1
|
Elmitwalli O, Darwish R, Al-Jabery L, Algahiny A, Roy S, Butler AE, Hasan AS. The Emerging Role of p21 in Diabetes and Related Metabolic Disorders. Int J Mol Sci 2024; 25:13209. [PMID: 39684919 DOI: 10.3390/ijms252313209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
In the context of cell cycle inhibition, anti-proliferation, and the dysregulation observed in certain cancer pathologies, the protein p21 assumes a pivotal role. p21 links DNA damage responses to cellular processes such as apoptosis, senescence, and cell cycle arrest, primarily functioning as a regulator of the cell cycle. However, accumulating empirical evidence suggests that p21 is both directly and indirectly linked to a number of different metabolic processes. Intriguingly, recent investigations indicate that p21 significantly contributes to the pathogenesis of diabetes. In this review, we present a comprehensive evaluation of the scientific literature regarding the involvement of p21 in metabolic processes, diabetes etiology, pancreatic function, glucose homeostasis, and insulin resistance. Furthermore, we provide an encapsulated overview of therapies that target p21 to alleviate metabolic disorders. A deeper understanding of the complex interrelationship between p21 and diabetes holds promise for informing current and future therapeutic strategies to address this rapidly escalating health crisis.
Collapse
Affiliation(s)
- Omar Elmitwalli
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Radwan Darwish
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Lana Al-Jabery
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Ahmed Algahiny
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Sornali Roy
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Alexandra E Butler
- Department of Postgraduate Studies and Research, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Ammar S Hasan
- Department of Postgraduate Studies and Research, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| |
Collapse
|
2
|
Krushkal J, Zhao Y, Roney K, Zhu W, Brooks A, Wilsker D, Parchment RE, McShane LM, Doroshow JH. Association of changes in expression of HDAC and SIRT genes after drug treatment with cancer cell line sensitivity to kinase inhibitors. Epigenetics 2024; 19:2309824. [PMID: 38369747 PMCID: PMC10878021 DOI: 10.1080/15592294.2024.2309824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 01/14/2024] [Indexed: 02/20/2024] Open
Abstract
Histone deacetylases (HDACs) and sirtuins (SIRTs) are important epigenetic regulators of cancer pathways. There is a limited understanding of how transcriptional regulation of their genes is affected by chemotherapeutic agents, and how such transcriptional changes affect tumour sensitivity to drug treatment. We investigated the concerted transcriptional response of HDAC and SIRT genes to 15 approved antitumor agents in the NCI-60 cancer cell line panel. Antitumor agents with diverse mechanisms of action induced upregulation or downregulation of multiple HDAC and SIRT genes. HDAC5 was upregulated by dasatinib and erlotinib in the majority of the cell lines. Tumour cell line sensitivity to kinase inhibitors was associated with upregulation of HDAC5, HDAC1, and several SIRT genes. We confirmed changes in HDAC and SIRT expression in independent datasets. We also experimentally validated the upregulation of HDAC5 mRNA and protein expression by dasatinib in the highly sensitive IGROV1 cell line. HDAC5 was not upregulated in the UACC-257 cell line resistant to dasatinib. The effects of cancer drug treatment on expression of HDAC and SIRT genes may influence chemosensitivity and may need to be considered during chemotherapy.
Collapse
Affiliation(s)
- Julia Krushkal
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - Yingdong Zhao
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - Kyle Roney
- Department of Biostatistics and Bioinformatics, George Washington University, Washington, DC, USA
| | - Weimin Zhu
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Alan Brooks
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Deborah Wilsker
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Ralph E. Parchment
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Lisa M. McShane
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - James H. Doroshow
- Division of Cancer Treatment and Diagnosis and Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
3
|
Alhalabi OT, Göttmann M, Gold MP, Schlue S, Hielscher T, Iskar M, Kessler T, Hai L, Lokumcu T, Cousins CC, Herold-Mende C, Heßling B, Horschitz S, Jabali A, Koch P, Baumgartner U, Day BW, Wick W, Sahm F, Krieg SM, Fraenkel E, Phillips E, Goidts V. Integration of transcriptomics, proteomics and loss-of-function screening reveals WEE1 as a target for combination with dasatinib against proneural glioblastoma. Cancer Lett 2024; 605:217265. [PMID: 39332586 DOI: 10.1016/j.canlet.2024.217265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
Glioblastoma is characterized by a pronounced resistance to therapy with dismal prognosis. Transcriptomics classify glioblastoma into proneural (PN), mesenchymal (MES) and classical (CL) subtypes that show differential resistance to targeted therapies. The aim of this study was to provide a viable approach for identifying combination therapies in glioblastoma subtypes. Proteomics and phosphoproteomics were performed on dasatinib inhibited glioblastoma stem cells (GSCs) and complemented by an shRNA loss-of-function screen to identify genes whose knockdown sensitizes GSCs to dasatinib. Proteomics and screen data were computationally integrated with transcriptomic data using the SamNet 2.0 algorithm for network flow learning to reveal potential combination therapies in PN GSCs. In vitro viability assays and tumor spheroid models were used to verify the synergy of identified therapy. Further in vitro and TCGA RNA-Seq data analyses were utilized to provide a mechanistic explanation of these effects. Integration of data revealed the cell cycle protein WEE1 as a potential combination therapy target for PN GSCs. Validation experiments showed a robust synergistic effect through combination of dasatinib and the WEE1 inhibitor, MK-1775, in PN GSCs. Combined inhibition using dasatinib and MK-1775 propagated DNA damage in PN GCSs, with GCSs showing a differential subtype-driven pattern of expression of cell cycle genes in TCGA RNA-Seq data. The integration of proteomics, loss-of-function screens and transcriptomics confirmed WEE1 as a target for combination with dasatinib against PN GSCs. Utilizing this integrative approach could be of interest for studying resistance mechanisms and revealing combination therapy targets in further tumor entities.
Collapse
Affiliation(s)
- Obada T Alhalabi
- Brain Tumor Translational Targets, DKFZ Junior Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurosurgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Mona Göttmann
- Brain Tumor Translational Targets, DKFZ Junior Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Maxwell P Gold
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Silja Schlue
- Brain Tumor Translational Targets, DKFZ Junior Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Hielscher
- Division of Biostatistics (C060), German Cancer Research Center, Germany
| | - Murat Iskar
- Division of Molecular Genetics, Heidelberg Center for Personalized Oncology, German Cancer Research Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tobias Kessler
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Ling Hai
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Tolga Lokumcu
- Brain Tumor Translational Targets, DKFZ Junior Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Clara C Cousins
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Bernd Heßling
- Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sandra Horschitz
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty Mannheim, Mannheim, Germany; Hector Institute for Translational Brain Research (HITBR gGmbH), Mannheim, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ammar Jabali
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty Mannheim, Mannheim, Germany; Hector Institute for Translational Brain Research (HITBR gGmbH), Mannheim, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Philipp Koch
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty Mannheim, Mannheim, Germany; Hector Institute for Translational Brain Research (HITBR gGmbH), Mannheim, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrich Baumgartner
- Cell and Molecular Biology Department, QIMR Berghofer Medical Research Institute, Sid Faithfull Brain Cancer Laboratory, Brisbane, QLD, 4006, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
| | - Bryan W Day
- Cell and Molecular Biology Department, QIMR Berghofer Medical Research Institute, Sid Faithfull Brain Cancer Laboratory, Brisbane, QLD, 4006, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
| | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Sandro M Krieg
- Department of Neurosurgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Emma Phillips
- Brain Tumor Translational Targets, DKFZ Junior Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Violaine Goidts
- Brain Tumor Translational Targets, DKFZ Junior Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
4
|
Kološa K, Žegura B, Štampar M, Filipič M, Novak M. Adverse Toxic Effects of Tyrosine Kinase Inhibitors on Non-Target Zebrafish Liver (ZFL) Cells. Int J Mol Sci 2023; 24:ijms24043894. [PMID: 36835302 PMCID: PMC9965539 DOI: 10.3390/ijms24043894] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023] Open
Abstract
Over the past 20 years, numerous tyrosine kinase inhibitors (TKIs) have been introduced for targeted therapy of various types of malignancies. Due to frequent and increasing use, leading to eventual excretion with body fluids, their residues have been found in hospital and household wastewaters as well as surface water. However, the effects of TKI residues in the environment on aquatic organisms are poorly described. In the present study, we investigated the cytotoxic and genotoxic effects of five selected TKIs, namely erlotinib (ERL), dasatinib (DAS), nilotinib (NIL), regorafenib (REG), and sorafenib (SOR), using the in vitro zebrafish liver cell (ZFL) model. Cytotoxicity was determined using the MTS assay and propidium iodide (PI) live/dead staining by flow cytometry. DAS, SOR, and REG decreased ZFL cell viability dose- and time-dependently, with DAS being the most cytotoxic TKI studied. ERL and NIL did not affect viability at concentrations up to their maximum solubility; however, NIL was the only TKI that significantly decreased the proportion of PI negative cells as determined by the flow cytometry. Cell cycle progression analyses showed that DAS, ERL, REG, and SOR caused the cell cycle arrest of ZFL cells in the G0/G1 phase, with a concomitant decrease of cells in the S-phase fraction. No data could be obtained for NIL due to severe DNA fragmentation. The genotoxic activity of the investigated TKIs was evaluated using comet and cytokinesis block micronucleus (CBMN) assays. The dose-dependent induction of DNA single strand breaks was induced by NIL (≥2 μM), DAS (≥0.006 μM), and REG (≥0.8 μM), with DAS being the most potent. None of the TKIs studied induced micronuclei formation. These results suggest that normal non-target fish liver cells are sensitive to the TKIs studied in a concentration range similar to those previously reported for human cancer cell lines. Although the TKI concentrations that induced adverse effects in exposed ZFL cells are several orders of magnitude higher than those currently expected in the aquatic environment, the observed DNA damage and cell cycle effects suggest that residues of TKIs in the environment may pose a hazard to non-intentionally exposed organisms living in environments contaminated with TKIs.
Collapse
Affiliation(s)
- Katja Kološa
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia
| | - Bojana Žegura
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia
- Jozef Stefan International Postgraduate School, 1000 Ljubljana, Slovenia
- Correspondence:
| | - Martina Štampar
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia
| | - Metka Filipič
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia
- Jozef Stefan International Postgraduate School, 1000 Ljubljana, Slovenia
| | - Matjaž Novak
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia
| |
Collapse
|
5
|
Senescent cells and SASP in cancer microenvironment: New approaches in cancer therapy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:115-158. [PMID: 36707199 DOI: 10.1016/bs.apcsb.2022.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cellular senescence was first described as a state characterized by telomere shortening, resulting in limiting cell proliferation in aging. Apart from this type of senescence, which is called replicative senescence, other senescence types occur after exposure to different stress factors. One of these types of senescence induced after adjuvant therapy (chemotherapy and radiotherapy) is called therapy-induced senescence. The treatment with chemotherapeutics induces cellular senescence in normal and cancer cells in the tumor microenvironment. Thus therapy-induced senescence in the cancer microenvironment is accepted one of the drivers of tumor progression. Recent studies have revealed that senescence-associated secretory phenotype induction has roles in pathological processes such as inducing epithelial-mesenchymal transition and promoting tumor vascularization. Thus senolytic drugs that specifically kill senescent cells and senomorphic drugs that inhibit the secretory activity of senescent cells are seen as a new approach in cancer treatment. Developing and discovering new senotherapeutic agents targeting senescent cells is also gaining importance. In this review, we attempt to summarize the signaling pathways regarding the metabolism, cell morphology, and organelles of the senescent cell. Furthermore, we also reviewed the effects of SASP in the cancer microenvironment and the senotherapeutics that have the potential to be used as adjuvant therapy in cancer treatment.
Collapse
|
6
|
Jin N, Xia Y, Gao Q. Combined PARP inhibitors and small molecular inhibitors in solid tumor treatment (Review). Int J Oncol 2023; 62:28. [PMID: 36601757 PMCID: PMC9851129 DOI: 10.3892/ijo.2023.5476] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/23/2022] [Indexed: 01/05/2023] Open
Abstract
With the development of precision medicine, targeted therapy has attracted extensive attention. Poly(ADP‑ribose) polymerase inhibitors (PARPi) are critical clinical drugs designed to induce cell death and are major antitumor targeted agents. However, preclinical and clinical data have revealed the limitations of PARPi monotherapy. Therefore, their combination with other targeted drugs has become a research hotspot in tumor treatment. Recent studies have demonstrated the critical role of small molecular inhibitors in multiple haematological cancers and solid tumors via cellular signalling modulation, exhibiting potential as a combined pharmacotherapy. In the present review, studies focused on small molecular inhibitors targeting the homologous recombination pathway were summarized and clinical trials evaluating the safety and efficacy of combined treatment were discussed.
Collapse
Affiliation(s)
- Ning Jin
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Yu Xia
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Qinglei Gao
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| |
Collapse
|
7
|
Mourkioti I, Angelopoulou A, Belogiannis K, Lagopati N, Potamianos S, Kyrodimos E, Gorgoulis V, Papaspyropoulos A. Interplay of Developmental Hippo-Notch Signaling Pathways with the DNA Damage Response in Prostate Cancer. Cells 2022; 11:cells11152449. [PMID: 35954292 PMCID: PMC9367915 DOI: 10.3390/cells11152449] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022] Open
Abstract
Prostate cancer belongs in the class of hormone-dependent cancers, representing a major cause of cancer incidence in men worldwide. Since upon disease onset almost all prostate cancers are androgen-dependent and require active androgen receptor (AR) signaling for their survival, the primary treatment approach has for decades relied on inhibition of the AR pathway via androgen deprivation therapy (ADT). However, following this line of treatment, cancer cell pools often become resistant to therapy, contributing to disease progression towards the significantly more aggressive castration-resistant prostate cancer (CRPC) form, characterized by poor prognosis. It is, therefore, of critical importance to elucidate the molecular mechanisms and signaling pathways underlying the progression of early-stage prostate cancer towards CRPC. In this review, we aim to shed light on the role of major signaling pathways including the DNA damage response (DDR) and the developmental Hippo and Notch pathways in prostate tumorigenesis. We recapitulate key evidence demonstrating the crosstalk of those pathways as well as with pivotal prostate cancer-related 'hubs' such as AR signaling, and evaluate the clinical impact of those interactions. Moreover, we attempt to identify molecules of the complex DDR-Hippo-Notch interplay comprising potentially novel therapeutic targets in the battle against prostate tumorigenesis.
Collapse
Affiliation(s)
- Ioanna Mourkioti
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
| | - Andriani Angelopoulou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
| | - Konstantinos Belogiannis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
| | - Nefeli Lagopati
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
| | - Spyridon Potamianos
- First ENT Department, Hippocration Hospital, University of Athens, 11527 Athens, Greece
| | - Efthymios Kyrodimos
- First ENT Department, Hippocration Hospital, University of Athens, 11527 Athens, Greece
| | - Vassilis Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
- Clinical Molecular Pathology, Medical School, University of Dundee, Dundee DD1 9SY, UK
- Molecular and Clinical Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4GJ, UK
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Faculty of Health and Medical Sciences, University of Surrey, Surrey GU2 7YH, UK
- Correspondence: (V.G.); (A.P.); Tel.: +30-210-7462352 (V.G.); +30-210-7462174 (A.P.)
| | - Angelos Papaspyropoulos
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
- Correspondence: (V.G.); (A.P.); Tel.: +30-210-7462352 (V.G.); +30-210-7462174 (A.P.)
| |
Collapse
|
8
|
Targeting the FAK-Src Complex in Desmoplastic Small Round Cell Tumors, Ewing Sarcoma, and Rhabdomyosarcoma. Sarcoma 2022; 2022:3089424. [PMID: 35655525 PMCID: PMC9153931 DOI: 10.1155/2022/3089424] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/31/2022] [Indexed: 11/21/2022] Open
Abstract
Desmoplastic small round cell tumors (DSRCTs), Ewing sarcoma (ES), and alveolar and embryonal rhabdomyosarcoma (ARMS and ERMS) are malignant sarcomas typically occurring at young age, with a poor prognosis in the metastatic setting. New treatment options are necessary. Src family kinase inhibitor dasatinib single-agent treatment has been investigated in a phase 2 study in patients with advanced sarcomas including ES and RMS but failed as a single agent in these subtypes. Since previous studies demonstrated high FAK and Src activities in RMS and ES tissue and cell lines, and dasatinib treatment was shown to upregulate activated FAK, we hypothesized that FAK-Src combination treatment could potentially be an interesting treatment option for these tumor types. We examined the effects of targeting the FAK-Src complex by addressing (p)FAK and (p)Src expressions in tumor sections of DSRCT (n = 13), ES (n = 68), ARMS (n = 21), and ERMS (n = 39) and by determining the antitumor effects of single and combined treatment with FAK inhibitor defactinib and multikinase (Abl/SFK) inhibitor dasatinib in vitro on cell lines of each subtype. In vivo effects were assessed in DSRCT and ERMS models. Concurrent pFAK and pSrc expressions (H-score >50) were observed in DSRCT (67%), ES (6%), ARMS (35%), and ERMS (19%) samples. Defactinib treatment decreased pFAK expression and reduced cell viability in all subtypes. Dasatinib treatment decreased pSrc expression and cell viability in each subtype. Combination treatment led to a complete reduction in pFAK and pSrc in each cell line and showed enhanced cell viability reduction, drug synergy, DNA damage induction, and a trend toward higher apoptosis induction in DSRCT, ERMS, and ARMS but not in ES cells. These promising in vitro results unfortunately do not translate into promising in vivo results as we did not observe a significant effect on tumor volume in vivo, and the combination did not show superior effects compared to dasatinib single-agent treatment.
Collapse
|
9
|
Zhang J, Li C, Zhang L, Heng Y, Wang S, Pan Y, Cai L, Zhang Y, Xu T, Chen X, Hoffman RM, Jia L. Andrographolide, a diterpene lactone from the Traditional Chinese Medicine Andrographis paniculate, induces senescence in human lung adenocarcinoma via p53/p21 and Skp2/p27. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 98:153933. [PMID: 35121394 DOI: 10.1016/j.phymed.2022.153933] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Senescence leads to permanent cell-cycle arrest and is a potential target for cancer therapy. Andrographolide (AD) is a diterpene lactone isolated from Traditional Chinese Medicine (TCM) Andrographis paniculate, which has been used as an anti-inflammatory drug in clinical practice with the potential to target senescence in recalcitrant lung cancer. PURPOSE To determine whether AD can induce senescence in human lung adenocarcinoma in vitro and in vivo and to elucidate the underlying mechanisms. METHODS SA-β-Gal staining was used to detect the expression of senescence-associated β-galactosidase (SA-β-Gal) in human lung adenocarcinoma cells A549 and NCI-H1795. DNA damage was examined by the detection of γH2AX foci. Cell cycle was analyzed by flow cytometry. Cancer cell proliferation was determined by ATPlite assay and clonogenic survival assay in vitro. Tumor growth was determined in a mouse model of A549. The expression level of proteins and mRNA was estimated by Western blotting and Quantitative RT-PCR, respectively. Small interfering RNA (siRNA) was used to knock down p21, p27 and p53 to explore the potential mechanism of AD-induced senescence in human lung adenocarcinoma cells. RESULTS AD-induced A549 and NCI-H1795 cell senescence determined by increased cell size, flattened morphology, DNA damage, cell cycle arrest as well as the increased expression of β-galactosidase. AD inhibited cell proliferation in lung cells in vitro and lung cells xenograft growth in nude mice. p21 and p27, the major cell cycle regulators and mediators of senescence, were upregulated at the protein level in AD-treated A549 lung adenocarcinoma in vitro and in vivo. Further studies demonstrated that AD induced cell senescence via p53/p21 and Skp2/p27. CONCLUSION In the present study, we found that the primary anti-inflammatory drug AD could have a potential antitumor effect in lung cancer. We demonstrated that AD induced lung adenocarcinoma senescence in vitro and in vivo via p53/p21 and Skp2/p27 for the first time. AD is therefore a promising senescence-inducing therapeutic for recalcitrant human lung adenocarcinoma.
Collapse
Affiliation(s)
- Junqian Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Chunjie Li
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Li Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yongqing Heng
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shiwen Wang
- Department of Laboratory Medicine, Huadong Hospital, Affiliated to Fudan University, Shanghai, China
| | - Yongfu Pan
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Lili Cai
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yunjing Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Tong Xu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Xihui Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Robert M Hoffman
- Department of Surgery, University of California, San Diego, CA, USA; Anticancer, Inc., San Diego, CA, USA
| | - Lijun Jia
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
10
|
Fakhri S, Zachariah Moradi S, DeLiberto LK, Bishayee A. Cellular senescence signaling in cancer: A novel therapeutic target to combat human malignancies. Biochem Pharmacol 2022; 199:114989. [DOI: 10.1016/j.bcp.2022.114989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 12/26/2022]
|
11
|
Subramaniyan V, Fuloria S, Gupta G, Kumar DH, Sekar M, Sathasivam KV, Sudhakar K, Alharbi KS, Al-Malki WH, Afzal O, Kazmi I, Al-Abbasi FA, Altamimi ASA, Fuloria NK. A review on epidermal growth factor receptor's role in breast and non-small cell lung cancer. Chem Biol Interact 2022; 351:109735. [PMID: 34742684 DOI: 10.1016/j.cbi.2021.109735] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/28/2021] [Accepted: 11/01/2021] [Indexed: 12/11/2022]
Abstract
Epithelial growth factor receptor (EGFR) is a cell surface transmembrane receptor that mediates the tyrosine signaling pathway to carry the extracellular messages inside the cell and thereby alter the function of nucleus. This leads to the generation of various protein products to up or downregulate the cellular function. It is encoded by cell erythroblastosis virus oncogene B1, so called C-erb B1/ERBB2/HER-2 gene that acts as a proto-oncogene. It belongs to the HER-2 receptor-family in breast cancer and responds best with anti-Herceptin therapy (anti-tyrosine kinase monoclonal antibody). HER-2 positive breast cancer patient exhibits worse prognosis without Herceptin therapy. Similar incidence and prognosis are reported in other epithelial neoplasms like EGFR + lung non-small cell carcinoma and glioblastoma (grade IV brain glial tumor). Present study highlights the role and connectivity of EGF with various cancers via signaling pathways, cell surface receptors mechanism, macromolecules, mitochondrial genes and neoplasm. Present study describes the EGFR associated gene expression profiling (in breast cancer and NSCLC), relation between mitrochondrial genes and carcinoma, and several in vitro and in vivo models to screen the synergistic effect of various combination treatments. According to this study, although clinical studies including targeted treatments, immunotherapies, radiotherapy, TKi-EGFR combined targeted therapy have been carried out to investigate the synergism of combination therapy; however still there is a gap to apply the scenarios of experimental and clinical studies for further developments. This review will give an idea about the transition from experimental to most advanced clinical studies with different combination drug strategies to treat cancer.
Collapse
Affiliation(s)
- Vetriselvan Subramaniyan
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jalan SP 2, Bandar Saujana Putra, 42610, Jenjarom, Selangor, Malaysia
| | - Shivkanya Fuloria
- Faculty of Pharmacy & Centre of Excellence for Biomaterials Engineering, AIMST University, Bedong 08100, Kedah, Malaysia
| | - Gaurav Gupta
- Department of Pharmacology, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India; Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical Sciences, Saveetha University, Chennai, India
| | - Darnal Hari Kumar
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selngor, 47500, Malaysia
| | - Mahendran Sekar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur Royal College of Medicine Perak, Ipoh, 30450, Malaysia
| | - Kathiresan V Sathasivam
- Faculty of Applied Science & Centre of Excellence for Biomaterials Engineering, AIMST University, Bedong 08100, Kedah, Malaysia
| | - Kalvatala Sudhakar
- School of Pharmaceutical Sciences (LIT-Pharmacy), Lovely Professional University, Jalandhar, 144411, India
| | - Khalid Saad Alharbi
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Waleed Hassan Al-Malki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam BinAbdulaziz University, AlKharj, 11942, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | | | - Neeraj Kumar Fuloria
- Faculty of Pharmacy & Centre of Excellence for Biomaterials Engineering, AIMST University, Bedong 08100, Kedah, Malaysia.
| |
Collapse
|
12
|
Mechanisms of Hydroxyurea-Induced Cellular Senescence: An Oxidative Stress Connection? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7753857. [PMID: 34707779 PMCID: PMC8545575 DOI: 10.1155/2021/7753857] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/09/2021] [Accepted: 09/25/2021] [Indexed: 01/10/2023]
Abstract
Hydroxyurea (HU) is a water-soluble antiproliferative agent used for decades in neoplastic and nonneoplastic conditions. HU is considered an essential medicine because of its cytoreduction functions. HU is an antimetabolite that inhibits ribonucleotide reductase, which causes a depletion of the deoxyribonucleotide pool and dramatically reduces cell proliferation. The proliferation arrest, depending on drug concentration and exposure, may promote a cellular senescence phenotype associated with cancer cell therapy resistance and inflammation, influencing neighboring cell functions, immunosuppression, and potential cancer relapse. HU can induce cellular senescence in both healthy and transformed cells in vitro, in part, because of increased reactive oxygen species (ROS). Here, we analyze the main molecular mechanisms involved in cytotoxic/genotoxic HU function, the potential to increase intracellular ROS levels, and the principal features of cellular senescence induction. Understanding the mechanisms involved in HU's ability to induce cellular senescence may help to improve current chemotherapy strategies and control undesirable treatment effects in cancer patients and other diseases.
Collapse
|
13
|
Yun HS, Lee J, Kil WJ, Kramp TR, Tofilon PJ, Camphausen K. The Radiosensitizing Effect of AZD0530 in Glioblastoma and Glioblastoma Stem-Like Cells. Mol Cancer Ther 2021; 20:1672-1679. [PMID: 34158343 PMCID: PMC8419151 DOI: 10.1158/1535-7163.mct-20-0883] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/23/2021] [Accepted: 06/11/2021] [Indexed: 11/16/2022]
Abstract
AZD0530, a potent small-molecule inhibitor of the Src kinase family, is an anticancer drug used in the treatment of various cancers. In the case of glioblastoma (GBM), where resistance to radiotherapy frequently occurs, Src kinase is known as one of the molecules responsible for imparting radioresistance to GBM. Thus, we evaluated the effect of AZD0530 on the radiosensitivity of human GBM cells and human glioblastoma stem-like cells (GSCs). We show that Src activity of GBM and GSC is increased by radiation and inhibited by AZD0530, and using clonogenic assays, AZD0530 enhances the radiosensitivity of GBM and GSCs. Also, AZD0530 induced a prolongation of radiation-induced γH2AX without specific cell cycle and mitotic index changes, suggesting that AZD0530-induced radiosensitization in GBM cells and GSCs results from the inhibition of DNA repair. In addition, AZD0530 was shown to inhibit the radiation-induced EGFR/PI3K/AKT pathway, which is known to promote and regulate radioresistance and survival of GBM cells by radiation. Finally, mice bearing orthotopic xenografts initiated from GBM cells were then used to evaluate the in vivo response to AZD0530 and radiation. The combination of AZD0530 and radiation showed the longest median survival compared with any single modality. Thus, these results show that AZD0530 enhances the radiosensitivity of GBM cells and GSCs and suggest the possibility of AZD0530 as a clinical radiosensitizer for treatment of GBM.
Collapse
Affiliation(s)
- Hong Shik Yun
- Radiation Oncology Branch, NCI, NIH, Bethesda, Maryland
| | - Jennifer Lee
- Radiation Oncology Branch, NCI, NIH, Bethesda, Maryland
| | | | | | | | | |
Collapse
|
14
|
Patel NH, Bloukh S, Alwohosh E, Alhesa A, Saleh T, Gewirtz DA. Autophagy and senescence in cancer therapy. Adv Cancer Res 2021; 150:1-74. [PMID: 33858594 DOI: 10.1016/bs.acr.2021.01.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Tumor cells can undergo diverse responses to cancer therapy. While apoptosis represents the most desirable outcome, tumor cells can alternatively undergo autophagy and senescence. Both autophagy and senescence have the potential to make complex contributions to tumor cell survival via both cell autonomous and cell non-autonomous pathways. The induction of autophagy and senescence in tumor cells, preclinically and clinically, either individually or concomitantly, has generated interest in the utilization of autophagy modulating and senolytic therapies to target autophagy and senescence, respectively. This chapter summarizes the current evidence for the promotion of autophagy and senescence as fundamental responses to cancer therapy and discusses the complexity of their functional contributions to cell survival and disease outcomes. We also highlight current modalities designed to exploit autophagy and senescence in efforts to improve the efficacy of cancer therapy.
Collapse
Affiliation(s)
- Nipa H Patel
- Department of Pharmacology and Toxicology and Medicine, Virginia Commonwealth University, Richmond, VA, United States; Massey Cancer Center, Goodwin Research Laboratories, Virginia Commonwealth University, Richmond, VA, United States
| | - Sarah Bloukh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Enas Alwohosh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Ahmad Alhesa
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - David A Gewirtz
- Department of Pharmacology and Toxicology and Medicine, Virginia Commonwealth University, Richmond, VA, United States; Massey Cancer Center, Goodwin Research Laboratories, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
15
|
Transcriptomic Changes Associated with Loss of Cell Viability Induced by Oxysterol Treatment of a Retinal Photoreceptor-Derived Cell Line: An In Vitro Model of Smith-Lemli-Opitz Syndrome. Int J Mol Sci 2021; 22:ijms22052339. [PMID: 33652836 PMCID: PMC7956713 DOI: 10.3390/ijms22052339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 11/17/2022] Open
Abstract
Smith–Lemli–Opitz Syndrome (SLOS) results from mutations in the gene encoding the enzyme DHCR7, which catalyzes conversion of 7-dehydrocholesterol (7DHC) to cholesterol (CHOL). Rats treated with a DHCR7 inhibitor serve as a SLOS animal model, and exhibit progressive photoreceptor-specific cell death, with accumulation of 7DHC and oxidized sterols. To understand the basis of this cell type specificity, we performed transcriptomic analyses on a photoreceptor-derived cell line (661W), treating cells with two 7DHC-derived oxysterols, which accumulate in tissues and bodily fluids of SLOS patients and in the rat SLOS model, as well as with CHOL (negative control), and evaluated differentially expressed genes (DEGs) for each treatment. Gene enrichment analysis and compilation of DEG sets indicated that endoplasmic reticulum stress, oxidative stress, DNA damage and repair, and autophagy were all highly up-regulated pathways in oxysterol-treated cells. Detailed analysis indicated that the two oxysterols exert their effects via different molecular mechanisms. Changes in expression of key genes in highlighted pathways (Hmox1, Ddit3, Trib3, and Herpud1) were validated by immunofluorescence confocal microscopy. The results extend our understanding of the pathobiology of retinal degeneration and SLOS, identifying potential new druggable targets for therapeutic intervention into these and other related orphan diseases.
Collapse
|
16
|
Mongiardi MP, Pellegrini M, Pallini R, Levi A, Falchetti ML. Cancer Response to Therapy-Induced Senescence: A Matter of Dose and Timing. Cancers (Basel) 2021; 13:484. [PMID: 33513872 PMCID: PMC7865402 DOI: 10.3390/cancers13030484] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/18/2021] [Accepted: 01/23/2021] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence participates to fundamental processes like tissue remodeling in embryo development, wound healing and inhibition of preneoplastic cell growth. Most senescent cells display common hallmarks, among which the most characteristic is a permanent (or long lasting) arrest of cell division. However, upon senescence, different cell types acquire distinct phenotypes, which also depend on the specific inducing stimuli. Senescent cells are metabolically active and secrete a collection of growth factors, cytokines, proteases, and matrix-remodeling proteins collectively defined as senescence-associated secretory phenotype, SASP. Through SASP, senescent cells modify their microenvironment and engage in a dynamic dialog with neighbor cells. Senescence of neoplastic cells, at least temporarily, reduces tumor expansion, but SASP of senescent cancer cells as well as SASP of senescent stromal cells in the tumor microenvironment may promote the growth of more aggressive cancer subclones. Here, we will review recent data on the mechanisms and the consequences of cancer-therapy induced senescence, enlightening the potentiality and the risk of senescence inducing treatments.
Collapse
Affiliation(s)
- Maria Patrizia Mongiardi
- CNR-Institute of Biochemistry and Cell Biology, Campus Adriano Buzzati Traverso, Via Ercole Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy; (M.P.M.); (M.P.); (A.L.)
| | - Manuela Pellegrini
- CNR-Institute of Biochemistry and Cell Biology, Campus Adriano Buzzati Traverso, Via Ercole Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy; (M.P.M.); (M.P.); (A.L.)
| | - Roberto Pallini
- Institute of Neurosurgery, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy;
| | - Andrea Levi
- CNR-Institute of Biochemistry and Cell Biology, Campus Adriano Buzzati Traverso, Via Ercole Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy; (M.P.M.); (M.P.); (A.L.)
| | - Maria Laura Falchetti
- CNR-Institute of Biochemistry and Cell Biology, Campus Adriano Buzzati Traverso, Via Ercole Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy; (M.P.M.); (M.P.); (A.L.)
| |
Collapse
|
17
|
Saleh T, Bloukh S, Carpenter VJ, Alwohoush E, Bakeer J, Darwish S, Azab B, Gewirtz DA. Therapy-Induced Senescence: An "Old" Friend Becomes the Enemy. Cancers (Basel) 2020; 12:cancers12040822. [PMID: 32235364 PMCID: PMC7226427 DOI: 10.3390/cancers12040822] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/21/2020] [Accepted: 03/25/2020] [Indexed: 01/10/2023] Open
Abstract
For the past two decades, cellular senescence has been recognized as a central component of the tumor cell response to chemotherapy and radiation. Traditionally, this form of senescence, termed Therapy-Induced Senescence (TIS), was linked to extensive nuclear damage precipitated by classical genotoxic chemotherapy. However, a number of other forms of therapy have also been shown to induce senescence in tumor cells independently of direct genomic damage. This review attempts to provide a comprehensive summary of both conventional and targeted anticancer therapeutics that have been shown to induce senescence in vitro and in vivo. Still, the utility of promoting senescence as a therapeutic endpoint remains under debate. Since senescence represents a durable form of growth arrest, it might be argued that senescence is a desirable outcome of cancer therapy. However, accumulating evidence suggesting that cells have the capacity to escape from TIS would support an alternative conclusion, that senescence provides an avenue whereby tumor cells can evade the potentially lethal action of anticancer drugs, allowing the cells to enter a temporary state of dormancy that eventually facilitates disease recurrence, often in a more aggressive state. Furthermore, TIS is now strongly connected to tumor cell remodeling, potentially to tumor dormancy, acquiring more ominous malignant phenotypes and accounts for several untoward adverse effects of cancer therapy. Here, we argue that senescence represents a barrier to effective anticancer treatment, and discuss the emerging efforts to identify and exploit agents with senolytic properties as a strategy for elimination of the persistent residual surviving tumor cell population, with the goal of mitigating the tumor-promoting influence of the senescent cells and to thereby reduce the likelihood of cancer relapse.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan; (T.S.); (S.D.)
| | - Sarah Bloukh
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
| | - Valerie J. Carpenter
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Enas Alwohoush
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
| | - Jomana Bakeer
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
| | - Sarah Darwish
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan; (T.S.); (S.D.)
| | - Belal Azab
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA;
- Correspondence:
| |
Collapse
|
18
|
Santos-de-Frutos K, Segrelles C, Lorz C. Hippo Pathway and YAP Signaling Alterations in Squamous Cancer of the Head and Neck. J Clin Med 2019; 8:jcm8122131. [PMID: 31817001 PMCID: PMC6947155 DOI: 10.3390/jcm8122131] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 12/20/2022] Open
Abstract
Head and neck cancer affects the upper aerodigestive tract and is the sixth leading cancer worldwide by incidence and the seventh by cause of death. Despite significant advances in surgery and chemotherapy, molecularly targeted therapeutic options for this type of cancer are scarce and long term survival rates remain low. Recently, comprehensive genomic studies have highlighted the most commonly altered genes and signaling pathways in this cancer. The Hippo-YAP pathway has been identified as a key oncogenic pathway in multiple tumors. Expression of genes controlled by the Hippo downstream transcriptional coactivators YAP (Yes-associated protein 1) and TAZ (WWTR1, WW domain containing transcription regulator 1) is widely deregulated in human cancer including head and neck squamous cell carcinoma (HNSCC). Interestingly, YAP/TAZ signaling might not be as essential for the normal homeostasis of adult tissues as for oncogenic growth, altogether making the pathway an amenable therapeutic target in cancer. Recent advances in the role of Hippo-YAP pathway in HNSCC have provided evidence that genetic alterations frequent in this type of cancer such as PIK3CA (phosphatidylinositide 3-kinase catalytic subunit alpha) overexpression or FAT1 (FAT atypical cadherin 1) functional loss can result in YAP activation. We discuss current therapeutic options targeting this pathway which are currently in use for other tumor types.
Collapse
Affiliation(s)
- Karla Santos-de-Frutos
- Molecular Oncology Unit, CIEMAT (ed 70A), Ave Complutense 40, 28040 Madrid, Spain; (K.S.-d.-F.); (C.S.)
- Molecular Oncology, Research Institute 12 de Octubre i+12, University Hospital 12 de Octubre, Ave Córdoba s/n, 28041 Madrid, Spain
| | - Carmen Segrelles
- Molecular Oncology Unit, CIEMAT (ed 70A), Ave Complutense 40, 28040 Madrid, Spain; (K.S.-d.-F.); (C.S.)
- Molecular Oncology, Research Institute 12 de Octubre i+12, University Hospital 12 de Octubre, Ave Córdoba s/n, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Ave Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Corina Lorz
- Molecular Oncology Unit, CIEMAT (ed 70A), Ave Complutense 40, 28040 Madrid, Spain; (K.S.-d.-F.); (C.S.)
- Molecular Oncology, Research Institute 12 de Octubre i+12, University Hospital 12 de Octubre, Ave Córdoba s/n, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Ave Monforte de Lemos 3-5, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-91-4962-521; Fax: +34-91-3466-484
| |
Collapse
|
19
|
4,5-Diphenyl-2-methyl picolinate induces cellular senescence by accumulating DNA damage and activating associated signaling pathways in gastric cancer. Life Sci 2019; 238:116973. [PMID: 31639393 DOI: 10.1016/j.lfs.2019.116973] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/13/2019] [Accepted: 10/14/2019] [Indexed: 01/28/2023]
Abstract
AIMS Gastric cancer (GC) is a common cancer with a relatively low survival rate. Cellular senescence, a potent anti-cancer mechanism, is naturally occurred, and can be induced by chemotherapeutic agents. We sought to explore new compounds against GC cells by inducing cellular senescence. MAIN METHODS Primary screening of a library of N-heterocyclic compounds identified some with potent inhibitory effects on GC cells. Furthermore, in vitro effects of the most potent candidate compound on the proliferation and senescence of GC cells were studied by classical assays, including senescence-associated (SA)-β-galactosidase staining, and immunofluorescence; and in vivo effects of this compound was evaluated in a xenograft tumor mouse model. KEY FINDINGS Among 43 tested compounds, 4,5-diphenyl-2-methyl picolinate (DMP) showed the highest inhibition effects on the growth of GC cells. In vitro experiments showed that DMP inhibited the proliferation by inducing senescence and DNA-damage associated protein markers and signaling pathways. In vivo experiment confirmed that DMP treatment inhibited tumor growth by promoting DNA-damage signaling. SIGNIFICANCE This study set up a platform to identify senescence-inducing anti-cancer compounds, and uncovers that DMP exerted anticancer effects by inducing cellular senescence through targeting DNA damage and associated signaling pathways in GC cancer.
Collapse
|
20
|
Wen Z, Jiang R, Huang Y, Wen Z, Rui D, Liao X, Ling Z. Inhibition of lung cancer cells and Ras/Raf/MEK/ERK signal transduction by ectonucleoside triphosphate phosphohydrolase-7 (ENTPD7). Respir Res 2019; 20:194. [PMID: 31443651 PMCID: PMC6708200 DOI: 10.1186/s12931-019-1165-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 08/13/2019] [Indexed: 12/11/2022] Open
Abstract
Background The aim of this study was to investigate the effects and mechanisms of ectonucleoside triphosphate phosphohydrolase-7 (ENTPD7) on lung cancer cells. Methods The expression characteristics of ENTPD7 and its effect on the survival of lung cancer patients were analyzed by referring to The Cancer Genome Atlas (TCGA). Streptavidin-peroxidase (SP) staining was performed to detect the ENTPD7 protein in tumor tissues and adjacent tissues. Plasmid transfection technology was also applied to silence ENTPD7 gene. Crystal violet staining and flow cytometry were performed to determine cell proliferation and apoptosis. Tumor-bearing nude mice model was established to investigate the effect of sh-ENTPD7 on tumors. Results The results showed that patients with low levels of ENTPD7 had higher survival rates. ENTPD7 was up-regulated in lung cancer tissues and cells. Down-regulation of the expression of ENTPD7 inhibited proliferation but promoted apoptosis of lung cancer cell. Silencing ENTPD7 also inhibited the expression levels of Ras and Raf proteins and the phosphorylation of mitogen-activated protein kinase (MEK) and extracellular signal-regulated kinase (ERK). Tumor-bearing nude mice experiments showed that silencing ENTPD7 had an inhibitory effect on lung cancer cells. Conclusions ENTPD7 was overexpressed in lung cancer cells. Down-regulating ENTPD7 could inhibit lung cancer cell proliferation and promote apoptosis via inhibiting the Ras/Raf/MEK/ERK pathway.
Collapse
Affiliation(s)
- Zhongwei Wen
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, No. 1 Liushi Road, Liuzhou, 545005, Guangxi Province, China
| | - Rongfang Jiang
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, No. 1 Liushi Road, Liuzhou, 545005, Guangxi Province, China
| | - Ying Huang
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, No. 1 Liushi Road, Liuzhou, 545005, Guangxi Province, China
| | - Zhineng Wen
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, No. 1 Liushi Road, Liuzhou, 545005, Guangxi Province, China
| | - Dong Rui
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, No. 1 Liushi Road, Liuzhou, 545005, Guangxi Province, China
| | - Xiaoxiao Liao
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, No. 1 Liushi Road, Liuzhou, 545005, Guangxi Province, China
| | - Zhougui Ling
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, No. 1 Liushi Road, Liuzhou, 545005, Guangxi Province, China.
| |
Collapse
|
21
|
van der Feen DE, Berger RMF, Bartelds B. Converging Paths of Pulmonary Arterial Hypertension and Cellular Senescence. Am J Respir Cell Mol Biol 2019; 61:11-20. [DOI: 10.1165/rcmb.2018-0329tr] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Diederik E. van der Feen
- Center for Congenital Heart Diseases, Department of Paediatric Cardiology, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rolf M. F. Berger
- Center for Congenital Heart Diseases, Department of Paediatric Cardiology, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Beatrijs Bartelds
- Center for Congenital Heart Diseases, Department of Paediatric Cardiology, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
22
|
Singh R, Peng S, Viswanath P, Sambandam V, Shen L, Rao X, Fang B, Wang J, Johnson FM. Non-canonical cMet regulation by vimentin mediates Plk1 inhibitor-induced apoptosis. EMBO Mol Med 2019; 11:e9960. [PMID: 31040125 PMCID: PMC6505578 DOI: 10.15252/emmm.201809960] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/28/2019] [Accepted: 03/12/2019] [Indexed: 12/26/2022] Open
Abstract
To address the need for improved systemic therapy for non-small-cell lung cancer (NSCLC), we previously demonstrated that mesenchymal NSCLC was sensitive to polo-like kinase (Plk1) inhibitors, but the mechanisms of resistance in epithelial NSCLC remain unknown. Here, we show that cMet was differentially regulated in isogenic pairs of epithelial and mesenchymal cell lines. Plk1 inhibition inhibits cMet phosphorylation only in mesenchymal cells. Constitutively active cMet abrogates Plk1 inhibitor-induced apoptosis. Likewise, cMet silencing or inhibition enhances Plk1 inhibitor-induced apoptosis. Cells with acquired resistance to Plk1 inhibitors are more epithelial than their parental cells and maintain cMet activation after Plk1 inhibition. In four animal NSCLC models, mesenchymal tumors were more sensitive to Plk1 inhibition alone than were epithelial tumors. The combination of cMet and Plk1 inhibition led to regression of tumors that did not regrow when drug treatment was stopped. Plk1 inhibition did not affect HGF levels but did decrease vimentin phosphorylation, which regulates cMet phosphorylation via β1-integrin. This research defines a heretofore unknown mechanism of ligand-independent activation of cMet downstream of Plk1 and an effective combination therapy.
Collapse
Affiliation(s)
- Ratnakar Singh
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shaohua Peng
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pavitra Viswanath
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Vaishnavi Sambandam
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Shen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiayu Rao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Faye M Johnson
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
23
|
Oku Y, Nishiya N, Tazawa T, Kobayashi T, Umezawa N, Sugawara Y, Uehara Y. Augmentation of the therapeutic efficacy of WEE1 kinase inhibitor AZD1775 by inhibiting the YAP-E2F1-DNA damage response pathway axis. FEBS Open Bio 2018; 8:1001-1012. [PMID: 29928579 PMCID: PMC5986022 DOI: 10.1002/2211-5463.12440] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/26/2018] [Accepted: 04/25/2018] [Indexed: 01/10/2023] Open
Abstract
The main reasons for failure of cancer chemotherapy are intrinsic and acquired drug resistance. The Hippo pathway effector Yes‐associated protein (YAP) is associated with resistance to both cytotoxic and molecular targeted drugs. Several lines of evidence indicate that YAP activates transcriptional programmes to promote cell cycle progression and DNA damage responses. Therefore, we hypothesised that YAP is involved in the sensitivity of cancer cells to small‐molecule agents targeting cell cycle‐related proteins. Here, we report that the inactivation of YAP sensitises the OVCAR‐8 ovarian cancer cell line to AZD1775, a small‐molecule WEE1 kinase inhibitor. The accumulation of DNA damage and mitotic failures induced by AZD1775‐based therapy were further enhanced by YAP depletion. YAP depletion reduced the expression of the Fanconi anaemia (FA) pathway components required for DNA repair and their transcriptional regulator E2F1. These results suggest that YAP activates the DNA damage response pathway, exemplified by the FA pathway and E2F1. Furthermore, we aimed to apply this finding to combination chemotherapy against ovarian cancers. The regimen containing dasatinib, which inhibits the nuclear localisation of YAP, improved the response to AZD1775‐based therapy in the OVCAR‐8 ovarian cancer cell line. We propose that dasatinib acts as a chemosensitiser for a subset of molecular targeted drugs, including AZD1775, by targeting YAP.
Collapse
Affiliation(s)
- Yusuke Oku
- Department of Integrated Information for Pharmaceutical Sciences Iwate Medical University School of Pharmacy Yahaba-cho Japan
| | - Naoyuki Nishiya
- Department of Integrated Information for Pharmaceutical Sciences Iwate Medical University School of Pharmacy Yahaba-cho Japan
| | - Takaaki Tazawa
- Department of Integrated Information for Pharmaceutical Sciences Iwate Medical University School of Pharmacy Yahaba-cho Japan
| | - Takaya Kobayashi
- Department of Integrated Information for Pharmaceutical Sciences Iwate Medical University School of Pharmacy Yahaba-cho Japan
| | - Nanami Umezawa
- Department of Integrated Information for Pharmaceutical Sciences Iwate Medical University School of Pharmacy Yahaba-cho Japan
| | - Yasuyo Sugawara
- Department of Integrated Information for Pharmaceutical Sciences Iwate Medical University School of Pharmacy Yahaba-cho Japan
| | - Yoshimasa Uehara
- Department of Integrated Information for Pharmaceutical Sciences Iwate Medical University School of Pharmacy Yahaba-cho Japan
| |
Collapse
|
24
|
Shang D, Wu Y, Ding Y, Lu Z, Shen Y, Zhu F, Liu H, Zhu C, Tu Z. Identification of a pyridine derivative inducing senescence in ovarian cancer cell lines via P21 activation. Clin Exp Pharmacol Physiol 2017; 45:452-460. [DOI: 10.1111/1440-1681.12891] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 10/04/2017] [Accepted: 10/26/2017] [Indexed: 01/15/2023]
Affiliation(s)
- Dongsheng Shang
- School of Pharmacy; Jiangsu University; Zhenjiang China
- Institute of Life Sciences; Jiangsu University; Zhenjiang China
| | - Yanfang Wu
- Institute of Life Sciences; Jiangsu University; Zhenjiang China
| | - Ya Ding
- School of Chemistry & Chemical Engineering; Jiangsu University; Zhenjiang China
| | - Ziwen Lu
- School of Pharmacy; Jiangsu University; Zhenjiang China
| | - Yanting Shen
- School of Pharmacy; Jiangsu University; Zhenjiang China
| | - Feifei Zhu
- Institute of Life Sciences; Jiangsu University; Zhenjiang China
| | - Hanqing Liu
- School of Pharmacy; Jiangsu University; Zhenjiang China
| | - Chunyin Zhu
- School of Chemistry & Chemical Engineering; Jiangsu University; Zhenjiang China
| | - Zhigang Tu
- Institute of Life Sciences; Jiangsu University; Zhenjiang China
| |
Collapse
|
25
|
Phase I study of the combination of crizotinib (as a MET inhibitor) and dasatinib (as a c-SRC inhibitor) in patients with advanced cancer. Invest New Drugs 2017; 36:416-423. [PMID: 29047029 DOI: 10.1007/s10637-017-0513-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 09/20/2017] [Indexed: 01/19/2023]
Abstract
Background Both MET and c-SRC are important mediators of cancer progression and there is cross talk between the two molecules. Preclinical studies have demonstrated combination of MET and c-SRC inhibitors is effective in multiple cancer types. Methods We analyzed the safety and efficacy of administering a c-SRC inhibitor (dasatinib) in combination with a MET inhibitor (crizotinib) in a two-arm concurrent phase I study. Arm A consisted of crizotinib fixed at 250 mg twice per day with escalation of dasatinib. Arm B consisted of dasatinib fixed at 140 mg daily with escalation of crizotinib. Endpoints included dose-limiting toxicities (DLTs), recommended phase II dose (RP2D), and response (RECIST 1.1). Results We enrolled 61 patients (arm A: 31, arm B: 30). The most common cancers were sarcoma (21%) and prostate cancer (16%). In Arm A, at dose level 2 (DL2), 40% (2/5) experienced DLTs. In the expanded DL1, 21% (4/19) experienced DLTs (all grade 3). In Arm B, at DL2, 50% (2/4) experienced DLTs. In the expanded DL1, 22% (4/18) experienced DLTs (all grade 3). RP2D was determined to be arm A, DL1 (250 mg crizotinib orally twice per day plus 50 mg dasatinib orally daily). Partial response (N = 1) and stable disease for ≥6 months (N = 3) were seen. Conclusions The combination of crizotinib and dasatinib is safe to administer but tolerability is limited given the high rate of adverse events. Responses and durable stable disease were limited. Further precision therapy approach using this specific combination may be difficult given the toxicity.
Collapse
|
26
|
Cardnell RJ, Li L, Sen T, Bara R, Tong P, Fujimoto J, Ireland AS, Guthrie MR, Bheddah S, Banerjee U, Kalu NN, Fan YH, Dylla SJ, Johnson FM, Wistuba II, Oliver TG, Heymach JV, Glisson BS, Wang J, Byers LA. Protein expression of TTF1 and cMYC define distinct molecular subgroups of small cell lung cancer with unique vulnerabilities to aurora kinase inhibition, DLL3 targeting, and other targeted therapies. Oncotarget 2017; 8:73419-73432. [PMID: 29088717 PMCID: PMC5650272 DOI: 10.18632/oncotarget.20621] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 08/14/2017] [Indexed: 01/09/2023] Open
Abstract
Small cell lung cancer (SCLC) is a recalcitrant cancer for which no new treatments have been approved in over 30 years. While molecular subtyping now guides treatment selection for patients with non-small cell lung cancer and other cancers, SCLC is still treated as a single disease entity. Using model-based clustering, we found two major proteomic subtypes of SCLC characterized by either high thyroid transcription factor-1 (TTF1)/low cMYC protein expression or high cMYC/low TTF1. Applying "drug target constellation" (DTECT) mapping, we further show that protein levels of TTF1 and cMYC predict response to targeted therapies including aurora kinase, Bcl2, and HSP90 inhibitors. Levels of TTF1 and DLL3 were also highly correlated in preclinical models and patient tumors. TTF1 (used in the diagnosis lung cancer) could therefore be used as a surrogate of DLL3 expression to identify patients who may respond to the DLL3 antibody-drug conjugate rovalpituzumab tesirine. These findings suggest that TTF1, cMYC or other protein markers identified here could be used to identify subgroups of SCLC patients who may respond preferentially to several emerging targeted therapies.
Collapse
Affiliation(s)
- Robert J Cardnell
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lerong Li
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Triparna Sen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rasha Bara
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pan Tong
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Junya Fujimoto
- Department of Molecular Translational Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Abbie S Ireland
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Matthew R Guthrie
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | | | - Upasana Banerjee
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nene N Kalu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - You-Hong Fan
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Faye M Johnson
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Ignacio I Wistuba
- Department of Molecular Translational Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Trudy G Oliver
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bonnie S Glisson
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Lauren A Byers
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
27
|
Lee CS, Baek J, Han SY. The Role of Kinase Modulators in Cellular Senescence for Use in Cancer Treatment. Molecules 2017; 22:molecules22091411. [PMID: 28841181 PMCID: PMC6151769 DOI: 10.3390/molecules22091411] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/22/2017] [Accepted: 08/24/2017] [Indexed: 12/27/2022] Open
Abstract
Recently, more than 30 small molecules and eight monoclonal antibodies that modulate kinase signaling have been approved for the treatment of several pathological conditions, including cancer, idiopathic pulmonary fibrosis, and rheumatoid arthritis. Among them, kinase modulators have been a primary focus for use in cancer treatment. Cellular senescence is believed to protect cells from tumorigenesis by irreversibly halting cell cycle progression and avoiding the growth of damaged cells and tissues. Senescence can also contribute to tumor suppression and be utilized as a mechanism by anti-cancer agents. Although the role of kinase modulators in cancer treatment and their effects on senescence in tumor development have been extensively studied, the relationship between kinase modulators for cancer treatment and senescence has not been fully discussed. In this review, we discuss the pro- and anti-tumorigenesis functions of senescence and summarize the key roles of kinase modulators in the regulation of senescence against tumors.
Collapse
Affiliation(s)
- Chang Sup Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju-daero, Jinju, Gyeongnam 52828, Korea.
| | - Juhwa Baek
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju-daero, Jinju, Gyeongnam 52828, Korea.
| | - Sun-Young Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju-daero, Jinju, Gyeongnam 52828, Korea.
| |
Collapse
|
28
|
Ubiquitin-specific protease 21 stabilizes BRCA2 to control DNA repair and tumor growth. Nat Commun 2017; 8:137. [PMID: 28743957 PMCID: PMC5526993 DOI: 10.1038/s41467-017-00206-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 06/13/2017] [Indexed: 01/23/2023] Open
Abstract
Tumor growth relies on efficient DNA repair to mitigate the detrimental impact of DNA damage associated with excessive cell division. Modulating repair factor function, thus, provides a promising strategy to manipulate malignant growth. Here, we identify the ubiquitin-specific protease USP21 as a positive regulator of BRCA2, a key mediator of DNA repair by homologous recombination. USP21 interacts with, deubiquitinates and stabilizes BRCA2 to promote efficient RAD51 loading at DNA double-strand breaks. As a result, depletion of USP21 decreases homologous recombination efficiency, causes an increase in DNA damage load and impairs tumor cell survival. Importantly, BRCA2 overexpression partially restores the USP21-associated survival defect. Moreover, we show that USP21 is overexpressed in hepatocellular carcinoma, where it promotes BRCA2 stability and inversely correlates with patient survival. Together, our findings identify deubiquitination as a means to regulate BRCA2 function and point to USP21 as a potential therapeutic target in BRCA2-proficient tumors.BRCA2 is essential for the repair of DNA damage; therefore, defects in BRCA2 are associated with tumorigenesis but also with increased susceptibility to genotoxic stress. Here the authors show that USP21 regulates the ability of tumor cells to repair damaged DNA by regulating BRCA2 stability.
Collapse
|
29
|
Shi J, Yuan M, Wang ZD, Xu XL, Hong L, Sun S. Comprehensive profiling and quantitation of oncogenic mutations in non-small cell lung carcinoma using single-molecule amplification and re-sequencing technology. Tumour Biol 2017; 39:1010428317691413. [PMID: 28218040 DOI: 10.1177/1010428317691413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The carcinogenesis of non-small cell lung carcinoma has been found to associate with activating and resistant mutations in the tyrosine kinase domain of specific oncogenes. Here, we assessed the type, frequency, and abundance of epithelial growth factor receptor, KRAS, BRAF, and ALK mutations in 154 non-small cell lung carcinoma specimens using single-molecule amplification and re-sequencing technology. We found that epithelial growth factor receptor mutations were the most prevalent (44.2%), followed by KRAS (18.8%), ALK (7.8%), and BRAF (5.8%) mutations. The type and abundance of the mutations in tumor specimens appeared to be heterogeneous. Thus, we conclude that identification of clinically significant oncogenic mutations may improve the classification of patients and provide valuable information for determination of the therapeutic strategies.
Collapse
Affiliation(s)
- Jian Shi
- 1 Department of Medical Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Meng Yuan
- 2 College of Science and Technology, China Three Gorges University, Yichang, China
| | - Zhan-Dong Wang
- 3 Department of Pathology, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiao-Li Xu
- 4 Department of Medical Record Library, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lei Hong
- 1 Department of Medical Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shenglin Sun
- 1 Department of Medical Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
30
|
Zhang R, Zhao J, Xu J, Liu F. Association of peripheral leukocyte telomere length and its variation with pancreatic cancer and colorectal cancer risk in Chinese population. Oncotarget 2016; 7:38579-85. [PMID: 27509261 PMCID: PMC5122412 DOI: 10.18632/oncotarget.9536] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 04/27/2016] [Indexed: 11/29/2022] Open
Abstract
There is increasing evidence supporting the role of telomeres in cancer pathogenesis. However, limited studies have investigated the association between telomere length features and risk of pancreatic cancer and colorectal cancer (CRC), and little was conducted in Asians. To help clarify this issue, We measured relative peripheral leukocytes telomere length (LTL) and telomere length variation (TLV) in a prospective study of 900 pancreatic cancer cases, 300 CRC cases, and 900 controls. Both subjects with longer LTL (quartile 4: adjusted OR=1.51, 95% CI: 1.14-1.99, P=0.004) and shorter LTL (quartile 1: adjusted OR=3.12, 95% CI: 1.89-5.14, P=8.50x10-6) showed increased risk of pancreatic cancer. A linear increased risk was detected For TLV (adjusted OR=1.60, 95% CI: 1.14-2.24, P=0.006). We also identified significant interaction for relative LTL, TLV on pancreatic cancer risk (P interaction =0.009). Significant relationship between shorter RTL and increased CRC risk were also detected. This findings provide insights into telomere dynamics and highlight the complex relationship between relative LTL, TLV and cancer risk.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Colorectal surgery, Liaoning cancer hospital & institute, Shenyang, Liaoning Province, 110042, P.R. China
| | - Jian Zhao
- Department of Colorectal surgery, Liaoning cancer hospital & institute, Shenyang, Liaoning Province, 110042, P.R. China
| | - Jian Xu
- Department of Colorectal surgery, Liaoning cancer hospital & institute, Shenyang, Liaoning Province, 110042, P.R. China
| | - Fang Liu
- Department of Colorectal surgery, Liaoning cancer hospital & institute, Shenyang, Liaoning Province, 110042, P.R. China
| |
Collapse
|
31
|
Emergence of a daptomycin-non-susceptible Enterococcus faecium strain that encodes mutations in DNA repair genes after high-dose daptomycin therapy. BMC Res Notes 2016; 9:197. [PMID: 27036708 PMCID: PMC4818489 DOI: 10.1186/s13104-016-2003-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/22/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND An increasing number of reports have documented the emergence of daptomycin-nonsusceptible Enterococcus in patients during daptomycin therapy. Even though several mechanisms for daptomycin-nonsusceptibility have been suggested, the potential genetic mutations which might contribute to the daptomycin-nonsusceptibility are not fully understood. CASE PRESENTATION We isolated a vancomycin-susceptible, daptomycin nonsusceptible Enterococcus faecium strain from a patient with acute lymphocytic leukemia who received high-dose daptomycin therapy for E. faecium endocarditis. Whole-genome sequencing analysis revealed mutations within genes encoding DNA repair proteins MutL and RecJ of the daptomycin-nonsusceptible Enterococcus strain which might have facilitated its emergence. CONCLUSIONS We identified the mutations of DNA mismatch repair genes in a clinical isolate of daptomycin nonsusceptible E. faecium which emerged in spite of high-dose daptomycin therapy. The finding implicates the possible association of DNA repair mechanism and daptomycin resistance. Careful monitoring is necessary to avoid the emergence of daptomycin non-susceptible isolates of E. faecium and particularly in cases of long-term daptomycin use or in immunocompromised patients.
Collapse
|