1
|
Siragusa G, Tomasello L, Giordano C, Pizzolanti G. Survivin (BIRC5): Implications in cancer therapy. Life Sci 2024; 350:122788. [PMID: 38848940 DOI: 10.1016/j.lfs.2024.122788] [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: 01/26/2024] [Revised: 03/13/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Inhibitors of Apoptosis proteins (IAPs) were discovered through experiments aimed at rescuing apoptosis in insects. Classically associated with the inhibition of apoptosis, the IAP member Survivin also regulates cell cycle progression and is an essential component of the Chromosomal Passenger Complex (CPC), responsible for chromosomal segregation. Although undetectable in most adult tissues, Survivin is expressed in Adult Stem Cells (ASCs) and plays a crucial role in their maintenance. Survivin is overexpressed in most cancers, contributing to their clonal expansion. As a result, it has been proposed as a possible anticancer target for nearly two decades. In this discussion, we will explore the rationale behind Survivin as a therapeutic target, focusing on common cancer types such as carcinomas, sarcomas, and leukemias. We will delve into the modulation of Survivin by cancer pro-survival cell signaling, the association between SNPs and tumorigenesis, and its regulation by miRNAs. Finally, we will compare cell growth, clonogenic capacity, and apoptosis, along with different strategies for Survivin inhibition, including gene expression and protein activity modulation.
Collapse
Affiliation(s)
- Giuseppe Siragusa
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Italy
| | - Laura Tomasello
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Italy
| | - Carla Giordano
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Italy
| | - Giuseppe Pizzolanti
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Italy; Advanced Technologies Network Center (ATEN Center), University of Palermo, Italy.
| |
Collapse
|
2
|
Cui Q, Huang C, Liu JY, Zhang JT. Small Molecule Inhibitors Targeting the "Undruggable" Survivin: The Past, Present, and Future from a Medicinal Chemist's Perspective. J Med Chem 2023; 66:16515-16545. [PMID: 38092421 DOI: 10.1021/acs.jmedchem.3c01130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Survivin, a homodimeric protein and a member of the IAP family, plays a vital function in cell survival and cycle progression by interacting with various proteins and complexes. Its expression is upregulated in cancers but not detectable in normal tissues. Thus, it has been regarded and validated as an ideal cancer target. However, survivin is "undruggable" due to its lack of enzymatic activities or active sites for small molecules to bind/inhibit. Academic and industrial laboratories have explored different strategies to overcome this hurdle over the past two decades, with some compounds advanced into clinical testing. These strategies include inhibiting survivin expression, its interaction with binding partners and homodimerization. Here, we provide comprehensive analyses of these strategies and perspective on different small molecule survivin inhibitors to help drug discovery targeting "undruggable" proteins in general and survivin specifically with a true survivin inhibitor that will prevail in the foreseeable future.
Collapse
Affiliation(s)
- Qingbin Cui
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Caoqinglong Huang
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Jing-Yuan Liu
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Jian-Ting Zhang
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| |
Collapse
|
3
|
Serafini RA, Frere JJ, Zimering J, Giosan IM, Pryce KD, Golynker I, Panis M, Ruiz A, tenOever BR, Zachariou V. SARS-CoV-2 airway infection results in the development of somatosensory abnormalities in a hamster model. Sci Signal 2023; 16:eade4984. [PMID: 37159520 PMCID: PMC10422867 DOI: 10.1126/scisignal.ade4984] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 04/06/2023] [Indexed: 05/11/2023]
Abstract
Although largely confined to the airways, SARS-CoV-2 infection has been associated with sensory abnormalities that manifest in both acute and chronic phenotypes. To gain insight on the molecular basis of these sensory abnormalities, we used the golden hamster model to characterize and compare the effects of infection with SARS-CoV-2 and influenza A virus (IAV) on the sensory nervous system. We detected SARS-CoV-2 transcripts but no infectious material in the cervical and thoracic spinal cord and dorsal root ganglia (DRGs) within the first 24 hours of intranasal virus infection. SARS-CoV-2-infected hamsters exhibited mechanical hypersensitivity that was milder but prolonged compared with that observed in IAV-infected hamsters. RNA sequencing analysis of thoracic DRGs 1 to 4 days after infection suggested perturbations in predominantly neuronal signaling in SARS-CoV-2-infected animals as opposed to type I interferon signaling in IAV-infected animals. Later, 31 days after infection, a neuropathic transcriptome emerged in thoracic DRGs from SARS-CoV-2-infected animals, which coincided with SARS-CoV-2-specific mechanical hypersensitivity. These data revealed potential targets for pain management, including the RNA binding protein ILF3, which was validated in murine pain models. This work elucidates transcriptomic signatures in the DRGs triggered by SARS-CoV-2 that may underlie both short- and long-term sensory abnormalities.
Collapse
Affiliation(s)
- Randal A. Serafini
- Nash Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Justin J. Frere
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jeffrey Zimering
- Nash Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ilinca M. Giosan
- Nash Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kerri D. Pryce
- Nash Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ilona Golynker
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Maryline Panis
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Anne Ruiz
- Nash Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Benjamin R. tenOever
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Venetia Zachariou
- Nash Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
4
|
Jafarzadeh A, Bazargan N, Chatrabnous N, Jafarzadeh S, Nemati M. Contribution of survivin to the immune system, allergies and autoimmune diseases. Hum Immunol 2023; 84:301-310. [PMID: 36754653 DOI: 10.1016/j.humimm.2023.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/30/2022] [Accepted: 01/30/2023] [Indexed: 02/09/2023]
Abstract
In addition to malignancies, survivin (a member of the apoptosis inhibitor family) has been implicated in the pathogenesis of inflammatory disorders, including autoimmune and allergic diseases. Survivin is constantly expressed in the proliferating hematopoietic progenitor cells, and it is re-expressed in the mature cells of the innate and adaptive immunity, upon activation. Survivin enhances the expression of co-stimulatory molecules and MHC class II molecules in dendritic cells, and promotes the lifespan of macrophages, neutrophils, and eosinophils, while suppressing natural killer (NK) cell activity. Survivin has been implicated in T cell maturation, T cell expansion, effector CD4+ T cell differentiation, maintenance of memory CD4+ T and CD8+ T cells, as well as antibody production. Upregulated expression of survivin was indicated in the T cells as well as various samples collected from allergic patients. Survivin can contribute to the pathogenesis of allergic diseases via the promotion of the Th2 polarization, promoting IL-4 expression, compromising activation-induced cell death (AICD) in Th2 cells, and preventing apoptosis of eosinophils, as well as, amplification of eosinophilia. Moreover, survivin can interfere with clonal deletion of autoreactive T and B cells, as well as suppress Treg cell development and activity supporting the development of autoimmune diseases. This review discusses the role of survivin in immunity, allergy and autoimmunity as well as provides evidence that survivin may be considered as a novel therapeutic target for the treatment of allergic and autoimmune diseases.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Nasrin Bazargan
- Department of Internal Medicine, Medical School, Kerman University of Medical Sciences, Kerman, Iran
| | - Nazanin Chatrabnous
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Jafarzadeh
- Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Nemati
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Haematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
5
|
Survivin Small Molecules Inhibitors: Recent Advances and Challenges. Molecules 2023; 28:molecules28031376. [PMID: 36771042 PMCID: PMC9919791 DOI: 10.3390/molecules28031376] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/24/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Survivin, as a member of the inhibitor of apoptosis proteins (IAPs) family, acts as a suppressor of apoptosis and plays a central role in cell division. Survivin has been considered as an important cancer drug target because it is highly expressed in many types of human cancers, while it is effectively absent from terminally differentiated normal tissues. Moreover, survivin is involved in tumor cell resistance to chemotherapy and radiation. Preclinically, downregulation of survivin expression or function reduced tumor growth induced apoptosis and sensitized tumor cells to radiation and chemotherapy in different human tumor models. This review highlights the role of survivin in promoting cellular proliferation and inhibiting apoptosis and summarizes the recent advances in and challenges of developing small-molecule survivin inhibitors.
Collapse
|
6
|
Serafini RA, Frere JJ, Zimering J, Giosan IM, Pryce KD, Golynker I, Panis M, Ruiz A, tenOever B, Zachariou V. SARS-CoV-2 Airway Infection Results in Time-dependent Sensory Abnormalities in a Hamster Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.08.19.504551. [PMID: 36032984 PMCID: PMC9413707 DOI: 10.1101/2022.08.19.504551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Despite being largely confined to the airways, SARS-CoV-2 infection has been associated with sensory abnormalities that manifest in both acute and long-lasting phenotypes. To gain insight on the molecular basis of these sensory abnormalities, we used the golden hamster infection model to characterize the effects of SARS-CoV-2 versus Influenza A virus (IAV) infection on the sensory nervous system. Efforts to detect the presence of virus in the cervical/thoracic spinal cord and dorsal root ganglia (DRGs) demonstrated detectable levels of SARS-CoV-2 by quantitative PCR and RNAscope uniquely within the first 24 hours of infection. SARS-CoV-2-infected hamsters demonstrated mechanical hypersensitivity during acute infection; intriguingly, this hypersensitivity was milder, but prolonged when compared to IAV-infected hamsters. RNA sequencing (RNA-seq) of thoracic DRGs from acute infection revealed predominantly neuron-biased signaling perturbations in SARS-CoV-2-infected animals as opposed to type I interferon signaling in tissue derived from IAV-infected animals. RNA-seq of 31dpi thoracic DRGs from SARS-CoV-2-infected animals highlighted a uniquely neuropathic transcriptomic landscape, which was consistent with substantial SARS-CoV-2-specific mechanical hypersensitivity at 28dpi. Ontology analysis of 1, 4, and 30dpi RNA-seq revealed novel targets for pain management, such as ILF3. Meta-analysis of all SARS-CoV-2 RNA-seq timepoints against preclinical pain model datasets highlighted both conserved and unique pro-nociceptive gene expression changes following infection. Overall, this work elucidates novel transcriptomic signatures triggered by SARS-CoV-2 that may underlie both short- and long-term sensory abnormalities while also highlighting several therapeutic targets for alleviation of infection-induced hypersensitivity. One Sentence Summary SARS-CoV-2 infection results in an interferon-associated transcriptional response in sensory tissues underlying time-dependent hypersensitivity.
Collapse
Affiliation(s)
- Randal A. Serafini
- Nash Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box #1022, New York, NY, 10029
| | - Justin J. Frere
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box #1124, New York, NY, 10029
- Department of Microbiology, New York University Langone, 430-450 E. 29 St., New York, NY 10016
| | - Jeffrey Zimering
- Nash Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box #1022, New York, NY, 10029
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box #1136, New York, NY, 10029
| | - Ilinca M. Giosan
- Nash Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box #1022, New York, NY, 10029
| | - Kerri D. Pryce
- Nash Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box #1022, New York, NY, 10029
| | - Ilona Golynker
- Department of Microbiology, New York University Langone, 430-450 E. 29 St., New York, NY 10016
| | - Maryline Panis
- Department of Microbiology, New York University Langone, 430-450 E. 29 St., New York, NY 10016
| | - Anne Ruiz
- Nash Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box #1022, New York, NY, 10029
| | - Benjamin tenOever
- Department of Microbiology, New York University Langone, 430-450 E. 29 St., New York, NY 10016
| | - Venetia Zachariou
- Nash Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box #1022, New York, NY, 10029
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box #1677, New York, New York 10029
| |
Collapse
|
7
|
Vay C, Schlünder PM, Dizdar L, Esposito I, Ghadimi MPH, Knoefel WT, Krieg A. Targeting abundant survivin expression in liposarcoma: subtype dependent therapy responses to YM155 treatment. J Cancer Res Clin Oncol 2021; 148:633-645. [PMID: 34860309 PMCID: PMC8881260 DOI: 10.1007/s00432-021-03871-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/25/2021] [Indexed: 12/23/2022]
Abstract
Purpose Liposarcoma (LPS) represent the largest group of malignant soft tissue tumours comprising a heterogeneous group of subtypes in which the degrees of chemoresistance and radiosensitivity strongly vary. Consequently, it is of utmost interest to establish novel therapeutic regimens based on molecular targets. Methods Immunohistochemical staining of survivin was performed in tissue microarrays comprising 49 primary LPS specimens. LPS cell lines were treated with survivin antagonist YM155 and doxorubicin or etoposide alone as well as in combination. Changes in cell viability were investigated and the synergistic effect of a combined therapy analysed. Results Immunohistochemistry revealed an abundant expression of survivin in LPS that significantly concurred with less-differentiated tumour subtypes and grading. In vitro, we demonstrated the impact of the survivin inhibitor YM155 on dedifferentiated LPS (DDLPS) and, even more imposing, pleomorphic LPS (PLS) tumour cell viability with a strong induction of apoptosis. A combined treatment of doxorubicin or etoposide with YM155 augmented the cytotoxic effects on DDLPS and PLS cells. Conclusion These findings support the significant role of survivin in the oncogenesis and progression of LPS subtypes providing a rationale to target survivin in eligible in-vivo models and to pioneer clinical applications of survivin-specific substances unfolding their therapeutic potential in LPS patients prospectively. Supplementary Information The online version contains supplementary material available at 10.1007/s00432-021-03871-5.
Collapse
Affiliation(s)
- Christian Vay
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Bldg. 12.46, 40225, Duesseldorf, Germany
| | - Philipp M Schlünder
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Bldg. 12.46, 40225, Duesseldorf, Germany
| | - Levent Dizdar
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Bldg. 12.46, 40225, Duesseldorf, Germany
| | - Irene Esposito
- Institute of Pathology, Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Markus P H Ghadimi
- Department of General, Visceral, Tumour, and Transplant Surgery, University of Cologne, Kerpener Strasse 62, 50931, Cologne, Germany
| | - Wolfram T Knoefel
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Bldg. 12.46, 40225, Duesseldorf, Germany
| | - Andreas Krieg
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Bldg. 12.46, 40225, Duesseldorf, Germany.
| |
Collapse
|
8
|
Chiou JT, Lee YC, Huang CH, Wang LJ, Shi YJ, Chang LS. Inhibition of Sp1-mediated survivin and MCL1 expression cooperates with SLC35F2 and myeloperoxidase to modulate YM155 cytotoxicity to human leukemia cells. Biochem Pharmacol 2021; 188:114544. [PMID: 33831396 DOI: 10.1016/j.bcp.2021.114544] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 11/17/2022]
Abstract
Although YM155 is reported to suppress survivin (also known as BIRC5) expression in cancer cells, its cytotoxic mechanism in human acute myeloid leukemia (AML) cells has not been clearly resolved. In this study, we analyzed the mechanistic pathways that modulate the sensitivity of human AML U937 and HL-60 cells to YM155. YM155 induced apoptosis in AML cells, which was characterized by p38 MAPK phosphorylation and downregulation of survivin and MCL1 expression. Phosphorylated p38 MAPK causes autophagy-mediated Sp1 degradation, thereby inhibiting the transcription of survivin and MCL1. The reduction of survivin and MCL1 levels further facilitated Sp1 protein degradation through autophagy. The restoration of Sp1, survivin, or MCL1 expression protected U937 and HL-60 cells from YM155-mediated cytotoxicity. U937 and HL-60 cells were continuously exposed to hydroquinone (HQ) to generate U937/HQ and HL-60/HQ cells, which showed increased SLC35F2 expression. The increase in SLC35F2 expression led to an increase in the sensitivity of U937/HQ cells to YM155-mediated cytotoxicity, whereas no such effect was observed in HL-60/HQ cells. Of note, myeloperoxidase (MPO) activity in HL-60 and HL-60/HQ cells enhanced YM155 cytotoxicity in these cells, and the enforced expression of MPO also increased the sensitivity of U937 cells to YM155. Taken together, we conclude that p38 MAPK-modulated autophagy inhibits Sp1-mediated survivin and MCL1 expression, which, in turn, leads to the death of U937 and HL-60 cells following YM155 treatment. In addition, our data indicate that SLC35F2 increases the sensitivity of U937 cells to YM155-mediated cytotoxicity, whereas MPO enhances YM155 cytotoxicity in U937 and HL-60 cells.
Collapse
Affiliation(s)
- Jing-Ting Chiou
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Yuan-Chin Lee
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Chia-Hui Huang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Liang-Jun Wang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Yi-Jun Shi
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
9
|
Qiao Y, Pei Y, Luo M, Rajasekaran M, Hui KM, Chen J. Cytokinesis regulators as potential diagnostic and therapeutic biomarkers for human hepatocellular carcinoma. Exp Biol Med (Maywood) 2021; 246:1343-1354. [PMID: 33899543 DOI: 10.1177/15353702211008380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cytokinesis, the final step of mitosis, is critical for maintaining the ploidy level of cells. Cytokinesis is a complex, highly regulated process and its failure can lead to genetic instability and apoptosis, contributing to the development of cancer. Human hepatocellular carcinoma is often accompanied by a high frequency of aneuploidy and the DNA ploidy pattern observed in human hepatocellular carcinoma results mostly from impairments in cytokinesis. Many key regulators of cytokinesis are abnormally expressed in human hepatocellular carcinoma, and their expression levels are often correlated with patient prognosis. Moreover, preclinical studies have demonstrated that the inhibition of key cytokinesis regulators can suppress the growth of human hepatocellular carcinoma. Here, we provide an overview of the current understanding of the signaling networks regulating cytokinesis, the key cytokinesis regulators involved in the initiation and development of human hepatocellular carcinoma, and their applications as potential diagnostic and therapeutic biomarkers.
Collapse
Affiliation(s)
- Yiting Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, P. R. China
| | - Yunxin Pei
- Pharmacy Institute and Department of Hepatology, Institute of Hepatology and Metabolic Diseases, Institute of Integrated Chinese and Western Medicine for Oncology, The affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Collaborative Innovation Center of Traditional Chinese Medicines from Zhejiang Province, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China
| | - Miao Luo
- Pharmacy Institute and Department of Hepatology, Institute of Hepatology and Metabolic Diseases, Institute of Integrated Chinese and Western Medicine for Oncology, The affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Collaborative Innovation Center of Traditional Chinese Medicines from Zhejiang Province, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China
| | - Muthukumar Rajasekaran
- Laboratory of Cancer Genomics, Division of Cellular and Molecular Research, National Cancer Centre, Singapore 169610, Singapore
| | - Kam M Hui
- Pharmacy Institute and Department of Hepatology, Institute of Hepatology and Metabolic Diseases, Institute of Integrated Chinese and Western Medicine for Oncology, The affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Collaborative Innovation Center of Traditional Chinese Medicines from Zhejiang Province, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China.,Laboratory of Cancer Genomics, Division of Cellular and Molecular Research, National Cancer Centre, Singapore 169610, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore.,Institute of Molecular and Cell Biology, A*STAR, Singapore 138673, Singapore.,Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jianxiang Chen
- Pharmacy Institute and Department of Hepatology, Institute of Hepatology and Metabolic Diseases, Institute of Integrated Chinese and Western Medicine for Oncology, The affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Collaborative Innovation Center of Traditional Chinese Medicines from Zhejiang Province, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China.,Laboratory of Cancer Genomics, Division of Cellular and Molecular Research, National Cancer Centre, Singapore 169610, Singapore
| |
Collapse
|
10
|
Identification of RNA-Binding Proteins as Targetable Putative Oncogenes in Neuroblastoma. Int J Mol Sci 2020; 21:ijms21145098. [PMID: 32707690 PMCID: PMC7403987 DOI: 10.3390/ijms21145098] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 12/26/2022] Open
Abstract
Neuroblastoma is a common childhood cancer with almost a third of those affected still dying, thus new therapeutic strategies need to be explored. Current experimental therapies focus mostly on inhibiting oncogenic transcription factor signalling. Although LIN28B, DICER and other RNA-binding proteins (RBPs) have reported roles in neuroblastoma development and patient outcome, the role of RBPs in neuroblastoma is relatively unstudied. In order to elucidate novel RBPs involved in MYCN-amplified and other high-risk neuroblastoma subtypes, we performed differential mRNA expression analysis of RBPs in a large primary tumour cohort (n = 498). Additionally, we found via Kaplan–Meier scanning analysis that 685 of the 1483 tested RBPs have prognostic value in neuroblastoma. For the top putative oncogenic candidates, we analysed their expression in neuroblastoma cell lines, as well as summarised their characteristics and existence of chemical inhibitors. Moreover, to help explain their association with neuroblastoma subtypes, we reviewed candidate RBPs’ potential as biomarkers, and their mechanistic roles in neuronal and cancer contexts. We found several highly significant RBPs including RPL22L1, RNASEH2A, PTRH2, MRPL11 and AFF2, which remain uncharacterised in neuroblastoma. Although not all RBPs appear suitable for drug design, or carry prognostic significance, we show that several RBPs have strong rationale for inhibition and mechanistic studies, representing an alternative, but nonetheless promising therapeutic strategy in neuroblastoma treatment.
Collapse
|
11
|
Shimizu T, Nishio K, Sakai K, Okamoto I, Okamoto K, Takeda M, Morishita M, Nakagawa K. Phase I safety and pharmacokinetic study of YM155, a potent selective survivin inhibitor, in combination with erlotinib in patients with EGFR TKI refractory advanced non-small cell lung cancer. Cancer Chemother Pharmacol 2020; 86:211-219. [PMID: 32638093 DOI: 10.1007/s00280-020-04112-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 06/30/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE This phase I study was conducted to evaluate the safety and pharmacokinetics of YM155, a potent, selective survivin inhibitor, in combination with erlotinib in patients with EGFR TKI refractory advanced non-small cell lung cancer (NSCLC). METHODS The pimary objectives were to evaluate the safety and tolerability of YM155 at escalating doses (3.6, 4.8, 6.0, and 8.0 mg/m2/days) administered every 3 weeks as continuous intravenous infusion over 168 h in combination with erlotinib at a fixed dose (150 mg, once a day). Secondary objectives were to assess the pharmacokinetics of YM155, antitumor activity, and the relationship between biomarkers and efficacy. The changes in survivin expression in biopsied tumor pre- and post-YM155 administration and serum cytokine levels were also analyzed. RESULTS Fifteen patients were treated. The most common YM155-related adverse event was the presence of urine microalbumin, whereas grades 3/4 toxicities were rare. One patient who received 4.8 mg/m2/days YM155 developed a dose-limiting grade 2 serum creatinine elevation. YM155 exposure in plasma showed dose proportionality across all dose ranges tested. No pharmacokinetic interaction occurred between YM155 and erlotinib. The serum cytokines IL-8, G-CSF, and MIP-1b showed decreasing trends in patients who achieved progression-free survival of ≥ 12 weeks. Durable stable disease for ≥ 24 weeks was observed in two patients. CONCLUSION Up to 8.0 mg/m2/days YM155 administered every 3 weeks in combination with erlotinib exhibited a favorable safety profile and moderate clinical efficacy. These results suggest that inhibiting survivin is a potential therapeutic strategy for select patients with EGFR TKI refractory NSCLC. TRIAL REGISTRATION UMIN000031912 at UMIN Clinical Trials Registry (UMIN-CTR).
Collapse
Affiliation(s)
- Toshio Shimizu
- Department of Medical Oncology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama City, Osaka, 5898511, Japan. .,Department of Experimental Therapeutics (Early Phase 1 Drug Development Service), National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 1040045, Japan.
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka, 5898511, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka, 5898511, Japan
| | - Isamu Okamoto
- Department of Medical Oncology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama City, Osaka, 5898511, Japan.,Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 8128582, Japan
| | - Kunio Okamoto
- Department of Medical Oncology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama City, Osaka, 5898511, Japan.,Department of Medical Oncology, Kagawa Prefectural Central Hospital, Kagawa, 7608557, Japan
| | - Masayuki Takeda
- Department of Medical Oncology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama City, Osaka, 5898511, Japan
| | - Maiko Morishita
- Division of Clinical Development, Astellas Pharma Inc., 2-5-1 Nihonbashi-Honcho, Chuo-ku, Tokyo, 1038411, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama City, Osaka, 5898511, Japan
| |
Collapse
|
12
|
Liu Y, Wang JX, Nie ZY, Wen Y, Jia XJ, Zhang LN, Duan HJ, Shi YH. Upregulation of ERp57 promotes clear cell renal cell carcinoma progression by initiating a STAT3/ILF3 feedback loop. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:439. [PMID: 31747963 PMCID: PMC6864981 DOI: 10.1186/s13046-019-1453-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 10/16/2019] [Indexed: 01/09/2023]
Abstract
Background ERp57 dysfunction has been shown to contribute to tumorigenesis in multiple malignances. However, the role of ERp57 in clear cell renal carcinoma (ccRCC) remains unclear. Methods Cell proliferation ability was measured by MTT and colony forming assays. Western blotting and quantitative real-time PCR (qRT-PCR) were performed to measure protein and mRNA expression. Co-immunoprecipitation (CoIP) and proximity ligation assay (PLA) were performed to detect protein-protein interaction. Chromatin immunoprecipitation (ChIP), ribonucleoprotein immunoprecipitation (RIP), and oligo pull-down were used to confirm DNA–protein and RNA–protein interactions. Promoter luciferase analysis was used to detect transcription factor activity. Results Here we found ERp57 was overexpressed in ccRCC tissues, and the higher levels of ERp57 were correlated with poor survival in patients with ccRCC. In vivo and in vitro experiments showed that ccRCC cell proliferation was enhanced by ERp57 overexpression and inhibited by ERp57 deletion. Importantly, we found ERp57 positively regulated ILF3 expression in ccRCC cells. Mechanically, ERp57 was shown to bind to STAT3 protein and enhance the STAT3-mediated transcriptional activity of ILF3. Furthermore, ILF3 levels were increased in ccRCC tissues and associated with poor prognosis. Interestingly, we revealed that ILF3 could bind to ERp57 and positively regulate its expression by enhancing its mRNA stability. Furthermore, ccRCC cell proliferation was moderated via the ERp57/STAT3/ILF3 feedback loop. Conclusions In summary, our results indicate that the ERp57/STAT3/ILF3 feedback loop plays a key role in the oncogenesis of ccRCC and provides a potential therapeutic target for ccRCC treatment.
Collapse
Affiliation(s)
- Yan Liu
- Department of Pathology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China.,Department of Anesthesiology, The 4th Hospital of Hebei Medical University, 169 Tianshan Street , 050000, Shijiazhuang, People's Republic of China
| | - Jian-Xing Wang
- Department of Pathology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China.,Department of Otolaryngology, The Second Hospital of Hebei Medical University, 215 Heping West Road Shijiazhuang, 050000, Shijiazhuang, People's Republic of China
| | - Zi-Yuan Nie
- Department of Hematology, The Second Hospital of Hebei Medical University, 215 Heping West Road Shijiazhuang, 050000, Shijiazhuang, People's Republic of China
| | - Yue Wen
- Department of Pathology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Xin-Ju Jia
- Department of Endocrinology, The First Hospital of Hebei Medical University, 89 Donggang Road Shijiazhuang, 050000, Shijiazhuang, People's Republic of China
| | - Li-Na Zhang
- Department of Pathology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Hui-Jun Duan
- Department of Pathology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China.
| | - Yong-Hong Shi
- Department of Pathology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China.
| |
Collapse
|
13
|
Sepantronium Bromide (YM155), A Small Molecule Survivin Inhibitor, Promotes Apoptosis by Induction of Oxidative Stress, Worsens the Behavioral Deficits and Develops an Early Model of Toxic Demyelination: In Vivo and In-Silico Study. Neurochem Res 2019; 44:2482-2498. [DOI: 10.1007/s11064-019-02865-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/21/2019] [Accepted: 08/23/2019] [Indexed: 12/14/2022]
|
14
|
Li F, Aljahdali I, Ling X. Cancer therapeutics using survivin BIRC5 as a target: what can we do after over two decades of study? JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:368. [PMID: 31439015 PMCID: PMC6704566 DOI: 10.1186/s13046-019-1362-1] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023]
Abstract
Survivin (also named BIRC5) is a well-known cancer therapeutic target. Since its discovery more than two decades ago, the use of survivin as a target for cancer therapeutics has remained a central goal of survivin studies in the cancer field. Many studies have provided intriguing insight into survivin's functional role in cancers, thus providing promise for survivin as a cancer therapeutic target. Despite this, moving survivin-targeting agents into and through the clinic remains a challenge. In order to address this challenge, we may need to rethink current strategies in order to develop a new mindset for targeting survivin. In this Review, we will first summarize the current survivin mechanistic studies, and then review the status of survivin cancer therapeutics, which is classified into five categories: (i) survivin-partner protein interaction inhibitors, (ii) survivin homodimerization inhibitors, (iii) survivin gene transcription inhibitors, (iv) survivin mRNA inhibitors and (v) survivin immunotherapy. We will then provide our opinions on cancer therapeutics using survivin as a target, with the goal of stimulating discussion that might facilitate translational research for discovering improved strategies and/or more effective anticancer agents that target survivin for cancer therapy.
Collapse
Affiliation(s)
- Fengzhi Li
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York, 14263, USA. .,Developmental Therapeutics Program, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York, 14263, USA.
| | - Ieman Aljahdali
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York, 14263, USA.,Department of Cellular & Molecular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York, 14263, USA
| | - Xiang Ling
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York, 14263, USA.,Canget BioTekpharma LLC, Buffalo, New York, USA
| |
Collapse
|
15
|
Mika A, Luelling SE, Pavek A, Nartker C, Heyneman AL, Jones KB, Barrott JJ. Epigenetic Changes at the Birc5 Promoter Induced by YM155 in Synovial Sarcoma. J Clin Med 2019; 8:jcm8030408. [PMID: 30909651 PMCID: PMC6463023 DOI: 10.3390/jcm8030408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 12/21/2022] Open
Abstract
YM155 is an anti-cancer therapy that has advanced into 11 different human clinical trials to treat various cancers. This apoptosis-inducing therapy indirectly affects the protein levels of survivin (gene: Birc5), but the molecular underpinnings of the mechanism remain largely unknown. Synovial sarcoma is a rare soft-tissue malignancy with high protein expression of survivin. We investigated whether YM155 would be a viable therapeutic option to treat synovial sarcoma. YM155 therapy was applied to human synovial sarcoma cell lines and to a genetically engineered mouse model of synovial sarcoma. We discovered that YM155 exhibited nanomolar potency against human synovial sarcoma cell lines and the treated mice with synovial sarcoma demonstrated a 50% reduction in tumor volume compared to control treated mice. We further investigated the mechanism of action of YM155 by looking at the change of lysine modifications of the histone tails that were within 250 base pairs of the Birc5 promoter. Using chromatin immunoprecipitation (ChIP)-qPCR, we discovered that the histone epigenetic marks of H3K27 for the Birc5 promoter changed upon YM155 treatment. H3K27me3 and H3K27ac increased, but the net result was decreased Birc5/survivin expression. Furthermore, the combination of molecular events resulted in caspase 3/7/8 upregulation and death of the sarcoma cells.
Collapse
Affiliation(s)
- Aleksander Mika
- Departments of Orthopedics and Oncological Sciences, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| | - Sarah E Luelling
- Department of Biomedical and Pharmaceutical Sciences, Idaho State University, Pocatello, ID 83209, USA.
| | - Adriene Pavek
- Department of Biomedical and Pharmaceutical Sciences, Idaho State University, Pocatello, ID 83209, USA.
| | - Christopher Nartker
- Department of Biomedical and Pharmaceutical Sciences, Idaho State University, Pocatello, ID 83209, USA.
| | - Alexandra L Heyneman
- Department of Biomedical and Pharmaceutical Sciences, Idaho State University, Pocatello, ID 83209, USA.
| | - Kevin B Jones
- Departments of Orthopedics and Oncological Sciences, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| | - Jared J Barrott
- Departments of Orthopedics and Oncological Sciences, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
- Department of Biomedical and Pharmaceutical Sciences, Idaho State University, Pocatello, ID 83209, USA.
| |
Collapse
|
16
|
Xin X, Wu Y, Zang R, Yang ST. A fluorescent 3D cell culture assay for high throughput screening of cancer drugs down-regulating survivin. J Biotechnol 2019; 289:80-87. [DOI: 10.1016/j.jbiotec.2018.11.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 11/09/2018] [Accepted: 11/21/2018] [Indexed: 12/13/2022]
|
17
|
Canine squamous cell carcinoma cell lines with high expression of survivin are sensitive to survivin inhibitor YM155. Vet J 2018; 240:31-36. [DOI: 10.1016/j.tvjl.2018.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 08/30/2018] [Accepted: 09/02/2018] [Indexed: 01/06/2023]
|
18
|
Sanglard LP, Nascimento M, Moriel P, Sommer J, Ashwell M, Poore MH, Duarte MDS, Serão NVL. Impact of energy restriction during late gestation on the muscle and blood transcriptome of beef calves after preconditioning. BMC Genomics 2018; 19:702. [PMID: 30253751 PMCID: PMC6156876 DOI: 10.1186/s12864-018-5089-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 09/19/2018] [Indexed: 02/03/2023] Open
Abstract
Background Maternal nutrition has been highlighted as one of the main factors affecting intra-uterine environment. The increase in nutritional requirements by beef cows during late gestation can cause nutritional deficiency in the fetus and impact the fetal regulation of genes associated with myogenesis and immune response. Methods Forty days before the expected calving date, cows were assigned to one of two diets: 100% (control) or 70% (restricted group) of the daily energy requirement. Muscle samples were collected from 12 heifers and 12 steers, and blood samples were collected from 12 steers. The objective of this work was to identify and to assess the biological relevance of differentially expressed genes (DEG) in the skeletal muscle and blood of beef calves born from cows that experienced [or not] a 30% energy restriction during the last 40 days of gestation. Results A total of 160, 164, and 346 DEG (q-value< 0.05) were identified in the skeletal muscle for the effects of diet, sex, and diet-by-sex interaction, respectively. For blood, 452, 1392, and 155 DEG were identified for the effects of diet, time, and diet-by-time interaction, respectively. For skeletal muscle, results based on diet identified genes involved in muscle metabolism. In muscle, from the 10 most DEG down-regulated in the energy-restricted group (REST), we identified 5 genes associated with muscle metabolism and development: SLCO3A1, ATP6V0D1, SLC2A1, GPC4, and RASD2. In blood, among the 10 most DEG, we found genes related to response to stress up-regulated in the REST after weaning, such as SOD3 and INO80D, and to immune response down-regulated in the REST after vaccination, such as OASL, KLRF1, and LOC104968634. Conclusion In conclusion, maternal energy restriction during late gestation may limit the expression of genes in the muscle and increase expression in the blood of calves. In addition, enrichment analysis showed that a short-term maternal energy restriction during pregnancy affects the expression of genes related to energy metabolism and muscle contraction, and immunity and stress response in the blood. Therefore, alterations in the intra-uterine environment can modify prenatal development with lasting consequences to adult life. Electronic supplementary material The online version of this article (10.1186/s12864-018-5089-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Leticia P Sanglard
- Department of Animal Science, Iowa State University, Ames, 50011, USA.,Department of Animal Science, North Carolina State University, Raleigh, 27695, USA
| | - Moysés Nascimento
- Department of Animal Science, North Carolina State University, Raleigh, 27695, USA.,Department of Statistics, Universidade Federal de Viçosa, Viçosa, 36570-000, Brazil
| | - Philipe Moriel
- Range Cattle Research and Education Center, University of Florida, Ona, Florida, 33865, USA
| | - Jeffrey Sommer
- Department of Animal Science, North Carolina State University, Raleigh, 27695, USA
| | - Melissa Ashwell
- Department of Animal Science, North Carolina State University, Raleigh, 27695, USA
| | - Matthew H Poore
- Department of Animal Science, North Carolina State University, Raleigh, 27695, USA
| | - Márcio de S Duarte
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, 36570-000, Brazil.,Instituto Nacional de Ciência e Tecnologia - Ciência Animal, Viçosa, 36570-000, Brazil
| | - Nick V L Serão
- Department of Animal Science, Iowa State University, Ames, 50011, USA. .,Department of Animal Science, North Carolina State University, Raleigh, 27695, USA.
| |
Collapse
|
19
|
Li D, Chen Y, Mei H, Jiao W, Song H, Ye L, Fang E, Wang X, Yang F, Huang K, Zheng L, Tong Q. Ets-1 promoter-associated noncoding RNA regulates the NONO/ERG/Ets-1 axis to drive gastric cancer progression. Oncogene 2018; 37:4871-4886. [PMID: 29773901 PMCID: PMC6117270 DOI: 10.1038/s41388-018-0302-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 03/20/2018] [Accepted: 04/16/2018] [Indexed: 01/12/2023]
Abstract
Emerging studies have indicated the essential functions of long noncoding RNAs (lncRNAs) during cancer progression. However, whether lncRNAs contribute to the upregulation of v-ets erythroblastosis virus E26 oncogene homolog 1 (Ets-1), an established oncogenic protein facilitating tumor invasion and metastasis, in gastric cancer remains elusive. Herein, we identified Ets-1 promoter-associated noncoding RNA (pancEts-1) as a novel lncRNA associated with the gastric cancer progression via mining of publicly available datasets and rapid amplification of cDNA ends. RNA pull-down, RNA immunoprecipitation, in vitro binding, and RNA electrophoretic mobility shift assays indicated the binding of pancEts-1 to non-POU domain containing octamer binding (NONO) protein. Mechanistically, pancEts-1 facilitated the physical interaction between NONO and Ets related gene (ERG), resulting in increased ERG transactivation and transcription of Ets-1 associated with gastric cancer progression. In addition, pancEts-1 facilitated the growth and aggressiveness of gastric cancer cells via interacting with NONO. In gastric cancer tissues, pancEts-1, NONO, and ERG were upregulated and significantly correlated with Ets-1 levels. High levels of pancEts-1, NONO, ERG, or Ets-1 were respectively associated with poor survival of gastric cancer patients, whereas simultaneous expression of all of them (HR = 3.012, P = 0.105) was not an independent prognostic factor for predicting clinical outcome. Overall, these results demonstrate that lncRNA pancEts-1 exhibits oncogenic properties that drive the progression of gastric cancer via regulating the NONO/ERG/Ets-1 axis.
Collapse
Affiliation(s)
- Dan Li
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei Province, China
| | - Yajun Chen
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei Province, China
| | - Hong Mei
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei Province, China
| | - Wanju Jiao
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei Province, China
| | - Huajie Song
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei Province, China
| | - Lin Ye
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei Province, China
| | - Erhu Fang
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei Province, China
| | - Xiaojing Wang
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei Province, China
| | - Feng Yang
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei Province, China
| | - Kai Huang
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei Province, China
| | - Liduan Zheng
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei Province, China. .,Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei Province, China.
| | - Qiangsong Tong
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei Province, China. .,Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei Province, China.
| |
Collapse
|
20
|
YM155 sensitizes TRAIL-induced apoptosis through cathepsin S-dependent down-regulation of Mcl-1 and NF-κB-mediated down-regulation of c-FLIP expression in human renal carcinoma Caki cells. Oncotarget 2018; 7:61520-61532. [PMID: 27528031 PMCID: PMC5308669 DOI: 10.18632/oncotarget.11137] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/27/2016] [Indexed: 11/25/2022] Open
Abstract
YM155, a small-molecule survivin inhibitor, has been reported for its anti-cancer activity in various cancer cells. In this study, we investigated the effect of YM155 to enhance TRAIL-mediated apoptosis in human renal carcinoma cells. We found that YM155 alone had no effect on apoptosis, however, combined treatment with YM155 and TRAIL markedly induced apoptosis in human renal carcinoma cells (Caki, ACHN, and A498), breast cancer cells (MDA-MB231), and glioma cells (U251MG), but not normal cells [mesangial cell (MC) and human skin fibroblast (HSF)]. YM155 induced down-regulation of Mcl-1 expression at the post-translational levels, and the overexpression of Mcl-1 markedly inhibited YM155 plus TRAIL-induced apoptosis. Furthermore, YM155 induced down-regulation of c-FLIP mRNA expression through inhibition of NF-κB transcriptional activity. Ectopic expression of c-FLIP markedly blocked YM155-induced TRAIL sensitization. Taken together, our results suggested that YM155 sensitizes TRAIL-mediated apoptosis via down-regulation of Mcl-1 and c-FLIP expression in renal carcinoma Caki cells.
Collapse
|
21
|
Falkenhorst J, Grunewald S, Mühlenberg T, Marino-Enriquez A, Reis AC, Corless C, Heinrich M, Treckmann J, Podleska LE, Schuler M, Fletcher JA, Bauer S. Inhibitor of Apoptosis Proteins (IAPs) are commonly dysregulated in GIST and can be pharmacologically targeted to enhance the pro-apoptotic activity of imatinib. Oncotarget 2018; 7:41390-41403. [PMID: 27167336 PMCID: PMC5173067 DOI: 10.18632/oncotarget.9159] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 04/14/2016] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal stromal tumors (GIST) exhibit a strong oncogenic dependency on KIT and KIT inhibitors confer long lasting disease stabilization in the majority of patients. Nonetheless, KIT inhibition alone does not cure GIST as a subset of GIST cells evade apoptosis and eventually develop resistance. Inhibitors of Apoptosis Proteins (IAPs) may confer resistance to drug-induced apoptosis. We observed that the mRNA and protein of IAPs XIAP (BIRC4) and survivin (BIRC5) were highly expressed in primary GIST tumors and cell line models. Amplification of the respective gene loci (BIRC2, BIRC3, BIRC4, BIRC5) was detected in 47% of GIST studied by SNP arrays. Whole exome analyses revealed a mutation of SMAC(DIABLO) in a heavily pretreated patient. Both, survivin (rank 62-92/11.194 tested proteins) and XIAP (rank 106-557/11.194) were found to be essential proteins for survival in a synthetic lethality screen. Expression of XIAP and survivin decreased upon KIT inhibition and may play a role in KIT-regulated pro-survival signaling. SMAC-mimetic treatment with LCL161 and TL32711 reduced cIAP1 and XIAP expression. Survivin inhibitor YM155 lead to transcriptional repression of BIRC5/survivin (YM155) and induced apoptosis. Combinational treatment with KIT inhibitors (imatinib, regorafenib) enhanced the proapoptotic effect. These findings support the combination of KIT inhibition with IAP antagonists in GIST.
Collapse
Affiliation(s)
- Johanna Falkenhorst
- Sarcoma Center, Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Susanne Grunewald
- Sarcoma Center, Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Thomas Mühlenberg
- Sarcoma Center, Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | | | - Anna-Carina Reis
- Sarcoma Center, Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Pathology and Neuropathology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Christopher Corless
- Department of Pathology, Oregon Health and Science University Knight Cancer Institute, Portland, OR, USA
| | - Michael Heinrich
- Department of Medical Oncology, Oregon Health and Science University Knight Cancer Institute, Portland, OR, USA
| | - Jürgen Treckmann
- Sarcoma Center, Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Surgery, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Lars Erik Podleska
- Sarcoma Center, Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Surgery, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Martin Schuler
- Sarcoma Center, Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | | | - Sebastian Bauer
- Sarcoma Center, Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
22
|
Dizdar L, Jünemann LM, Werner TA, Verde PE, Baldus SE, Stoecklein NH, Knoefel WT, Krieg A. Clinicopathological and functional implications of the inhibitor of apoptosis proteins survivin and XIAP in esophageal cancer. Oncol Lett 2018; 15:3779-3789. [PMID: 29467895 DOI: 10.3892/ol.2018.7755] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 11/21/2017] [Indexed: 12/17/2022] Open
Abstract
Based on their overexpression and important roles in progression and therapy-resistance in malignant diseases, the inhibitor of apoptosis protein family (IAP) members, survivin and X-linked inhibitor of apoptosis protein (XIAP), represent attractive candidates for targeted therapy. The present study investigated the prognostic and biological relevance of survivin and XIAP in esophageal squamous-cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC). Survivin and XIAP expression was analyzed by immunohistochemistry using tissue microarrays containing 120 ESCC and 90 EAC samples as well as the corresponding non-neoplastic esophageal mucosa samples. IAP expression levels were then correlated to clinicopathological parameters and overall survival to identify any associations. In addition, esophageal cancer cell lines were treated with the survivin inhibitor YM155, and the XIAP inhibitors Birinapant and GDC-0152 in vitro. Survivin and XIAP expression were significantly increased in EAC and ESCC when compared with tumor-adjacent mucosa. In patients with ESCC XIAP expression was associated with female gender and advanced tumor stages, and nuclear survivin expression was associated with poor grading. High XIAP expression was identified as an independent negative prognostic marker in ESCC. By contrast, XIAP inhibitors did not affect cancer cell viability in vitro, and the small molecule survivin inhibitor YM155 significantly reduced cell viability and proliferation in esophageal cancer cell lines. Western blot analysis revealed a dose dependent decrease of survivin accompanied by an increased poly (adenosine diphosphate-ribose) polymerase cleavage following YM155 treatment. These findings underline the potential role of survivin and XIAP in the oncogenesis of esophageal cancer and provide a rationale for future clinical studies investigating the therapeutic efficacy of IAP directed therapies in patients with esophageal cancer.
Collapse
Affiliation(s)
- Levent Dizdar
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Düsseldorf, D-40225 Düsseldorf, Germany
| | - Lisa M Jünemann
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Düsseldorf, D-40225 Düsseldorf, Germany
| | - Thomas A Werner
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Düsseldorf, D-40225 Düsseldorf, Germany
| | - Pablo E Verde
- Coordination Centre for Clinical Trials, Heinrich-Heine-University and University Hospital Düsseldorf, D-40225 Düsseldorf, Germany
| | - Stephan E Baldus
- Institute of Pathology, Cytology and Molecular Pathology, D-51465 Bergisch Gladbach, Germany
| | - Nikolas H Stoecklein
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Düsseldorf, D-40225 Düsseldorf, Germany
| | - Wolfram T Knoefel
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Düsseldorf, D-40225 Düsseldorf, Germany
| | - Andreas Krieg
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Düsseldorf, D-40225 Düsseldorf, Germany
| |
Collapse
|
23
|
Wei Q, Li J, Tang F, Yin Y, Zhao Y, Yao Q. Synthesis and biological evaluation of novel 2-arylvinyl-substituted naphtho[2,3-d]imidazolium halide derivatives as potent antitumor agents. Eur J Med Chem 2017; 144:504-516. [PMID: 29288947 DOI: 10.1016/j.ejmech.2017.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 12/02/2017] [Accepted: 12/02/2017] [Indexed: 11/27/2022]
Abstract
Two series of novel 2-arylvinyl-naphtho[2,3-d]imidazol-3-ium iodide derivatives and 2-arylvinyl-naphtho[2,3-d]imidazol-3-ium bromide derivatives were designed and synthesized by the structural combination of YM155 with stilbenoids. All compounds were tested for anti-proliferative activity against PC-3, A375 and HeLa human cancer cell lines. Two of the compounds were selected for further investigation: 12b, which showed potent cytotoxicity against the three tested cell lines with IC50 values in the range of 0.06-0.21 μM, and 7l, which displayed excellent selectivity for PC-3 cells with an IC50 of only 22 nM. Western blot analysis results indicated that both 12b and 7l suppress the expression of Bcl-2 and Survivin proteins, which helps induce apoptosis. As determined by the percent of Annexin V-FITC-positive apoptotic cells, 12b was not only significantly more effective than 7l at a concentration of 100 nM in PC-3 cells but also induced apoptosis in a dose-dependent manner with more potency than 7l at a concentration of 1000 nM in A375 cells. Therefore, compound 12b was chosen for further in-depth studies investigating the mechanism of apoptosis. The results showed that it could activate caspase-3, hydrolyze PARP, and even inactivate ERK. Moreover, 12b arrested A375 cells at S phase in a time-dependent and dose-dependent manner, while having a visible effect on microtubule dynamics. In addition, (E)-2-(2-(1H-indol-3-yl)vinyl)-1-benzyl-3-(2-methoxyethyl)-4,9-dioxo-4,9-dihydro-1H-naphtho[2,3-d]imidazol-3-ium bromide (12b) exhibited significant antitumor activity when evaluated in a subcutaneous solid tumor model. Our study reveals that 2-arylvinyl-substituted naphtho[2,3-d]imidazolium scaffolding is a promising new entity for the development of multi-target anticancer drugs.
Collapse
Affiliation(s)
- Qingyun Wei
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Ju Li
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China
| | - Feng Tang
- MtC Biopharma, Co., Ltd, Nanjing 210042, PR China
| | - Yin Yin
- MtC Biopharma, Co., Ltd, Nanjing 210042, PR China
| | - Yong Zhao
- MtC Biopharma, Co., Ltd, Nanjing 210042, PR China.
| | - Qizheng Yao
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
24
|
Ookura M, Fujii T, Yagi H, Ogawa T, Kishi S, Hosono N, Shigemi H, Yamauchi T, Ueda T, Yoshida A. YM155 exerts potent cytotoxic activity against quiescent (G 0/G 1) multiple myeloma and bortezomib resistant cells via inhibition of survivin and Mcl-1. Oncotarget 2017; 8:111535-111550. [PMID: 29340073 PMCID: PMC5762341 DOI: 10.18632/oncotarget.22871] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 11/13/2017] [Indexed: 12/25/2022] Open
Abstract
YM155, a novel small molecule inhibitor of survivin, shows broad anticancer activity. Here, we have focused on the cytotoxic activity of YM155 against multiple myeloma (MM) including cytokinetically quiescent (G0/G1) cells and bortezomib resistant cells. YM155 strongly inhibited the growth of MM cell lines with the IC50 value of below 10 nM. YM155 also showed potent anti-myeloma activity in mouse xenograft model. YM155 suppressed the expression of survivin and rapidly directed Mcl-1 protein for proteasome degradation. YM155 abrogated the interleukin-6-induced STAT3 phosphorylation, subsequently blocked Mcl-1 expression and induced apoptosis in MM cells. Triple-color flow cytometric analysis revealed that YM155 potently induced cell death of MM cells in G0 phase. Quiescent primary MM cells were also sensitive to YM155. We established bortezomib-resistant MM cell line, U266/BTZR1, which possess a point mutation G322A. YM155 exhibited similar cytotoxic potency against U266/BTZR1 compared with parental cells. Interestingly, survivin expression was markedly elevated in U266/BTZR1 cells. Treatment with YM155 significantly down-regulated this increased survivin and Mcl-1 expression in U266/BTZR1 cells. In conclusion, our data indicate that YM155 exhibits potent cytotoxicity against quiescent (G0/G1) MM cells and bortezomib-resistant cells. These unique features of YM155 may be beneficial for the development of new therapeutic strategies to eliminate quiescent MM cells and overcome bortezomib resistance.
Collapse
Affiliation(s)
- Miyuki Ookura
- Department of Hematology and Oncology, University of Fukui, Matsuoka, Fukui 910-1193, Japan
| | - Tatsuya Fujii
- Department of Hematology and Oncology, University of Fukui, Matsuoka, Fukui 910-1193, Japan
| | - Hideki Yagi
- Department of Pharmaceutical Sciences, International University of Health and Welfare, Otawara, Tochigi 324-8501, Japan
| | - Takuya Ogawa
- Department of Pharmaceutical Sciences, International University of Health and Welfare, Otawara, Tochigi 324-8501, Japan
| | - Shinji Kishi
- Department of Hematology and Oncology, University of Fukui, Matsuoka, Fukui 910-1193, Japan
| | - Naoko Hosono
- Department of Hematology and Oncology, University of Fukui, Matsuoka, Fukui 910-1193, Japan
| | - Hiroko Shigemi
- Department of Hematology and Oncology, University of Fukui, Matsuoka, Fukui 910-1193, Japan
| | - Takahiro Yamauchi
- Department of Hematology and Oncology, University of Fukui, Matsuoka, Fukui 910-1193, Japan
| | - Takanori Ueda
- Department of Hematology and Oncology, University of Fukui, Matsuoka, Fukui 910-1193, Japan
| | - Akira Yoshida
- Department of Hematology, International University of Health and Welfare Hospital, Iguchi, Nasushiobara, Tochigi, 329-2763, Japan
| |
Collapse
|
25
|
Mazur J, Roy K, Kanwar JR. Recent advances in nanomedicine and survivin targeting in brain cancers. Nanomedicine (Lond) 2017; 13:105-137. [PMID: 29161215 DOI: 10.2217/nnm-2017-0286] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Brain cancer is a highly lethal disease, especially devastating toward both the elderly and children. This cancer has no therapeutics available to combat it, predominately due to the blood-brain barrier (BBB) preventing treatments from maintaining therapeutic levels within the brain. Recently, nanoparticle technology has entered the forefront of cancer therapy due to its ability to deliver therapeutic effects while potentially passing physiological barriers. Key nanoparticles for brain cancer treatment include glutathione targeted PEGylated liposomes, gold nanoparticles, superparamagnetic iron oxide nanoparticles and nanoparticle-albumin bound drugs, with these being discussed throughout this review. Recently, the survivin protein has gained attention as it is over-expressed in a majority of tumors. This review will briefly discuss the properties of survivin, while focusing on how both nanoparticles and survivin-targeting treatments hold potential as brain cancer therapies. This review may provide useful insight into new brain cancer treatment options, particularly survivin inhibition and nanomedicine.
Collapse
Affiliation(s)
- Jake Mazur
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research, Centre for Molecular and Medical Research (CMMR), School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong VIC 3217, Australia
| | - Kislay Roy
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research, Centre for Molecular and Medical Research (CMMR), School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong VIC 3217, Australia
| | - Jagat R Kanwar
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research, Centre for Molecular and Medical Research (CMMR), School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong VIC 3217, Australia
| |
Collapse
|
26
|
Chia S, Low JL, Zhang X, Kwang XL, Chong FT, Sharma A, Bertrand D, Toh SY, Leong HS, Thangavelu MT, Hwang JSG, Lim KH, Skanthakumar T, Tan HK, Su Y, Hui Choo S, Hentze H, Tan IBH, Lezhava A, Tan P, Tan DSW, Periyasamy G, Koh JLY, Gopalakrishna Iyer N, DasGupta R. Phenotype-driven precision oncology as a guide for clinical decisions one patient at a time. Nat Commun 2017; 8:435. [PMID: 28874669 PMCID: PMC5585361 DOI: 10.1038/s41467-017-00451-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 06/30/2017] [Indexed: 12/22/2022] Open
Abstract
Genomics-driven cancer therapeutics has gained prominence in personalized cancer treatment. However, its utility in indications lacking biomarker-driven treatment strategies remains limited. Here we present a "phenotype-driven precision-oncology" approach, based on the notion that biological response to perturbations, chemical or genetic, in ex vivo patient-individualized models can serve as predictive biomarkers for therapeutic response in the clinic. We generated a library of "screenable" patient-derived primary cultures (PDCs) for head and neck squamous cell carcinomas that reproducibly predicted treatment response in matched patient-derived-xenograft models. Importantly, PDCs could guide clinical practice and predict tumour progression in two n = 1 co-clinical trials. Comprehensive "-omics" interrogation of PDCs derived from one of these models revealed YAP1 as a putative biomarker for treatment response and survival in ~24% of oral squamous cell carcinoma. We envision that scaling of the proposed PDC approach could uncover biomarkers for therapeutic stratification and guide real-time therapeutic decisions in the future.Treatment response in patient-derived models may serve as a biomarker for response in the clinic. Here, the authors use paired patient-derived mouse xenografts and patient-derived primary culture models from head and neck squamous cell carcinomas, including metastasis, as models for high-throughput screening of anti-cancer drugs.
Collapse
Affiliation(s)
- Shumei Chia
- Genome Institute of Singapore, A*STAR, Cancer Therapeutics & Stratified Oncology, PerkinElmer-GIS Centre for Precision Oncology, 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore
| | - Joo-Leng Low
- Genome Institute of Singapore, A*STAR, Cancer Therapeutics & Stratified Oncology, PerkinElmer-GIS Centre for Precision Oncology, 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore
| | - Xiaoqian Zhang
- Genome Institute of Singapore, A*STAR, Cancer Therapeutics & Stratified Oncology, PerkinElmer-GIS Centre for Precision Oncology, 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore
| | - Xue-Lin Kwang
- National Cancer Centre Singapore, Cancer Therapeutics Research Laboratory, 11 Hospital Drive, Singapore, 169610, Singapore
| | - Fui-Teen Chong
- National Cancer Centre Singapore, Cancer Therapeutics Research Laboratory, 11 Hospital Drive, Singapore, 169610, Singapore
| | - Ankur Sharma
- Genome Institute of Singapore, A*STAR, Cancer Therapeutics & Stratified Oncology, PerkinElmer-GIS Centre for Precision Oncology, 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore
| | - Denis Bertrand
- Genome Institute of Singapore, A*STAR, Cancer Therapeutics & Stratified Oncology, PerkinElmer-GIS Centre for Precision Oncology, 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore
| | - Shen Yon Toh
- National Cancer Centre Singapore, Cancer Therapeutics Research Laboratory, 11 Hospital Drive, Singapore, 169610, Singapore
| | - Hui-Sun Leong
- National Cancer Centre Singapore, Cancer Therapeutics Research Laboratory, 11 Hospital Drive, Singapore, 169610, Singapore
| | - Matan T Thangavelu
- Genome Institute of Singapore, A*STAR, Cancer Therapeutics & Stratified Oncology, PerkinElmer-GIS Centre for Precision Oncology, 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore
| | - Jacqueline S G Hwang
- Department of Anatomical Pathology, Singapore General Hospital, Outram Road, Singapore, 169608, Singapore
| | - Kok-Hing Lim
- Department of Anatomical Pathology, Singapore General Hospital, Outram Road, Singapore, 169608, Singapore
| | - Thakshayeni Skanthakumar
- National Cancer Centre Singapore, Cancer Therapeutics Research Laboratory, 11 Hospital Drive, Singapore, 169610, Singapore
| | - Hiang-Khoon Tan
- Department of Anatomical Pathology, Singapore General Hospital, Outram Road, Singapore, 169608, Singapore
| | - Yan Su
- Genome Institute of Singapore, A*STAR, Cancer Therapeutics & Stratified Oncology, PerkinElmer-GIS Centre for Precision Oncology, 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore
| | - Siang Hui Choo
- Genome Institute of Singapore, A*STAR, Cancer Therapeutics & Stratified Oncology, PerkinElmer-GIS Centre for Precision Oncology, 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore
| | - Hannes Hentze
- Biological Resource Centre (BRC), A*STAR, 20 Biopolis Way, #07-01 Centros, Singapore, 138668, Singapore
| | - Iain B H Tan
- Genome Institute of Singapore, A*STAR, Cancer Therapeutics & Stratified Oncology, PerkinElmer-GIS Centre for Precision Oncology, 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore
- National Cancer Centre Singapore, Cancer Therapeutics Research Laboratory, 11 Hospital Drive, Singapore, 169610, Singapore
| | - Alexander Lezhava
- Genome Institute of Singapore, A*STAR, Cancer Therapeutics & Stratified Oncology, PerkinElmer-GIS Centre for Precision Oncology, 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore
| | - Patrick Tan
- Genome Institute of Singapore, A*STAR, Cancer Therapeutics & Stratified Oncology, PerkinElmer-GIS Centre for Precision Oncology, 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore
| | - Daniel S W Tan
- National Cancer Centre Singapore, Cancer Therapeutics Research Laboratory, 11 Hospital Drive, Singapore, 169610, Singapore
| | - Giridharan Periyasamy
- Genome Institute of Singapore, A*STAR, Cancer Therapeutics & Stratified Oncology, PerkinElmer-GIS Centre for Precision Oncology, 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore
| | - Judice L Y Koh
- Genome Institute of Singapore, A*STAR, Cancer Therapeutics & Stratified Oncology, PerkinElmer-GIS Centre for Precision Oncology, 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore
| | - N Gopalakrishna Iyer
- National Cancer Centre Singapore, Cancer Therapeutics Research Laboratory, 11 Hospital Drive, Singapore, 169610, Singapore.
| | - Ramanuj DasGupta
- Genome Institute of Singapore, A*STAR, Cancer Therapeutics & Stratified Oncology, PerkinElmer-GIS Centre for Precision Oncology, 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore.
| |
Collapse
|
27
|
Sim MY, Huynh H, Go ML, Yuen JSP. Action of YM155 on clear cell renal cell carcinoma does not depend on survivin expression levels. PLoS One 2017; 12:e0178168. [PMID: 28582447 PMCID: PMC5459331 DOI: 10.1371/journal.pone.0178168] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 05/08/2017] [Indexed: 12/12/2022] Open
Abstract
The dioxonapthoimidazolium YM155 is a survivin suppressant which has been investigated as an anticancer agent in clinical trials. Here, we investigated its growth inhibitory properties on a panel of immortalized and patient derived renal cell carcinoma (RCC) cell lines which were either deficient in the tumour suppressor von Hippel-Lindau (VHL) protein or possessed a functional copy. Neither the VHL status nor the survivin expression levels of these cell lines influenced their susceptibility to growth inhibition by YM155. Of the various RCC lines, the papillary subtype was more resistant to YM155, suggesting that the therapeutic efficacy of YM155 may be restricted to clear cell subtypes. YM155 was equally potent in cells (RCC786.0) in which survivin expression had been stably silenced or overexpressed, implicating a limited reliance on survivin in the mode of action of YM155. A follow-up in-vitro high throughput RNA microarray identified possible targets of YM155 apart from survivin. Selected genes (ID1, FOXO1, CYLD) that were differentially expressed in YM155-sensitive RCC cells and relevant to RCC pathology were validated with real-time PCR and western immunoblotting analyses. Thus, there is corroboratory evidence that the growth inhibitory activity of YM155 in RCC cell lines is not exclusively mediated by its suppression of survivin. In view of the growing importance of combination therapy in oncology, we showed that a combination of YM155 and sorafenib at ½ x IC50 concentrations was synergistic on RCC786.0 cells. However, when tested intraperitoneally on a murine xenograft model derived from a nephrectomised patient with clear cell RCC, a combination of suboptimal doses of both drugs failed to arrest tumour progression. The absence of synergy in vivo highlighted the need to further optimize the dosing schedules of YM155 and sorafenib, as well as their routes of administration. It also implied that the expression of other oncogenic proteins which YM155 may target is either low or absent in this clear cell RCC.
Collapse
Affiliation(s)
- Mei Yi Sim
- Department of Urology, Singapore General Hospital, Republic of Singapore
- * E-mail:
| | - Hung Huynh
- Laboratory of Molecular Endocrinology, Division of Molecular and Cellular Research, National Cancer Centre, Republic of Singapore
| | - Mei Lin Go
- Department of Pharmacy, National University of Singapore, Republic of Singapore
| | | |
Collapse
|
28
|
Peery RC, Liu JY, Zhang JT. Targeting survivin for therapeutic discovery: past, present, and future promises. Drug Discov Today 2017; 22:1466-1477. [PMID: 28577912 DOI: 10.1016/j.drudis.2017.05.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/12/2017] [Accepted: 05/23/2017] [Indexed: 12/11/2022]
Abstract
Survivin, the smallest member of the inhibitor of apoptosis protein (IAP) family, is overexpressed in cells of almost all cancers but not in most normal tissues in adults. Survivin expression is required for cancer cell survival and knocking down its expression or inhibiting its function using molecular approaches results in spontaneous apoptosis. Thus, survivin is an attractive and perhaps ideal target for cancer drug discovery. However, a US Food and Drug Administration (FDA)-approved drug targeting survivin has yet to emerge. In this Foundation Review, we examine and evaluate various strategies that have been used to target survivin and the stages of each survivin inhibitor to help understand this lack of success. We also provide future perspectives moving forward in targeting survivin for drug discovery.
Collapse
Affiliation(s)
- Robert C Peery
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jing-Yuan Liu
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Computer and Information Science, Indiana University Purdue University, Indianapolis, IN 46202, USA
| | - Jian-Ting Zhang
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
29
|
Abstract
Survivin is a protein functionally important for cell division, apoptosis, and possibly, for micro-RNA biogenesis. It is an established marker of malignant cell transformation. In non-malignant conditions, the unique properties of survivin make it indispensable for homeostasis of the immune system. Indeed, it is required for the innate and adaptive immune responses, controlling differentiation and maintenance of CD4+ and CD8+ memory T-cells, and in B cell maturation. Recently, survivin has emerged as an important player in the pathogenesis of autoimmune diseases. Under the conditions of unreserved inflammation, survivin enhances antigen presentation, maintains persistence of autoreactive cells, and supports production of autoantibodies. In this context, survivin takes its place as a diagnostic and prognostic marker in rheumatoid arthritis, psoriasis, systemic sclerosis and pulmonary arterial hypertension, neuropathology and multiple sclerosis, inflammatory bowel diseases and oral lichen planus. In this review, we summarise the knowledge about non-malignant properties of survivin and focus on its engagement in cellular and molecular pathology of autoimmune diseases. The review highlights utility of survivin measures for clinical applications. It provides rational for the survivin inhibiting strategies and presents results of recent reports on survivin inhibition in modern therapies of cancers and autoimmune diseases.
Collapse
|
30
|
Nyquist MD, Corella A, Burns J, Coleman I, Gao S, Tharakan R, Riggan L, Cai C, Corey E, Nelson PS, Mostaghel EA. Exploiting AR-Regulated Drug Transport to Induce Sensitivity to the Survivin Inhibitor YM155. Mol Cancer Res 2017; 15:521-531. [PMID: 28465296 PMCID: PMC5471626 DOI: 10.1158/1541-7786.mcr-16-0315-t] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/03/2016] [Accepted: 12/22/2016] [Indexed: 12/12/2022]
Abstract
Androgen receptor (AR) signaling is fundamental to prostate cancer and is the dominant therapeutic target in metastatic disease. However, stringent androgen deprivation therapy regimens decrease quality of life and have been largely unsuccessful in curtailing mortality. Recent clinical and preclinical studies have taken advantage of the dichotomous ability of AR signaling to elicit growth-suppressive and differentiating effects by administering hyperphysiologic levels of testosterone. In this study, high-throughput drug screening identified a potent synergy between high-androgen therapy and YM155, a transcriptional inhibitor of survivin (BIRC5). This interaction was mediated by the direct transcriptional upregulation of the YM155 transporter SLC35F2 by the AR. Androgen-mediated YM155-induced cell death was completely blocked by the overexpression of multidrug resistance transporter ABCB1. SLC35F2 expression was significantly correlated with intratumor androgen levels in four distinct patient-derived xenograft models, and with AR activity score in a large gene expression dataset of castration-resistant metastases. A subset of tumors had significantly elevated SLC35F2 expression and, therefore, may identify patients who are highly responsive to YM155 treatment. IMPLICATIONS The combination of androgen therapy with YM155 represents a novel drug synergy, and SLC35F2 may serve as a clinical biomarker of response to YM155.
Collapse
Affiliation(s)
- Michael D Nyquist
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Alexandra Corella
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - John Burns
- Virginia Mason Medical Center, Seattle, Washington
| | - Ilsa Coleman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Shuai Gao
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Robin Tharakan
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Luke Riggan
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Changmeng Cai
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Urology, University of Washington, Seattle, Washington
- Division of Oncology, Department of Medicine, University of Washington, Seattle, Washington
| | - Elahe A Mostaghel
- Division of Oncology, Department of Medicine, University of Washington, Seattle, Washington.
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
31
|
Khan Z, Khan AA, Yadav H, Prasad GBKS, Bisen PS. Survivin, a molecular target for therapeutic interventions in squamous cell carcinoma. Cell Mol Biol Lett 2017; 22:8. [PMID: 28536639 PMCID: PMC5415770 DOI: 10.1186/s11658-017-0038-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 03/27/2017] [Indexed: 12/14/2022] Open
Abstract
Squamous cell carcinoma (SCC) is the most common cancer worldwide. The treatment of locally advanced disease generally requires various combinations of radiotherapy, surgery, and systemic therapy. Despite aggressive multimodal treatment, most of the patients relapse. Identification of molecules that sustain cancer cell growth and survival has made molecular targeting a feasible therapeutic strategy. Survivin is a member of the Inhibitor of Apoptosis Protein (IAP) family, which is overexpressed in most of the malignancies including SCC and totally absent in most of the normal tissues. This feature makes survivin an ideal target for cancer therapy. It orchestrates several important mechanisms to support cancer cell survival including inhibition of apoptosis and regulation of cell division. Overexpression of survivin in tumors is also associated with poor prognosis, aggressive tumor behavior, resistance to therapy, and high tumor recurrence. Various strategies have been developed to target survivin expression in cancer cells, and their effects on apoptosis induction and tumor growth attenuation have been demonstrated. In this review, we discuss recent advances in therapeutic potential of survivin in cancer treatment.
Collapse
Affiliation(s)
- Zakir Khan
- School of Studies in Biotechnology, Jiwaji University, Gwalior, 474001 MP India.,Department of Biomedical Sciences, Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Abdul Arif Khan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hariom Yadav
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | | | - Prakash Singh Bisen
- School of Studies in Biotechnology, Jiwaji University, Gwalior, 474001 MP India
| |
Collapse
|
32
|
Nyquist MD, Prasad B, Mostaghel EA. Harnessing Solute Carrier Transporters for Precision Oncology. Molecules 2017; 22:E539. [PMID: 28350329 PMCID: PMC5570559 DOI: 10.3390/molecules22040539] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 12/11/2022] Open
Abstract
Solute Carrier (SLC) transporters are a large superfamily of transmembrane carriers involved in the regulated transport of metabolites, nutrients, ions and drugs across cellular membranes. A subset of these solute carriers play a significant role in the cellular uptake of many cancer therapeutics, ranging from chemotherapeutics such as antimetabolites, topoisomerase inhibitors, platinum-based drugs and taxanes to targeted therapies such as tyrosine kinase inhibitors. SLC transporters are co-expressed in groups and patterns across normal tissues, suggesting they may comprise a coordinated regulatory circuit serving to mediate normal tissue functions. In cancer however, there are dramatic changes in expression patterns of SLC transporters. This frequently serves to feed the increased metabolic demands of the tumor cell for amino acids, nucleotides and other metabolites, but also presents a therapeutic opportunity, as increased transporter expression may serve to increase intracellular concentrations of substrate drugs. In this review, we examine the regulation of drug transporters in cancer and how this impacts therapy response, and discuss novel approaches to targeting therapies to specific cancers via tumor-specific aberrations in transporter expression. We propose that among the oncogenic changes in SLC transporter expression there exist emergent vulnerabilities that can be exploited therapeutically, extending the application of precision medicine from tumor-specific drug targets to tumor-specific determinants of drug uptake.
Collapse
Affiliation(s)
- Michael D Nyquist
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA.
| | - Elahe A Mostaghel
- Division of Oncology, Department of Medicine, University of Washington, Seattle, WA 98195 USA.
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| |
Collapse
|
33
|
YM155 enhances ABT-737-mediated apoptosis through Mcl-1 downregulation in Mcl-1-overexpressed cancer cells. Mol Cell Biochem 2017; 429:91-102. [PMID: 28120212 DOI: 10.1007/s11010-016-2938-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/23/2016] [Indexed: 10/20/2022]
Abstract
ABT-737 is a BH3 mimetic inhibitor of Bcl-xL, Bcl-2, and Bcl-w, and it has been reported for anti-cancer effects in various types of cancer cells. However, ABT-737 fails to induce apoptosis in cancer cell with high levels of Mcl-1 expression. The pharmacological survivin inhibitor YM155 has been reported to induce downregulation of Mcl-1 expression. Therefore, we investigated the effect of YM155 to sensitize resistance against ABT-737 in Mcl-1-overexpressed human renal carcinoma Caki cells. We found that ABT-737 alone and YM155 alone did not induce apoptosis, but YM155 markedly sensitized ABT-737-mediated apoptosis in Mcl-1-overexpressed Caki cells, human glioma cells (U251MG), and human lung carcinoma cells (A549). In contrast, combined treatment with ABT-737 and YM155 did not increase apoptosis in normal mouse kidney cells (TCMK-1) and human mesangial cells (MC). YM155 induced lysosome-dependent downregulation of Mcl-1 expression in Mcl-1-overexpressed Caki cells. In addition, combined treatment with ABT-737 and YM155 induced loss of mitochondrial membrane potential and inhibited interaction of Bcl-xL and Bax. Taken together, our results suggested that YM155 effectively improves sensitivity to ABT-737 through downregulation of Mcl-1 expression.
Collapse
|
34
|
Zhao N, Mao Y, Han G, Ju Q, Zhou L, Liu F, Xu Y, Zhao X. YM155, a survivin suppressant, triggers PARP-dependent cell death (parthanatos) and inhibits esophageal squamous-cell carcinoma xenografts in mice. Oncotarget 2016; 6:18445-59. [PMID: 26090615 PMCID: PMC4621902 DOI: 10.18632/oncotarget.4315] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 06/02/2015] [Indexed: 01/09/2023] Open
Abstract
Here we demonstrated that sepantronium bromide (YM155), a survivin suppressant, inhibited esophageal squamous-cell carcinoma (ESCC) growth in mice bearing human ESCC xenografts without affecting body weight. In cell culture, YM155 decreased survivin levels and caused PARP-1 activation, poly-ADP polymer formation, and AIF translocation from the cytosol to the nucleus. Genetic knockdown of PARP-1 or AIF abrogated YM155-induced parthanatos cell death. Furthermore, FOS, JUN and c-MYC gene transcription, which is stimulated by activated PARP-1, was increased following YM155 treatment. Our data demonstrate that YM155 did not trigger apoptosis, but induced parthanatos, a cell death dependent on PARP-1 hyper-activation, and support clinical development of YM155 in ESCC.
Collapse
Affiliation(s)
- Nan Zhao
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yousheng Mao
- Department of Thoracic Surgical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Gaijing Han
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qiang Ju
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lanping Zhou
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fang Liu
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yang Xu
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaohang Zhao
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
35
|
Murphy J, Hall WW, Ratner L, Sheehy N. Novel interactions between the HTLV antisense proteins HBZ and APH-2 and the NFAR protein family: Implications for the HTLV lifecycles. Virology 2016; 494:129-42. [PMID: 27110706 DOI: 10.1016/j.virol.2016.04.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 04/07/2016] [Accepted: 04/08/2016] [Indexed: 01/17/2023]
Abstract
The human T-cell leukaemia virus type 1 and type 2 (HTLV-1/HTLV-2) antisense proteins HBZ and APH-2 play key roles in the HTLV lifecycles and persistence in the host. Nuclear Factors Associated with double-stranded RNA (NFAR) proteins NF90/110 function in the lifecycles of several viruses and participate in host innate immunity against infection and oncogenesis. Using GST pulldown and co-immunoprecipitation assays we demonstrate specific novel interactions between HBZ/APH-2 and NF90/110 and characterised the protein domains involved. Moreover we show that NF90/110 significantly enhance Tax mediated LTR activation, an effect that was abolished by HBZ but enhanced by APH-2. Additionally we found that HBZ and APH-2 modulate the promoter activity of survivin and are capable of antagonising NF110-mediated survivin activation. Thus interactions between HTLV antisense proteins and the NFAR protein family have an overall positive impact on HTLV infection. Hence NFARs may represent potential therapeutic targets in HTLV infected cells.
Collapse
Affiliation(s)
- Jane Murphy
- Centre for Research in Infectious Diseases, School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - William W Hall
- Centre for Research in Infectious Diseases, School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Lee Ratner
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Noreen Sheehy
- Centre for Research in Infectious Diseases, School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
36
|
Knott GJ, Bond CS, Fox AH. The DBHS proteins SFPQ, NONO and PSPC1: a multipurpose molecular scaffold. Nucleic Acids Res 2016; 44:3989-4004. [PMID: 27084935 PMCID: PMC4872119 DOI: 10.1093/nar/gkw271] [Citation(s) in RCA: 206] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/05/2016] [Indexed: 12/23/2022] Open
Abstract
Nuclear proteins are often given a concise title that captures their function, such as 'transcription factor,' 'polymerase' or 'nuclear-receptor.' However, for members of the Drosophila behavior/human splicing (DBHS) protein family, no such clean-cut title exists. DBHS proteins are frequently identified engaging in almost every step of gene regulation, including but not limited to, transcriptional regulation, RNA processing and transport, and DNA repair. Herein, we present a coherent picture of DBHS proteins, integrating recent structural insights on dimerization, nucleic acid binding modalities and oligomerization propensity with biological function. The emerging paradigm describes a family of dynamic proteins mediating a wide range of protein-protein and protein-nucleic acid interactions, on the whole acting as a multipurpose molecular scaffold. Overall, significant steps toward appreciating the role of DBHS proteins have been made, but we are only beginning to understand the complexity and broader importance of this family in cellular biology.
Collapse
Affiliation(s)
- Gavin J Knott
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia, WA 6009, Australia
| | - Charles S Bond
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia, WA 6009, Australia
| | - Archa H Fox
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, WA 6009, Australia Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia
| |
Collapse
|
37
|
Kiyohara Y, Yoshino K, Kubota S, Okuyama H, Endo H, Ueda Y, Kimura T, Kimura T, Kamiura S, Inoue M. Drug screening and grouping by sensitivity with a panel of primary cultured cancer spheroids derived from endometrial cancer. Cancer Sci 2016; 107:452-60. [PMID: 26825848 PMCID: PMC4832863 DOI: 10.1111/cas.12898] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 01/06/2016] [Accepted: 01/26/2016] [Indexed: 12/17/2022] Open
Abstract
Several molecular targeting drugs are being evaluated for endometrial cancer; selecting patients whose cancers are sensitive to these agents is of paramount importance. Previously, we developed the cancer tissue-originated spheroid method for primary cancer cells taken from patients' tumors as well as patient-derived xenografts. In this study, we successfully prepared and cultured cancer tissue-originated spheroids from endometrial cancers. Characteristics of the original tumors were well retained in cancer tissue-originated spheroids including morphology and expression of p53 or neuroendocrine markers. We screened 79 molecular targeting drugs using two cancer tissue-originated spheroid lines derived from endometrioid adenocarcinoma grade 3 and serous adenocarcinoma. Among several hits, we focused on everolimus, a mammalian target of rapamycin complex 1 inhibitor, and YM155, a survivin inhibitor. When sensitivity to everolimus or YM155 was assessed in 12 or 11 cancer tissue-originated spheroids, respectively, from different endometrial cancer patients, the sensitivity varied substantially. The cancer tissue-originated spheroids sensitive to everolimus showed remarkable suppression of proliferation. The phosphorylation status of the mammalian target of rapamycin complex 1 downstream molecules before and after everolimus treatment did not predict the effect of the drug. In contrast, the cancer tissue-originated spheroids sensitive to YM155 showed remarkable cell death. The effect of YM155 was also confirmed in vivo. The histological type correlated with YM155 sensitivity; non-endometrioid adenocarcinomas were sensitive and endometrioid adenocarcinomas were resistant. Non-canonical autophagic cell death was the most likely cause of cell death in a sensitive cancer tissue-originated spheroid. Thus, sensitivity assays using cancer tissue-originated spheroids from endometrial cancers may be useful for screening drugs and finding biomarkers.
Collapse
Affiliation(s)
- Yumiko Kiyohara
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan.,Department of Gynecology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan.,Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kiyoshi Yoshino
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Satoshi Kubota
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan.,Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroaki Okuyama
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Hiroko Endo
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Yutaka Ueda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Toshihiro Kimura
- Department of Gynecology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shoji Kamiura
- Department of Gynecology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Masahiro Inoue
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| |
Collapse
|
38
|
Tothill R, Estall V, Rischin D. Merkel cell carcinoma: emerging biology, current approaches, and future directions. Am Soc Clin Oncol Educ Book 2016:e519-26. [PMID: 25993218 DOI: 10.14694/edbook_am.2015.35.e519] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Merkel cell carcinoma (MCC) is an aggressive neuroendocrine cutaneous cancer that predominantly occurs in patients who are older, and is associated with a high rate of distant failure and mortality. Current management strategies that incorporate surgery and radiotherapy achieve high rates of locoregional control, but distant failure rates remain problematic, highlighting the need for new effective systemic therapies. Chemotherapy can achieve high response rates of limited duration in the metastatic setting, but its role in definitive management remains unproven. Recent developments in our knowledge about the biology of MCC have led to the identification of new potential therapeutic targets and treatments. A key finding has been the discovery that a human polyomavirus may be a causative agent. However, emerging data suggests that MCC may actually be two distinct entities, viral-associated and viral-negative MCC, which is likely to have implications for the management of MCC in the future and for the development of new treatments. In this review, we discuss recent discoveries about the biology of MCC, current approaches to management, and new therapeutic strategies that are being investigated.
Collapse
Affiliation(s)
- Richard Tothill
- From the Division of Research, Peter MacCallum Cancer Centre, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Department of Radiation Oncology and Skin and Melanoma Tumour Stream, Peter MacCallum Cancer Centre, and the Department of Pathology, University of Melbourne, Melbourne, Australia; Division of Cancer Medicine, and Head and Neck Tumour Stream, Peter MacCallum Cancer Centre, the Sir Peter MacCallum Department of Oncology and Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Vanessa Estall
- From the Division of Research, Peter MacCallum Cancer Centre, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Department of Radiation Oncology and Skin and Melanoma Tumour Stream, Peter MacCallum Cancer Centre, and the Department of Pathology, University of Melbourne, Melbourne, Australia; Division of Cancer Medicine, and Head and Neck Tumour Stream, Peter MacCallum Cancer Centre, the Sir Peter MacCallum Department of Oncology and Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Danny Rischin
- From the Division of Research, Peter MacCallum Cancer Centre, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Department of Radiation Oncology and Skin and Melanoma Tumour Stream, Peter MacCallum Cancer Centre, and the Department of Pathology, University of Melbourne, Melbourne, Australia; Division of Cancer Medicine, and Head and Neck Tumour Stream, Peter MacCallum Cancer Centre, the Sir Peter MacCallum Department of Oncology and Department of Medicine, University of Melbourne, Melbourne, Australia
| |
Collapse
|
39
|
Arai M, Kawachi T, Kotoku N, Nakata C, Kamada H, Tsunoda SI, Tsutsumi Y, Endo H, Inoue M, Sato H, Kobayashi M. Furospinosulin-1, Marine Spongean Furanosesterterpene, Suppresses the Growth of Hypoxia-Adapted Cancer Cells by Binding to Transcriptional Regulators p54(nrb) and LEDGF/p75. Chembiochem 2015; 17:181-9. [PMID: 26561285 DOI: 10.1002/cbic.201500519] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Indexed: 11/09/2022]
Abstract
Hypoxia-adapted cancer cells in tumors contribute to the pathological progression of cancer. Cancer research has therefore focused on the identification of molecules responsible for hypoxia adaptation in cancer cells, as well as the development of new compounds with action against hypoxia-adapted cancer cells. The marine natural product furospinosulin-1 (1) has displayed hypoxia-selective growth inhibition against cultured cancer cells, and has shown in vivo anti-tumor activity, although its precise mode of action and molecular targets remain unclear. In this study, we found that 1 is selectively effective against hypoxic regions of tumors, and that it directly binds to the transcriptional regulators p54(nrb) and LEDGF/p75, which have not been previously identified as mediators of hypoxia adaptation in cancer cells.
Collapse
Affiliation(s)
- Masayoshi Arai
- Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka 1-6, Suita, Osaka, 565-0871, Japan.
| | - Takashi Kawachi
- Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka 1-6, Suita, Osaka, 565-0871, Japan
| | - Naoyuki Kotoku
- Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka 1-6, Suita, Osaka, 565-0871, Japan
| | - Chiaki Nakata
- Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka 1-6, Suita, Osaka, 565-0871, Japan
| | - Haruhiko Kamada
- Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka 1-6, Suita, Osaka, 565-0871, Japan.,National Institute of Biomedical Innovation, 7-6-8 Saitoasagi, Ibaraki, Osaka, 567-0085, Japan
| | - Shin-ichi Tsunoda
- Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka 1-6, Suita, Osaka, 565-0871, Japan.,National Institute of Biomedical Innovation, 7-6-8 Saitoasagi, Ibaraki, Osaka, 567-0085, Japan
| | - Yasuo Tsutsumi
- Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka 1-6, Suita, Osaka, 565-0871, Japan.,National Institute of Biomedical Innovation, 7-6-8 Saitoasagi, Ibaraki, Osaka, 567-0085, Japan
| | - Hiroko Endo
- Osaka Medical Center for Cancer and Cardiovascular Diseases, Higashinari-ku, Osaka, 537-8511, Japan
| | - Masahiro Inoue
- Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka 1-6, Suita, Osaka, 565-0871, Japan.,Osaka Medical Center for Cancer and Cardiovascular Diseases, Higashinari-ku, Osaka, 537-8511, Japan
| | - Hiroki Sato
- Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka 1-6, Suita, Osaka, 565-0871, Japan
| | - Motomasa Kobayashi
- Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka 1-6, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
40
|
The microRNA-218~Survivin axis regulates migration, invasion, and lymph node metastasis in cervical cancer. Oncotarget 2015; 6:1090-100. [PMID: 25473903 PMCID: PMC4359219 DOI: 10.18632/oncotarget.2836] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/24/2014] [Indexed: 11/26/2022] Open
Abstract
Cervical cancer is the third most common cancer in women worldwide. In the present study, global microRNA profiling for 79 cervical cancer patient samples led to the identification of miR-218 down-regulation in cervical cancer tissues compared to normal cervical tissues. Lower miR-218 expression was associated significantly with worse overall survival (OS), disease-free survival (DFS), and pelvic/aortic lymph node recurrence. In vitro, miR-218 over-expression decreased clonogenicity, migration, and invasion. Survivin (BIRC5) was subsequently identified as an important cervical cancer target of miR-218 using in silico prediction, mRNA profiling, and quantitative real-time PCR (qRT-PCR). Concordant with miR-218 over-expression, survivin knockdown by siRNA decreased clonogenicity, migration, and invasion. YM155, a small molecule survivin inhibitor, significantly suppressed tumor growth and lymph node metastasis in vivo. Our findings demonstrate that the miR-218~survivin axis inhibits cervical cancer progression by regulating clonogenicity, migration, and invasion, and suggest that the inhibition of survivin could be a potential therapeutic strategy to improve outcome in this disease.
Collapse
|
41
|
Ho SHS, Sim MY, Yee WLS, Yang T, Yuen SPJ, Go ML. Antiproliferative, DNA intercalation and redox cycling activities of dioxonaphtho[2,3-d]imidazolium analogs of YM155: A structure-activity relationship study. Eur J Med Chem 2015; 104:42-56. [PMID: 26433618 DOI: 10.1016/j.ejmech.2015.09.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/17/2015] [Accepted: 09/21/2015] [Indexed: 10/23/2022]
Abstract
The anticancer agent YM155 is widely investigated as a specific survivin suppressant. More recently, YM155 was found to induce DNA damage and this has raised doubts as to whether survivin is its primary target. In an effort to assess the contribution of DNA damage to the anticancer activity of YM155, several analogs were prepared and evaluated for antiproliferative activity on malignant cells, participation in DNA intercalation and free radical generation by redox cycling. The intact positively charged scaffold was found to be essential for antiproliferative activity and intercalation but was less critical for redox cycling where the minimal requirement was a pared down bicyclic quinone. Side chain requirements at the N(1) and N(3) positions of the scaffold were more alike for redox cycling and intercalation than antiproliferative activity, underscoring yet again, the limited structural overlaps for these activities. Furthermore, antiproliferative activities were poorly correlated to DNA intercalation and redox cycling. Potent antiproliferative activity (IC50 9-23 nM), exceeding that of YM155, was found for a minimally substituted methyl analog AB7. Like YM155 and other dioxonaphthoimidazoliums, AB7 was a modest DNA intercalator but with weak redox cycling activity. Thus, the capacity of this scaffold to inflict direct DNA damage leading to cell death may not be significant and YM155 should not be routinely classified as a DNA damaging agent.
Collapse
Affiliation(s)
- Si-Han Sherman Ho
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, 117543, Republic of Singapore
| | - Mei-Yi Sim
- Department of Urology, Singapore General Hospital, 20 College Road, 169856, Republic of Singapore
| | - Wei-Loong Sherman Yee
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, 117543, Republic of Singapore
| | - Tianming Yang
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, 117543, Republic of Singapore
| | - Shyi-Peng John Yuen
- Department of Urology, Singapore General Hospital, 20 College Road, 169856, Republic of Singapore
| | - Mei-Lin Go
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, 117543, Republic of Singapore.
| |
Collapse
|
42
|
Mitobe Y, Yasunaga JI, Furuta R, Matsuoka M. HTLV-1 bZIP Factor RNA and Protein Impart Distinct Functions on T-cell Proliferation and Survival. Cancer Res 2015; 75:4143-52. [PMID: 26383166 DOI: 10.1158/0008-5472.can-15-0942] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 07/20/2015] [Indexed: 11/16/2022]
Abstract
Infection of T cells with human T-cell leukemia virus type-1 (HTLV-1) induces clonal proliferation and is closely associated with the onset of adult T-cell leukemia-lymphoma (ATL) and inflammatory diseases. Although Tax expression is frequently suppressed in HTLV-1-infected cells, the accessory gene, HTLV-1 bZIP factor (HBZ), is continuously expressed and has been implicated in HTLV-1 pathogenesis. Here, we report that transduction of mouse T cells with specific mutants of HBZ that distinguish between its RNA and protein activity results in differential effects on T-cell proliferation and survival. HBZ RNA increased cell number by attenuating apoptosis, whereas HBZ protein induced apoptosis. However, both HBZ RNA and protein promoted S-phase entry of T cells. We further identified that the first 50 bp of the HBZ coding sequence are required for RNA-mediated cell survival. Transcriptional profiling of T cells expressing wild-type HBZ, RNA, or protein revealed that HBZ RNA is associated with genes involved in cell cycle, proliferation, and survival, while HBZ protein is more closely related to immunological properties of T cells. Specifically, HBZ RNA enhances the promoter activity of survivin, an inhibitor of apoptosis, to upregulate its expression. Inhibition of survivin using YM155 resulted in impaired proliferation of several ATL cell lines as well as a T-cell line expressing HBZ RNA. The distinct functions of HBZ RNA and protein may have several implications for the development of strategies to control the proliferation and survival mechanisms associated with HTLV-1 infection and ATL.
Collapse
Affiliation(s)
- Yuichi Mitobe
- Laboratory of Virus Control, Institute for Virus Research, Kyoto University, Sakyo-ku, Kyoto, Japan. Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Jun-ichirou Yasunaga
- Laboratory of Virus Control, Institute for Virus Research, Kyoto University, Sakyo-ku, Kyoto, Japan.
| | - Rie Furuta
- Laboratory of Virus Control, Institute for Virus Research, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Masao Matsuoka
- Laboratory of Virus Control, Institute for Virus Research, Kyoto University, Sakyo-ku, Kyoto, Japan.
| |
Collapse
|
43
|
Wagner V, Hose D, Seckinger A, Weiz L, Meißner T, Rème T, Breitkreutz I, Podar K, Ho AD, Goldschmidt H, Krämer A, Klein B, Raab MS. Preclinical efficacy of sepantronium bromide (YM155) in multiple myeloma is conferred by down regulation of Mcl-1. Oncotarget 2015; 5:10237-50. [PMID: 25296978 PMCID: PMC4279369 DOI: 10.18632/oncotarget.2529] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 09/25/2014] [Indexed: 12/11/2022] Open
Abstract
The inhibitor-of-apoptosis family member survivin has been reported to inhibit apoptosis and regulate mitosis and cytokinesis. In multiple myeloma, survivin has been described to be involved in downstream sequelae of various therapeutic agents. We assessed 1093 samples from previously untreated patients, including two independent cohorts of 392 and 701 patients, respectively. Survivin expression was associated with cell proliferation, adverse prognostic markers, and inferior event-free and overall survival, supporting the evaluation of survivin as a therapeutic target in myeloma. The small molecule suppressant of survivin - YM155 - is in clinical development for the treatment of solid tumors. YM155 potently inhibited proliferation and induced apoptosis in primary myeloma cells and cell lines. Gene expression and protein profiling revealed the critical roles of IL6/STAT3-signaling and the unfolded protein response in the efficacy of YM155. Both pathways converged to down regulate anti-apoptotic Mcl-1 in myeloma cells. Conversely, growth inhibition and apoptotic cell death by YM155 was rescued by ectopic expression of Mcl-1 but not survivin, identifying Mcl-1 as the pivotal downstream target of YM155 in multiple myeloma. Mcl-1 expression was likewise associated with adverse prognostic markers, and inferior survival. Our results strongly support the clinical evaluation of YM155 in patients with multiple myeloma.
Collapse
Affiliation(s)
- Verena Wagner
- Max-Eder Group Experimental Therapies for Hematologic Malignancies, German Cancer Research Center (DKFZ) and Dept. of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Dirk Hose
- Dept. of Internal Medicine V, University of Heidelberg, Heidelberg, Germany. National Center of Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Anja Seckinger
- Dept. of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Ludmila Weiz
- Max-Eder Group Experimental Therapies for Hematologic Malignancies, German Cancer Research Center (DKFZ) and Dept. of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Tobias Meißner
- Dept. of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | - Iris Breitkreutz
- Max-Eder Group Experimental Therapies for Hematologic Malignancies, German Cancer Research Center (DKFZ) and Dept. of Internal Medicine V, University of Heidelberg, Heidelberg, Germany. National Center of Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Klaus Podar
- National Center of Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Anthony D Ho
- Dept. of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Hartmut Goldschmidt
- Dept. of Internal Medicine V, University of Heidelberg, Heidelberg, Germany. National Center of Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Dept. of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Bernard Klein
- INSERM U1040, Montpellier, France. CHU Montpellier, Institute of Research in Biotherapy, Montpellier, France
| | - Marc S Raab
- Max-Eder Group Experimental Therapies for Hematologic Malignancies, German Cancer Research Center (DKFZ) and Dept. of Internal Medicine V, University of Heidelberg, Heidelberg, Germany. Dept. of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
44
|
Influence of survivin-targeted therapy on chemosensitivity in the treatment of acute myeloid leukemia. Cancer Lett 2015; 366:160-72. [PMID: 26123662 DOI: 10.1016/j.canlet.2015.05.033] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/12/2015] [Accepted: 05/14/2015] [Indexed: 11/23/2022]
Abstract
Overexpression of survivin is observed in various hematological malignancies, including acute myeloid leukemia (AML). Studies show that elevated expression of survivin correlates with a worse clinic outcome in AML patients. It remains unclear whether inhibition of survivin may alter the efficacy of chemotherapy against AML. Here, we evaluate the effects of specific knockdown of survivin on AML cells' sensitivity to chemotherapy, and investigate the therapeutic potential of the transcription inhibitor of survivin YM155 either alone or in combination with chemotherapeutic agents. We found Kasumi-1 and HL-60 cells had relatively higher expression levels of survivin among all AML cell lines tested. Specific knockdown of survivin in Kasumi-1 and HL-60 cells resulted in: inhibition of cell proliferation; cell cycle G2/M arrest; induction of DNA damage response and apoptosis. Downregulation of survivin enhanced etoposide- or doxorubicin-induced anti-proliferative/anti-survival activity in AML cells. The small molecule inhibitor YM155 reduced survivin in a dose- and time-dependent manner and trigged apoptosis in Kasumi-1 and HL-60 cells. The combinatorial effects of YM155 and chemotherapeutics were either synergetic or antagonistic, depending upon the drugs used for combination and the type of AML cells being treated. Collectively, our data demonstrate that survivin plays an important role in the maintenance and proliferation of AML cells. While specific knockdown of survivin enhances chemosensitivity, the combinations of YM155 and chemotherapeutic agents exhibit synergetic or antagonistic effects on AML cells. Our findings provide a rationale for further assessment of survivin-targeted therapy in the treatment of patients with AML.
Collapse
|
45
|
de Necochea-Campion R, Diaz Osterman CJ, Hsu HW, Fan J, Mirshahidi S, Wall NR, Chen CS. AML sensitivity to YM155 is modulated through AKT and Mcl-1. Cancer Lett 2015; 366:44-51. [PMID: 26118775 DOI: 10.1016/j.canlet.2015.05.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 05/29/2015] [Accepted: 05/29/2015] [Indexed: 10/23/2022]
Abstract
HL60 and U937 (acute myeloid leukemia (AML) cell lines) were assessed for sensitivity to YM155, and found to have distinct sensitive and resistant phenotypes, respectively. In HL60 cells, YM155 inhibition of growth proliferation was due to apoptosis which was measured by annexin V/PI staining. YM155 induced apoptosis through activation of intrinsic and extrinsic pathways that also culminated in caspase-3 activity and PARP cleavage. YM155 sensitivity was partially associated with this compound's ability to down-regulate survivin transcription since this was more pronounced in the HL60 cell line. However, marked differences were also observed in XIAP, Bcl-2, and Mcl-1L, and Mcl-1s. Furthermore, YM155 treatment completely inhibited production of total Akt protein in HL60, but not U937 cells. Importantly, Akt activity (pAkt-Ser473) levels were maintained in YM155 treated U937 cells which may help stabilize other anti-apoptotic proteins. Combination treatments with an Akt inhibitor, MK-2206, reduced levels of pAkt-Ser473 in U937 cells and synergistically sensitized them to YM155 cytotoxicity. Collectively our results indicate that Akt signaling may be an important factor mediating YM155 response in AML, and combinatorial therapies with Akt inhibitors could improve treatment efficacy in YM155-resistant cells.
Collapse
Affiliation(s)
- Rosalia de Necochea-Campion
- Department of Internal Medicine, Division of Hematology and Medical Oncology & Biospecimen Laboratory, Loma Linda University, Loma Linda, CA 92350, USA
| | - Carlos J Diaz Osterman
- Center for Health Disparities & Molecular Medicine, Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92350, USA
| | - Heng-Wei Hsu
- Department of Internal Medicine, Division of Hematology and Medical Oncology & Biospecimen Laboratory, Loma Linda University, Loma Linda, CA 92350, USA
| | - Junjie Fan
- Department of Internal Medicine, Division of Hematology and Medical Oncology & Biospecimen Laboratory, Loma Linda University, Loma Linda, CA 92350, USA
| | - Saied Mirshahidi
- Department of Internal Medicine, Division of Hematology and Medical Oncology & Biospecimen Laboratory, Loma Linda University, Loma Linda, CA 92350, USA
| | - Nathan R Wall
- Center for Health Disparities & Molecular Medicine, Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Chien-Shing Chen
- Department of Internal Medicine, Division of Hematology and Medical Oncology & Biospecimen Laboratory, Loma Linda University, Loma Linda, CA 92350, USA.
| |
Collapse
|
46
|
Jung SA, Park YM, Hong SW, Moon JH, Shin JS, Lee HR, Ha SH, Lee DH, Kim JH, Kim SM, Kim JE, Kim KP, Hong YS, Choi EK, Lee JS, Jin DH, Kim T. Cellular inhibitor of apoptosis protein 1 (cIAP1) stability contributes to YM155 resistance in human gastric cancer cells. J Biol Chem 2015; 290:9974-85. [PMID: 25635055 DOI: 10.1074/jbc.m114.600874] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Indexed: 01/02/2023] Open
Abstract
YM155, which blocks the expression of survivin, a member of the inhibitor of apoptosis (IAP) family, induces cell death in a variety of cancer types, including prostate, bladder, breast, leukemia, and non-small lung cancer. However, the mechanism underlying gastric cancer susceptibility and resistance to YM155 is yet to be specified. Here, we demonstrate that cIAP1 stability dictates resistance to YM155 in human gastric cancer cells. Treatment of human gastric cancer cells with YM155 differentially induced cell death dependent on the stability of cIAP1 as well as survivin. Transfection with cIAP1 expression plasmids decreased cell sensitivity to YM155, whereas knockdown of endogenous cIAP1 using RNA interference enhanced sensitivity to YM155. In addition, double knockdown of survivin and cIAP1 significantly induced cell death in the YM155-resistant cell line, MKN45. We also showed that YM155 induced autoubiquitination and proteasome-dependent degradation of cIAP1. Surprisingly, survivin affected the stability of cIAP1 through binding, contributing to cell sensitivity to YM155. Thus, our findings reveal that YM155 sensitizes human gastric cancer cells to apoptotic cell death by degrading cIAP1, and furthermore, cIAP1 in gastric cancer cells may act as a PD marker for YM155 treatment.
Collapse
Affiliation(s)
- Soo-A Jung
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology
| | - Yong-Man Park
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology
| | - Seung-Woo Hong
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology
| | - Jai-Hee Moon
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology
| | - Jae-Sik Shin
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology
| | - Ha-Reum Lee
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology, the Department of Life Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Seung-Hee Ha
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology
| | - Dae-Hee Lee
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology
| | - Jeong Hee Kim
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology
| | - Seung-Mi Kim
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology
| | - Jeong Eun Kim
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology
| | - Kyu-pyo Kim
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology
| | - Yong Sang Hong
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology
| | - Eun Kyung Choi
- From the Innovative Cancer Research, Asan Institute for Life Science, Radiation Oncology, and
| | - Jung Shin Lee
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology
| | - Dong-Hoon Jin
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology, Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul and
| | - TaeWon Kim
- From the Innovative Cancer Research, Asan Institute for Life Science, Departments of Oncology,
| |
Collapse
|
47
|
Abstract
Survivin is a well-established target in experimental cancer therapy. The molecule is over-expressed in most human tumors, but hardly detectable in normal tissues. Multiple functions in different subcellular compartments have been assigned. It participates in the control of cell division, apoptosis, the cellular stress response, and also in the regulation of cell migration and metastasis. Survivin expression has been recognized as a biomarker: high expression indicates an unfavorable prognosis and resistance to chemotherapeutic agents and radiation treatment. Survivin is an unconventional drug target and several indirect approaches have been exploited to affect its function and the phenotype of survivin-expressing cells. Interference with the expression of the survivin gene, the utilization of its messenger RNA, the intracellular localization, the interaction with binding partners, the stability of the survivin protein, and the induction of survivin-specific immune responses have been taken into consideration. A direct strategy to inhibit survivin has been based on the identification of a specifically interacting peptide. This peptide can recognize survivin intracellularly and cause the degradation of the ligand–survivin complex. Technology is being developed that might allow the derivation of small molecular-weight, drug-like compounds that are functionally equivalent to the peptide ligand.
Collapse
Affiliation(s)
- Bernd Groner
- Georg Speyer Haus, Institute for Biomedical Research, Paul Ehrlich Str. 42, 60322, Frankfurt am Main, Germany,
| | | |
Collapse
|
48
|
Castella S, Bernard R, Corno M, Fradin A, Larcher JC. Ilf3 and NF90 functions in RNA biology. WILEY INTERDISCIPLINARY REVIEWS-RNA 2014; 6:243-56. [PMID: 25327818 DOI: 10.1002/wrna.1270] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 09/09/2014] [Accepted: 09/17/2014] [Indexed: 12/24/2022]
Abstract
Double-stranded RNA-binding proteins (DRBPs) are known to regulate many processes of RNA metabolism due, among others, to the presence of double-stranded RNA (dsRNA)-binding motifs (dsRBMs). Among these DRBPs, Interleukin enhancer-binding factor 3 (Ilf3) and Nuclear Factor 90 (NF90) are two ubiquitous proteins generated by mutually exclusive and alternative splicings of the Ilf3 gene. They share common N-terminal and central sequences but display specific C-terminal regions. They present a large heterogeneity generated by several post-transcriptional and post-translational modifications involved in their subcellular localization and biological functions. While Ilf3 and NF90 were first identified as activators of gene expression, they are also implicated in cellular processes unrelated to RNA metabolism such as regulation of the cell cycle or of enzymatic activites. The implication of Ilf3 and NF90 in RNA biology will be discussed with a focus on eukaryote transcription and translation regulation, on viral replication and translation as well as on noncoding RNA field.
Collapse
Affiliation(s)
- Sandrine Castella
- Laboratoire de Biologie du développement, Institut de Biologie Paris-Seine, Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Laboratoire de Biologie du développement, Institut de Biologie Paris-Seine, CNRS, UMR 7622, Paris, France
| | | | | | | | | |
Collapse
|
49
|
Brun SN, Markant SL, Esparza LA, Garcia G, Terry D, Huang JM, Pavlyukov MS, Li XN, Grant GA, Crawford JR, Levy ML, Conway EM, Smith LH, Nakano I, Berezov A, Greene MI, Wang Q, Wechsler-Reya RJ. Survivin as a therapeutic target in Sonic hedgehog-driven medulloblastoma. Oncogene 2014; 34:3770-9. [PMID: 25241898 PMCID: PMC4369477 DOI: 10.1038/onc.2014.304] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/22/2014] [Accepted: 07/31/2014] [Indexed: 12/31/2022]
Abstract
Medulloblastoma (MB) is a highly malignant brain tumor that occurs primarily in children. Although surgery, radiation and high-dose chemotherapy have led to increased survival, many MB patients still die from their disease, and patients who survive suffer severe long-term side effects as a consequence of treatment. Thus, more effective and less toxic therapies for MB are critically important. Development of such therapies depends in part on identification of genes that are necessary for growth and survival of tumor cells. Survivin is an inhibitor of apoptosis protein that regulates cell cycle progression and resistance to apoptosis, is frequently expressed in human MB and when expressed at high levels predicts poor clinical outcome. Therefore, we hypothesized that Survivin may have a critical role in growth and survival of MB cells and that targeting it may enhance MB therapy. Here we show that Survivin is overexpressed in tumors from patched (Ptch) mutant mice, a model of Sonic hedgehog (SHH)-driven MB. Genetic deletion of survivin in Ptch mutant tumor cells significantly inhibits proliferation and causes cell cycle arrest. Treatment with small-molecule antagonists of Survivin impairs proliferation and survival of both murine and human MB cells. Finally, Survivin antagonists impede growth of MB cells in vivo. These studies highlight the importance of Survivin in SHH-driven MB, and suggest that it may represent a novel therapeutic target in patients with this disease.
Collapse
Affiliation(s)
- S N Brun
- 1] Tumor Initiation and Maintenance Program, National Cancer Institute (NCI)-Designated Cancer Center, Sanford-Burnham Medical Research Institute (SBMRI), La Jolla, CA, USA [2] Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA [3] Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - S L Markant
- 1] Tumor Initiation and Maintenance Program, National Cancer Institute (NCI)-Designated Cancer Center, Sanford-Burnham Medical Research Institute (SBMRI), La Jolla, CA, USA [2] Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA [3] Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - L A Esparza
- 1] Tumor Initiation and Maintenance Program, National Cancer Institute (NCI)-Designated Cancer Center, Sanford-Burnham Medical Research Institute (SBMRI), La Jolla, CA, USA [2] Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - G Garcia
- Histopathology Core SBMRI, La Jolla, CA, USA
| | - D Terry
- Conrad Prebys Center for Chemical Genomics, SBMRI, Lake Nona, FL, USA
| | - J-M Huang
- Cedars-Sinai Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
| | - M S Pavlyukov
- 1] Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA [2] James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - X-N Li
- Brain Tumor Program, Texas Children's Cancer Center, and Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - G A Grant
- Department of Neurosurgery, Stanford University/Lucile Packard Children's Hospital, Stanford, CA, USA
| | - J R Crawford
- 1] Department of Pediatrics, University of California San Diego, San Diego, CA, USA [2] Departments of Neurosciences, University of California San Diego, San Diego, CA, USA [3] Rady Children's Hospital, San Diego, CA, USA
| | - M L Levy
- 1] Rady Children's Hospital, San Diego, CA, USA [2] Department of Neurosurgery, University of California San Diego, La Jolla, CA, USA
| | - E M Conway
- Centre for Blood Research, Division of Hematology, Department of Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
| | - L H Smith
- 1] Conrad Prebys Center for Chemical Genomics, SBMRI, Lake Nona, FL, USA [2] Cardiopathobiology Program, Sanford Burnham Medical Research Institute, Lake Nona, FL, USA
| | - I Nakano
- 1] Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA [2] James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - A Berezov
- Department of Biomedical Sciences at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - M I Greene
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Q Wang
- Cedars-Sinai Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
| | - R J Wechsler-Reya
- 1] Tumor Initiation and Maintenance Program, National Cancer Institute (NCI)-Designated Cancer Center, Sanford-Burnham Medical Research Institute (SBMRI), La Jolla, CA, USA [2] Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA [3] Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| |
Collapse
|
50
|
Systemic Therapy for Merkel Cell Carcinoma: What's on the Horizon? Cancers (Basel) 2014; 6:1180-94. [PMID: 24840048 PMCID: PMC4074823 DOI: 10.3390/cancers6021180] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/08/2014] [Accepted: 05/09/2014] [Indexed: 12/23/2022] Open
Abstract
Merkel cell carcinoma is an aggressive neuroendocrine skin cancer that usually affects elderly patients. Despite being uncommon, incidence has been steadily increasing over the last two decades, likely due to increased awareness, better diagnostic methods and aging of the population. It is currently one of the most lethal cutaneous malignancies, with a five-year overall survival of approximately 50%. With the better understanding of the molecular pathways that lead to the development of Merkel cell carcinoma, there has been an increasing excitement and optimism surrounding novel targeted therapies, in particular to immunotherapy. Some of the concepts surrounding the novel targeted therapies and currently ongoing clinical trials are reviewed here.
Collapse
|