1
|
Molecular perspective on targeted therapy in breast cancer: a review of current status. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:149. [PMID: 35834030 PMCID: PMC9281252 DOI: 10.1007/s12032-022-01749-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/11/2022] [Indexed: 12/24/2022]
Abstract
Breast cancer is categorized at the molecular level according to the status of certain hormone and growth factor receptors, and this classification forms the basis of current diagnosis and treatment. The development of resistance to treatment and recurrence of the disease have led researchers to develop new therapies. In recent years, most of the research in the field of oncology has focused on the development of targeted therapies, which are treatment methods developed directly against molecular abnormalities. Promising advances have been made in clinical trials investigating the effect of these new treatment modalities and their combinations with existing therapeutic treatments in the treatment of breast cancer. Monoclonal antibodies, tyrosine kinase inhibitors, antibody–drug conjugates, PI3K/Akt/mTOR pathway inhibitors, cyclin-dependent kinase 4/6 inhibitors, anti-angiogenic drugs, PARP inhibitors are among the targeted therapies used in breast cancer treatment. In this review, we aim to present a molecular view of recently approved target agents used in breast cancer.
Collapse
|
2
|
Metformin inhibits the development and metastasis of colorectal cancer. Med Oncol 2022; 39:136. [PMID: 35780231 DOI: 10.1007/s12032-022-01722-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/29/2022] [Indexed: 10/17/2022]
Abstract
Metformin is a commonly used drug for the treatment of diabetes. Accumulating evidence suggests that it exerts anti-cancer effects in many cancers, including colorectal cancer. However, the underlying molecular mechanisms of colorectal cancer metastasis remain unclear. Colorectal cancer cell lines were treated with metformin, and cell proliferation, invasion, and migration were analyzed in vitro. The relationship between metformin and the AMPK-mTOR axis was assessed by Western blot analysis and transfection with small interfering RNA. A colorectal cancer xenograft mouse model was used to observe the effects of metformin on liver metastasis. Immunohistochemical analysis was performed on liver metastatic tumors. In in vitro experiments, metformin significantly inhibited the proliferation, migration, and invasion only in HCT116 and SW837 cells, but not in HCT8 and Lovo cells. Only in HCT116 and SW837, a change in AMPK-mTOR expression was observed in a dose-dependent manner. In colorectal cancer xenograft mice, the liver metastatic rate (10% vs. 50%, p = 0.05) and the number of liver metastatic nodules (0.1/body vs. 1.2/body, p = 0.04) were significantly lower in the metformin group. Tumor proliferation and EMT were decreased and apoptosis was promoted only in metastatic liver tumors of mice treated with metformin. The molecular mechanism of the anti-cancer effects of metformin involves repression of mTOR pathways via AMPK activation. Moreover, the differences in metformin sensitivity depend on the response of the AMPK-mTOR pathway to metformin. Our study provides a theoretical basis for the anti-metastatic treatment of colorectal cancer using metformin.
Collapse
|
3
|
Deng S, Leong HC, Datta A, Gopal V, Kumar AP, Yap CT. PI3K/AKT Signaling Tips the Balance of Cytoskeletal Forces for Cancer Progression. Cancers (Basel) 2022; 14:1652. [PMID: 35406424 PMCID: PMC8997157 DOI: 10.3390/cancers14071652] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/13/2022] [Accepted: 03/21/2022] [Indexed: 02/01/2023] Open
Abstract
The PI3K/AKT signaling pathway plays essential roles in multiple cellular processes, which include cell growth, survival, metabolism, and motility. In response to internal and external stimuli, the PI3K/AKT signaling pathway co-opts other signaling pathways, cellular components, and cytoskeletal proteins to reshape individual cells. The cytoskeletal network comprises three main components, which are namely the microfilaments, microtubules, and intermediate filaments. Collectively, they are essential for many fundamental structures and cellular processes. In cancer, aberrant activation of the PI3K/AKT signaling cascade and alteration of cytoskeletal structures have been observed to be highly prevalent, and eventually contribute to many cancer hallmarks. Due to their critical roles in tumor progression, pharmacological agents targeting PI3K/AKT, along with cytoskeletal components, have been developed for better intervention strategies against cancer. In our review, we first discuss existing evidence in-depth and then build on recent advances to propose new directions for therapeutic intervention.
Collapse
Affiliation(s)
- Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
| | - Hin Chong Leong
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Arpita Datta
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
| | - Vennila Gopal
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
| | - Alan Prem Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| | - Celestial T. Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| |
Collapse
|
4
|
Wikan N, Hankittichai P, Thaklaewphan P, Potikanond S, Nimlamool W. Oxyresveratrol Inhibits TNF-α-Stimulated Cell Proliferation in Human Immortalized Keratinocytes (HaCaT) by Suppressing AKT Activation. Pharmaceutics 2021; 14:63. [PMID: 35056961 PMCID: PMC8781909 DOI: 10.3390/pharmaceutics14010063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 12/17/2022] Open
Abstract
Psoriasis is a complex inflammatory disease characterized by hyperproliferative keratinocyte caused by active PI3K/AKT signaling. TNF-α concentrated in the psoriatic lesions stimulates AKT activation. We previously discovered that oxyresveratrol inhibited inflammation via suppressing AKT phosphorylation, therefore oxyresveratrol may possess a conserved property to block AKT activation and proliferation in keratinocyte in response to TNF-α. Our current study proved that oxyresveratrol exhibited potent anti-proliferative effects against TNF-α. These effects are explained by the findings that oxyresveratrol could potentially inhibit TNF-α-stimulated AKT and GSK3-β activation in a dose-dependent manner, and its inhibitory pattern was comparable to that of a specific PI3K inhibitor. Results from immunofluorescence supported that oxyresveratrol effectively inhibited AKT and GSK3-β activation in individual cells upon TNF-α stimulation. Furthermore, functional assay confirmed that oxyresveratrol repressed the expansion of the HaCaT colony over 3 days, and this was caused by the ability of oxyresveratrol to induce cell cycle arrest at S and G2/M phases and the reduction in the expression of a proliferative marker (Ki-67) and a survival marker (MCL-1). Given the importance of TNF-α and the PI3K/AKT pathway in the psoriatic phenotype, we anticipate that oxyresveratrol, which targets the TNF-α-stimulated PI3K/AKT pathway, would represent a promising psoriasis therapy in the near future.
Collapse
Affiliation(s)
- Nitwara Wikan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (N.W.); (P.H.); (P.T.); (S.P.)
| | - Phateep Hankittichai
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (N.W.); (P.H.); (P.T.); (S.P.)
| | - Phatarawat Thaklaewphan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (N.W.); (P.H.); (P.T.); (S.P.)
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (N.W.); (P.H.); (P.T.); (S.P.)
- Research Center for Development of Local Lanna Rice and Rice Products, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (N.W.); (P.H.); (P.T.); (S.P.)
- Research Center for Development of Local Lanna Rice and Rice Products, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
5
|
High-Fat Diet-Induced Fatty Liver Is Associated with Immunosuppressive Response during Sepsis in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5833857. [PMID: 34925696 PMCID: PMC8674062 DOI: 10.1155/2021/5833857] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022]
Abstract
High-fat diet-induced fatty liver is an indolent and chronic disease accompanied by immune dysfunction and metabolic disturbances involving numerous biological pathways. This study investigated how this abnormal metabolic disorder influences sepsis in mice. Mice were fed with normal chow (NC) or high-fat diet (HFD), and palmitic acid (PA) was used to treat hepatocytes to mimic fat accumulation in vitro. Lipopolysaccharide (LPS) was used to induce sepsis and related immune responses. Mice fed on a high-fat diet displayed higher mortality and more severe liver damage but compromised immunoreaction. The supernatant from PA-treated primary hepatocytes markedly diminished the inflammatory cytokine expression of macrophages after LPS stimulation, which showed a state of immunosuppression. Metabolomics analysis indicated the level of many key metabolites with possible roles in immunoreaction was altered in the HFD and PA groups compared with corresponding controls; specifically, β-hydroxybutyric acid (BHB) showed an immunosuppressive effect on Raw264.7 cells during the LPS stimulation. Transcriptomic analysis suggested that several differential signaling pathways may be associated with the alteration of immune function between the NC and HFD groups, as well as in the in vitro model. Our study suggests that the consumption of HFD may alter the hepatic metabolic profile, and that certain metabolites may remold the immune system to immunosuppressive state in the context of sepsis.
Collapse
|
6
|
Cha BH, Jung M, Kim AS, Lepak VC, Colson BA, Bull DA, Won Y. AZD2014, a dual mTOR inhibitor, attenuates cardiac hypertrophy in vitro and in vivo. J Biol Eng 2021; 15:24. [PMID: 34674743 PMCID: PMC8529824 DOI: 10.1186/s13036-021-00276-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 10/04/2021] [Indexed: 11/17/2022] Open
Abstract
Cardiac hypertrophy is one of the most common genetic heart disorders and considered a risk factor for cardiac morbidity and mortality. The mammalian target of rapamycin (mTOR) pathway plays a key regulatory function in cardiovascular physiology and pathology in hypertrophy. AZD2014 is a small-molecule ATP competitive mTOR inhibitor working on both mTORC1 and mTORC2 complexes. Little is known about the therapeutic effects of AZD2014 in cardiac hypertrophy and its underlying mechanism. Here, AZD2014 is examined in in vitro model of phenylephrine (PE)-induced human cardiomyocyte hypertrophy and a myosin-binding protein-C (Mybpc3)-targeted knockout (KO) mouse model of cardiac hypertrophy. Our results demonstrate that cardiomyocytes treated with AZD2014 retain the normal phenotype and AZD2014 attenuates cardiac hypertrophy in the Mybpc3-KO mouse model through inhibition of dual mTORC1 and mTORC2, which in turn results in the down-regulation of the Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Byung-Hyun Cha
- Division of Cardio-Thoracic Surgery, Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Minjin Jung
- Division of Cardio-Thoracic Surgery, Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Angela S Kim
- Division of Cardio-Thoracic Surgery, Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Victoria C Lepak
- Department of Cellular & Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Brett A Colson
- Department of Cellular & Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - David A Bull
- Division of Cardio-Thoracic Surgery, Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Youngwook Won
- Division of Cardio-Thoracic Surgery, Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, 85724, USA.
| |
Collapse
|
7
|
Colardo M, Segatto M, Di Bartolomeo S. Targeting RTK-PI3K-mTOR Axis in Gliomas: An Update. Int J Mol Sci 2021; 22:4899. [PMID: 34063168 PMCID: PMC8124221 DOI: 10.3390/ijms22094899] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/01/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022] Open
Abstract
Gliomas are the most common and challenging malignancies of the central nervous system (CNS), due to their infiltrative nature, tendency to recurrence, and poor response to treatments. Indeed, despite the advances in neurosurgical techniques and in radiation therapy, the modest effects of therapy are still challenging. Moreover, tumor recurrence is associated with the onset of therapy resistance; it is therefore critical to identify effective and well-tolerated pharmacological approaches capable of inducing durable responses in the appropriate patient groups. Molecular alterations of the RTK/PI3K/Akt/mTOR signaling pathway are typical hallmarks of glioma, and several clinical trials targeting one or more players of this axis have been launched, showing disappointing results so far, due to the scarce BBB permeability of certain compounds or to the occurrence of resistance/tolerance mechanisms. However, as RTK/PI3K/mTOR is one of the pivotal pathways regulating cell growth and survival in cancer biology, targeting still remains a strong rationale for developing strategies against gliomas. Future rigorous clinical studies, aimed at addressing the tumor heterogeneity, the interaction with the microenvironment, as well as diverse posology adjustments, are needed-which might unravel the therapeutic efficacy and response prediction of an RTK/PI3K/mTOR-based approach.
Collapse
Affiliation(s)
| | | | - Sabrina Di Bartolomeo
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, IS, Italy; (M.C.); (M.S.)
| |
Collapse
|
8
|
Duan Y, Haybaeck J, Yang Z. Therapeutic Potential of PI3K/AKT/mTOR Pathway in Gastrointestinal Stromal Tumors: Rationale and Progress. Cancers (Basel) 2020; 12:2972. [PMID: 33066449 PMCID: PMC7602170 DOI: 10.3390/cancers12102972] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/03/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal stromal tumor (GIST) originates from interstitial cells of Cajal (ICCs) in the myenteric plexus of the gastrointestinal tract. Most GISTs arise due to mutations of KIT and PDGFRA gene activation, encoding the receptor tyrosine kinase (RTK). The clinical use of the RTK inhibitor imatinib has significantly improved the management of GIST patients; however, imatinib resistance remains a challenge. The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway is a critical survival pathway for cell proliferation, apoptosis, autophagy and translation in neoplasms. Constitutive autophosphorylation of RTKs has an impact on the activation of the PI3K/AKT/mTOR pathway. In several preclinical and early-stage clinical trials PI3K/AKT/mTOR signaling inhibition has been considered as a promising targeted therapy strategy for GISTs. Various inhibitory drugs targeting different parts of the PI3K/AKT/mTOR pathway are currently being investigated in phase I and phase II clinical trials. This review highlights the progress for PI3K/AKT/mTOR-dependent mechanisms in GISTs, and explores the relationship between mTOR downstream signals, in particular, eukaryotic initiation factors (eIFs) and the development of GISTs, which may be instrumental for identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Yi Duan
- Department of Pathology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China;
| | - Johannes Haybaeck
- Department of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Diagnostic & Research Center for Molecular BioMedicine, Institute of Pathology, Medical University of Graz, 8010 Graz, Austria
| | - Zhihui Yang
- Department of Pathology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China;
| |
Collapse
|
9
|
Proteomic Analysis of Human Endometrial Tissues Reveals the Roles of PI3K/AKT/mTOR Pathway and Tumor Angiogenesis Molecules in the Pathogenesis of Endometrial Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5273969. [PMID: 32908897 PMCID: PMC7463404 DOI: 10.1155/2020/5273969] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023]
Abstract
As one major gynecological malignancy, endometrial cancer (EC) has been widely studied recently. However, its pathogenesis is still unclear to date. In this study, we identified differentially expressed proteins between 30 endometrial cancer tissues and 30 matched normal controls using 2D LC-MS/MS quantitative proteomics. As a result, we identified 619 differentially expressed proteins among all 2521 proteins being quantified. Further analyses suggested that the changes of fat, amino acid metabolism, peroxisome, extracellular signal, cytoskeleton, and other signaling or metabolic pathways may be closely related to the development of this cancer. Particularly, the PI3K/AKT/mTOR pathway-related molecules including PI3K and mTOR, ERK (the molecule of the ERK pathway), SPP1, and ANGPT2 (angiogenesis-related molecules) are highly associated with the pathogenesis of EC, which were reconfirmed by western blot and immunohistochemistry (IHC) analysis. In summary, our study revealed that the PI3K/AKT/mTOR pathway and tumor angiogenesis molecules contribute to the pathogenesis of endometrial cancer.
Collapse
|
10
|
Yao J, Xu M, Liu Z. Rapamycin inhibits proliferation and apoptosis of retinoblastoma cells through PI3K/AKT signaling pathway. Oncol Lett 2020; 19:2950-2956. [PMID: 32218850 PMCID: PMC7068238 DOI: 10.3892/ol.2020.11363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/03/2019] [Indexed: 11/23/2022] Open
Abstract
Effects of Rapamycin on the proliferation and apoptosis of retinoblastoma cells through the phosphatidylinositol 3-hydroxy kinase (PI3K)/protein kinase B (AKT) signaling pathway were studied. The retinoblastoma Y79 cells were selected and divided into negative control group (NC group), 0.2 µM Rapamycin group and 0.4 µM Rapamycin group. Then the proliferative activity of Y79 cells was detected using Cell Counting Kit-8 (CCK8) assay, the content of reactive oxygen species (ROS), malondialdehyde (MDA) and superoxide dismutase (SOD) in cells in each group was detected using enzyme-linked immunosorbent assay (ELISA), and the apoptosis of Y79 cells was detected via terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay. Moreover, the changes in Y79 cell cycle and apoptosis were determined through flow cytometry, and apoptosis and PI3K/AKT pathway were detected using reverse transcription-polymerase chain reaction (RT-PCR) and western blotting. It was found that the number of cells and the proliferative activity were significantly reduced in 0.2 µM Rapamycin group and 0.4 µM Rapamycin group. In 0.2 µM Rapamycin group and 0.4 µM Rapamycin group, the content of ROS and MDA was significantly decreased, while that of SOD was notably increased. TUNEL assay and flow cytometry showed that in 0.2 µM Rapamycin group and 0.4 µM Rapamycin group, the number of apoptotic cells was obviously increased, and the cell cycle was basically arrested in S phase. The expression levels of Bcl-2, PI3K and AKT declined in 0.2 µM Rapamycin group and 0.4 µM Rapamycin group, whereas the expression of Caspase 8 increased. Similar results were also obtained in the protein assay. The above results were significantly superior in 0.4 µM Rapamycin group to those in 0.2 µM Rapamycin group. Rapamycin inhibits proliferation and promotes apoptosis of retinoblastoma cells through inhibiting the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Jun Yao
- Department of Ophthalmology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Min Xu
- Department of Otorhinolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Xian Jiaotong University, Xian, Shaanxi 710004, P.R. China
| | - Ziyao Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Xian Jiaotong University, Xian, Shaanxi 710004, P.R. China
| |
Collapse
|
11
|
Jiang P, Huang S, Fu Z, Sun Z, Lakowski TM, Hu P. Deep graph embedding for prioritizing synergistic anticancer drug combinations. Comput Struct Biotechnol J 2020; 18:427-438. [PMID: 32153729 PMCID: PMC7052513 DOI: 10.1016/j.csbj.2020.02.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 02/07/2020] [Accepted: 02/09/2020] [Indexed: 12/11/2022] Open
Abstract
Drug combinations are frequently used for the treatment of cancer patients in order to increase efficacy, decrease adverse side effects, or overcome drug resistance. Given the enormous number of drug combinations, it is cost- and time-consuming to screen all possible drug pairs experimentally. Currently, it has not been fully explored to integrate multiple networks to predict synergistic drug combinations using recently developed deep learning technologies. In this study, we proposed a Graph Convolutional Network (GCN) model to predict synergistic drug combinations in particular cancer cell lines. Specifically, the GCN method used a convolutional neural network model to do heterogeneous graph embedding, and thus solved a link prediction task. The graph in this study was a multimodal graph, which was constructed by integrating the drug-drug combination, drug-protein interaction, and protein-protein interaction networks. We found that the GCN model was able to correctly predict cell line-specific synergistic drug combinations from a large heterogonous network. The majority (30) of the 39 cell line-specific models show an area under the receiver operational characteristic curve (AUC) larger than 0.80, resulting in a mean AUC of 0.84. Moreover, we conducted an in-depth literature survey to investigate the top predicted drug combinations in specific cancer cell lines and found that many of them have been found to show synergistic antitumor activity against the same or other cancers in vitro or in vivo. Taken together, the results indicate that our study provides a promising way to better predict and optimize synergistic drug pairs in silico.
Collapse
Key Words
- ACC, accuracy
- AUC, area under the curve
- CNN, convolutional neural network
- Cancer
- Cell line
- DDS, drug-drug synergy
- DNN, deep neural network
- DTI, drug-target interaction
- ER, estrogen receptor
- FPR, false positive rate
- GBM, glioblastoma multiforme
- GCN, graph convolutional network
- Graph convolutional network
- HTS, high throughput screening
- Heterogenous network
- PPI, protein–protein interaction
- RF, random forest
- ROC, receiver operating characteristic
- SD, standard deviation
- SVM, support vector machine
- Synergistic drug combination
- TNBC, triple negative breast cancer
- TPR, true positive rate
- XGBoost, extreme gradient boosting
Collapse
Affiliation(s)
- Peiran Jiang
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
- Department of Bioinformatics & Systems Biology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shujun Huang
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada
| | - Zhenyuan Fu
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zexuan Sun
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
- School of Mathematics and Statistic, Wuhan University, Wuhan 430072, China
| | - Ted M. Lakowski
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
- Research Institute in Oncology and Hematology, CancerCare Manitoba, Winnipeg R3E 0V9, Canada
| |
Collapse
|
12
|
Chatani PD, Agarwal SK, Sadowski SM. Molecular Signatures and Their Clinical Utility in Pancreatic Neuroendocrine Tumors. Front Endocrinol (Lausanne) 2020; 11:575620. [PMID: 33537001 PMCID: PMC7848028 DOI: 10.3389/fendo.2020.575620] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/30/2020] [Indexed: 12/23/2022] Open
Abstract
Pancreatic neuroendocrine tumors (PNETs) are classified based on their histologic differentiation and proliferative indices, which have been used extensively to determine prognosis. Advances in next-generation sequencing and other high-throughput techniques have allowed researchers to objectively explore tumor specimens and learn about the genetic alterations associated with malignant transformation in PNETs. As a result, targeted, pathway-specific therapies have been emerging for the treatment of unresectable and metastatic disease. As we continue to trial various pharmaceutical products, evidence from studies using multi-omics approaches indicates that clinical aggressiveness stratifies along other genotypic and phenotypic demarcations, as well. In this review, we explore the clinically relevant and potentially targetable molecular signatures of PNETs, their associated trials, and the overall differences in reported prognoses and responses to existing therapies.
Collapse
Affiliation(s)
- Praveen Dilip Chatani
- Endocrine Surgery Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sunita Kishore Agarwal
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Samira Mercedes Sadowski
- Endocrine Surgery Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Samira Mercedes Sadowski,
| |
Collapse
|
13
|
Munster P, Mita M, Mahipal A, Nemunaitis J, Massard C, Mikkelsen T, Cruz C, Paz-Ares L, Hidalgo M, Rathkopf D, Blumenschein G, Smith DC, Eichhorst B, Cloughesy T, Filvaroff EH, Li S, Raymon H, de Haan H, Hege K, Bendell JC. First-In-Human Phase I Study Of A Dual mTOR Kinase And DNA-PK Inhibitor (CC-115) In Advanced Malignancy. Cancer Manag Res 2019; 11:10463-10476. [PMID: 31853198 PMCID: PMC6916675 DOI: 10.2147/cmar.s208720] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 09/16/2019] [Indexed: 11/23/2022] Open
Abstract
Purpose This first-in-human Phase I study investigated the safety, pharmacokinetics (PK), pharmacodynamic profile, and preliminary efficacy of CC-115, a dual inhibitor of mammalian target of rapamycin (mTOR) kinase and DNA-dependent protein kinase. Patients and Methods Patients with advanced solid or hematologic malignancies were enrolled in dose-finding and cohort expansion phases. In dose-finding, once-daily or twice-daily (BID) ascending oral doses of CC-115 (range: 0.5-40 mg/day) in 28-day continuous cycles identified the maximum-tolerated dose for cohort expansion in 5 specified tumor types. Twelve additional patients with mixed solid tumors participated in a bioavailability substudy. Results Forty-four patients were enrolled in the dose-finding cohort. Dose-limiting toxicity included thrombocytopenia, stomatitis, hyperglycemia, asthenia/fatigue, and increased transaminases. CC-115 10 mg BID was selected for cohort expansion (n=74) in which fatigue, nausea, and decreased appetite were the most frequent toxicities. Dose-proportional PK was found. CC-115 distributed to glioblastoma tissue (mean tumor/plasma concentration ratio: 0.713). Total exposure of CC-115 was similar under fasting and fed conditions. A patient with endometrial carcinoma remained in complete remission >4 years. Partial response (PR; n=2) and stable disease (SD; n=4) were reported in the bioavailability substudy; SD was reached in 53%, 22%, 21%, and 64% of patients with head and neck squamous cell carcinoma, Ewing sarcoma, glioblastoma multiforme, and castration-resistant prostate cancer, respectively. Chronic lymphocytic leukemia/small lymphocytic lymphoma showed 38% PR and 25% SD. Conclusion CC-115 was well-tolerated, with toxicities consistent with mTOR inhibitors. Together with biomarker inhibition and preliminary efficacy, oral CC-115 10 mg BID is a promising novel anticancer treatment. Clinical trial registration NCT01353625.
Collapse
Affiliation(s)
- Pamela Munster
- UCSF Helen Diller Family Comprehensive Cancer Center, Department of Medicine, San Francisco, CA, USA
| | - Monica Mita
- Samuel Oschin Comprehensive Cancer Institute, Internal Medicine Medical Oncology Department, Los Angeles, CA, USA
| | - Amit Mahipal
- Mayo Clinic, Medical Oncology Department, Rochester, MN, USA
| | - John Nemunaitis
- University of Toledo College of Medicine and Life Sciences, Hematology/Oncology Department, Toledo, OH, USA
| | | | - Tom Mikkelsen
- Henry Ford Health System, Neurology Department, Detroit, MI, USA
| | - Cristina Cruz
- Vall d'Hebron University Hospital, Medical Oncology Department, Barcelona, Spain
| | - Luis Paz-Ares
- University Hospital 12 de Octubre, CNIO, Universidad Complutense & Ciberonc, Medical Oncology Department, Madrid, Spain
| | - Manuel Hidalgo
- Centro Integral Oncológico Clara Campal, Oncology Department, Madrid, Spain
| | - Dana Rathkopf
- Memorial Sloan Kettering Cancer Center, Department of Medicine, New York, NY, USA
| | - George Blumenschein
- The University of Texas MD Anderson Cancer Center, Department of Thoracic/Head and Neck Medical Oncology, Houston, TX, USA
| | - David C Smith
- University of Michigan, Urology Oncology, Ann Arbor, MI, USA
| | - Barbara Eichhorst
- University Hospital of Cologne, Department I for Internal Medicine, Cologne, Germany
| | - Tim Cloughesy
- David Geffen School of Medicine, Neurology Department, UCLA, Los Angeles, CA, USA
| | - Ellen H Filvaroff
- Celgene Corporation, Translational Development Department, San Francisco, CA, USA
| | - Shaoyi Li
- Celgene Corporation, Department of Statistics, Summit, NJ, USA
| | - Heather Raymon
- Celgene Corporation, Department of Pharmacology, San Diego, CA, USA
| | - Hans de Haan
- Celgene Corporation, Translational Development Department, San Francisco, CA, USA
| | - Kristen Hege
- Celgene Corporation, Translational Development Department, San Francisco, CA, USA
| | - Johanna C Bendell
- Sarah Cannon Research Institute, Drug Development Unit, Tennessee Oncology, Nashville, TN, USA
| |
Collapse
|
14
|
Hadavi R, Mohammadi-Yeganeh S, Razaviyan J, Koochaki A, Kokhaei P, Bandegi A. Expression of Bioinformatically Candidate miRNAs including, miR-576-5p, miR-501-3p and miR-3143, Targeting PI3K Pathway in Triple-Negative Breast Cancer. Galen Med J 2019; 8:e1646. [PMID: 34466540 PMCID: PMC8343935 DOI: 10.31661/gmj.v8i0.1646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/03/2019] [Accepted: 09/01/2019] [Indexed: 12/21/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) is an invasive and lethal form of breast cancer. PI3K pathway, which often activated in TNBC patients, can be a target of miRNAs. The purpose of this study was bioinformatic prediction of miRNAs targeting the key genes of this pathway and evaluation of the expression of them and their targets in TNBC. Materials and Methods We predicted miRNAs targeting PIK3CA and AKT1 genes using bioinformatics tools. Extraction of total RNA, synthesis of cDNA and quantitative real-time polymerase chain reaction were performed from 18 TNBC samples and normal adjacent tissues and cell lines. Results Our results demonstrated that miR-576-5p, miR-501-3p and miR-3143 were predicted to target PIK3CA, AKT1 and both of these mRNAs, respectively and were down-regulated while their target mRNAs were up-regulated in clinical samples and cell lines. The analysis of the receiver operating characteristic curve was done for the evaluation of the diagnostic value of predicted miRNAs in TNBC patients. Conclusion The findings of our study demonstrated the reverse correlation between miRNAs and their target genes and therefore the possibility of these miRNAs to be proposed as new candidates for TNBC targeted therapies.
Collapse
Affiliation(s)
- Razie Hadavi
- Department of Biochemistry, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Samira Mohammadi-Yeganeh
- Medical Nanotechnology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Razaviyan
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Molecular Biology and Genetic Engineering, Stem Cell Technology Research Center, Tehran, Iran
| | - Ameneh Koochaki
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parviz Kokhaei
- Cancer Research Center and Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Ahmadreza Bandegi
- Department of Biochemistry, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Research Center of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Correspondence to: Ahmadreza Bandegi, Damghan Road, Semnan University of Medical Sciences, Semnan, Iran Telephone Number: 33654162-023 Email Address: .
| |
Collapse
|
15
|
Huang T, Wang M, Huang B, Chang A, Liu F, Zhang Y, Jiang B. Long noncoding RNAs in the mTOR signaling network: biomarkers and therapeutic targets. Apoptosis 2019; 23:255-264. [PMID: 29556906 DOI: 10.1007/s10495-018-1453-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
As an evolutionarily conserved serine/threonine kinase of the phosphoinositide 3-kinase (PI3K) related kinase family, the mechanistic/mammalian target of rapamycin (mTOR) plays vital roles in the PI3K/AKT/mTOR pathway, participating in different cellular processes including cell survival, metabolism and proliferation. Aberrant activity of this signaling pathway may lead to oncogenesis. Over the last two decades, great progress has been made in the understanding of mTOR activation and how its response is counteracted for maintaining tissue homeostasis. Besides regulatory proteins and microRNAs, long noncoding RNA (lncRNA) is another emerging critical layer of the intricate modulatory architecture for the control of the mTOR signaling circuit. Also, the production of numerous lncRNAs is induced by mTOR treatment. These findings offer new perspectives for designing novel diagnostic and therapeutic strategies. In this review, we summarize the interactions between the mTOR signaling pathway and lncRNAs in the development and progression of various types of tumors, focusing on the mechanisms of these interactions, and also discuss the potential use of lncRNAs as biomarkers and therapeutic targets for malignancies.
Collapse
Affiliation(s)
- Tinglei Huang
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201900, China
| | - Meiling Wang
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201900, China
| | - Bo Huang
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201900, China
| | - Augustus Chang
- Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA
| | - Feng Liu
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201900, China
| | - Yanjie Zhang
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201900, China.
| | - Bin Jiang
- Oncology Department, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201900, China.
| |
Collapse
|
16
|
Rosenberg Y, Doniger T, Levy O. Sustainability of coral reefs are affected by ecological light pollution in the Gulf of Aqaba/Eilat. Commun Biol 2019; 2:289. [PMID: 31396569 PMCID: PMC6683144 DOI: 10.1038/s42003-019-0548-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 07/15/2019] [Indexed: 01/12/2023] Open
Abstract
As human populations grow and lighting technologies improve, artificial light gradually alters natural cycles of light and dark that have been consistent over long periods of geological and evolutionary time. While considerable ecological implications of artificial light have been identified in both terrestrial and aquatic habitats, knowledge about the physiological and molecular effects of light pollution is vague. To determine if ecological light pollution (ELP) impacts coral biological processes, we characterized the transcriptome of the coral Acropora eurystoma under two different light regimes: control conditions and treatment with light at night. Here we show that corals exposed to ELP have approximately 25 times more differentially expressed genes that regulate cell cycle, cell proliferation, cell growth, protein synthesis and display changes in photo physiology. The finding of this work confirms that ELP acts as a chronic disturbance that may impact the future of coral reefs.
Collapse
Affiliation(s)
- Yael Rosenberg
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 52900 Israel
| | - Tirza Doniger
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 52900 Israel
| | - Oren Levy
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 52900 Israel
| |
Collapse
|
17
|
Chang MM, Pan BS, Wang CY, Huang BM. Cordycepin-induced unfolded protein response-dependent cell death, and AKT/MAPK-mediated drug resistance in mouse testicular tumor cells. Cancer Med 2019; 8:3949-3964. [PMID: 31145545 PMCID: PMC6639181 DOI: 10.1002/cam4.2285] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 04/19/2019] [Accepted: 05/10/2019] [Indexed: 02/06/2023] Open
Abstract
Testicular cancer is the most commonly diagnosed cancer in men at 15-44 years of age, and radical orchidectomy combined with chemotherapy is currently considered as the standard treatment. However, drugs resistance and side effects that impact the quality of life for patients with testicular cancer have not been markedly improved in recent decades. In this study, we characterized the pharmacological exacerbation of the unfolded protein response (UPR), which is an effective approach to kill testicular cancer cells, by carrying out a clustering analysis of mRNA expression profiles and the immunobloting examination of cordycepin-treated MA-10 cells. The UPR is executed in response to endoplasmic reticulum stress to complement by an apoptotic response if the defect cannot be resolved. Results showed that cordycepin significantly modulated FoxO/P15/P27, PERK-eIF2α (apoptotic), and the IRE1-XBP1 (adaptive) UPR pathways. Interestingly, a fraction of MA-10 cells survived after cordycepin treatment, the AKT, LC3 I/II, and MAPK signaling pathways were highly induced in attached cells as compared to the suspended cells, illustrating the drug resistance to cordycepin via activating AKT and MAPK pathways in MA-10 cells. In summary, PERK-eIF2α signaling pathway is required for pro-apoptotic UPR in MA-10 cell death following cordycepin treatment, suggesting a potential therapeutic application in treating testicular cancer. However, activation of AKT and MAPK pathways could possibly result in drug resistance to cordycepin in MA-10 cells.
Collapse
Affiliation(s)
- Ming-Min Chang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Bo-Syong Pan
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Chia-Yih Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Bu-Miin Huang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, Republic of China
| |
Collapse
|
18
|
Gupta I, Sareyeldin RM, Al-Hashimi I, Al-Thawadi HA, Al Farsi H, Vranic S, Al Moustafa AE. Triple Negative Breast Cancer Profile, from Gene to microRNA, in Relation to Ethnicity. Cancers (Basel) 2019; 11:cancers11030363. [PMID: 30871273 PMCID: PMC6468678 DOI: 10.3390/cancers11030363] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the most frequent cause of cancer-related deaths among women worldwide. It is classified into four major molecular subtypes. Triple-negative breast cancers (TNBCs), a subgroup of breast cancer, are defined by the absence of estrogen and progesterone receptors and the lack of HER-2 expression; this subgroup accounts for ~15% of all breast cancers and exhibits the most aggressive metastatic behavior. Currently, very limited targeted therapies exist for the treatment of patients with TNBCs. On the other hand, it is important to highlight that knowledge of the molecular biology of breast cancer has recently changed the decision-making process regarding the course of cancer therapies. Thus, a number of new techniques, such as gene profiling and sequencing, proteomics, and microRNA analysis have been used to explore human breast carcinogenesis and metastasis including TNBC, which consequently could lead to new therapies. Nevertheless, based on evidence thus far, genomics profiles (gene and miRNA) can differ from one geographic location to another as well as in different ethnic groups. This review provides a comprehensive and updated information on the genomics profile alterations associated with TNBC pathogenesis associated with different ethnic backgrounds.
Collapse
Affiliation(s)
- Ishita Gupta
- College of Medicine, Qatar University, Doha P. O. Box:2713, Qatar.
| | | | - Israa Al-Hashimi
- College of Medicine, Qatar University, Doha P. O. Box:2713, Qatar.
| | | | - Halema Al Farsi
- College of Medicine, Qatar University, Doha P. O. Box:2713, Qatar.
| | - Semir Vranic
- College of Medicine, Qatar University, Doha P. O. Box:2713, Qatar.
| | - Ala-Eddin Al Moustafa
- College of Medicine, Qatar University, Doha P. O. Box:2713, Qatar.
- Biomedical Research Centre, Qatar University, Doha P.O Box: 2713, Qatar.
| |
Collapse
|
19
|
Wu CE, Chen MH, Yeh CN. mTOR Inhibitors in Advanced Biliary Tract Cancers. Int J Mol Sci 2019; 20:E500. [PMID: 30682771 PMCID: PMC6386826 DOI: 10.3390/ijms20030500] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 01/19/2019] [Accepted: 01/22/2019] [Indexed: 12/19/2022] Open
Abstract
Patients with advanced biliary tract cancers (BTCs), including cholangiocarcinoma (CCA), have poor prognosis so novel treatment is warranted for advanced BTC. In current review, we discuss the limitations of current treatment in BTC, the importance of mTOR signalling in BTC, and the possible role of mTOR inhibitors as a future treatment in BTC. Chemotherapy with gemcitabine-based chemotherapy is still the standard of care and no targeted therapy has been established in advanced BTC. PI3K/AKT/mTOR signaling pathway linking to several other pathways and networks regulates cancer proliferation and progression. Emerging evidences reveal mTOR activation is associated with tumorigenesis and drug-resistance in BTC. Rapalogs, such as sirolimus and everolimus, partially inhibit mTOR complex 1 (mTORC1) and exhibit anti-cancer activity in vitro and in vivo in BTC. Rapalogs in clinical trials demonstrate some activity in patients with advanced BTC. New-generation mTOR inhibitors against ATP-binding pocket inhibit both TORC1 and TORC2 and demonstrate more potent anti-tumor effects in vitro and in vivo, however, prospective clinical trials are warranted to prove its efficacy in patients with advanced BTC.
Collapse
Affiliation(s)
- Chao-En Wu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou branch, Chang Gung University, Taoyuan 333, Taiwan.
| | - Ming-Huang Chen
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan.
- Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan.
| | - Chun-Nan Yeh
- Department of General Surgery and Liver Research Center, Chang Gung Memorial Hospital, Linkou branch, Chang Gung University, Taoyuan 333, Taiwan.
| |
Collapse
|
20
|
Li XG, Du JH, Lu Y, Lin XJ. Neuroprotective effects of rapamycin on spinal cord injury in rats by increasing autophagy and Akt signaling. Neural Regen Res 2019; 14:721-727. [PMID: 30632514 PMCID: PMC6352584 DOI: 10.4103/1673-5374.247476] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Rapamycin treatment has been shown to increase autophagy activity and activate Akt phosphorylation, suppressing apoptosis in several models of ischemia reperfusion injury. However, little has been studied on the neuroprotective effects on spinal cord injury by activating Akt phosphorylation. We hypothesized that both effects of rapamycin, the increased autophagy activity and Akt signaling, would contribute to its neuroprotective properties. In this study, a compressive spinal cord injury model of rat was created by an aneurysm clip with a 30 g closing force. Rat models were intraperitoneally injected with rapamycin 1 mg/kg, followed by autophagy inhibitor 3-methyladenine 2.5 mg/kg and Akt inhibitor IV 1 µg/kg. Western blot assay, immunofluorescence staining and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay were used to observe the expression of neuronal autophagy molecule Beclin 1, apoptosis-related molecules Bcl-2, Bax, cytochrome c, caspase-3 and Akt signaling. Our results demonstrated that rapamycin inhibited the expression of mTOR in injured spinal cord tissue and up-regulated the expression of Beclin 1 and phosphorylated-Akt. Rapamycin prevented the decrease of bcl-2 expression in injured spinal cord tissue, reduced Bax, cytochrome c and caspase-3 expression levels and reduced the number of apoptotic neurons in injured spinal cord tissue 24 hours after spinal cord injury. 3-Methyladenine and Akt inhibitor IV intervention suppressed the expression of Beclin-1 and phosphorylated-Akt in injured spinal cord tissue and reduced the protective effect of rapamycin on apoptotic neurons. The above results indicate that the neuroprotective effect of rapamycin on spinal cord injury rats can be achieved by activating autophagy and the Akt signaling pathway.
Collapse
Affiliation(s)
- Xi-Gong Li
- Department of Orthopedic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jun-Hua Du
- Department of Orthopedic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yang Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Xiang-Jin Lin
- Department of Orthopedic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
21
|
Schram AM, Gandhi L, Mita MM, Damstrup L, Campana F, Hidalgo M, Grande E, Hyman DM, Heist RS. A phase Ib dose-escalation and expansion study of the oral MEK inhibitor pimasertib and PI3K/MTOR inhibitor voxtalisib in patients with advanced solid tumours. Br J Cancer 2018; 119:1471-1476. [PMID: 30425349 PMCID: PMC6288157 DOI: 10.1038/s41416-018-0322-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/17/2018] [Accepted: 10/12/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND This phase Ib study evaluated the safety, maximum-tolerated dose (MTD), pharmacokinetics, pharmacodynamics, and preliminary efficacy of pimasertib (MSC1936369B), a MEK1/2 inhibitor, in combination with voxtalisib (SAR245409), a pan-PI3K and mTORC1/mTORC2 inhibitor, in patients with advanced solid tumours. METHODS This study included a dose escalation and expansion in patients with select tumour types and alterations in the MAPK or PI3K pathways. A 3 + 3 design was used to determine MTD. Patients were evaluated for adverse events and tumour response. RESULTS 146 patients were treated, including 63 in dose escalation and 83 in expansion. The MTD was pimasertib 90 mg and voxtalisib 70 mg daily. Based on the safety profile, the recommended phase 2 dose (RP2D) was pimasertib 60 mg and voxtalisib 70 mg. The most frequent treatment-emergent adverse events (TEAEs) were diarrhoea (75%), fatigue (57%), and nausea (50%). Responses included a complete response in one patient (1%), partial response in five (5%), and stable disease in 51 (46%). At the RP2D, 74 patients required dose interruption (73%), 20 required dose reduction (20%), and 26 discontinued treatment due to TEAEs (26%). CONCLUSIONS The combination of pimasertib and voxtalisib showed poor long-term tolerability and limited anti-tumour activity in patients with advanced solid tumours.
Collapse
Affiliation(s)
| | - Leena Gandhi
- New York University Perlmutter Cancer Center, New York, NY, USA
| | | | | | | | | | | | - David M Hyman
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
22
|
Gao JX, Li Y, Wang SN, Chen XC, Lin LL, Zhang H. Overexpression of microRNA-183 promotes apoptosis of substantia nigra neurons via the inhibition of OSMR in a mouse model of Parkinson's disease. Int J Mol Med 2018; 43:209-220. [PMID: 30431059 PMCID: PMC6257840 DOI: 10.3892/ijmm.2018.3982] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 10/24/2018] [Indexed: 12/13/2022] Open
Abstract
The present study aimed to investigate the effect of microRNA-183 (miR-183) on substantia nigra neurons by targeting oncostatin M receptor (OSMR) in a mouse model of Parkinson’s disease (PD). The positive expression rates of OSMR and the apoptosis of substantia nigra neurons were detected by immunohistochemistry and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling, respectively. Substantia nigra neurons in normal and PD mice were cultured in vitro. The association between miR-183 and OSMR was verified using a dual luciferase reporter gene assay. The expression of miR-183 and the phosphoinositide 3-kinase-Akt signaling pathway-associated genes were detected by reverse transcription-quantitative polymerase chain reaction and western blot analysis, respectively. Cell apoptosis was detected by flow cytometry. OSMR is the target gene of miR-183. The number of OSMR-positive cells and the apoptotic rate of substantia nigra neurons were increased in the PD group. Neurons transfected with miR-183 mimic exhibited elevated expression levels of miR-183, B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax) and caspase-9 and increased apoptotic rate, and reduced expression levels of OSMR, Akt, phosphorylated (p-)Akt, glycogen synthase kinase-3 (GSK-3β), p-GSK-3β, Bcl-2, insulin-like growth factor 1 (IGF-1), mammalian target of rapamycin (mTOR) and p-mTOR. The miR-183 inhibitor decreased the expression levels of miR-183, Bax and caspase-9 and the apoptotic rate; however, increased the expression of OSMR, Akt, p-Akt, GSK-3β, p-GSK-3β, Bcl-2, IGF-1, mTOR and p-mTOR. The results of the present study provide evidence that the overexpression of miR-183 promotes the apoptosis of substantia nigra neurons by inhibiting the expression of OSMR.
Collapse
Affiliation(s)
- Jin-Xia Gao
- Department of Anesthesiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Yu Li
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Sai-Nan Wang
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Xing-Chi Chen
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Lu-Lu Lin
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Hui Zhang
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| |
Collapse
|
23
|
Son JY, Yoon S, Tae IH, Park YJ, De U, Jeon Y, Park YJ, Rhyu IJ, Lee BM, Chung K, Lim JE, Lee SJ, Lee HW, Kwak JH, Kim HS, Choi HY. Novel therapeutic roles of MC-4 in combination with everolimus against advanced renal cell carcinoma by dual targeting of Akt/pyruvate kinase muscle isozyme M2 and mechanistic target of rapamycin complex 1 pathways. Cancer Med 2018; 7:5083-5095. [PMID: 30160042 PMCID: PMC6198234 DOI: 10.1002/cam4.1748] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/08/2018] [Accepted: 08/05/2018] [Indexed: 12/11/2022] Open
Abstract
Current clinical trials of new anticancer therapies against metastatic renal cell carcinoma (RCC), including molecular-targeted therapies, have not shown promise. The purpose of this study was to preclinically assess the antitumor effects of MC-4, a partially purified material of Artemisia annua L., as a monotherapy or in combination with the known mechanistic target of rapamycin complex 1 (mTORC1) inhibitor, everolimus, against Caki-1 (Von Hippel-Lindau (VHL)+/+) and 786-O (VHL-/-) human RCC cells. MC-4 monotherapy significantly increased tumor growth inhibition and autophagic cell death in RCC cells in vitro and in vivo. Everolimus led to compensatory Akt activation by inhibiting only mTORC1 signaling pathway. In contrast to everolimus, MC-4 enhanced phosphatase and tensin homolog expression and reduced its downstream effector, Akt/pyruvate kinase muscle isozyme M2 (PKM2), leading to decreased expression of glucose transporter 1, which is associated with cancer cell metabolism. The synergistic antitumor and anti-metastatic effects induced by co-administration of MC-4 and everolimus involve cell growth inhibition and autophagic cell death via dual targeting of phosphatidylinositol 3-kinase (PI3K)/Akt/PKM2 and mTORC1. These findings suggest that MC-4 is a novel Akt/PKM2 inhibitor that can overcome the limitation of existing mTOR inhibitors and can be considered a novel strategy to treat patients with rapidly progressing advanced RCC.
Collapse
Affiliation(s)
- Ji Yeon Son
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | - Sungpil Yoon
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | - In Hwan Tae
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | - Yu Jin Park
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | - Umasankar De
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | | | | | - Im Joo Rhyu
- Department of AnatomyKorea University College of MedicineSeoulKorea
| | - Byung Mu Lee
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | | | - Joung Eun Lim
- Departments of UrologySamsung Medical CenterSungkyunkwan University School of MedicineSeoulKorea
| | - Se Jeong Lee
- Department of AnatomyKorea University College of MedicineSeoulKorea
| | - Hye Won Lee
- Departments of UrologySamsung Medical CenterSungkyunkwan University School of MedicineSeoulKorea
- Department of Anatomy and Cell BiologySungkyunkwan University School of MedicineSuwonKorea
- Single Cell Network Research CenterSungkyunkwan University School of MedicineSuwonKorea
| | | | | | - Han Yong Choi
- Department of UrologyKangbuk Samsung HospitalSeoulKorea
| |
Collapse
|
24
|
Li X, Tang Y, Yu F, Sun Y, Huang F, Chen Y, Yang Z, Ding G. Inhibition of Prostate Cancer DU-145 Cells Proliferation by Anthopleura anjunae Oligopeptide (YVPGP) via PI3K/AKT/mTOR Signaling Pathway. Mar Drugs 2018; 16:E325. [PMID: 30208576 PMCID: PMC6165336 DOI: 10.3390/md16090325] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022] Open
Abstract
We investigated the antitumor mechanism of Anthopleura anjunae oligopeptide (AAP-H, YVPGP) in prostate cancer DU-145 cells in vitro and in vivo. Results indicated that AAP-H was nontoxic and exhibited antitumor activities. Cell cycle analysis indicated that AAP-H may arrest DU-145 cells in the S phase. The role of the phosphatidylinositol 3-kinase/protein kinase B/mammalian rapamycin target protein (PI3K/AKT/mTOR) signaling pathway in the antitumor mechanism of APP-H was investigated. Results showed that AAP-H treatment led to dose-dependent reduction in the levels of p-AKT (Ser473), p-PI3K (p85), and p-mTOR (Ser2448), whereas t-AKT and t-PI3K levels remained unaltered compared to the untreated DU-145 cells. Inhibition of PI3K/AKT/mTOR signaling pathway in the DU-145 cells by employing inhibitor LY294002 (10 μM) or rapamycin (20 nM) effectively attenuated AAP-H-induced phosphorylation of AKT and mTOR. At the same time, inhibitor addition further elevated AAP-H-induced cleaved-caspase-3 levels. Furthermore, the effect of AAP-H on tumor growth and the role of the PI3K/AKT/mTOR signaling pathway in nude mouse model were also investigated. Immunohistochemical analysis showed that activated AKT, PI3K, and mTOR levels were reduced in DU-145 xenografts. Western blotting showed that AAP-H treatment resulted in dose-dependent reduction in p-AKT (Ser473), p-PI3K (p85), and p-mTOR (Ser2448) levels, whereas t-AKT and t-PI3K levels remained unaltered. Similarly, Bcl-xL levels decreased, whereas that of Bax increased after AAP-H treatment. AAP-H also increased initiator (caspase 8 and 9) and executor caspase (caspase 3 and 7) levels. Therefore, the antitumor mechanism of APP-H on DU-145 cells may involve regulation of the PI3K/AKT/mTOR signaling pathway, which eventually promotes apoptosis via mitochondrial and death receptor pathways. Thus, the hydrophobic oligopeptide (YVPGP) can be developed as an adjuvant for the prevention or treatment of prostate cancer in the future.
Collapse
Affiliation(s)
- Xiaojuan Li
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Yunping Tang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Fangmiao Yu
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Yu Sun
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Zhejiang Ocean University Donghai Science and Technology College, Zhoushan 316000, China.
| | - Fangfang Huang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Yan Chen
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Zuisu Yang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Guofang Ding
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
- Zhejiang Marine Fisheries Research Institution, Zhoushan 316021, China.
| |
Collapse
|
25
|
Li XG, Wang YB. SRPK1 gene silencing promotes vascular smooth muscle cell proliferation and vascular remodeling via inhibition of the PI3K/Akt signaling pathway in a rat model of intracranial aneurysms. CNS Neurosci Ther 2018; 25:233-244. [PMID: 30101479 DOI: 10.1111/cns.13043] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/20/2018] [Accepted: 07/15/2018] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Intracranial aneurysm (IA) is a life threatening cerebrovascular disease characterized by phenotypic modulation of vascular smooth muscle cells (VSMCs) and loss of vessel cells. In addition to environmental factors, genetic factors have been proposed to be a critical factor in the onset and progression of IA. The present study investigates the effects of serine-arginine protein kinase 1 (SRPK1) on VSMC proliferation and apoptosis both in vivo and in vitro, as well as its role in vascular remodeling in vivo through PI3 K/Akt signaling in IA. METHODS Differentially expressed genes related to IA were initially identified using microarray analysis. Immunohistochemistry was conducted to determine SRPK1 expression in the vascular walls in IA and normal cerebral vascular walls. TUNEL staining were applied to observe cell apoptosis patterns of VSMCs. VSMC proliferation and apoptosis in vitro were detected by cell counting kit-8 (CCK8) assay and flow cytometry. The expressions of SRPK1, PI3 K/Akt signaling pathway- and apoptosis-related genes were evaluated by RT-qPCR and Western blot analysis. RESULTS Microarray data of GSE36791 and GSE54083 were analyzed to determine the selection of SRPK1 gene. The vascular walls in IA rat models produced high levels of SRPK1 expression and an activated PI3 K/Akt signaling pathway. VSMCs treated with siRNA-SRPK1 exhibited enhanced cell proliferation, repressed cell apoptosis, and increased vascular remodeling, all of which suggest the inhibition of the PI3 K/AKT pathway. Notably, PI3 K/AKT pathway reversed the effect of SRPK1 silencing. CONCLUSION Our results show that siRNA-mediated silencing of SRPK1 gene inhibits VSMC apoptosis, and increases VSMCs proliferation and vascular remodeling in IA via the PI3 K/Akt signaling pathway. Our findings provide a novel intervention target for the molecular treatment of IA.
Collapse
Affiliation(s)
- Xin-Guo Li
- Department of Neurosurgery, the First Hospital of China Medical University, Shenyang, China
| | - Yi-Bao Wang
- Department of Neurosurgery, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
26
|
KRAS mutant allele-specific expression knockdown in pancreatic cancer model with systemically delivered bi-shRNA KRAS lipoplex. PLoS One 2018; 13:e0193644. [PMID: 29851957 PMCID: PMC5979018 DOI: 10.1371/journal.pone.0193644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 02/15/2018] [Indexed: 12/18/2022] Open
Abstract
The KRAS oncogene, present in over 90% of pancreatic ductal adenocarcinomas, is most frequently the result of one of three gain-of-function substitution mutations of codon 12 glycine. Thus far, RAS mutations have been clinically refractory to both direct and selective inhibition by systemic therapeutics. This report presents the results of pre-clinical assessment of a lipoplex comprising a plasmid-encoded, modular bi-functional shRNA (bi-shRNA), which executes selective and multi-mutant allelic KRASG12mut gene silencing, encased within a fusogenic liposome systemic delivery vehicle. Using both a dual luciferase reporter system and a Restriction Fragment Length Polymorphism (RFLP) assay, selective discrimination of KRASG12mut from KRASwt was confirmed in vitro in PANC1 cells. Subsequently, systemic administration of the bi-shRNAKRAS fusogenic lipoplex into female athymic Nu/Nu mice bearing PANC1 xenografts demonstrated intratumoral plasmid delivery, KRASG12mut knockdown, and inhibition of tumor growth, without adverse effect. Clinical trials with the bi-shRNA lipoplex have been implemented.
Collapse
|
27
|
Abstract
The ribosome has long been considered as a consistent molecular factory, with a rather passive role in the translation process. Recent findings have shifted this obsolete view, revealing a remarkably complex and multifaceted machinery whose role is to orchestrate spatiotemporal control of gene expression. Ribosome specialization discovery has raised the interesting possibility of the existence of its malignant counterpart, an 'oncogenic' ribosome, which may promote tumor progression. Here we weigh the arguments supporting the existence of an 'oncogenic' ribosome and evaluate its role in cancer evolution. In particular, we provide an analysis and perspective on how the ribosome may play a critical role in the acquisition and maintenance of cancer stem cell phenotype.
Collapse
|
28
|
Mansini AP, Peixoto E, Thelen KM, Gaspari C, Jin S, Gradilone SA. The cholangiocyte primary cilium in health and disease. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1245-1253. [PMID: 28625917 PMCID: PMC5732091 DOI: 10.1016/j.bbadis.2017.06.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 06/08/2017] [Indexed: 12/14/2022]
Abstract
Cholangiocytes, like most cells, express primary cilia extending from their membranes. These organelles function as antennae which detect stimuli from bile and transmit the information into cells regulating several signaling pathways involved in secretion, proliferation and apoptosis. The ability of primary cilia to detect different signals is provided by ciliary associated proteins which are expressed in its membrane. Defects in the structure and/or function of these organelles lead to cholangiociliopathies that result in cholangiocyte hyperproliferation, altered fluid secretion and absorption. Since primary cilia dysfunction has been observed in several epithelial tumors, including cholangiocarcinoma (CCA), primary cilia have been proposed as tumor suppressor organelles. In addition, the loss of cilia is associated with dysregulation of several molecular pathways resulting in CCA development and progression. Thus, restoration of the primary cilia may be a potential therapeutic approach for several ciliopathies and CCA.
Collapse
Affiliation(s)
| | | | | | - Cesar Gaspari
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Sujeong Jin
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Sergio A Gradilone
- The Hormel Institute, University of Minnesota, Austin, MN, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
29
|
Kang FC, Wang SC, Chang MM, Pan BS, Wong KL, Cheng KS, So EC, Huang BM. Midazolam activates caspase, MAPKs and endoplasmic reticulum stress pathways, and inhibits cell cycle and Akt pathway, to induce apoptosis in TM3 mouse Leydig progenitor cells. Onco Targets Ther 2018; 11:1475-1490. [PMID: 29588601 PMCID: PMC5858853 DOI: 10.2147/ott.s154442] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Midazolam (MDZ) has powerful hypnosis, amnesia, anti-anxiety and anticonvulsant effects. Studies have shown that prenatally developmental toxicity of diazepam can be observed in many organs/tissues. However, it remains elusive in male reproductive system. Materials and methods TM3 mouse Leydig progenitor cell line was used to determine whether MDZ has any unfavorable effects. Results Midazolam significantly decreased cell viability in dose- and time-dependent manners in TM3 cells. In flow cytometry analysis, midazolam significantly increased subG1 phase cell numbers, and annexin V/PI double staining assay further confirmed that MDZ induced apoptosis in TM3 cells. Moreover, MDZ significantly induced the expression of caspase-8 and -3 proteins and the phosphorylation of JNK, ERK1/2 and p38. Besides, MDZ didn’t activate Akt pathway in TM3 cells. Furthermore, the expressions of p-EIF2α, ATF4, ATF3 and CHOP were induced by midazolam, suggesting that midazolam could induce apoptosis through endoplasmic reticulum (ER) stress in TM3 cells. Additionally, the expressions of cyclin A, cyclin B and CDK1 were inhibited by midazolam through the regulation of p53 in TM3 cells, indicating that midazolam could regulate cell cycle to induce apoptosis. Conclusion Midazolam could activate caspase, MAPKs and ER stress pathways and impede Akt pathway and cell cycle to induce apoptosis in TM3 mouse Leydig progenitor cells.
Collapse
Affiliation(s)
- Fu-Chi Kang
- Department of Anesthesia, Chi Mei Medical Center, Chiali, Tainan, Taiwan, Republic of China
| | - Shu-Chun Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Ming-Min Chang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Bo-Syong Pan
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Kar-Lok Wong
- Department of Anesthesia, China Medical University, Taichung, Taiwan, Republic of China
| | - Ka-Shun Cheng
- Department of Anesthesia, China Medical University, Taichung, Taiwan, Republic of China.,Department of Anesthesiology, The Qingdao University Yuhuangding Hospital, Yantai, Shandong, China
| | - Edmund Cheung So
- Department of Anesthesia, China Medical University, Taichung, Taiwan, Republic of China.,Department of Anesthesia, An Nan Hospital, China Medical University, Tainan, Taiwan, Republic of China
| | - Bu-Miin Huang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, Republic of China
| |
Collapse
|
30
|
Synergistic antitumor activity of the combination of salubrinal and rapamycin against human cholangiocarcinoma cells. Oncotarget 2018; 7:85492-85501. [PMID: 27863431 PMCID: PMC5356752 DOI: 10.18632/oncotarget.13408] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 10/27/2016] [Indexed: 12/20/2022] Open
Abstract
Less is known about the roles of eukaryotic initiation factor alpha (eIF2α) in cholangiocarcinoma (CCA). Here, we report that eIF2α inhibitor salubrinal inhibits the proliferation of human CCA cells. Clinical application of mammalian target of rapamycin (mTOR) inhibitors only has moderate antitumor efficacy. Therefore, combination approaches may be required for effective clinical use of mTOR inhibitors. Here, we investigated the efficacy of the combination of salubrinal and rapamycin in the treatment of CCA. Our data demonstrate a synergistic antitumor effect of the combination of salubrinal and rapamycin against CCA cells. Rapamycin significantly inhibits the proliferation of CCA cells. However, rapamycin initiates a negative feedback activation of Akt. Inhibition of Akt by salubrinal potentiates the efficacy of rapamycin both in vitro and in vivo. Additionally, rapamycin treatment results in the up-regulation of Bcl-xL in a xenograft mouse model. It is notable that salubrinal inhibits rapamycin-induced Bcl-xL up-regulation in vivo. Taken together, our data suggest that salubrinal and rapamycin combination might be a new and effective strategy for the treatment of CCA.
Collapse
|
31
|
Wang R, Lu Y, Li H, Sun L, Yang N, Zhao M, Zhang M, Shi Q. Antitumor activity of the Ailanthus altissima bark phytochemical ailanthone against breast cancer MCF-7 cells. Oncol Lett 2018; 15:6022-6028. [PMID: 29552229 DOI: 10.3892/ol.2018.8039] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 11/16/2017] [Indexed: 11/06/2022] Open
Abstract
Ailanthone is isolated from the bark of Ailanthus altissima (Mill.) Swingle (Simaroubaceae). The mechanism that underlies the activity of ailanthone on MCF-7 cells was investigated by MTT assay. Breast cancer MCF-7 cells were treated with 0.5, 1.0, 2.0, 4.0 and 8.0 µg/ml ailanthone for 24, 48 and 72 h. The inhibition of proliferation induced by treatment with ailanthone was assessed by MTT assay. Apoptosis and cell cycle distribution in MCF-7 cells with the same doses of ailanthone for 48 h were determined by flow cytometry. Expression of apoptosis-associated genes and proteins were analyzed by reverse transcription-polymerase chain reaction (RT-PCR) and western blot analysis, respectively. The results revealed that ailanthone inhibited MCF-7 cell proliferation. Flow cytometry assay demonstrated that ailanthone induced apoptosis and G0/G1 cell cycle arrest in MCF-7 cells. Western blotting and RT-PCR assays demonstrated that upregulation of pro-apoptotic caspase-3 and Bcl-associated X, and downregulation of anti-apoptotic apoptosis regulator B-cell lymphoma-2 in MCF-7 cells may be associated with the induction of apoptosis and inhibition of proliferation. To the best of our knowledge, the present study is the first to investigate the antitumor activity of ailanthone from A. altissima on MCF-7 cells and to attempt to elucidate the underlying mechanism. The present study revealed the presence of ailanthone-mediated antitumor effects, indicating that ailanthone may be a novel phytomedicine with potential use in breast cancer therapy.
Collapse
Affiliation(s)
- Ruxing Wang
- Department of Medicinal Natural Product Chemistry, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Hebei Province Key Laboratory of Research and Development for Chinese Medicine, Institute of Chinese Materia Medica, Chengde, Hebei 067000, P.R. China
| | - Yanjie Lu
- Department of Pathology, Institute of Cancer Research, Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Hong Li
- Department of Central Laboratory, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Lixin Sun
- Department of Central Laboratory, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Ning Yang
- Department of Central Laboratory, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Mingzhen Zhao
- Department of Central Laboratory, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Manli Zhang
- Department of Medicinal Natural Product Chemistry, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Qingwen Shi
- Department of Medicinal Natural Product Chemistry, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| |
Collapse
|
32
|
Alameen AAM, Simioni C, Martelli AM, Zauli G, Ultimo S, McCubrey JA, Gonelli A, Marisi G, Ulivi P, Capitani S, Neri LM. Healthy CD4+ T lymphocytes are not affected by targeted therapies against the PI3K/Akt/mTOR pathway in T-cell acute lymphoblastic leukemia. Oncotarget 2018; 7:55690-55703. [PMID: 27494886 PMCID: PMC5342446 DOI: 10.18632/oncotarget.10984] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/11/2016] [Indexed: 12/19/2022] Open
Abstract
An attractive molecular target for novel anti-cancer therapies is the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway which is commonly deregulated in many types of cancer. Nevertheless, the effects of PI3K/Akt/mTOR inhibitors on T lymphocytes, a key component of immune responses, have been seldom explored. In this study we investigated the effects on human CD4+ T-cells of a panel of PI3K/Akt/mTOR inhibitors: BGT226, Torin-2, MK-2206, and ZSTK474. We also assessed their efficacy against two acute leukemia T cell lines. T lymphocytes were stimulated with phytohemagglutinin. Inhibitor effects on cell cycle and apoptosis were analyzed by flow cytometry, while cytotoxicity was assessed by MTT assays. In addition, the activation status of the pathway as well as induction of autophagy were analyzed by Western blotting. Quiescent healthy T lymphocytes were unaffected by the drugs whereas mitogen-stimulated lymphocytes as well as leukemic cell lines displayed a cell cycle block, caspase-dependent apoptosis, and dephosphorylation of key components of the signaling pathway. Autophagy was also induced in proliferating lymphocytes and in JURKAT and MOLT-4 cell lines. When autophagy was inhibited by 3-methyladenine or Bafilomycin A1, drug cytotoxicity was increased, indicating that autophagy is a protective mechanism. Therefore, our findings suggest that PI3K/Akt/mTOR inhibitors preserve lymphocyte viability. This is a valuable result to be taken into account when selecting drugs for targeted cancer therapy in order to minimize detrimental effects on immune function.
Collapse
Affiliation(s)
- Ayman A M Alameen
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.,Department of Chemical Pathology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, Sudan
| | - Carolina Simioni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giorgio Zauli
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Simona Ultimo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Arianna Gonelli
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Giorgia Marisi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Silvano Capitani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.,LTTA Center, University of Ferrara, Ferrara, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
33
|
Raimondo L, D'Amato V, Servetto A, Rosa R, Marciano R, Formisano L, Di Mauro C, Orsini RC, Cascetta P, Ciciola P, De Maio AP, Di Renzo MF, Cosconati S, Bruno A, Randazzo A, Napolitano F, Montuori N, Veneziani BM, De Placido S, Bianco R. Everolimus induces Met inactivation by disrupting the FKBP12/Met complex. Oncotarget 2018; 7:40073-40084. [PMID: 27223077 PMCID: PMC5129993 DOI: 10.18632/oncotarget.9484] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 04/26/2016] [Indexed: 12/03/2022] Open
Abstract
Inhibition of the mechanistic target of rapamycin (mTOR) is a promising treatment strategy for several cancer types. Rapamycin derivatives such as everolimus are allosteric mTOR inhibitors acting through interaction with the intracellular immunophilin FKBP12, a prolyl isomerase with different cellular functions. Although mTOR inhibitors have significantly improved survival of different cancer patients, resistance and lack of predictive factors of response remain unsolved issues. To elucidate the mechanisms of resistance to everolimus, we evaluated Met activation in everolimus-sensitive/resistant human cancer cells, in vitro and in vivo. Biochemical and computational analyses were performed. Everolimus-resistant cells were xenografted into mice (10/group) and studied for their response to everolimus and Met inhibitors. The statistical significance of the in vitro results was evaluated by Student's t test. Everolimus reduced Met phosphorylation in everolimus-sensitive cells. This event was mediated by the formation of a Met-FKBP12 complex, which in turn is disrupted by everolimus. Aberrant Met activation in everolimus-resistant cells and overexpression of wild-type/mutant Met caused everolimus resistance. Pharmacological inhibition and RNA silencing of Met are effective in condition of everolimus resistance (P<0.01). In mice xenografted with everolimus-resistant cells, the combination of everolimus with the Met inhibitor PHA665752 reduced tumor growth and induced a statistically significant survival advantage (combination vs control P=0.0005). FKBP12 binding is required for full Met activation and everolimus can inhibit Met. Persistent Met activation might sustain everolimus resistance. These results identify a novel everolimus mechanism of action and suggest the development of clinical strategies based on Met inhibitors in everolimus-resistant cancers.
Collapse
Affiliation(s)
- Lucia Raimondo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Valentina D'Amato
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Alberto Servetto
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Roberta Rosa
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Roberta Marciano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Concetta Di Mauro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Roberta Clara Orsini
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Priscilla Cascetta
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Paola Ciciola
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Ana Paula De Maio
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Maria Flavia Di Renzo
- Department of Oncology, University of Turin, Candiolo Cancer Institute - FPO IRCCS, Turin, Italy
| | | | - Agostino Bruno
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | - Antonio Randazzo
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | - Filomena Napolitano
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Nunzia Montuori
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Bianca Maria Veneziani
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
34
|
Zhang J, Liu C, Shi W, Yang L, Zhang Q, Cui J, Fang Y, Li Y, Ren G, Yang S, Xiang R. The novel VEGF receptor 2 inhibitor YLL545 inhibits angiogenesis and growth in breast cancer. Oncotarget 2018; 7:41067-41080. [PMID: 27203384 PMCID: PMC5173043 DOI: 10.18632/oncotarget.9392] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/16/2016] [Indexed: 11/25/2022] Open
Abstract
Their antiangiogenic effects make vascular endothelial growth factor receptor 2 (VEGFR2) inhibitors useful for cancer treatment. However, most of these drugs have unexpected adverse side effects. Here, we show that the novel VEGFR2 inhibitor YLL545 suppressed tumor angiogenesis and growth in triple-negative breast cancer without adverse effects. YLL545 treatment also markedly inhibited proliferation, migration, invasion, and tube formation by human umbilical vascular endothelial cells (HUVECs) in vitro. These effects of YLL545 were equal to or greater than those seen with sorafenib. In addition, YLL545 inhibited VEGF-induced phosphorylation of VEGFR2 and activation of downstream signaling regulators, such as phospho-STAT3 and phospho-ERK1/2, in HUVECs. Embryonic angiogenesis assays in zebrafish and Matrigel plug assays in mice demonstrated that YLL545 inhibits angiogenesis in vivo. YLL545 also inhibited proliferation and induced apoptosis in MDA-MB-231 breast cancer cells both in vitro and in vivo, and 50 mg/kg/d YLL545 inhibited human tumor xenograft growth by more than 50% in BALB/c nude mice. These observations suggest YLL545 is a potentially useful anticancer drug candidate.
Collapse
Affiliation(s)
- Jianbo Zhang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chen Liu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wen Shi
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, Tianjin, China
| | - Lingling Yang
- School of Food and Bioengineering, Xihua University, Sichuan, China
| | - Quansheng Zhang
- Tianjin Key Laboratory of Organ Transplantation, Tianjin First Center Hospital, Tianjin, China
| | - Jianlin Cui
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, Tianjin, China
| | - Yangwu Fang
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, Tianjin, China
| | - Yuhao Li
- School of Food and Bioengineering, Xihua University, Sichuan, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuang Yang
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, Tianjin, China
| | - Rong Xiang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
35
|
PI3K/Akt/mTOR signaling pathway and targeted therapy for glioblastoma. Oncotarget 2017; 7:33440-50. [PMID: 26967052 PMCID: PMC5078108 DOI: 10.18632/oncotarget.7961] [Citation(s) in RCA: 385] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 02/24/2016] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiform (GBM) is the most common malignant glioma of all the brain tumors and currently effective treatment options are still lacking. GBM is frequently accompanied with overexpression and/or mutation of epidermal growth factor receptor (EGFR), which subsequently leads to activation of many downstream signal pathways such as phosphatidylinositol 3-kinase (PI3K)/Akt/rapamycin-sensitive mTOR-complex (mTOR) pathway. Here we explored the reason why inhibition of the pathway may serve as a compelling therapeutic target for the disease, and provided an update data of EFGR and PI3K/Akt/mTOR inhibitors in clinical trials.
Collapse
|
36
|
Salazar R, Garcia-Carbonero R, Libutti SK, Hendifar AE, Custodio A, Guimbaud R, Lombard-Bohas C, Ricci S, Klümpen HJ, Capdevila J, Reed N, Walenkamp A, Grande E, Safina S, Meyer T, Kong O, Salomon H, Tavorath R, Yao JC. Phase II Study of BEZ235 versus Everolimus in Patients with Mammalian Target of Rapamycin Inhibitor-Naïve Advanced Pancreatic Neuroendocrine Tumors. Oncologist 2017; 23:766-e90. [PMID: 29242283 PMCID: PMC6058330 DOI: 10.1634/theoncologist.2017-0144] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/30/2017] [Indexed: 12/26/2022] Open
Abstract
Lessons Learned. Treatment with BEZ235 has not been shown to demonstrate increased efficacy compared with everolimus and may be associated with a poorer tolerability profile. The hypothesis of dual targeting of the phosphatidylinositol 3‐kinase and mammalian target of rapamycin pathways in patients with advanced pancreatic neuroendocrine tumors may warrant further study using other agents.
Background. This phase II study investigated whether targeting the phosphatidylinositol 3‐kinase (PI3K)/mammalian target of rapamycin (mTOR) pathway via PI3K, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2) inhibition using BEZ235 may be more effective than mTORC1 inhibition with everolimus in patients with advanced pancreatic neuroendocrine tumors (pNET) who are naïve to mTOR inhibitor therapy. Methods. Patients with advanced pNET were randomized (1:1) to oral BEZ235 400 mg twice daily or oral everolimus 10 mg once daily on a continuous dosing schedule. The primary endpoint was progression‐free survival (PFS). Secondary endpoints included safety, overall response rate (ORR), overall survival (OS), and time to treatment failure. Results. Enrollment in this study was terminated early (62 enrolled of the 140 planned). The median PFS was 8.2 months (95% confidence interval [CI]: 5.3 to not evaluable [NE]) with BEZ235 versus 10.8 months (95% CI: 8.1–NE) with everolimus (hazard ratio 1.53; 95% CI: 0.72–3.25). The most commonly reported all‐grade adverse events (>50% of patients regardless of study treatment relationship) with BEZ235 were diarrhea (90.3%), stomatitis (74.2%), and nausea (54.8%). Conclusion. BEZ235 treatment in mTOR inhibitor‐naïve patients with advanced pNET did not demonstrate increased efficacy compared with everolimus and may be associated with a poorer tolerability profile.
Collapse
Affiliation(s)
- Ramon Salazar
- Department of Medical Oncology, Institut Català d'Oncologia-IDIBELL-CIBERONC, Universitat de Barcelona, Barcelona, Spain
| | | | - Steven K Libutti
- Albert Einstein College of Medicine, New York City, New York, USA
| | - Andrew E Hendifar
- David Geffen School of Medicine and Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Ana Custodio
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain
| | - Rosine Guimbaud
- Department of Digestive Medical Oncology (IUCT-RL), Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | | | - Sergio Ricci
- Division of Medical Oncology, S Chiara University Hospital, Pisa, Italy
| | - Heinz-Josef Klümpen
- Department of Medical Oncology, Academic Medical Center, Amsterdam, The Netherlands
| | - Jaume Capdevila
- Vall Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Nicholas Reed
- Beatson West of Scotland Cancer Centre, Glasgow, Scotland
| | - Annemiek Walenkamp
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Enrique Grande
- Department of Medical Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Sufiya Safina
- Department of Biochemistry, Kazan State Medical University, Kazan, Russia
| | - Tim Meyer
- Royal Free Hospital, London, United Kingdom
| | - Oliver Kong
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | - Ranjana Tavorath
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - James C Yao
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
37
|
Li Z, Qiu R, Qiu X, Tian T. EYA2 promotes lung cancer cell proliferation by downregulating the expression of PTEN. Oncotarget 2017; 8:110837-110848. [PMID: 29340020 PMCID: PMC5762288 DOI: 10.18632/oncotarget.22860] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/29/2017] [Indexed: 12/13/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide. Despite advances have been made in diagnosis and therapeutic strategies, the prognosis of lung cancer is still very poor. Eyes absent transcriptional cofactor EYA2 has been shown to promote lung cancer cell growth, however, the underlying molecular mechanism is still not fully understood. In the present study, we found that EYA2 was up-regulated in lung cancer, and EYA2 led to increased cell proliferation by inhibiting Phosphatase and tensin homologue (PTEN) expression via modulation of miR-93. Additionally, survival analysis showed that lung cancer patients with higher EYA2 expression predicted a worse prognosis. Therefore, these findings demonstrate that EYA2 may play an important role in lung cancer occurrence and progression. Targeting EYA2 may provide a feasible approach in developing novel anticancer therapeutics.
Collapse
Affiliation(s)
- Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ran Qiu
- Wuhan Institute of Bioengineering, Wuhan, China
| | - Xia Qiu
- Department of Medicine, Shangqiu Medical School, Shangqiu, China
| | - Tian Tian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
38
|
Abstract
Rapamycin slows organismal aging and delays age-related diseases, extending lifespan in numerous species. In cells, rapamycin and other rapalogs such as everolimus suppress geroconversion from quiescence to senescence. Rapamycin inhibits some, but not all, activities of mTOR. Recently we and others demonstrated that pan-mTOR inhibitors, known also as dual mTORC1/C2 inhibitors, suppress senescent phenotype. As a continuation of these studies, here we investigated in detail a panel of pan-mTOR inhibitors, to determine their optimal gerosuppressive concentrations. During geroconversion, cells become hypertrophic and flat, accumulate lysosomes (SA-beta-Gal staining) and lipids (Oil Red staining) and lose their re-proliferative potential (RPP). We determined optimal gerosuppressive concentrations: Torin1 (30 nM), Torin 2 (30 nM), AZD8055 (100 nM), PP242 (300 nM), both KU-006379 and GSK1059615 (1000 nM). These agents decreased senescence-associated hypertrophy with IC50s: 20, 18, 15, 200 and 400 nM, respectively. Preservation of RPP by pan-mTOR inhibitors was associated with inhibition of the pS6K/pS6 axis. Inhibition of rapamycin-insensitive functions of mTOR further contributed to anti-hypertrophic and cytostatic effects. Torin 1 and PP242 were more "rapamycin-like" than Torin 2 and AZD8055. Pan-mTOR inhibitors were superior to rapamycin in suppressing hypertrophy, senescent morphology, Oil Red O staining and in increasing so-called "chronological life span (CLS)". We suggest that, at doses lower than anti-cancer concentrations, pan-mTOR inhibitors can be developed as anti-aging drugs.
Collapse
|
39
|
Bhardwaj A, Singh H, Rajapakshe K, Tachibana K, Ganesan N, Pan Y, Gunaratne PH, Coarfa C, Bedrosian I. Regulation of miRNA-29c and its downstream pathways in preneoplastic progression of triple-negative breast cancer. Oncotarget 2017; 8:19645-19660. [PMID: 28160548 PMCID: PMC5386711 DOI: 10.18632/oncotarget.14902] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 01/04/2017] [Indexed: 12/21/2022] Open
Abstract
Little is understood about the early molecular drivers of triple-negative breast cancer (TNBC), making the identification of women at risk and development of targeted therapy for prevention significant challenges. By sequencing a TNBC cell line-based breast cancer progression model we have found that miRNA-29c is progressively lost during TNBC tumorigenesis. In support of the tumor suppressive role of miRNA 29c, we found that low levels predict poor overall patient survival and, conversely, that ectopic expression of miRNA-29c in preneoplastic cell models inhibits growth. miRNA-29c exerts its growth inhibitory effects through direct binding and regulation of TGFB-induced factor homeobox 2 (TGIF2), CAMP-responsive element binding protein 5 (CREB5), and V-Akt murine thymoma viral oncogene homolog 3 (AKT3). miRNA-29c regulation of these gene targets seems to be functionally relevant, as TGIF2, CREB5, and AKT3 were able to rescue the inhibition of cell proliferation and colony formation caused by ectopic expression of miRNA-29c in preneoplastic cells. AKT3 is an oncogene of known relevance in breast cancer, and as a proof of principle we show that inhibition of phosphoinositide 3-kinase (PI3K) activity, a protein upstream of AKT3, suppressed proliferation in TNBC preneoplastic cells. We explored additional opportunities for prevention of TNBC by studying the regulation of miRNA-29c and identified DNA methylation to have a role in the inhibition of miRNA-29c during TNBC tumorigenesis. Consistent with these observations, we found 5 aza-cytadine to relieve the suppression of miRNA-29c. Together, these results demonstrate that miRNA-29c loss plays a key role in the early development of TNBC.
Collapse
Affiliation(s)
- Anjana Bhardwaj
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Harpreet Singh
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Kazunoshin Tachibana
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nivetha Ganesan
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yinghong Pan
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Preethi H Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Isabelle Bedrosian
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
40
|
Control of leucine-dependent mTORC1 pathway through chemical intervention of leucyl-tRNA synthetase and RagD interaction. Nat Commun 2017; 8:732. [PMID: 28963468 PMCID: PMC5622079 DOI: 10.1038/s41467-017-00785-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 07/27/2017] [Indexed: 01/19/2023] Open
Abstract
Leucyl-tRNA synthetase (LRS) is known to function as leucine sensor in the mammalian target of rapamycin complex 1 (mTORC1) pathway. However, the pathophysiological significance of its activity is not well understood. Here, we demonstrate that the leucine sensor function for mTORC1 activation of LRS can be decoupled from its catalytic activity. We identified compounds that inhibit the leucine-dependent mTORC1 pathway by specifically inhibiting the GTPase activating function of LRS, while not affecting the catalytic activity. For further analysis, we selected one compound, BC-LI-0186, which binds to the RagD interacting site of LRS, thereby inhibiting lysosomal localization of LRS and mTORC1 activity. It also effectively suppressed the activity of cancer-associated MTOR mutants and the growth of rapamycin-resistant cancer cells. These findings suggest new strategies for controlling tumor growth that avoid the resistance to existing mTOR inhibitors resulting from cancer-associated MTOR mutations.Leucyl-tRNA synthetase (LRS) is a leucine sensor of the mTORC1 pathway. Here, the authors identify inhibitors of the GTPase activating function of LRS, not affecting its catalytic activity, and demonstrate that the leucine sensor function of LRS can be a new target for mTORC1 inhibition.
Collapse
|
41
|
Byun S, Lee E, Lee KW. Therapeutic Implications of Autophagy Inducers in Immunological Disorders, Infection, and Cancer. Int J Mol Sci 2017; 18:ijms18091959. [PMID: 28895911 PMCID: PMC5618608 DOI: 10.3390/ijms18091959] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/10/2017] [Accepted: 09/11/2017] [Indexed: 12/19/2022] Open
Abstract
Autophagy is an essential catabolic program that forms part of the stress response and enables cells to break down their own intracellular components within lysosomes for recycling. Accumulating evidence suggests that autophagy plays vital roles in determining pathological outcomes of immune responses and tumorigenesis. Autophagy regulates innate and adaptive immunity affecting the pathologies of infectious, inflammatory, and autoimmune diseases. In cancer, autophagy appears to play distinct roles depending on the context of the malignancy by either promoting or suppressing key determinants of cancer cell survival. This review covers recent developments in the understanding of autophagy and discusses potential therapeutic interventions that may alter the outcomes of certain diseases.
Collapse
Affiliation(s)
- Sanguine Byun
- Division of Bioengineering, Incheon National University, Incheon 22012, Korea.
| | - Eunjung Lee
- Traditional Alcoholic Beverage Research Team, Korea Food Research Institute, Seongnam 13539, Korea.
| | - Ki Won Lee
- Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16495, Korea.
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
42
|
Maynard J, Emmas SA, Ble FX, Barjat H, Lawrie E, Hancox U, Polanska UM, Pritchard A, Hudson K. The use of 18F-Fluoro-deoxy-glucose positron emission tomography (18F-FDG PET) as a non-invasive pharmacodynamic biomarker to determine the minimally pharmacologically active dose of AZD8835, a novel PI3Kα inhibitor. PLoS One 2017; 12:e0183048. [PMID: 28806782 PMCID: PMC5555689 DOI: 10.1371/journal.pone.0183048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/30/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The phosphatidyl inositol 3 kinase (PI3K), AKT and mammalian target of rapamycin (mTOR) signal transduction pathway is frequently de-regulated and activated in human cancer and is an important therapeutic target. AZD8835 is a PI3K inhibitor, with selectivity against PI3K α and δ isoforms, which is currently in Phase 1 clinical trials. 18F-Fluoro-deoxy-glucose positron emission tomography (18F-FDG PET) is a non-invasive pharmacodynamic imaging biomarker that has become an integral part of drug development. It has been used widely with PI3K inhibitors both clinically and pre-clinically because of the role of the PI3K pathway in glucose metabolism. In this study we investigated the potential of 18F-FDG PET as a non-invasive pharmacodynamic biomarker for AZD8835. We sought to understand if 18F-FDG PET could determine the minimally effective dose of AZD8835 and correlate with other pharmacodynamic biomarkers for validation of its use in clinical development. 18F-FDG PET scans were performed in nude mice in the BT474C breast xenograft model. Mice were fasted prior to imaging and static 18F-FDG PET was performed. Treatment groups received AZD8835 by oral gavage at a dose volume of 10ml/kg. Treatment groups received either 3, 6, 12.5, 25 or 50mg/kg AZD8835. Tumour growth was monitored throughout the study, and at the end of the imaging procedure, tumours were taken and a full pharmacodynamic analysis was performed. RESULTS Results showed that AZD8835 reduced 18F-FDG uptake at a dose of 12.5, 25 and 50mg/kg with no significant reduction at doses of 3 and 6mg/kg. These results were consistent with other pharmacodynamics biomarkers measured and show 18F-FDG PET as a sensitive biomarker with the ability to determine the minimal effective dose of AZD8835. CONCLUSIONS Our pre-clinical studies support the use of 18F-FDG PET imaging as a sensitive and non- invasive pharmacodynamic biomarker (understanding the role of PI3K signalling in glucose uptake) for AZD8835 with a decrease in 18F-FDG uptake observed at only two hours post treatment. The decrease in 18F-FDG uptake was dose dependent and data showed excellent PK/PD correlation. This data supports and parallels observations obtained with this class of compounds in patients.
Collapse
Affiliation(s)
- Juliana Maynard
- Personalised Healthcare & Biomarkers, AstraZeneca, Cheshire, United Kingdom
- * E-mail:
| | - Sally-Ann Emmas
- Personalised Healthcare & Biomarkers, AstraZeneca, Cheshire, United Kingdom
| | | | - Herve Barjat
- Personalised Healthcare & Biomarkers, AstraZeneca, Cheshire, United Kingdom
| | - Emily Lawrie
- Drug Safety and Metabolism iMED, AstraZeneca, Cheshire, United Kingdom
| | - Urs Hancox
- Oncology Imed, Astrazenenca, Cheshire, United Kingdom
| | | | | | - Kevin Hudson
- Oncology Imed, Astrazenenca, Cheshire, United Kingdom
| |
Collapse
|
43
|
Gallo M, Malandrino P, Fanciulli G, Rota F, Faggiano A, Colao A. Everolimus as first line therapy for pancreatic neuroendocrine tumours: current knowledge and future perspectives. J Cancer Res Clin Oncol 2017; 143:1209-1224. [PMID: 28405826 DOI: 10.1007/s00432-017-2407-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 03/24/2017] [Indexed: 01/21/2023]
Abstract
PURPOSE Everolimus has been shown to be effective for advanced pancreatic neuroendocrine tumours (pNETs), but its positioning in the therapeutic algorithm for pNETs is matter of debate. METHODS With the aim to shed light on this point, we performed an up-to-date critical review taking into account the results of both retrospective and prospective published studies, and the recommendations of international guidelines. In addition, we performed an extensive search on the Clinical Trial Registries databases worldwide, to gather information on the ongoing clinical trials related to this specific topic. RESULTS We identified eight retrospective published studies, two prospective published studies, and five registered clinical trials. Moreover, we analyzed the content of four widespread international guidelines. CONCLUSIONS Our critical review confirms the lack of high-quality data to recommend everolimus as the first line therapy for pNETs. The ongoing clinical trials reported in this review will hopefully help clinicians, in the near future, to better evaluate the role of everolimus as the first line therapy for pNETs. However, at the moment, there is already enough evidence to recommend everolimus as the first line therapy for patients with symptomatic malignant unresectable insulin-secreting pNETs, to control the endocrine syndrome regardless of tumour growth.
Collapse
Affiliation(s)
- Marco Gallo
- Oncological Endocrinology Unit, Department of Medical Sciences, University of Turin, AOU Città della Salute e della Scienza di Torino, Via Genova 3, 10126, Turin, Italy.
| | | | - Giuseppe Fanciulli
- Neuroendocrine Tumours Unit, Department of Clinical and Experimental Medicine, University of Sassari, AOU Sassari, Sassari, Italy
| | - Francesca Rota
- Endocrinology Unit, Regina Elena National Cancer Institute, Rome, Italy
| | - Antongiulio Faggiano
- Thyroid and Parathyroid Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Annamaria Colao
- Department of Clinical Medicine and Surgery, University "Federico II", Naples, Italy
| |
Collapse
|
44
|
FKBP51 decreases cell proliferation and increases progestin sensitivity of human endometrial adenocarcinomas by inhibiting Akt. Oncotarget 2017; 8:80405-80415. [PMID: 29113312 PMCID: PMC5655207 DOI: 10.18632/oncotarget.18903] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 06/15/2017] [Indexed: 12/23/2022] Open
Abstract
In this study, we investigated the role of FK506 binding protein 51 (FKBP51) in human endometrial adenocarcinoma progression. Immunohistochemical analysis showed decreased FKBP51 expression in endometrial adenocarcinoma tissues. Moreover, higher FKBP51 expression was observed in the normal secretory phase than in proliferative-phase endometrial tissues. FKBP51-shRNA transfected KLE cells showed high Ser473-phospho Akt with decreased p21 and p27 levels, which promoted S-G2/M phase cell cycle progression and proliferation. Conversely, FKBP51 overexpressing Ishikawa cells showed low Ser473-phospho Akt, which led to increased p21 and p27 levels and, in turn, G0/G1 cell cycle arrest and decreased cell proliferation. FKBP51 overexpression in progesterone receptor-positive Ishikawa cells sensitized them to medroxyprogesterone acetate (MPA; progestin) treatment by repressing Akt signaling. Conversely, FKBP51-shRNA knockdown in RL95-2 cells attenuated progestin sensitivity. These findings indicate FKBP51 inhibits cell proliferation and promotes progestin sensitivity in endometrial adenocarcinoma by decreasing Akt signaling.
Collapse
|
45
|
Adeno-Associated Viral Vector-Mediated mTOR Inhibition by Short Hairpin RNA Suppresses Laser-Induced Choroidal Neovascularization. MOLECULAR THERAPY-NUCLEIC ACIDS 2017; 8:26-35. [PMID: 28918027 PMCID: PMC5477068 DOI: 10.1016/j.omtn.2017.05.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/29/2017] [Accepted: 05/29/2017] [Indexed: 12/20/2022]
Abstract
Choroidal neovascularization (CNV) is the defining characteristic feature of the wet subtype of age-related macular degeneration (AMD) and may result in irreversible blindness. Based on anti-vascular endothelial growth factor (anti-VEGF), the current therapeutic approaches to CNV are fraught with difficulties, and mammalian target of rapamycin (mTOR) has recently been proposed as a possible therapeutic target, although few studies have been conducted. Here, we show that a recombinant adeno-associated virus-delivered mTOR-inhibiting short hairpin RNA (rAAV-mTOR shRNA), which blocks the activity of both mTOR complex 1 and 2, represents a promising therapeutic approach for the treatment of CNV. Eight-week-old male C57/B6 mice were treated with the short hairpin RNA (shRNA) after generating CNV lesions in the eyes via laser photocoagulation. The recombinant adeno-associated virus (rAAV) delivery vehicle was able to effectively transduce cells in the inner retina, and significantly fewer inflammatory cells and less extensive CNV were observed in the animals treated with rAAV-mTOR shRNA when compared with control- and rAAV-scrambled shRNA-treated groups. Presumably related to the reduction of CNV, increased autophagy was detected in CNV lesions treated with rAAV-mTOR shRNA, whereas significantly fewer apoptotic cells detected in the outer nuclear layer around the CNV indicate that mTOR inhibition may also have neuroprotective effects. Taken together, these results demonstrate the therapeutic potential of mTOR inhibition, resulting from rAAV-mTOR shRNA activity, in the treatment of AMD-related CNV.
Collapse
|
46
|
Mateo F, Arenas EJ, Aguilar H, Serra-Musach J, de Garibay GR, Boni J, Maicas M, Du S, Iorio F, Herranz-Ors C, Islam A, Prado X, Llorente A, Petit A, Vidal A, Català I, Soler T, Venturas G, Rojo-Sebastian A, Serra H, Cuadras D, Blanco I, Lozano J, Canals F, Sieuwerts AM, de Weerd V, Look MP, Puertas S, García N, Perkins AS, Bonifaci N, Skowron M, Gómez-Baldó L, Hernández V, Martínez-Aranda A, Martínez-Iniesta M, Serrat X, Cerón J, Brunet J, Barretina MP, Gil M, Falo C, Fernández A, Morilla I, Pernas S, Plà MJ, Andreu X, Seguí MA, Ballester R, Castellà E, Nellist M, Morales S, Valls J, Velasco A, Matias-Guiu X, Figueras A, Sánchez-Mut JV, Sánchez-Céspedes M, Cordero A, Gómez-Miragaya J, Palomero L, Gómez A, Gajewski TF, Cohen EEW, Jesiotr M, Bodnar L, Quintela-Fandino M, López-Bigas N, Valdés-Mas R, Puente XS, Viñals F, Casanovas O, Graupera M, Hernández-Losa J, Ramón y Cajal S, García-Alonso L, Saez-Rodriguez J, Esteller M, Sierra A, Martín-Martín N, Matheu A, Carracedo A, González-Suárez E, Nanjundan M, Cortés J, Lázaro C, Odero MD, Martens JWM, Moreno-Bueno G, Barcellos-Hoff MH, Villanueva A, Gomis RR, Pujana MA. Stem cell-like transcriptional reprogramming mediates metastatic resistance to mTOR inhibition. Oncogene 2017; 36:2737-2749. [PMID: 27991928 PMCID: PMC5442428 DOI: 10.1038/onc.2016.427] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 08/31/2016] [Accepted: 10/10/2016] [Indexed: 01/16/2023]
Abstract
Inhibitors of the mechanistic target of rapamycin (mTOR) are currently used to treat advanced metastatic breast cancer. However, whether an aggressive phenotype is sustained through adaptation or resistance to mTOR inhibition remains unknown. Here, complementary studies in human tumors, cancer models and cell lines reveal transcriptional reprogramming that supports metastasis in response to mTOR inhibition. This cancer feature is driven by EVI1 and SOX9. EVI1 functionally cooperates with and positively regulates SOX9, and promotes the transcriptional upregulation of key mTOR pathway components (REHB and RAPTOR) and of lung metastasis mediators (FSCN1 and SPARC). The expression of EVI1 and SOX9 is associated with stem cell-like and metastasis signatures, and their depletion impairs the metastatic potential of breast cancer cells. These results establish the mechanistic link between resistance to mTOR inhibition and cancer metastatic potential, thus enhancing our understanding of mTOR targeting failure.
Collapse
Affiliation(s)
- F Mateo
- Breast Cancer and Systems Biology Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - E J Arenas
- Oncology Program, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - H Aguilar
- Breast Cancer and Systems Biology Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - J Serra-Musach
- Breast Cancer and Systems Biology Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - G Ruiz de Garibay
- Breast Cancer and Systems Biology Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - J Boni
- Breast Cancer and Systems Biology Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - M Maicas
- Centre for Applied Medical Research (CIMA) and Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain
| | - S Du
- Department of Radiation Oncology, New York University School of Medicine, New York, NY, USA
| | - F Iorio
- European Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Cambridge, UK
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - C Herranz-Ors
- Breast Cancer and Systems Biology Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - A Islam
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - X Prado
- Breast Cancer and Systems Biology Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - A Llorente
- Breast Cancer and Systems Biology Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - A Petit
- Department of Pathology, University Hospital of Bellvitge, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - A Vidal
- Department of Pathology, University Hospital of Bellvitge, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - I Català
- Department of Pathology, University Hospital of Bellvitge, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - T Soler
- Department of Pathology, University Hospital of Bellvitge, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - G Venturas
- Department of Pathology, University Hospital of Bellvitge, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - A Rojo-Sebastian
- Department of Pathology, MD Anderson Cancer Center, Madrid, Spain
| | - H Serra
- Angiogenesis Research Group, ProCURE, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - D Cuadras
- Statistics Unit, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - I Blanco
- Hereditary Cancer Programme, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - J Lozano
- Department of Molecular Biology and Biochemistry, Málaga University, and Molecular Oncology Laboratory, Mediterranean Institute for the Advance of Biotechnology and Health Research (IBIMA), University Hospital Virgen de la Victoria, Málaga, Spain
| | - F Canals
- ProteoRed-Instituto de Salud Carlos III, Proteomic Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona, Spain
| | - A M Sieuwerts
- Department of Medical Oncology, Erasmus University Medical Center, Daniel den Hoed Cancer Center, Cancer Genomics Centre, Rotterdam, The Netherlands
| | - V de Weerd
- Department of Medical Oncology, Erasmus University Medical Center, Daniel den Hoed Cancer Center, Cancer Genomics Centre, Rotterdam, The Netherlands
| | - M P Look
- Department of Medical Oncology, Erasmus University Medical Center, Daniel den Hoed Cancer Center, Cancer Genomics Centre, Rotterdam, The Netherlands
| | - S Puertas
- Chemoresistance and Predictive Factors Laboratory, ProCURE, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - N García
- Breast Cancer and Systems Biology Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - A S Perkins
- University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, NY, USA
| | - N Bonifaci
- Breast Cancer and Systems Biology Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - M Skowron
- Breast Cancer and Systems Biology Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - L Gómez-Baldó
- Breast Cancer and Systems Biology Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - V Hernández
- Biological Clues of the Invasive and Metastatic Phenotype Laboratory, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - A Martínez-Aranda
- Biological Clues of the Invasive and Metastatic Phenotype Laboratory, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - M Martínez-Iniesta
- Chemoresistance and Predictive Factors Laboratory, ProCURE, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - X Serrat
- Cancer and Human Molecular Genetics, IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - J Cerón
- Cancer and Human Molecular Genetics, IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - J Brunet
- Hereditary Cancer Programme, ICO, Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - M P Barretina
- Department of Medical Oncology, ICO, IDIBGI, Girona, Spain
| | - M Gil
- Department of Medical Oncology, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - C Falo
- Department of Medical Oncology, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - A Fernández
- Department of Medical Oncology, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - I Morilla
- Department of Medical Oncology, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - S Pernas
- Department of Medical Oncology, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - M J Plà
- Department of Gynecology, University Hospital of Bellvitge, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - X Andreu
- Department of Pathology, Parc Taulí Hospital Consortium, Sabadell, Barcelona, Spain
| | - M A Seguí
- Medical Oncology Service, Parc Taulí Hospital Consortium, Sabadell, Barcelona, Spain
| | - R Ballester
- Department of Radiation Oncology, University Hospital Germans Trias i Pujol, ICO, Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - E Castellà
- Department of Pathology, University Hospital Germans Trias i Pujol, ICO, IGTP, Badalona, Barcelona, Spain
| | - M Nellist
- Department of Clinical Genetics, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - S Morales
- Hospital Arnau de Vilanova, University of Lleida, Biomedical Research Institute of Lleida (IRB Lleida), Lleida, Spain
| | - J Valls
- Hospital Arnau de Vilanova, University of Lleida, Biomedical Research Institute of Lleida (IRB Lleida), Lleida, Spain
| | - A Velasco
- Hospital Arnau de Vilanova, University of Lleida, Biomedical Research Institute of Lleida (IRB Lleida), Lleida, Spain
| | - X Matias-Guiu
- Hospital Arnau de Vilanova, University of Lleida, Biomedical Research Institute of Lleida (IRB Lleida), Lleida, Spain
| | - A Figueras
- Angiogenesis Research Group, ProCURE, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - J V Sánchez-Mut
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - M Sánchez-Céspedes
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - A Cordero
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - J Gómez-Miragaya
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - L Palomero
- Breast Cancer and Systems Biology Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - A Gómez
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - T F Gajewski
- Departments of Pathology and Medicine, University of Chicago, Chicago, IL, USA
| | - E E W Cohen
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - M Jesiotr
- Department of Pathology, Military Institute of Medicine, Warsaw, Poland
| | - L Bodnar
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - M Quintela-Fandino
- Breast Cancer Clinical Research Unit, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - N López-Bigas
- Department of Experimental and Health Sciences, Barcelona Biomedical Research Park, Pompeu Fabra University (UPF), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - R Valdés-Mas
- Department of Biochemistry and Molecular Biology, University Institute of Oncology of Asturias, University of Oviedo, Oviedo, Spain
| | - X S Puente
- Department of Biochemistry and Molecular Biology, University Institute of Oncology of Asturias, University of Oviedo, Oviedo, Spain
| | - F Viñals
- Angiogenesis Research Group, ProCURE, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - O Casanovas
- Angiogenesis Research Group, ProCURE, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - M Graupera
- Angiogenesis Research Group, ProCURE, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - J Hernández-Losa
- Department of Pathology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - S Ramón y Cajal
- Department of Pathology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - L García-Alonso
- European Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Cambridge, UK
| | - J Saez-Rodriguez
- European Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Cambridge, UK
| | - M Esteller
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Physiological Sciences II, School of Medicine, University of Barcelona, Barcelona, Spain
| | - A Sierra
- Molecular and Translational Oncology Laboratory, Biomedical Research Center CELLEX-CRBC, Biomedical Research Institute ‘August Pi i Sunyer' (IDIBAPS), and Systems Biology Department, Faculty of Science and Technology, University of Vic, Central University of Catalonia, Barcelona, Spain
| | - N Martín-Martín
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Derio, Spain
| | - A Matheu
- Neuro-Oncology Section, Oncology Department, Biodonostia Research Institute, San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - A Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - E González-Suárez
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - M Nanjundan
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - J Cortés
- Department of Medical Oncology, VHIO, Vall d'Hebron University Hospital, Barcelona, Spain
| | - C Lázaro
- Hereditary Cancer Programme, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - M D Odero
- Centre for Applied Medical Research (CIMA) and Department of Biochemistry and Genetics, University of Navarra, Pamplona, Spain
| | - J W M Martens
- Department of Medical Oncology, Erasmus University Medical Center, Daniel den Hoed Cancer Center, Cancer Genomics Centre, Rotterdam, The Netherlands
| | - G Moreno-Bueno
- Department of Biochemistry, Autonomous University of Madrid (UAM), Biomedical Research Institute ‘Alberto Sols' (Spanish National Research Council (CSIC)-UAM), Translational Research Laboratory, Hospital La Paz Institute for Health Research (IdiPAZ), and MD Anderson International Foundation, Madrid, Spain
| | - M H Barcellos-Hoff
- Department of Radiation Oncology, New York University School of Medicine, New York, NY, USA
| | - A Villanueva
- Chemoresistance and Predictive Factors Laboratory, ProCURE, ICO, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - R R Gomis
- Oncology Program, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - M A Pujana
- Breast Cancer and Systems Biology Laboratory, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| |
Collapse
|
47
|
A phase I dose-escalation study of the safety and pharmacokinetics of a tablet formulation of voxtalisib, a phosphoinositide 3-kinase inhibitor, in patients with solid tumors. Invest New Drugs 2017; 36:36-44. [PMID: 28417284 DOI: 10.1007/s10637-017-0467-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 04/05/2017] [Indexed: 12/13/2022]
Abstract
Background Voxtalisib, a PI3K/mTOR inhibitor, has shown antitumor activity in capsule formulation in patients with solid tumors. This Phase I study assessed safety and pharmacokinetics of voxtalisib administered as immediate-release tablets in patients with solid tumors (NCT01596270). Methods A "3 + 3" dose escalation design was used. Adverse events (AEs), pharmacokinetics (PK), food effect and tumor response were evaluated. Results Thirty-two patients received voxtalisib doses ranging from 50 mg to 70 mg once daily (QD) and 17 patients received voxtalisib doses ranging from 30 mg to 50 mg twice daily (BID), for two 28-day cycles. Dose-limiting toxicities (DLTs) were Grade 3 fatigue (two patients at 70 mg QD, one patient at 40 mg BID) and Grade 3 rash (two patients at 50 mg BID). The maximum tolerated dose (MTD) was 60 mg for QD and 40 mg for BID regimens. Common treatment-emergent AEs were diarrhea (41%), nausea (37%) and fatigue (33%). Voxtalisib appeared to follow linear PK, with a general increase in plasma exposure with dose and no significant accumulation. Administration with food caused a slight decrease in exposure; however, given the high variability observed in the exposure parameters, this should be interpreted with caution. Best response was stable disease in 29% and 50% of patients (QD and BID regimens, respectively). Conclusions The safety profile of voxtalisib tablets at the MTD in patients with solid tumors was consistent with that observed with voxtalisib capsules. Given the limited activity observed across multiple clinical trials, no further trials of voxtalisib are planned.
Collapse
|
48
|
Kim ST, Lee J, Park SH, Park JO, Park YS, Kang WK, Lim HY. Prospective phase II trial of everolimus in PIK3CA amplification/mutation and/or PTEN loss patients with advanced solid tumors refractory to standard therapy. BMC Cancer 2017; 17:211. [PMID: 28330462 PMCID: PMC5363054 DOI: 10.1186/s12885-017-3196-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 03/14/2017] [Indexed: 02/08/2023] Open
Abstract
Background We designed a single-arm, open-label phase II trial of everolimus in PIK3CA amplification/mutation and/or PTEN loss patients with advanced solid tumors refractory to standard therapy (#NCT02449538). Methods Everolimus was administered orally at a daily dose of 10 mg continuously (28-day cycles). Treatment was continued until progression of the disease or intolerable toxicity was observed. Based on Simon’s two-stage optimal design, 10 patients were treated with everolimus during the first stage. Results The median age of the patients was 55.5 years (range, 42–72), and the median Eastern Cooperative Oncology Group (ECOG) performance status (PS) was 2 (range, 1–2). Most of the patients (50.0%) had gastric cancer (GC) as the site of their primary tumor followed by colorectal cancer (CRC), pancreatic cancer, and cholangiocarcinoma. Patients received everolimus as a third-line (3 patients), fourth-line (4 patients), fifth-line (1 patient) or sixth-line (2 patients) treatment. Complete or partial responses were not observed in any of the patients. Four patients showed stable disease, resulting in a disease control rate of 40%. The median PFS was 1.6 months (95% CI, 0.8–2.4 months). Grade 3 or greater hematologic/non-hematologic toxicity was not observed. Grade 2 diarrhea and stomatitis were reported in one patient each. There were no treatment-related deaths. There was less than one response out of the 10 initial patients during the first stage, and the study did not progress to the second stage. Conclusions The study did not meet its primary objective of demonstrating the anti-tumor activity of everolimus in PIK3CA amplification/mutation and/or PTEN loss patients with advanced solid tumors refractory to standard therapy. Further investigation using other genomic candidates and new-generation mTOR inhibitors is warranted in patients with treatment-refractory cancer. Trial registration #NCT02449538, April 2015.
Collapse
Affiliation(s)
- Seung Tae Kim
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro Gangnam-gu, Seoul, 135-710, Korea
| | - Jeeyun Lee
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro Gangnam-gu, Seoul, 135-710, Korea
| | - Se Hoon Park
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro Gangnam-gu, Seoul, 135-710, Korea
| | - Joon Oh Park
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro Gangnam-gu, Seoul, 135-710, Korea
| | - Young Suk Park
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro Gangnam-gu, Seoul, 135-710, Korea
| | - Won Ki Kang
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro Gangnam-gu, Seoul, 135-710, Korea
| | - Ho Yeong Lim
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro Gangnam-gu, Seoul, 135-710, Korea.
| |
Collapse
|
49
|
Qin Y, Meng L, Fu Y, Quan Z, Ma M, Weng M, Zhang Z, Gao C, Shi X, Han K. SNORA74B gene silencing inhibits gallbladder cancer cells by inducing PHLPP and suppressing Akt/mTOR signaling. Oncotarget 2017; 8:19980-19996. [PMID: 28212545 PMCID: PMC5386738 DOI: 10.18632/oncotarget.15301] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/08/2017] [Indexed: 01/17/2023] Open
Abstract
Small nucleolar RNAs (snoRNAs) have been implicated in the development of many cancers. We therefore examined the differential expression of snoRNAs between gallbladder cancer (GBC) tissues and matched adjacent non-tumor tissues using expression microarray analysis with confirmation by quantitative real-time PCR (qRT-PCR). Western blot analysis showed that SNORA74B levels were higher in GBC than non-tumor tissues. SNORA74B expression was positively associated with local invasion, advanced TNM stage, CA19-9 level, and Ki67 expression in patients with GBC, while it was negatively associated with expression of PHLPP, an endogenous Akt inhibitor. Moreover, SNORA74B expression was prognostic for overall survival (OS) and disease-free survival (DFS). Functional studies revealed that silencing SNORA74B in GBC cells using sh-SNORA74B suppressed cell proliferation, induced G1 arrest, and promoted apoptosis. Preliminary molecular investigation revealed that SNORA74B silencing inhibited activation of the AKT/mTOR signaling pathway, while increasing PHLPP expression. PHLPP depletion using shRNA abrogated sh-SNORA74B suppression of GBC cell proliferation, indicating that the antitumor effects of SNORA74B silencing were mediated by PHLPP. These findings define the important role of SNORA74B in cell proliferation, cell cycle, and apoptosis of GBC, and suggest that it may serve as a novel target for GBC treatment.
Collapse
Affiliation(s)
- Yiyu Qin
- Clinical College, Yancheng Institute of Health Sciences, Yancheng, Jiangsu 224000, China
- Research Centre of Biomedical Technology, Yancheng Institute of Health Sciences, Yancheng, Jiangsu 224000, China
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Li Meng
- Research Centre of Biomedical Technology, Yancheng Institute of Health Sciences, Yancheng, Jiangsu 224000, China
| | - Yang Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhiwei Quan
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Mingzhe Ma
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Mingzhe Weng
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Zhengdong Zhang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Cuixiang Gao
- Research Centre of Biomedical Technology, Yancheng Institute of Health Sciences, Yancheng, Jiangsu 224000, China
| | - Xinghua Shi
- Research Centre of Biomedical Technology, Yancheng Institute of Health Sciences, Yancheng, Jiangsu 224000, China
| | - Koulan Han
- Clinical College, Yancheng Institute of Health Sciences, Yancheng, Jiangsu 224000, China
| |
Collapse
|
50
|
Zhu CZ, Liu D, Kang WM, Yu JC, Ma ZQ, Ye X, Li K. Ghrelin and gastrointestinal stromal tumors. World J Gastroenterol 2017; 23:1758-1763. [PMID: 28348480 PMCID: PMC5352915 DOI: 10.3748/wjg.v23.i10.1758] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/28/2016] [Accepted: 01/11/2017] [Indexed: 02/06/2023] Open
Abstract
Ghrelin, as a kind of multifunctional protein polypeptide, is mainly produced in the fundus of the stomach and can promote occurrence and development of many tumors, including gastrointestinal tumors, which has been proved by the relevant researches. Most gastrointestinal stromal tumors (GISTs, about 80%), as the most common mesenchymal tumor, also develop in the fundus. Scientific research has confirmed that ghrelin, its receptors and mRNA respectively can be found in GISTs, which demonstrated the existence of a ghrelin autocrine/paracrine loop in GIST tissues. However, no reports to date have specified the mechanism whether ghrelin can promote the occurrence and development of GISTs. Studies of pulmonary artery endothelial cells in a low-oxygen environment and cardiac muscle cells in an ischemic environment have shown that ghrelin can activate the phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway. Moreover, some studies of GISTs have confirmed that activation of the PI3K/AKT/mTOR pathway can indeed promote the growth and progression of GISTs. Whether ghrelin is involved in the development or progression of GISTs through certain pathways remains unknown. Can we find a new target for the treatment of GISTs? This review explores and summaries the relationship among ghrelin, the PI3K/AKT/mTOR pathway and the development of GISTs.
Collapse
|