00607] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Successful treatment and diagnosis of neurological diseases depend on reliable delivery of molecules across the blood-brain barrier (BBB), which restricts penetration of pharmaceutical drugs and diagnostic agents into the brain. Thus, developing new noninvasive strategies to improve drug delivery across the BBB is critically needed. This study was aimed at evaluating the activity of HAV6 peptide (Ac-SHAVSS-NH2) in improving brain delivery of camptothecin-glutamate (CPT-Glu) conjugate and gadolinium-diethylenetriaminepentaacetate (Gd-DTPA) contrast agent in Sprague-Dawley rats. Brain delivery of both CPT-Glu and Gd-DTPA was evaluated in an in situ rat brain perfusion model in the presence and absence of HAV6 peptide (1.0 mM). Gd-DTPA (0.6 mmol/kg) was intravenously (iv) administered with and without HAV6 peptide (0.019 mmol/kg) in rats. The detection and quantification of CPT-Glu and Gd-DTPA in the brain were carried out by LC-MS/MS and quantitative magnetic resonance imaging (MRI), respectively. Rats perfused with CPT-Glu in combination with HAV6 had significantly higher deposition of drug in the brain compared to CPT-Glu alone. MRI results also showed that administration of Gd-DTPA in the presence of HAV6 peptide led to significant accumulation of Gd-DTPA in various regions of the brain in both the in situ rat brain perfusion and in vivo studies. All observations taken together indicate that HAV6 peptide can disrupt the BBB and enhance delivery of small molecules into the brain.
Collapse
Affiliation(s)
- Kayann Tabanor
- Department of Pharmaceutical Chemistry, The University of Kansas , Lawrence, Kansas 66047, United States
| | | | - Paul Kiptoo
- Department of Pharmaceutical Chemistry, The University of Kansas , Lawrence, Kansas 66047, United States
| | | | | | - Cheyenne Sun Eagle
- Department of Pharmaceutical Chemistry, The University of Kansas , Lawrence, Kansas 66047, United States
| | - Todd D Williams
- Mass Spectrometry Laboratory, The University of Kansas , Lawrence, Kansas 66047, United States
| | - Teruna J Siahaan
- Department of Pharmaceutical Chemistry, The University of Kansas , Lawrence, Kansas 66047, United States
| |
Collapse
|
35
|
Jović DS, Seke MN, Djordjevic AN, Mrđanović JŽ, Aleksić LD, Bogdanović GM, Pavić AB, Plavec J. Fullerenol nanoparticles as a new delivery system for doxorubicin. RSC Adv 2016. [DOI: 10.1039/c6ra03879d] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Fullerenol as a promising intracellular targeting carrier for the efficient delivery of antitumor drugs into tumor cells.
Collapse
Affiliation(s)
- Danica S. Jović
- Department of Chemistry
- Biochemistry and Environmental Protection
- Faculty of Sciences
- University of Novi Sad
- Novi Sad
| | - Mariana N. Seke
- Institute of Nuclear Sciences “Vinča”
- University of Belgrade
- Belgrade
- Serbia
| | - Aleksandar N. Djordjevic
- Department of Chemistry
- Biochemistry and Environmental Protection
- Faculty of Sciences
- University of Novi Sad
- Novi Sad
| | - Jasminka Ž. Mrđanović
- Oncology Institute of Vojvodina
- Faculty of Medicine
- University of Novi Sad
- Sremska Kamenica
- Serbia
| | - Lidija D. Aleksić
- Oncology Institute of Vojvodina
- Faculty of Medicine
- University of Novi Sad
- Sremska Kamenica
- Serbia
| | - Gordana M. Bogdanović
- Oncology Institute of Vojvodina
- Faculty of Medicine
- University of Novi Sad
- Sremska Kamenica
- Serbia
| | - Aleksandar B. Pavić
- Institute of Molecular Genetics and Genetic Engineering
- University of Belgrade
- Belgrade
- Serbia
| | - Janez Plavec
- Slovenian NMR Centre
- National Institute of Chemistry
- Ljubljana
- Slovenia
| |
Collapse
|
36
|
Prados MD, Byron SA, Tran NL, Phillips JJ, Molinaro AM, Ligon KL, Wen PY, Kuhn JG, Mellinghoff IK, de Groot JF, Colman H, Cloughesy TF, Chang SM, Ryken TC, Tembe WD, Kiefer JA, Berens ME, Craig DW, Carpten JD, Trent JM. Toward precision medicine in glioblastoma: the promise and the challenges. Neuro Oncol 2015; 17:1051-63. [PMID: 25934816 DOI: 10.1093/neuonc/nov031] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 02/15/2015] [Indexed: 12/17/2022] Open
Abstract
Integrated sequencing strategies have provided a broader understanding of the genomic landscape and molecular classifications of multiple cancer types and have identified various therapeutic opportunities across cancer subsets. Despite pivotal advances in the characterization of genomic alterations in glioblastoma, targeted agents have shown minimal efficacy in clinical trials to date, and patient survival remains poor. In this review, we highlight potential reasons why targeting single alterations has yielded limited clinical efficacy in glioblastoma, focusing on issues of tumor heterogeneity and pharmacokinetic failure. We outline strategies to address these challenges in applying precision medicine to glioblastoma and the rationale for applying targeted combination therapy approaches that match genomic alterations with compounds accessible to the central nervous system.
Collapse
Affiliation(s)
- Michael D Prados
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Sara A Byron
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Nhan L Tran
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Joanna J Phillips
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Annette M Molinaro
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Keith L Ligon
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Patrick Y Wen
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - John G Kuhn
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Ingo K Mellinghoff
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - John F de Groot
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Howard Colman
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Timothy F Cloughesy
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Susan M Chang
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Timothy C Ryken
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Waibhav D Tembe
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Jeffrey A Kiefer
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Michael E Berens
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - David W Craig
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - John D Carpten
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| | - Jeffrey M Trent
- University of California San Francisco, San Francisco, California (M.D.P, J.J.P., A.M.M., S.M.C.); Translational Genomics Research Institute, Phoenix, Arizona (S.A.B., N.L.T., W.D.T., J.A.K., M.E.B., D.W.C., J.D.C., J.M.T.); Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (K.L.L., P.Y.W.); University of Texas Health Science Center, San Antonio, Texas (J.G.K.); Memorial Sloan-Kettering Cancer Center, New York, New York (I.K.M.); The University of Texas M.D. Anderson Cancer Center, Houston, Texas (J.F.d.G.); University of Utah Huntsman Cancer Institute, Salt Lake City, Utah (H.C.); University of California Los Angeles, Los Angeles, California (T.F.C.); Iowa Spine and Brain Institute, Waterloo, Iowa (T.C.R.)
| |
Collapse
|
37
|
Ohno S, Naito Y, Mukai S, Yabuta N, Nojima H. ELAS1-mediated inhibition of the cyclin G1-B'γ interaction promotes cancer cell apoptosis via stabilization and activation of p53. Oncogene 2015; 34:5983-96. [PMID: 25915850 DOI: 10.1038/onc.2015.47] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/09/2015] [Accepted: 01/27/2015] [Indexed: 12/16/2022]
Abstract
Radiation therapy (RT) is useful for selectively killing cancer cells. However, because high levels of ionizing radiation (IR) are toxic to normal cells, RT cannot be applied repeatedly to cancer patients. Therefore, novel chemicals that enhance the efficacy of chemoradiotherapy (CRT) would be valuable. Here, we report that ELAS1, a peptide corresponding to the protein phosphatase 2A (PP2A) association domain of cyclin G1 (CycG1), can enhance the efficacy of CRT. ELAS1 interacts with the PP2A B'γ-subunit and competitively inhibits association with CycG1, thereby preventing the PP2A holoenzyme from dephosphorylating target proteins, Mdm2 (pT218) and p53 (pS46), following DNA double-strand break (DSB) insults. Doxycycline (Dox)-induced overexpression of Myc-ELAS1 caused γ-irradiation to induce apoptosis in human osteosarcoma (U2OS) cells, at 1/10th the effective dosage of γ-irradiation required for apoptosis in Myc-vector-expressing cells; ELAS1 peptide incorporation into U2OS cells also showed similar apoptotic effects. Moreover, administration of DSB-inducing chemicals, camptothecin (CPT) or irinotecan, to Myc-ELAS1-expressing U2OS cells also induced efficient apoptosis with only 1/100th (CPT) or 1/5th (irinotecan) of the amounts of drugs required for this effect in Myc-vector-expressing cells. Taken together, ELAS1 may be important for the design of ELAS1-mimetic compounds to improve CRT efficacy.
Collapse
Affiliation(s)
- S Ohno
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Y Naito
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - S Mukai
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - N Yabuta
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - H Nojima
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
38
|
Zuco V, De Cesare M, Zaffaroni N, Lanzi C, Cassinelli G. PLK1 is a critical determinant of tumor cell sensitivity to CPT11 and its inhibition enhances the drug antitumor efficacy in squamous cell carcinoma models sensitive and resistant to camptothecins. Oncotarget 2015; 6:8736-49. [PMID: 25826089 PMCID: PMC4496180 DOI: 10.18632/oncotarget.3538] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/10/2015] [Indexed: 01/21/2023] Open
Abstract
Intrinsic and acquired tumor drug resistance limits the therapeutic efficacy of camptothecins (CPTs). Downregulation of the mitotic kinase PLK1 was found associated with apoptosis induced by SN38 (CPT11 active metabolite). We investigated the role of PLK1 in the cell response to CPTs in squamous cell carcinoma (SCC) and pediatric sarcoma cell lines and explored the therapeutic potential of the combination of CPT11 and the PLK1 inhibitor BI2536 in CPT-sensitive and -resistant tumor models. Gain- and loss-of-function experiments established a direct role for PLK1 in counteracting SN38 antiproliferative and pro-apoptotic effects. The ability to activate an efficient G2/M cell cycle checkpoint allowing PLK1 ubiquitination and degradation was found associated with SN38-induced apoptosis in SCC cells. However, the synergistic interaction between SN38 and BI2536 enhanced apoptosis in cell lines both sensitive and resistant to SN38-induced apoptotic cell death. A well-tolerated CPT11/BI2536 cotreatment resulted in improved antitumor effect against SCC xenografts in mice compared to single agent treatments. The increased apoptosis induction was reflected in a high rate of complete responses and cures in mice harboring SCC, including tumors with intrinsic or acquired resistance to CPTs. PLK1 inhibition represents a promising strategy to improve the antitumor efficacy of CPT11-based regimens.
Collapse
Affiliation(s)
- Valentina Zuco
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michelandrea De Cesare
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Cinzia Lanzi
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuliana Cassinelli
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
39
|
Ashour ME, Atteya R, El-Khamisy SF. Topoisomerase-mediated chromosomal break repair: an emerging player in many games. Nat Rev Cancer 2015; 15:137-51. [PMID: 25693836 DOI: 10.1038/nrc3892] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The mammalian genome is constantly challenged by exogenous and endogenous threats. Although much is known about the mechanisms that maintain DNA and RNA integrity, we know surprisingly little about the mechanisms that underpin the pathology and tissue specificity of many disorders caused by defective responses to DNA or RNA damage. Of the different types of endogenous damage, protein-linked DNA breaks (PDBs) are emerging as an important player in cancer development and therapy. PDBs can arise during the abortive activity of DNA topoisomerases, a class of enzymes that modulate DNA topology during several chromosomal transactions, such as gene transcription and DNA replication, recombination and repair. In this Review, we discuss the mechanisms underpinning topoisomerase-induced PDB formation and repair with a focus on their role during gene transcription and the development of tissue-specific cancers.
Collapse
Affiliation(s)
- Mohamed E Ashour
- 1] Krebs Institute, Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, S10 2TN, UK. [2] Center for Genomics, Helmy Institute, Zewail City of Science and Technology, Giza 12588, Egypt
| | - Reham Atteya
- Center for Genomics, Helmy Institute, Zewail City of Science and Technology, Giza 12588, Egypt
| | - Sherif F El-Khamisy
- 1] Krebs Institute, Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, S10 2TN, UK. [2] Center for Genomics, Helmy Institute, Zewail City of Science and Technology, Giza 12588, Egypt
| |
Collapse
|
40
|
Pavlidou A, Kroupis C, Dimas K. Association of survivin splice variants with prognosis and treatment of breast cancer. World J Clin Oncol 2014; 5:883-894. [PMID: 25493226 PMCID: PMC4259950 DOI: 10.5306/wjco.v5.i5.883] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 04/01/2014] [Accepted: 10/10/2014] [Indexed: 02/06/2023] Open
Abstract
The purpose of this study was the overview of current knowledge regarding the use of survivin and its isoforms in prognosis and treatment of breast cancer. An advanced search of Medline was performed using the following search strategy: “(survivin isoforms) OR (survivin transcript variants) AND (breast cancer) AND (neoplasm OR tumor OR cancer OR carcinoma)”. Relevant studies were retrieved and processed thoroughly in order to analyze the related data. Besides wild-type survivin full-length transcript, another six splice variants have been identified. Overexpression of survivin and its isoforms leads to shorter overall and disease-free survival; the transcript variants are correlated with apoptosis and could assist prognosis prediction. It has been proved through numerous studies that inhibiting survivin isoforms might become a promising target of drug therapy of carcinomas. Use of small molecule YM155 could offer new therapy for triple negative breast cancer patients, while, chemotherapy with 5-fluorouracil + epirubicin + cyclophosphamide and Tax-Epi could be guided by survivin splice variants measurements. Survivin transcript variants could become prognostic biomarkers and could provide information about clinical management of patients suffering from breast cancer.
Collapse
|
41
|
Heparin and liver heparan sulfate can rescue hepatoma cells from topotecan action. BIOMED RESEARCH INTERNATIONAL 2014; 2014:765794. [PMID: 25309924 PMCID: PMC4170749 DOI: 10.1155/2014/765794] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/23/2014] [Accepted: 08/12/2014] [Indexed: 01/08/2023]
Abstract
Topotecan (TpT) is a major inhibitory compound of topoisomerase (topo) I that plays important roles in gene transcription and cell division. We have previously reported that heparin and heparan sulfate (HS) might be transported to the cell nucleus and they can interact with topoisomerase I. We hypothesized that heparin and HS might interfere with the action of TpT. To test this hypothesis we isolated topoisomerase I containing cell nuclear protein fractions from normal liver, liver cancer tissues, and hepatoma cell lines. The enzymatic activity of these extracts was measured in the presence of heparin, liver HS, and liver cancer HS. In addition, topo I activity, cell viability, and apoptosis of HepG2 and Hep3B cells were investigated after heparin and TpT treatments. Liver cancer HS inhibited topo I activity in vitro. Heparin treatment abrogated topo I enzyme activity in Hep3B cells, but not in HepG2 cells, where the basal activity was higher. Heparin protected the two hepatoma cell lines from TpT actions and decreased the rate of TpT induced S phase block and cell death. These results suggest that heparin and HS might interfere with the function of TpT in liver and liver cancer.
Collapse
|
42
|
Panczyk M. Pharmacogenetics research on chemotherapy resistance in colorectal cancer over the last 20 years. World J Gastroenterol 2014; 20:9775-827. [PMID: 25110414 PMCID: PMC4123365 DOI: 10.3748/wjg.v20.i29.9775] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Revised: 01/17/2014] [Accepted: 04/21/2014] [Indexed: 02/07/2023] Open
Abstract
During the past two decades the first sequencing of the human genome was performed showing its high degree of inter-individual differentiation, as a result of large international research projects (Human Genome Project, the 1000 Genomes Project International HapMap Project, and Programs for Genomic Applications NHLBI-PGA). This period was also a time of intensive development of molecular biology techniques and enormous knowledge growth in the biology of cancer. For clinical use in the treatment of patients with colorectal cancer (CRC), in addition to fluoropyrimidines, another two new cytostatic drugs were allowed: irinotecan and oxaliplatin. Intensive research into new treatment regimens and a new generation of drugs used in targeted therapy has also been conducted. The last 20 years was a time of numerous in vitro and in vivo studies on the molecular basis of drug resistance. One of the most important factors limiting the effectiveness of chemotherapy is the primary and secondary resistance of cancer cells. Understanding the genetic factors and mechanisms that contribute to the lack of or low sensitivity of tumour tissue to cytostatics is a key element in the currently developing trend of personalized medicine. Scientists hope to increase the percentage of positive treatment response in CRC patients due to practical applications of pharmacogenetics/pharmacogenomics. Over the past 20 years the clinical usability of different predictive markers has been tested among which only a few have been confirmed to have high application potential. This review is a synthetic presentation of drug resistance in the context of CRC patient chemotherapy. The multifactorial nature and volume of the issues involved do not allow the author to present a comprehensive study on this subject in one review.
Collapse
|
43
|
Meisenberg C, Ward SE, Schmid P, El-Khamisy SF. TDP1/TOP1 Ratio as a Promising Indicator for the Response of Small Cell Lung Cancer to Topotecan. JOURNAL OF CANCER SCIENCE & THERAPY 2014; 6:258-267. [PMID: 25232464 PMCID: PMC4163653 DOI: 10.4172/1948-5956.1000280] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND AND OBJECTIVE Small cell lung cancer (SCLC) is one of the most challenging tumors to treat due to high proliferation rate, early metastatic dissemination and rapid development of chemotherapy resistance. The current treatment protocols involve the use of topoisomerase 1 (TOP1) poisons such as irinotecan and topotecan in combination with platinum-based compounds. TOP1 poisons kill cancer cells by trapping TOP1 on DNA, generating lethal DNA double-strand breaks. A potential mechanism employed by cancer cells to resist killing by TOP1 poisons is to overexpress enzymes involved in the repair of TOP1-DNA breaks. Tyrosyl DNA phosphodiesterase 1 (TDP1) is a key player in this process and despite its importance, no data is currently available to correlate TDP1 protein and mRNA levels with catalytic activity in SCLC. In addition, it is not known if TDP1 and TOP1 protein levels correlate with the cellular response of SCLC to TOP1 based therapies. METHODS AND RESULTS We report a remarkable variation in TDP1 and TOP1 protein levels in a panel of SCLC cell lines. TDP1 protein level correlates well with TDP1 mRNA and TDP1 catalytic activity, as measured by two newly developed independent activity assays, suggesting the potential utility of immunohistochemistry in assessing TDP1 levels in SCLC tissues. We further demonstrate that whilst TDP1 protein level alone does not correlate with topotecan sensitivity, TDP1/TOP1 ratio correlates well with sensitivity in 8 out of 10 cell lines examined. CONCLUSION This study provides the first cellular analyses of TDP1 and TOP1 in SCLC and suggests the potential utility of TDP1/TOP1 ratio to assess the response of SCLC to topotecan. The establishment and validation of an easy-to-use TDP1 enzymatic assay in cell extracts could be exploited as a diagnostic tool in the clinic. These findings may help in stratifying patients that are likely to benefit from TOP1 poisons and TDP1 inhibitors currently under development.
Collapse
Affiliation(s)
- Cornelia Meisenberg
- The Wellcome Trust DNA Repair Group, Genome Damage and Stability Centre, University of Sussex, Brighton BN1 9RQ, UK
| | - Simon E Ward
- The Sussex Translational Drug Discovery Group, University of Sussex, Brighton, UK
| | - Peter Schmid
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK (former affiliation)
- Centre of Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University London, UK
| | - Sherif F El-Khamisy
- The Wellcome Trust DNA Repair Group, Genome Damage and Stability Centre, University of Sussex, Brighton BN1 9RQ, UK
- Krebs Institute, Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield S10 2TN, UK
- Center of Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| |
Collapse
|
44
|
Hosono Y, Abe T, Higuchi M, Kajii K, Sakuraba S, Tada S, Enomoto T, Seki M. Tipin functions in the protection against topoisomerase I inhibitor. J Biol Chem 2014; 289:11374-11384. [PMID: 24573676 DOI: 10.1074/jbc.m113.531707] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The replication fork temporarily stalls when encountering an obstacle on the DNA, and replication resumes after the barrier is removed. Simultaneously, activation of the replication checkpoint delays the progression of S phase and inhibits late origin firing. Camptothecin (CPT), a topoisomerase I (Top1) inhibitor, acts as a DNA replication barrier by inducing the covalent retention of Top1 on DNA. The Timeless-Tipin complex, a component of the replication fork machinery, plays a role in replication checkpoint activation and stabilization of the replication fork. However, the role of the Timeless-Tipin complex in overcoming the CPT-induced replication block remains elusive. Here, we generated viable TIPIN gene knock-out (KO) DT40 cells showing delayed S phase progression and increased cell death. TIPIN KO cells were hypersensitive to CPT. However, homologous recombination and replication checkpoint were activated normally, whereas DNA synthesis activity was markedly decreased in CPT-treated TIPIN KO cells. Proteasome-dependent degradation of chromatin-bound Top1 was induced in TIPIN KO cells upon CPT treatment, and pretreatment with aphidicolin, a DNA polymerase inhibitor, suppressed both CPT sensitivity and Top1 degradation. Taken together, our data indicate that replication forks formed without Tipin may collide at a high rate with Top1 retained on DNA by CPT treatment, leading to CPT hypersensitivity and Top1 degradation in TIPIN KO cells.
Collapse
Affiliation(s)
- Yoshifumi Hosono
- Molecular Cell Biology Laboratory, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba 6-3, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Takuya Abe
- Instituto FIRC di Oncologia Molecolare (IFOM), Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute for Molecular Oncology Foundation, IFOM-Istituto Europeo di Oncologia Campus, Via Adamello 16, 20139 Milan, Italy
| | - Masato Higuchi
- Molecular Cell Biology Laboratory, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba 6-3, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Kosa Kajii
- Department of Biochemistry, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan
| | - Shuichi Sakuraba
- Molecular Cell Biology Laboratory, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba 6-3, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Shusuke Tada
- Faculty of Pharmaceutical Sciences, Teikyo Heisei University, 4-21-2 Nakano, Nakano-ku, Tokyo 164-8530, Japan, and
| | - Takemi Enomoto
- Molecular Cell Biology Laboratory, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | - Masayuki Seki
- Department of Biochemistry, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan,.
| |
Collapse
|
45
|
Synergism of cyclin-dependent kinase inhibitors with camptothecin derivatives in small cell lung cancer cell lines. Molecules 2014; 19:2077-88. [PMID: 24549232 PMCID: PMC6271949 DOI: 10.3390/molecules19022077] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 01/21/2014] [Accepted: 01/29/2014] [Indexed: 11/17/2022] Open
Abstract
Advanced small cell lung cancer (SCLC) has a dismal prognosis. Modulation of the camptothecin topotecan, approved for second-line therapy, may improve response. Our recent finding of synergistic enhancement of the cytotoxic activity of camptothecin (CPT) by cyclin-dependent kinase 4 inhibitors is extended here to a panel of camptothecin analogs comprising 10-hydroxy-CPT (HOCPT), topotecan (TPT; 9-[(dimethylamino)-methyl]-10-hydroxy-CPT), 9-amino-CPT (9AC), 9-nitrocamptothecin (rubitecan), SN38 (7-ethyl-10-hydroxycamptothecin) and 10-hydroxy-9-nitrocamptothecin (CPT109) in combination with PD0332991, CDK4I, roscovitine and olomoucine. SCLC cell lines employed are chemoresistant NCI-H417 and DMS153 and the chemosensitive SCLC26A line established at our institution. The CPT analogs exhibiting highest cytotoxicity towards the three SCLC lines tested were SN38 and 9AC, followed by rubitecan, HOCPT, TPT and CPT109. NCI-H417 and DMS153 revealed an approximately 25-fold and 7-fold higher resistance compared to the chemosensitive SCLC26A cell line. Whereas the CDK4/6 inhibitor PD0332991 proved less effective to chemosensitize SCLC cells to CPT analogs, the CDK inhibitors CDK4I, roscovitine and olomoucine gave comparable chemosensitization effects in combination with 9AC, SN38, rubitecan and to a lesser extent with TPT and CPT109, not directly related with topoisomerase mRNA expression. In conclusion, small chemical modifications of the parent CPT structure result in differing cytotoxicities and chemomodulatory effects in combination with CDKIs of the resulting analogs.
Collapse
|
46
|
Chemotherapy-mediated p53-dependent DNA damage response in clear cell renal cell carcinoma: role of the mTORC1/2 and hypoxia-inducible factor pathways. Cell Death Dis 2013; 4:e865. [PMID: 24136229 PMCID: PMC3920935 DOI: 10.1038/cddis.2013.395] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 09/01/2013] [Accepted: 09/03/2013] [Indexed: 12/23/2022]
Abstract
The DNA-damaging agent camptothecin (CPT) and its analogs demonstrate clinical utility for the treatment of advanced solid tumors, and CPT-based nanopharmaceuticals are currently in clinical trials for advanced kidney cancer; however, little is known regarding the effects of CPT on hypoxia-inducible factor-2α (HIF-2α) accumulation and activity in clear cell renal cell carcinoma (ccRCC). Here we assessed the effects of CPT on the HIF/p53 pathway. CPT demonstrated striking inhibition of both HIF-1α and HIF-2α accumulation in von Hippel–Lindau (VHL)-defective ccRCC cells, but surprisingly failed to inhibit protein levels of HIF-2α-dependent target genes (VEGF, PAI-1, ET-1, cyclin D1). Instead, CPT induced DNA damage-dependent apoptosis that was augmented in the presence of pVHL. Further analysis revealed CPT regulated endothelin-1 (ET-1) in a p53-dependent manner: CPT increased ET-1 mRNA abundance in VHL-defective ccRCC cell lines that was significantly augmented in their VHL-expressing counterparts that displayed increased phosphorylation and accumulation of p53; p53 siRNA suppressed CPT-induced increase in ET-1 mRNA, as did an inhibitor of ataxia telangiectasia mutated (ATM) signaling, suggesting a role for ATM-dependent phosphorylation of p53 in the induction of ET-1. Finally, we demonstrate that p53 phosphorylation and accumulation is partially dependent on mTOR activity in ccRCC. Consistent with this result, pharmacological inhibition of mTORC1/2 kinase inhibited CPT-mediated ET-1 upregulation, and p53-dependent responses in ccRCC. Collectively, these data provide mechanistic insight into the action of CPT in ccRCC, identify ET-1 as a p53-regulated gene and demonstrate a requirement of mTOR for p53-mediated responses in this tumor type.
Collapse
|
47
|
Myeloprotection by cytidine deaminase gene transfer in antileukemic therapy. Neoplasia 2013; 15:239-48. [PMID: 23479503 DOI: 10.1593/neo.121954] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 01/03/2013] [Accepted: 01/07/2013] [Indexed: 12/22/2022] Open
Abstract
Gene transfer of drug resistance (CTX-R) genes can be used to protect the hematopoietic system from the toxicity of anticancer chemotherapy and this concept recently has been proven by overexpression of a mutant O(6)-methylguaninemethyltransferase in the hematopoietic system of glioblastoma patients treated with temozolomide. Given its protection capacity against such relevant drugs as cytosine arabinoside (ara-C), gemcitabine, decitabine, or azacytidine and the highly hematopoiesis-specific toxicity profile of several of these agents, cytidine deaminase (CDD) represents another interesting candidate CTX-R gene and our group recently has established the myeloprotective capacity of CDD gene transfer in a number of murine transplant studies. Clinically, CDD overexpression appears particularly suited to optimize treatment strategies for acute leukemias and myelodysplasias given the efficacy of ara-C (and to a lesser degree decitabine and azacytidine) in these disease entities. This article will review the current state of the art with regard to CDD gene transfer and point out potential scenarios for a clinical application of this strategy. In addition, risks and potential side effects associated with this approach as well as strategies to overcome these problems will be highlighted.
Collapse
|
48
|
Afriat R, Horowitz S, Priel E. Mycoplasma fermentans inhibits the activity of cellular DNA topoisomerase I by activation of PARP1 and alters the efficacy of its anti-cancer inhibitor. PLoS One 2013; 8:e72377. [PMID: 24013388 PMCID: PMC3754970 DOI: 10.1371/journal.pone.0072377] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 07/09/2013] [Indexed: 01/02/2023] Open
Abstract
To understand the effects of the interaction between Mycoplasma and cells on the host cellular function, it is important to elucidate the influences of infection of cells with Mycoplasma on nuclear enzymes such as DNA Topoisomerase type I (Topo I). Human Topo I participates in DNA transaction processes and is the target of anti-cancer drugs, the camptothecins (CPTs). Here we investigated the mechanism by which infection of human tumor cells with Mycoplasma fermentans affects the activity and expression of cellular Topo I, and the anti-cancer efficacy of CPT. Human cancer cells were infected or treated with live or sonicated M. fermentans and the activity and expression of Topo I was determined. M. fermentans significantly reduced (by 80%) Topo I activity in the infected/treated tumor cells without affecting the level of Topo I protein. We demonstrate that this reduction in enzyme activity resulted from ADP-ribosylation of the Topo I protein by Poly-ADP-ribose polymerase (PARP-1). In addition, pERK was activated as a result of the induction of the MAPK signal transduction pathway by M. fermentans. Since PARP-1 was shown to be activated by pERK, we concluded that M. fermentans modified the cellular Topo I activity by activation of PARP-I via the induction of the MAPK signal transduction pathway. Moreover, the infection of tumor cells with M. fermentans diminished the inhibitory effect of CPT. The results of this study suggest that modification of Topo I activity by M. fermentans may alter cellular gene expression and the response of tumor cells to Topo I inhibitors, influencing the anti-cancer capacity of Topo I antagonists.
Collapse
Affiliation(s)
- Reuven Afriat
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, and the Soroka University Medical Center, Ben-Gurion University, Beer-Sheva, Israel
| | - Shulamith Horowitz
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, and the Soroka University Medical Center, Ben-Gurion University, Beer-Sheva, Israel
| | - Esther Priel
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, and the Soroka University Medical Center, Ben-Gurion University, Beer-Sheva, Israel
| |
Collapse
|
49
|
Substituted benzo[i]phenanthridines as promising topoisomerase-I non-camptothecin targeting agents: an experimental and theoretical study. Med Chem Res 2013. [DOI: 10.1007/s00044-013-0515-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|