1
|
Farinha D, Sarmento-Ribeiro AB, Faneca H. Combination of Gene Therapy and Chemotherapy in a New Targeted Hybrid Nanosystem to Hepatocellular Carcinoma. Int J Nanomedicine 2024; 19:12505-12527. [PMID: 39606562 PMCID: PMC11598603 DOI: 10.2147/ijn.s474665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Purpose Hepatocellular carcinoma is the most frequent liver cancer and constitutes one of the main causes of cancer mortality. The combination of targeted therapy drugs, such as selumetinib and perifosine that inhibit cell signaling pathways involved in cell survival and proliferation, with the expression of tumor suppressor transgenes, such as PTEN, may result in an efficient therapeutic approach against HCC. Thus, the main objective of this work was to develop a new lipid-polymer hybrid nanosystem (HNP), composed of a PLGA core coated with a pH-sensitive lipid bilayer functionalized with the targeting ligand GalNAc, in order to specifically and efficiently deliver this novel combination of therapeutic agents in HCC cells. Methods Transmission electron microscopy, zeta potential, Fourier transform infrared spectroscopy, and dynamic light scattering were used to determine the physicochemical properties of hybrid nanosystems and their components. The biological activity and specificity of nanosystems were evaluated using luminescence and flow cytometry. A variety of techniques were used to assess the therapeutic activity of hybrid nanosystems, including the Alamar Blue assay for cell viability; flow cytometry for cell death mechanisms, mitochondrial membrane potential and cell cycle; luminescence for caspase activity; flow cytometry and fluorescence microscopy for cell proliferation; and Western blot for molecular targets levels. Results The obtained results showed that this new hybrid nanosystem not only has a high loading capacity of both drugs, but also allows for substantial expression of the PTEN transgene. In addition, the developed formulation has high stability, adequate physicochemical properties and high specificity to HCC cells. Moreover, the achieved data revealed that this innovative nanosystem presents a high antitumor effect, demonstrated not only by the enhancement on the programmed cell death, but also by the reduction in cell proliferation capacity. Conclusion The generated formulation shows a high anticancer effect, demonstrating a high translational potential for future clinical application in HCC treatment.
Collapse
Affiliation(s)
- Dina Farinha
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Ana Bela Sarmento-Ribeiro
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Clinical Hematology Department, Centro Hospitalar Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - Henrique Faneca
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
2
|
Fang YT, Yang WW, Niu YR, Sun YK. Recent advances in targeted therapy for pancreatic adenocarcinoma. World J Gastrointest Oncol 2023; 15:571-595. [PMID: 37123059 PMCID: PMC10134207 DOI: 10.4251/wjgo.v15.i4.571] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/11/2022] [Accepted: 03/16/2023] [Indexed: 04/12/2023] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is a fatal disease with a 5-year survival rate of 8% and a median survival of 6 mo. In PDAC, several mutations in the genes are involved, with Kirsten rat sarcoma oncogene (90%), cyclin-dependent kinase inhibitor 2A (90%), and tumor suppressor 53 (75%–90%) being the most common. Mothers against decapentaplegic homolog 4 represents 50%. In addition, the self-preserving cancer stem cells, dense tumor microenvironment (fibrous accounting for 90% of the tumor volume), and suppressive and relatively depleted immune niche of PDAC are also constitutive and relevant elements of PDAC. Molecular targeted therapy is widely utilized and effective in several solid tumors. In PDAC, targeted therapy has been extensively evaluated; however, survival improvement of this aggressive disease using a targeted strategy has been minimal. There is currently only one United States Food and Drug Administration-approved targeted therapy for PDAC – erlotinib, but the absolute benefit of erlotinib in combination with gemcitabine is also minimal (2 wk). In this review, we summarize current targeted therapies and clinical trials targeting dysregulated signaling pathways and components of the PDAC oncogenic process, analyze possible reasons for the lack of positive results in clinical trials, and suggest ways to improve them. We also discuss emerging trends in targeted therapies for PDAC: combining targeted inhibitors of multiple pathways. The PubMed database and National Center for Biotechnology Information clinical trial website (www.clinicaltrials.gov) were queried to identify completed and published (PubMed) and ongoing (clinicaltrials.gov) clinical trials (from 2003-2022) using the keywords pancreatic cancer and targeted therapy. The PubMed database was also queried to search for information about the pathogenesis and molecular pathways of pancreatic cancer using the keywords pancreatic cancer and molecular pathways.
Collapse
Affiliation(s)
- Yu-Ting Fang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wen-Wei Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ya-Ru Niu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yong-Kun Sun
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Hebei Cancer Hospital, Chinese Academy of Medical Sciences, Langfang 065001, Hebei Province, China
| |
Collapse
|
3
|
Jin N, Xia Y, Gao Q. Combined PARP inhibitors and small molecular inhibitors in solid tumor treatment (Review). Int J Oncol 2023; 62:28. [PMID: 36601757 PMCID: PMC9851129 DOI: 10.3892/ijo.2023.5476] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/23/2022] [Indexed: 01/05/2023] Open
Abstract
With the development of precision medicine, targeted therapy has attracted extensive attention. Poly(ADP‑ribose) polymerase inhibitors (PARPi) are critical clinical drugs designed to induce cell death and are major antitumor targeted agents. However, preclinical and clinical data have revealed the limitations of PARPi monotherapy. Therefore, their combination with other targeted drugs has become a research hotspot in tumor treatment. Recent studies have demonstrated the critical role of small molecular inhibitors in multiple haematological cancers and solid tumors via cellular signalling modulation, exhibiting potential as a combined pharmacotherapy. In the present review, studies focused on small molecular inhibitors targeting the homologous recombination pathway were summarized and clinical trials evaluating the safety and efficacy of combined treatment were discussed.
Collapse
Affiliation(s)
- Ning Jin
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Yu Xia
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Qinglei Gao
- Key Laboratory of The Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Wuhan, Hubei 430000, P.R. China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| |
Collapse
|
4
|
Li Q, Tie Y, Alu A, Ma X, Shi H. Targeted therapy for head and neck cancer: signaling pathways and clinical studies. Signal Transduct Target Ther 2023; 8:31. [PMID: 36646686 PMCID: PMC9842704 DOI: 10.1038/s41392-022-01297-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/27/2022] [Accepted: 12/13/2022] [Indexed: 01/17/2023] Open
Abstract
Head and neck cancer (HNC) is malignant, genetically complex and difficult to treat and is the sixth most frequent cancer, with tobacco, alcohol and human papillomavirus being major risk factors. Based on epigenetic data, HNC is remarkably heterogeneous, and treatment remains challenging. There is a lack of significant improvement in survival and quality of life in patients with HNC. Over half of HNC patients experience locoregional recurrence or distal metastasis despite the current multiple traditional therapeutic strategies and immunotherapy. In addition, resistance to chemotherapy, radiotherapy and some targeted therapies is common. Therefore, it is urgent to explore more effective and tolerable targeted therapies to improve the clinical outcomes of HNC patients. Recent targeted therapy studies have focused on identifying promising biomarkers and developing more effective targeted therapies. A well understanding of the pathogenesis of HNC contributes to learning more about its inner association, which provides novel insight into the development of small molecule inhibitors. In this review, we summarized the vital signaling pathways and discussed the current potential therapeutic targets against critical molecules in HNC, as well as presenting preclinical animal models and ongoing or completed clinical studies about targeted therapy, which may contribute to a more favorable prognosis of HNC. Targeted therapy in combination with other therapies and its limitations were also discussed.
Collapse
Affiliation(s)
- Qingfang Li
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Tie
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Aqu Alu
- grid.13291.380000 0001 0807 1581Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Huashan Shi
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
5
|
Serwetnyk MA, Blagg BS. The disruption of protein-protein interactions with co-chaperones and client substrates as a strategy towards Hsp90 inhibition. Acta Pharm Sin B 2021; 11:1446-1468. [PMID: 34221862 PMCID: PMC8245820 DOI: 10.1016/j.apsb.2020.11.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/12/2020] [Accepted: 11/13/2020] [Indexed: 12/16/2022] Open
Abstract
The 90-kiloDalton (kD) heat shock protein (Hsp90) is a ubiquitous, ATP-dependent molecular chaperone whose primary function is to ensure the proper folding of several hundred client protein substrates. Because many of these clients are overexpressed or become mutated during cancer progression, Hsp90 inhibition has been pursued as a potential strategy for cancer as one can target multiple oncoproteins and signaling pathways simultaneously. The first discovered Hsp90 inhibitors, geldanamycin and radicicol, function by competitively binding to Hsp90's N-terminal binding site and inhibiting its ATPase activity. However, most of these N-terminal inhibitors exhibited detrimental activities during clinical evaluation due to induction of the pro-survival heat shock response as well as poor selectivity amongst the four isoforms. Consequently, alternative approaches to Hsp90 inhibition have been pursued and include C-terminal inhibition, isoform-selective inhibition, and the disruption of Hsp90 protein-protein interactions. Since the Hsp90 protein folding cycle requires the assembly of Hsp90 into a large heteroprotein complex, along with various co-chaperones and immunophilins, the development of small molecules that prevent assembly of the complex offers an alternative method of Hsp90 inhibition.
Collapse
Key Words
- ADP, adenosine diphosphate
- ATP, adenosine triphosphate
- Aha1, activator of Hsp90 ATPase homologue 1
- CTD, C-terminal domain
- Cdc37, cell division cycle 37
- Disruptors
- Grp94, 94-kD glucose-regulated protein
- HIF-1α, hypoxia-inducing factor-1α
- HIP, Hsp70-interaction protein
- HOP, Hsp70‒Hsp90 organizing protein
- HSQC, heteronuclear single quantum coherence
- Her-2, human epidermal growth factor receptor-2
- Hsp90
- Hsp90, 90-kD heat shock protein
- MD, middle domain
- NTD, N-terminal domain
- Natural products
- PPI, protein−protein interaction
- Peptidomimetics
- Protein−protein interactions
- SAHA, suberoylanilide hydroxamic acid
- SAR, structure–activity relationship
- SUMO, small ubiquitin-like modifier
- Small molecules
- TPR2A, tetratricopeptide-containing repeat 2A
- TRAP1, Hsp75tumor necrosis factor receptor associated protein 1
- TROSY, transverse relaxation-optimized spectroscopy
- hERG, human ether-à-go-go-related gene
Collapse
|
6
|
Karagul MI, Aktas S, Yilmaz SN, Yetkin D, Celikcan HD, Cevik OS. Perifosine and vitamin D combination induces apoptotic and non-apoptotic cell death in endometrial cancer cells. EXCLI JOURNAL 2020; 19:532-546. [PMID: 32483402 PMCID: PMC7257250 DOI: 10.17179/excli2019-1834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 04/23/2020] [Indexed: 12/13/2022]
Abstract
Endometrial cancer is the most common cancer of the female reproductive system. Combination treatment with specific agents has been widely used as a targeted therapy for cancer. In this study, we aimed to investigate the anti-proliferative and apoptotic effects of varying concentrations of perifosine and vitamin D on the human endometrial cancer cell line (HEC-1A). HEC-1A cells were exposed to perifosine (10 μM, 30 μM), vitamin D (50 nM, 200 nM) and combinations of both for 48 h and 72 h. Monitoring of cell proliferation in a time-dependent manner was performed with the xCELLigence RTCA DP system. The levels of BCL2, BAX and P53 mRNA expression were examined using RT-qPCR. Apoptosis was determined using Annexin V, which were followed by flow cytometry analysis. Ultra-structural morphology of cells was analyzed by transmission electron microscopy (TEM) for 72 h. The anti-proliferative and apoptotic effects of the perifosine+vitamin D combination (30 μM + 200 nM at 48 h and 10 μM + 200 nM at 72 h) on HEC-1A cells were higher than in perifosine and vitamin D alone. It was observed that perifosine has increased the expression of BAX mRNA in HEC-1A cells in a dose-dependent manner. While perifosine+vitamin D combinations increased P53 mRNA expression in HEC-1A cells we did not find any significant change in BCL2, BAX mRNA expression levels. In TEM examinations of HEC-1A cells, perifosine appeared to lead autophagic cell death, whereas vitamin D caused paraptosis-like cell death and combination of perifosine+vitamin D caused apoptotic and non-apoptotic (paraptotic, autophagic and necrotic) cell death. Therefore, it is considered that the combination of both drugs in the treatment of endometrial cancer might be an alternative and effective treatment option through activating the apoptotic and non-apoptotic cell death mechanisms in cancer cells.
Collapse
Affiliation(s)
- Meryem Ilkay Karagul
- Department of Histology and Embryology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Savas Aktas
- Department of Histology and Embryology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Sakir Necat Yilmaz
- Department of Histology and Embryology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Derya Yetkin
- Advanced Technology of Education, Research and Application Center, Mersin University, Mersin, Turkey
| | - Havva Didem Celikcan
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Ozge Selin Cevik
- Department of Physiology, Faculty of Medicine, Mersin University, Mersin, Turkey
| |
Collapse
|
7
|
Preclinical efficacy of a novel dual PI3K/mTOR inhibitor, CMG002, alone and in combination with sorafenib in hepatocellular carcinoma. Cancer Chemother Pharmacol 2019; 84:809-817. [PMID: 31385002 DOI: 10.1007/s00280-019-03918-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/29/2019] [Indexed: 12/23/2022]
Abstract
PURPOSE Sorafenib has been the only first systemic drug that improves survival of patients with advanced hepatocellular carcinoma (HCC). However, because the response rate of sorafenib is relatively low, novel therapeutic strategies are needed to improve survival in patients with HCC. This study investigated the effect of CMG002 alone and in combination with sorafenib on HCC in vitro and vivo. METHODS The effect of a newly developed dual PI3K/mTOR inhibitor, CMG002, on the proliferation of Huh-7 and HepG2 HCC cells was investigated using the MTT assay. Western blotting was performed to assess phosphorylation of the key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. HepG2 cells were inoculated into mice, which were treated with vehicle, sorafenib, CMG002, and their combinations. Tumor cell proliferation and tumor angiogenesis were evaluated by immunohistochemical analysis of Ki-67 and CD31, respectively. Tumor cell apoptosis was detected by the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Levels of key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways were evaluated by western blot analysis. RESULTS The combination of sorafenib and CMG002 additively inhibited Huh-7 and HepG2 cell proliferation compared to single-agent treatment. Sorafenib and CMG002 as single agents differentially inhibited or activated key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. The combination of sorafenib and CMG002 inhibited all key enzymes in the two pathways. Treatment with CMG002 for 4 weeks alone and in combination with sorafenib strongly inhibited tumor growth. CMG002 inhibited HCC cell proliferation, induced apoptosis, and decreased tumor angiogenesis. Furthermore, these effects were enhanced when CMG002 was combined with sorafenib. CONCLUSIONS The combination of CMG002 and sorafenib significantly inhibited HCC cell proliferation and tumorigenesis by inhibiting the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. These findings suggest that CMG002 to be a potential novel candidate treatment for HCC.
Collapse
|
8
|
Zhuang H, Qiang Z, Shao X, Wang H, Dang Y, Wang Z, Wu F, Wei W, Li Y. Integration of metabolomics and expression of enolase-phosphatase 1 links to hepatocellular carcinoma progression. Theranostics 2019; 9:3639-3652. [PMID: 31281503 PMCID: PMC6587162 DOI: 10.7150/thno.31693] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 04/19/2019] [Indexed: 02/07/2023] Open
Abstract
Reprogramming of cellular metabolism is one of the hallmarks for cancer, in which tumor cells rewire their metabolic fluxes to generate sufficient energy and biosynthetic intermediates. Therefore, elucidating the correlation between cellular metabolism and hepatocellular carcinoma (HCC) progression may provide insights into novel approaches to cancer therapy. Methods: We assembled an integrated pathway-level metabolic profiling by mining metabolomic, transcriptomic and proteomic data of three HCC cell lines with increasing metastatic potentials. Immunohistochemical staining was performed in a tissue microarray from 185 HCC clinical specimens. Kaplan-Meier survival and Cox regression analyses were applied to test the association between gene expression and survival outcome. In vitro assays were conducted to investigate the functional role of enolase-phosphatase 1 (ENOPH1) in HCC malignant behaviors. Reversed genetics analysis was performed to determine the function of ENOPH1 in HCC metastasis. An intrahepatic mouse model further confirmed the role of ENOPH1 in metastasis. Results: We have determined that HCC cell metastasis is associated with alterations in metabolite levels and expressions of metabolic enzymes in the cysteine/methionine metabolism pathway, and show that one of metabolic enzymes, enolase-phosphatase 1 (ENOPH1), is persistently upregulated with an increase in metastatic potential. The upregulation of ENOPH1 expression was observed as an independent prognostic factor for HCC patients. ENOPH1 overexpression promoted cell migration and invasion, whereas ENOPH1 downregulation inhibited cell migration and invasion. Furthermore, an enhanced phosphorylation of AKT with ENOPH1 upregulation was observed. ENOPH1-mediated malignant capacity in HCC cells can be rescued by an AKT inhibitor. Conclusion: Taken together, our findings illustrate that ENOPH1 promotes HCC progression and could serve as a novel biomarker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Hao Zhuang
- Department of Hepatic Biliary Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450000, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Key Lab of Immune Microenvironment and Disease (Ministry of Education) Tianjin Medical University, Tianjin 300070, China
| | - Zhaoyan Qiang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xiaowen Shao
- Department of Pathogen Biology, School of Basic Medical Sciences, Key Lab of Immune Microenvironment and Disease (Ministry of Education) Tianjin Medical University, Tianjin 300070, China
| | - Huan Wang
- School of Life Sciences, Chongqing University, Chongqing, 400044, China
| | - Yamei Dang
- Department of Pathogen Biology, School of Basic Medical Sciences, Key Lab of Immune Microenvironment and Disease (Ministry of Education) Tianjin Medical University, Tianjin 300070, China
| | - Zun Wang
- Department of Pathogen Biology, School of Basic Medical Sciences, Key Lab of Immune Microenvironment and Disease (Ministry of Education) Tianjin Medical University, Tianjin 300070, China
| | - Fei Wu
- Department of Pathogen Biology, School of Basic Medical Sciences, Key Lab of Immune Microenvironment and Disease (Ministry of Education) Tianjin Medical University, Tianjin 300070, China
| | - Wen Wei
- School of Life Sciences, Chongqing University, Chongqing, 400044, China
| | - Yongmei Li
- Department of Pathogen Biology, School of Basic Medical Sciences, Key Lab of Immune Microenvironment and Disease (Ministry of Education) Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
9
|
Barati Bagherabad M, Afzaljavan F, ShahidSales S, Hassanian SM, Avan A. Targeted therapies in pancreatic cancer: Promises and failures. J Cell Biochem 2018; 120:2726-2741. [PMID: 28703890 DOI: 10.1002/jcb.26284] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/11/2018] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an incidence rate nearly equal to its mortality rate. The poor prognosis of the disease can be explained by the absence of effective biomarkers for screening and early detection, together with the aggressive behavior and resistance to the currently available chemotherapy. The therapeutic failure can also be attributed to the inter-/intratumor genetic heterogeneity and the abundance of tumor stroma that occupies the majority of the tumor mass. Gemcitabine is used in the treatment of PDAC; however, the response rate is less than 12%. A recent phase III trial revealed that the combination of oxaliplatin, irinotecan, fluorouracil, and leucovorin could be an option for the treatment of metastatic PDAC patients with good performance status, although these approaches can result in high toxicity level. Further investigations are required to develop innovative anticancer agents that either improve gemcitabine activity, within novel combinatorial approaches or acts with a better efficacy than gemcitabine. The aim of the current review is to give an overview of preclinical and clinical studies targeting key dysregulated signaling pathways in PDAC.
Collapse
Affiliation(s)
- Matineh Barati Bagherabad
- Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Afzaljavan
- Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soodabeh ShahidSales
- Cancer Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, Mashhad, Iran.,Molecular Medicine group, Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Bernardes N, Fialho AM. Perturbing the Dynamics and Organization of Cell Membrane Components: A New Paradigm for Cancer-Targeted Therapies. Int J Mol Sci 2018; 19:E3871. [PMID: 30518103 PMCID: PMC6321595 DOI: 10.3390/ijms19123871] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/27/2018] [Accepted: 12/03/2018] [Indexed: 01/26/2023] Open
Abstract
Cancer is a multi-process disease where different mechanisms exist in parallel to ensure cell survival and constant adaptation to the extracellular environment. To adapt rapidly, cancer cells re-arrange their plasma membranes to sustain proliferation, avoid apoptosis and resist anticancer drugs. In this review, we discuss novel approaches based on the modifications and manipulations that new classes of molecules can exert in the plasma membrane lateral organization and order of cancer cells, affecting growth factor signaling, invasiveness, and drug resistance. Furthermore, we present azurin, an anticancer protein from bacterial origin, as a new approach in the development of therapeutic strategies that target the cell membrane to improve the existing standard therapies.
Collapse
Affiliation(s)
- Nuno Bernardes
- iBB-Institute for Bioengineering and Biosciences, Biological Sciences Research Group, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal.
| | - Arsenio M Fialho
- iBB-Institute for Bioengineering and Biosciences, Biological Sciences Research Group, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal.
- Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal.
| |
Collapse
|
11
|
Li H, Shi CX, Liu H, Zhang HH, Sang HM, Soyfoo MD, Cao JL, Xu SF, Jiang JX. Hiwi overexpression does not affect proliferation, migration or apoptosis of liver cancer cells in vitro or in vivo. Oncol Lett 2018; 15:9711-9718. [PMID: 29928347 PMCID: PMC6004705 DOI: 10.3892/ol.2018.8585] [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: 08/07/2016] [Accepted: 03/16/2018] [Indexed: 11/27/2022] Open
Abstract
Piwi like RNA-mediated gene silencing 1 (Hiwi) is a human homolog of the Piwi gene family that has been reported to be upregulated in hepatocellular carcinoma (HCC). The present study aimed to investigate the role of Hiwi in the initiation and development of HCC in vitro and in vivo. Adenovirus-mediated Hiwi overexpression was established in primary murine hepatocytes and SMMC7721 HCC cells. Cell viability and proliferation were assessed using MTT and EdU assays, respectively. Cell migration was measured using a scratch migration assay. The cell cycle was assessed using flow cytometry, and the expression of genes associated with the epithelial mesenchymal transition (EMT) was assessed using reverse transcription-quantitative polymerase chain reaction. SMMC7721 cells that stably express Hiwi were also generated and injected subcutaneously into the nude mice, and tumor growth was examined. Recombinant adenovirus encoding green fluorescent protein or Hiwi was delivered by injection into the tail vein, and its effect on murine hepatocyte gene expression was studied. The present study revealed that the overexpression of Hiwi did not affect the proliferation or migration of liver cancer cells and failed to suppress perifosine- or doxorubicin-induced apoptosis in vitro. The tumors of mice that were injected with Hiwi-expressing SMMC7721 cells were not significantly larger compared with mice that were injected with control SMMC7721 cells. Hiwi overexpression did not noticeably alter the expression of genes involved in EMT, either in vitro or in vivo. The results of the present study indicate that although expression of Hiwi is associated with HCC development and progression in the clinic, it does not act as an oncogene in liver cancer cells.
Collapse
Affiliation(s)
- Hao Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Chen-Xi Shi
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Hui Liu
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Hai-Han Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Huai-Ming Sang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Muhammad-Djaleel Soyfoo
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jiu-Liang Cao
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Shun-Fu Xu
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jian-Xia Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
12
|
Wang Y, Ning Z, Zhou X, Yang Z, Tang H, Xu M, Wang X, Zhao J, Bai Y. Neuregulin1 acts as a suppressor in human lung adenocarcinoma via AKT and ERK1/2 pathway. J Thorac Dis 2018; 10:3166-3179. [PMID: 30069312 DOI: 10.21037/jtd.2018.05.175] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Background Neuregulin1 (NRG1) is critical signaling protein that mediates the activation of downstream signaling pathways associated with malignancies. Multiple gene fusions related to NRG1 have been found in lung cancer. However, the underlying role NRG1 in lung cancer is yet unclear. Therefore, the present study investigated the biological functions on human lung adenocarcinoma (LUAD). Methods The expression of NRG1 was detected in LUAD tissues by Western blot (WB), quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC). The expression of NRG1 was upregulated by the addition of exogenous NRG1 and downregulated by small interfering RNA (siRNA), and the biological behaviors of LUAD cells were assessed: cell proliferation by MTT assay, cell cycle and apoptosis by flow cytometry analysis, and migration and invasion using Transwell system. Finally, the pathway underlying the cellular function was analyzed by WB. Results A lower expression of NRG1 was observed in LUAD cancer tissues (P<0.05). Moreover, the addition of exogenous NRG1 reduced the cell proliferation, migration, and invasion (P<0.001), while the downregulation of endogenous NRG1 promoted the three kinds of biological behaviors of LUAD cell lines (P<0.001); however, these manifestations did no effect on the distribution of cell cycle and apoptosis status (P>0.05). Furthermore, the deficiency of NRG1 reduced the expression of p-ERK1/2 and p-AKT at the protein level (P<0.001). Conclusions The current results suggested that NRG1 might be a suppressor in the development of LUAD, and its function was related to AKT and ERK1/2 pathway.
Collapse
Affiliation(s)
- Youya Wang
- Department of Cardiothoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.,Clinical Medical School, Hubei University of Science and Technology, Xianning 437100, China
| | - Zhifeng Ning
- Basic Medical School, Hubei University of Science and Technology, Xianning 437100, China
| | - Xuefeng Zhou
- Department of Cardiothoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zetian Yang
- Department of Cardiothoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hexiao Tang
- Department of Cardiothoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ming Xu
- Department of Cardiothoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xianguo Wang
- Department of Cardiothoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jinping Zhao
- Department of Cardiothoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yuting Bai
- Department of Cardiothoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.,Clinical Medical School, Hubei University of Science and Technology, Xianning 437100, China
| |
Collapse
|
13
|
Atractylenolide II Inhibits Proliferation, Motility and Induces Apoptosis in Human Gastric Carcinoma Cell Lines HGC-27 and AGS. Molecules 2017; 22:molecules22111886. [PMID: 29099789 PMCID: PMC6150195 DOI: 10.3390/molecules22111886] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 11/01/2017] [Indexed: 12/20/2022] Open
Abstract
Atractylenolide II (AT-II) exhibits several biological and pharmacological functions, especially anti-cancer activity as the major sesquiterpene lactones isolated from Atractylodes macrocephala (also named Baizhu in Chinese). However, the effects and mechanisms of AT-II on human gastric cancer remain unclear. Cell Counting Kit-8 (CCK-8) assay, morphological changes, flow cytometry, wound healing assay and Western blot analysis were used to investigate the effects of AT-II on cell proliferation, apoptosis and motility of human gastric carcinoma cell lines HGC-27 and AGS. Our results indicated that AT-II could significantly inhibit cell proliferation, motility and induce apoptosis in a dose and time-dependent manner. Western blot analysis showed that the expression level of Bax was upregulated and the expression levels of B-cell lymphoma-2 (Bcl-2), phosphorylated-protein kinase B (p-Akt) and phosphorylated-ERK (p-ERK) were downregulated compared to control group. In conclusion, the findings suggested that AT-II exerted significant anti-tumor effects on gastric carcinoma cells by modulating Akt/ERK signaling pathway, which might shed light on therapy of gastric carcinoma.
Collapse
|
14
|
Wu YL, Maachani UB, Schweitzer M, Singh R, Wang M, Chang R, Souweidane MM. Dual Inhibition of PI3K/AKT and MEK/ERK Pathways Induces Synergistic Antitumor Effects in Diffuse Intrinsic Pontine Glioma Cells. Transl Oncol 2017; 10:221-228. [PMID: 28189993 PMCID: PMC5302185 DOI: 10.1016/j.tranon.2016.12.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 12/18/2016] [Accepted: 12/20/2016] [Indexed: 01/09/2023] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a devastating disease with an extremely poor prognosis. Recent studies have shown that platelet-derived growth factor receptor (PDGFR) and its downstream effector pathway, PI3K/AKT/mTOR, are frequently amplified in DIPG, and potential therapies targeting this pathway have emerged. However, the addition of targeted single agents has not been found to improve clinical outcomes in DIPG, and targeting this pathway alone has produced insufficient clinical responses in multiple malignancies investigated, including lung, endometrial, and bladder cancers. Acquired resistance also seems inevitable. Activation of the Ras/Raf/MEK/ERK pathway, which shares many nodes of cross talk with the PI3K/AKT pathway, has been implicated in the development of resistance. In the present study, perifosine, a PI3K/AKT pathway inhibitor, and trametinib, a MEK inhibitor, were combined, and their therapeutic efficacy on DIPG cells was assessed. Growth delay assays were performed with each drug individually or in combination. Here, we show that dual inhibition of PI3K/AKT and MEK/ERK pathways synergistically reduced cell viability. We also reveal that trametinib induced AKT phosphorylation in DIPG cells that could not be effectively attenuated by the addition of perifosine, likely due to the activation of other compensatory mechanisms. The synergistic reduction in cell viability was through the pronounced induction of apoptosis, with some effect from cell cycle arrest. We conclude that the concurrent inhibition of the PI3K/AKT and MEK/ERK pathways may be a potential therapeutic strategy for DIPG.
Collapse
Affiliation(s)
- Y Linda Wu
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY
| | - Uday Bhanu Maachani
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY
| | - Melanie Schweitzer
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY
| | - Ranjodh Singh
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY
| | - Melinda Wang
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY
| | - Raymond Chang
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY
| | - Mark M Souweidane
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY.
| |
Collapse
|
15
|
Akt targeting as a strategy to boost chemotherapy efficacy in non-small cell lung cancer through metabolism suppression. Sci Rep 2017; 7:45136. [PMID: 28332584 PMCID: PMC5362809 DOI: 10.1038/srep45136] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/15/2017] [Indexed: 01/13/2023] Open
Abstract
Metabolic reprogramming is a hallmark of cancer development, mediated by genetic and epigenetic alterations that may be pharmacologically targeted. Among oncogenes, the kinase Akt is commonly overexpressed in tumors and favors glycolysis, providing a rationale for using Akt inhibitors. Here, we addressed the question of whether and how inhibiting Akt activity could improve therapy of non-small cell lung cancer (NSCLC) that represents more than 80% of all lung cancer cases. First, we demonstrated that Akt inhibitors interacted synergistically with Microtubule-Targeting Agents (MTAs) and specifically in cancer cell lines, including those resistant to chemotherapy agents and anti-EGFR targeted therapies. In vivo, we further revealed that the chronic administration of low-doses of paclitaxel - i.e. metronomic scheduling - and the anti-Akt perifosine was the most efficient and the best tolerated treatment against NSCLC. Regarding drug mechanism of action, perifosine potentiated the pro-apoptotic effects of paclitaxel, independently of cell cycle arrest, and combining paclitaxel/perifosine resulted in a sustained suppression of glycolytic and mitochondrial metabolism. This study points out that targeting cancer cell bioenergetics may represent a novel therapeutic avenue in NSCLC, and provides a strong foundation for future clinical trials of metronomic MTAs combined with Akt inhibitors.
Collapse
|
16
|
Ríos-Marco P, Marco C, Gálvez X, Jiménez-López JM, Carrasco MP. Alkylphospholipids: An update on molecular mechanisms and clinical relevance. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:1657-1667. [PMID: 28238819 DOI: 10.1016/j.bbamem.2017.02.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 02/20/2017] [Accepted: 02/21/2017] [Indexed: 11/16/2022]
Abstract
Alkylphospholipids (APLs) represent a new class of drugs which do not interact directly with DNA but act on the cell membrane where they accumulate and interfere with lipid metabolism and signalling pathways. This review summarizes the mode of action at the molecular level of these compounds. In this sense, a diversity of mechanisms has been suggested to explain the actions of clinically-relevant APLs, in particular, in cancer treatment. One consistently reported finding is that APLs reduce the biosynthesis of phosphatidylcholine (PC) by inhibiting the rate-limiting enzyme CTP:phosphocholine cytidylyltransferase (CT). APLs also alter intracellular cholesterol traffic and metabolism in human tumour-cell lines, leading to an accumulation of cholesterol inside the cell. An increase in cholesterol biosynthesis associated with a decrease in the synthesis of choline-containing phospholipids and cholesterol esterification leads to a change in the free-cholesterol:PC ratio in cells exposed to APLs. Akt phosphorylation status after APL exposure shows that this critical regulator for cell survival is modulated by changes in cholesterol levels induced in the plasma membrane by these lipid analogues. Furthermore, APLs produce cell ultrastructural alterations with an abundant autophagic vesicles and autolysosomes in treated cells, indicating an interference of autophagy process after APL exposure. Thus, antitumoural APLs interfere with the proliferation of tumour cells via a complex mechanism involving phospholipid and cholesterol metabolism, interfere with lipid-dependent survival-signalling pathways and autophagy. Although APLs also exert antiparasitic, antibacterial, and antifungal effects, in this review we provide a summary of the antileishmanial activity of these lipid analogues. This article is part of a Special Issue entitled: Membrane Lipid Therapy: Drugs Targeting Biomembranes edited by Pablo V. Escribá.
Collapse
Affiliation(s)
- Pablo Ríos-Marco
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain
| | - Carmen Marco
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain
| | - Xiomara Gálvez
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain
| | - José M Jiménez-López
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain.
| | - María P Carrasco
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain.
| |
Collapse
|
17
|
Massihnia D, Avan A, Funel N, Maftouh M, van Krieken A, Granchi C, Raktoe R, Boggi U, Aicher B, Minutolo F, Russo A, Leon LG, Peters GJ, Giovannetti E. Phospho-Akt overexpression is prognostic and can be used to tailor the synergistic interaction of Akt inhibitors with gemcitabine in pancreatic cancer. J Hematol Oncol 2017; 10:9. [PMID: 28061880 PMCID: PMC5219723 DOI: 10.1186/s13045-016-0371-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 12/08/2016] [Indexed: 12/19/2022] Open
Abstract
Background There is increasing evidence of a constitutive activation of Akt in pancreatic ductal adenocarcinoma (PDAC), associated with poor prognosis and chemoresistance. Therefore, we evaluated the expression of phospho-Akt in PDAC tissues and cells, and investigated molecular mechanisms influencing the therapeutic potential of Akt inhibition in combination with gemcitabine. Methods Phospho-Akt expression was evaluated by immunohistochemistry in tissue microarrays (TMAs) with specimens tissue from radically-resected patients (n = 100). Data were analyzed by Fisher and log-rank test. In vitro studies were performed in 14 PDAC cells, including seven primary cultures, characterized for their Akt1 mRNA and phospho-Akt/Akt levels by quantitative-RT-PCR and immunocytochemistry. Growth inhibitory effects of Akt inhibitors and gemcitabine were evaluated by SRB assay, whereas modulation of Akt and phospho-Akt was investigated by Western blotting and ELISA. Cell cycle perturbation, apoptosis-induction, and anti-migratory behaviors were studied by flow cytometry, AnnexinV, membrane potential, and migration assay, while pharmacological interaction with gemcitabine was determined with combination index (CI) method. Results Immunohistochemistry of TMAs revealed a correlation between phospho-Akt expression and worse outcome, particularly in patients with the highest phospho-Akt levels, who had significantly shorter overall and progression-free-survival. Similar expression levels were detected in LPC028 primary cells, while LPC006 were characterized by low phospho-Akt. Remarkably, Akt inhibitors reduced cancer cell growth in monolayers and spheroids and synergistically enhanced the antiproliferative activity of gemcitabine in LPC028, while this combination was antagonistic in LPC006 cells. The synergistic effect was paralleled by a reduced expression of ribonucleotide reductase, potentially facilitating gemcitabine cytotoxicity. Inhibition of Akt decreased cell migration and invasion, which was additionally reduced by the combination with gemcitabine. This combination significantly increased apoptosis, associated with induction of caspase-3/6/8/9, PARP and BAD, and inhibition of Bcl-2 and NF-kB in LPC028, but not in LPC006 cells. However, targeting the key glucose transporter Glut1 resulted in similar apoptosis induction in LPC006 cells. Conclusions These data support the analysis of phospho-Akt expression as both a prognostic and a predictive biomarker, for the rational development of new combination therapies targeting the Akt pathway in PDAC. Finally, inhibition of Glut1 might overcome resistance to these therapies and warrants further studies. Electronic supplementary material The online version of this article (doi:10.1186/s13045-016-0371-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniela Massihnia
- Department of Medical Oncology VU University Medical Center, Cancer Center Amsterdam, CCA room 1.52, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.,Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Amir Avan
- Metabolic syndrome Research center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niccola Funel
- Cancer Pharmacology Lab, AIRC Start Up Unit, University of Pisa, Pisa, Italy
| | - Mina Maftouh
- Department of Medical Oncology VU University Medical Center, Cancer Center Amsterdam, CCA room 1.52, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Anne van Krieken
- Department of Medical Oncology VU University Medical Center, Cancer Center Amsterdam, CCA room 1.52, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | | | - Rajiv Raktoe
- Department of Medical Oncology VU University Medical Center, Cancer Center Amsterdam, CCA room 1.52, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Ugo Boggi
- Department of Surgery, University of Pisa, Pisa, Italy
| | - Babette Aicher
- Æterna Zentaris GmbH, Frankfurt am Main, Frankfurt, Germany
| | | | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Leticia G Leon
- Cancer Pharmacology Lab, AIRC Start Up Unit, University of Pisa, Pisa, Italy
| | - Godefridus J Peters
- Department of Medical Oncology VU University Medical Center, Cancer Center Amsterdam, CCA room 1.52, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology VU University Medical Center, Cancer Center Amsterdam, CCA room 1.52, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands. .,Cancer Pharmacology Lab, AIRC Start Up Unit, University of Pisa, Pisa, Italy.
| |
Collapse
|
18
|
Huang Y, Liu G, Yang F, Xing X, Li Y, Huang Z, Yuan H. Induction of apoptosis and proliferation inhibition of hepatocellular carcinoma by 6-chloro-2-methoxy- N-(phenylmethyl)-9-acridinamine (BA): in vitro and vivo studies. Cancer Cell Int 2017; 17:66. [PMID: 28680363 PMCID: PMC5496258 DOI: 10.1186/s12935-017-0435-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 06/15/2017] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND 6-Chloro-2-methoxy-N-(phenylmethyl)-9-acridinamine (BA), a novel sponge-derived compound, has been reported to elicit a cytotoxic effect by inhibiting cell proliferation. METHODS In this study, we investigated the anti-tumor effect of BA in human hepatocellular carcinoma (HCC) in vitro and in vivo using SMMC-7721 cells. The impact of BA on SMMC-7721 cells was determined by proliferation (clonogenicity and MTT), apoptosis (flow cytometry with annexin V-FITC labeling) and tumor cell migration (Transwell). Apoptosis-related molecules in the PI3K/AKT signaling pathway were examined via Western blotting. We also evaluated the effects of BA on tumor growth using a xenograft nude mouse model. RESULTS The data showed that BA induced dose-dependent cytotoxicity, anti-proliferation, anti-migration and apoptosis in SMMC-7721 cells, accompanied by activation of caspase-3 and a decreased level of caspase-9. Moreover, BA decreased PI3K and p-AKT levels, which indicated the cytotoxicity of BA through the PI3K/Akt pathway. Finally, we confirmed that BA inhibited tumor growth in an HCC xenograft mouse model. CONCLUSIONS We concluded that BA induced apoptosis and decreased PI3K and p-AKT expression in human HCC with no effect on the liver, kidney, spleen or lungs. These findings suggest that BA could provide a novel strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Yun Huang
- Center of Clinical Pharmacology, The Third Xiang Ya Hospital, Central South University, Changsha, 410013 China
- Department of Pharmacy, Ningbo City Medical Treatment Center Lihuili Hospital, Ningbo, 315000 China
| | - Guohua Liu
- Center of Clinical Pharmacology, The Third Xiang Ya Hospital, Central South University, Changsha, 410013 China
- Department of Pharmacy, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041 China
| | - Feng Yang
- Center of Clinical Pharmacology, The Third Xiang Ya Hospital, Central South University, Changsha, 410013 China
| | - Xiaowei Xing
- Center of Clinical Pharmacology, The Third Xiang Ya Hospital, Central South University, Changsha, 410013 China
| | - Ying Li
- Center of Clinical Pharmacology, The Third Xiang Ya Hospital, Central South University, Changsha, 410013 China
| | - Zhijun Huang
- Center of Clinical Pharmacology, The Third Xiang Ya Hospital, Central South University, Changsha, 410013 China
- National & Local Joint Engineering Laboratory for the Technology of Clinical Drug Evaluation, Changsha, 410013 China
| | - Hong Yuan
- Center of Clinical Pharmacology, The Third Xiang Ya Hospital, Central South University, Changsha, 410013 China
- National & Local Joint Engineering Laboratory for the Technology of Clinical Drug Evaluation, Changsha, 410013 China
| |
Collapse
|
19
|
Sui CJ, Xu M, Li WQ, Yang JM, Yan HZ, Liu HM, Xia CY, Yu HY. Co-culture of hepatoma cells with hepatocytic precursor (stem-like) cells inhibits tumor cell growth and invasion by downregulating Akt/NF-κB expression. Oncol Lett 2016; 12:4054-4060. [PMID: 27895771 PMCID: PMC5104234 DOI: 10.3892/ol.2016.5128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 08/05/2015] [Indexed: 11/10/2022] Open
Abstract
Hepatocytic stem cells (HSCs) have inhibitory effects on hepatocarcinoma cells. The present study investigated the effects of HSC activity in hepatocarcinoma cells in vitro. A Transwell co-culture system of hepatocytic precursor (stem-like) WB-F344 cells and hepatoma CBRH-7919 cells was used to assess HSC activity in metastasized hepatoma cells in vitro. Nude mouse xenografts were used to assess HSC activity in vivo. Co-culture of hepatoma CBRH-7919 cells with WB-F344 cells suppressed the growth and colony formation, tumor cell migration and invasion capacity of CBRH-7919 cells. The nude mouse xenograft assay demonstrated that the xenograft size of CBRH-7919 cells following co-culture with WB-F344 cells was significantly smaller compared with that of control cells. Furthermore, the expression levels of the epithelial markers E-cadherin and β-catenin were downregulated, while the mesenchymal markers α-SMA and vimentin were upregulated. Co-culture of CBRH-7919 cells with WB-F344 cells downregulated NF-κB and phospho-Akt expression. In conclusion, hepatocytic precursor (stem-like) WB-F344 cells inhibited the growth, colony formation and invasion capacity of metastasized hepatoma CBRH-7919 cells in vitro and in vivo by downregulating Akt/NF-κB signaling.
Collapse
Affiliation(s)
- Cheng-Jun Sui
- Department of Special Medical Care Unit I and Liver Transplantation, The Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, P.R. China
| | - Miao Xu
- Department of Geratology, Changhai Hospital, Shanghai 200438, P.R. China
| | - Wei-Qing Li
- Department of Pathology, Changzheng Hospital, The Second Military Medical University, Shanghai 200003, P.R. China
| | - Jia-Mei Yang
- Department of Special Medical Care Unit I and Liver Transplantation, The Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, P.R. China
| | - Hong-Zhu Yan
- Department of Pathology, Changzheng Hospital, The Second Military Medical University, Shanghai 200003, P.R. China
| | - Hui-Min Liu
- Department of Pathology, Changzheng Hospital, The Second Military Medical University, Shanghai 200003, P.R. China
| | - Chun-Yan Xia
- Department of Pathology, Changzheng Hospital, The Second Military Medical University, Shanghai 200003, P.R. China
| | - Hong-Yu Yu
- Department of Pathology, Changzheng Hospital, The Second Military Medical University, Shanghai 200003, P.R. China
- Correspondence to: Dr Hong-Yu Yu, Department of Pathology, Changzheng Hospital, The Second Military Medical University, 415 Fengyang Road, Shanghai 200003, P.R. China, E-mail:
| |
Collapse
|
20
|
Biophysics in cancer: The relevance of drug-membrane interaction studies. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:2231-2244. [DOI: 10.1016/j.bbamem.2016.06.025] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 05/31/2016] [Accepted: 06/26/2016] [Indexed: 12/26/2022]
|
21
|
Lee D, Wang YH, Kalaitzidis D, Ramachandran J, Eda H, Sykes DB, Raje N, Scadden DT. Endogenous transmembrane protein UT2 inhibits pSTAT3 and suppresses hematological malignancy. J Clin Invest 2016; 126:1300-10. [PMID: 26927669 PMCID: PMC4811118 DOI: 10.1172/jci84620] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 01/14/2016] [Indexed: 12/27/2022] Open
Abstract
Regulation of STAT3 activation is critical for normal and malignant hematopoietic cell proliferation. Here, we have reported that the endogenous transmembrane protein upstream-of-mTORC2 (UT2) negatively regulates activation of STAT3. Specifically, we determined that UT2 interacts directly with GP130 and inhibits phosphorylation of STAT3 on tyrosine 705 (STAT3Y705). This reduces cytokine signaling including IL6 that is implicated in multiple myeloma and other hematopoietic malignancies. Modulation of UT2 resulted in inverse effects on animal survival in myeloma models. Samples from multiple myeloma patients also revealed a decreased copy number of UT2 and decreased expression of UT2 in genomic and transcriptomic analyses, respectively. Together, these studies identify a transmembrane protein that functions to negatively regulate cytokine signaling through GP130 and pSTAT3Y705 and is molecularly and mechanistically distinct from the suppressors of cytokine signaling (SOCS) family of genes. Moreover, this work provides evidence that perturbations of this activation-dampening molecule participate in hematologic malignancies and may serve as a key determinant of multiple myeloma pathophysiology. UT2 is a negative regulator shared across STAT3 and mTORC2 signaling cascades, functioning as a tumor suppressor in hematologic malignancies driven by those pathways.
Collapse
Affiliation(s)
- Dongjun Lee
- Center for Regenerative Medicine and
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology and
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Ying-Hua Wang
- Center for Regenerative Medicine and
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology and
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Demetrios Kalaitzidis
- Center for Regenerative Medicine and
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology and
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | | | - Homare Eda
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David B. Sykes
- Center for Regenerative Medicine and
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology and
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Noopur Raje
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David T. Scadden
- Center for Regenerative Medicine and
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology and
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
22
|
Jiang B, Liu F, Liu Z, Zhang T, Hua D. B7-H3 increases thymidylate synthase expression via the PI3k-Akt pathway. Tumour Biol 2016; 37:9465-72. [PMID: 26787540 DOI: 10.1007/s13277-015-4740-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 12/22/2015] [Indexed: 01/05/2023] Open
Abstract
B7-H3, a member of the B7 family, has been reported to be highly expressed in colorectal cancer and is associated with poor prognosis and overall survival. In this study, we found that overexpression of B7-H3 protected SW80 and HCT8 cells from 5-fluorouracil (5-FU) using CCK-8 assays by inducing resistance to 5-FU chemotherapy. Further investigation has revealed elevated expression of thymidylate synthase (TS) and upregulation of the PI3-kinase (PI3K)/Akt pathway in B7-H3 overexpressing cells. The effects of B7-H3 on activation of the PI3K/Akt pathway and elevation of TS expression could be blocked by LY294002, a specific inhibitor of the PI3K signaling pathway. These results implied that B7-H3 can induce colorectal cancer cell resistance to 5-FU by increasing TS expression and PI3K/Akt/TS signaling and plays an important role during these processes. This study provides more proof concerning the non-immunology effect of B7 molecules, a reminder that both co-stimulatory or inhibitory effects and non-immunology effects should be devoted equal attention.
Collapse
Affiliation(s)
- Bo Jiang
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
| | - Fen Liu
- Institute of Cancer, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
| | - ZhiHui Liu
- Institute of Cancer, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
| | - Ting Zhang
- Institute of Cancer, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
| | - Dong Hua
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China.
| |
Collapse
|
23
|
Popescu AM, Purcaru SO, Alexandru O, Dricu A. New perspectives in glioblastoma antiangiogenic therapy. Contemp Oncol (Pozn) 2015; 20:109-18. [PMID: 27358588 PMCID: PMC4925727 DOI: 10.5114/wo.2015.56122] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/15/2015] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GB) is highly vascularised tumour, known to exhibit enhanced infiltrative potential. One of the characteristics of glioblastoma is microvascular proliferation surrounding necrotic areas, as a response to a hypoxic environment, which in turn increases the expression of angiogenic factors and their signalling pathways (RAS/RAF/ERK/MAPK pathway, PI3K/Akt signalling pathway and WTN signalling cascade). Currently, a small number of anti-angiogenic drugs, extending glioblastoma patients survival, are available for clinical use. Most medications are ineffective in clinical therapy of glioblastoma due to acquired malignant cells or intrinsic resistance, angiogenic receptors cross-activation and redundant intracellular signalling, or the inability of the drug to cross the blood-brain barrier and to reach its target in vivo. Researchers have also observed that GB tumours are different in many aspects, even when they derive from the same tissue, which is the reason for personalised therapy. An understanding of the molecular mechanisms regulating glioblastoma angiogenesis and invasion may be important in the future development of curative therapeutic approaches for the treatment of this devastating disease.
Collapse
Affiliation(s)
| | - Stefana Oana Purcaru
- Unit of Biochemistry, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Oana Alexandru
- Department of Neurology, University of Medicine and Pharmacy of Craiova and Clinical Hospital of Neuropsychiatry Craiova, Craiova, Romania
| | - Anica Dricu
- Unit of Biochemistry, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| |
Collapse
|
24
|
Shen J, Hong Y, Zhao Q, Zhang JL. Preclinical evaluation of perifosine as a potential promising anti-rhabdomyosarcoma agent. Tumour Biol 2015; 37:1025-33. [PMID: 26269112 DOI: 10.1007/s13277-015-3740-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/30/2015] [Indexed: 11/24/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is a highly malignant and metastatic pediatric cancer that arises from the skeletal muscle. Recent studies have identified an important role of AKT signaling in RMS progression. In the current study, we investigated the activity of perifosine, an oral alkylphospholipid AKT inhibitor, against human RMS cells (RD and Rh-30 lines) both in vivo and in vitro, and studied the underlying mechanisms. We showed that perifosine significantly inhibited RMS cell growth in concentration- and time-dependent manners. Meanwhile, perifosine induced dramatic apoptosis in RMS cells. At the signaling level, perifosine blocked AKT activation, while inducing reactive oxygen species (ROS) production as well as JNK and P38 phosphorylations in RMS cells. Restoring AKT activation by introducing a constitutively active-AKT (CA-AKT) only alleviated (not abolished) perifosine-induced cytotoxicity in RD cells. Yet, the ROS scavenger N-acetyl cysteine (NAC) as well as pharmacological inhibitors against JNK (SP-600125) or P38 (SB-203580) suppressed perifosine-induced cytotoxicity in RMS cells. Thus, perifosine induces growth inhibition and apoptosis in RMS cells through mechanisms more than just blocking AKT. In vivo, oral administration of perifosine significantly inhibited growth of Rh-30 xenografts in severe combined immunodeficient (SCID) mice. Our data indicate that perifosine might be further investigated as a promising anti-RMS agent.
Collapse
Affiliation(s)
- Jie Shen
- Department of Respiratory Diseases, The First Affiliated Hospital of Zhejiang University, No. 79 Qing-chun Road, Hangzhou, Zhejiang, 310003, People's Republic of China
| | - Yue Hong
- Department of Respiratory Diseases, The First Affiliated Hospital of Zhejiang University, No. 79 Qing-chun Road, Hangzhou, Zhejiang, 310003, People's Republic of China
| | - Qiong Zhao
- Department of Thoracic Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China.
| | - Jian-Li Zhang
- Department of Respiratory Diseases, The First Affiliated Hospital of Zhejiang University, No. 79 Qing-chun Road, Hangzhou, Zhejiang, 310003, People's Republic of China.
| |
Collapse
|
25
|
Kim MN, Ro SW, Kim DY, Kim DY, Cho KJ, Park JH, Lim HY, Han KH. Efficacy of perifosine alone and in combination with sorafenib in an HrasG12V plus shp53 transgenic mouse model of hepatocellular carcinoma. Cancer Chemother Pharmacol 2015; 76:257-67. [PMID: 26037205 DOI: 10.1007/s00280-015-2787-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/21/2015] [Indexed: 12/21/2022]
Abstract
PURPOSE Perifosine has shown antitumor activity via inhibition of Akt phosphorylation in many advanced solid tumors. This study investigated the efficacy of perifosine alone and in combination with sorafenib in a transgenic mouse model of HCC. METHODS The mouse model of HCC was generated by hydrodynamic injection of transposons encoding HrasG12V and short-hairpin RNA downregulating p53. The transgenic mice were treated with perifosine alone and in combination with sorafenib to evaluate efficacy of drugs on tumor growth and survival. RESULTS Treatment with perifosine for 5 weeks, alone and in combination with sorafenib, strongly inhibited tumor growth and increased survival. Perifosine inhibited HCC cell proliferation, induced apoptosis, and decreased tumor angiogenesis. Furthermore, its combination with sorafenib enhanced these effects. In addition, Akt phosphorylation was decreased by perifosine and further decreased by combination treatment. Although perifosine alone did not appear to activate the caspase pathway, combination treatment increased the cleavage of caspase-3, caspase-9, and poly (ADP-ribose) polymerase. CONCLUSIONS The preclinical effect that current study showed represents a strong rationale for clinical trials using perifosine alone and in combination with sorafenib in the treatment of HCC patients.
Collapse
Affiliation(s)
- Mi Na Kim
- Department of Internal Medicine, Yonsei University College of Medicine, 250 Seongsanno, Seodaemun-gu, Seoul, 120-752, Korea
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Amantini C, Morelli MB, Santoni M, Soriani A, Cardinali C, Farfariello V, Eleuteri AM, Bonfili L, Mozzicafreddo M, Nabissi M, Cascinu S, Santoni G. Sorafenib induces cathepsin B-mediated apoptosis of bladder cancer cells by regulating the Akt/PTEN pathway. The Akt inhibitor, perifosine, enhances the sorafenib-induced cytotoxicity against bladder cancer cells. Oncoscience 2015; 2:395-409. [PMID: 26097873 PMCID: PMC4468325 DOI: 10.18632/oncoscience.147] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 03/16/2015] [Indexed: 01/08/2023] Open
Abstract
Sorafenib, a tyrosine kinase inhibitor, has been demonstrated to exert anti-tumor effects. However, the molecular mechanisms underlying its effects on bladder cancer remain unknown. Here, we evaluated the mechanisms responsible for the sorafenib-induced anti-tumor effects on 5637 and T24 bladder cancer cells. We demonstrated that sorafenib reduces cell viability, stimulates lysosome permeabilization and induces apoptosis of bladder cancer cells. These effects are dependent by the activation of cathepsin B released from lysosomes. The sorafenib-increased cathepsin B activity induced the proteolysis of Bid into tBid that stimulates the intrinsic pathway of apoptosis characterized by mitochondrial membrane depolarization, oxygen radical generation and cytochrome c release. Moreover, we found that cathepsin B enzymatic activity, induced by sorafenib, is dependent on its dephosphorylation via PTEN activation and Akt inactivation. Pretreatment with orthovanadate rescued bladder cancer cells from apoptosis. In addition, the Akt inhibitor perifosine increased the sensitivity of bladder cancer cells to sorafenib-induced cytotoxicity. Overall, our results show that apoptotic cell death induced by sorafenib in bladder cancer cells is dependent on cathepsin B activity and involved PTEN and Akt signaling pathways. The Akt inhibitor perifosine increased the cytotoxic effects of sorafenib in bladder cancer cells.
Collapse
Affiliation(s)
- Consuelo Amantini
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Maria Beatrice Morelli
- School of Pharmacy, Experimental Medicine Section, University of Camerino, Camerino, Italy ; Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Matteo Santoni
- Department of Medical Oncology, Polytechnic University of Marche, Ancona, Italy
| | | | - Claudio Cardinali
- School of Pharmacy, Experimental Medicine Section, University of Camerino, Camerino, Italy ; Department of Molecular Medicine, Sapienza University, Rome, Italy
| | | | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Matteo Mozzicafreddo
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Massimo Nabissi
- School of Pharmacy, Experimental Medicine Section, University of Camerino, Camerino, Italy
| | - Stefano Cascinu
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Giorgio Santoni
- School of Pharmacy, Experimental Medicine Section, University of Camerino, Camerino, Italy
| |
Collapse
|
27
|
Zhang J, Hong Y, Shen J. Combination treatment with perifosine and MEK-162 demonstrates synergism against lung cancer cells in vitro and in vivo. Tumour Biol 2015; 36:5699-706. [PMID: 25697899 DOI: 10.1007/s13277-015-3244-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 02/09/2015] [Indexed: 10/24/2022] Open
Abstract
Lung cancer is a global health problem. The search for new therapeutic approaches for the treatment of lung cancer is important. Here, we reported that the AKT inhibitor perifosine and the MEK\ERK inhibitor MEK-162 synergistically induced lung cancer cell (A549 and H460 lines) growth inhibition and apoptosis. The combined efficiency was significantly higher than either agent alone. For the molecular study, perifosine and MEK-162 worked together to concurrently block AKT, mammalian target of rapamycin (mTOR) complex 1 (mTORC1), and MEK-ERK signalings in lung cancer cells, while either agent alone only affected one or two signalings with lower efficiency. In vivo, MEK-162 and perifosine co-administration dramatically inhibited A549 lung cancer xenograft growth, without inducing apparent toxicities. The synergistic activity in vivo was again superior than either agent alone. Thus, perifosine and MEK-162 combination is biologically plausible by acting through effects on different proliferation and survival-related signaling pathways. Our in vitro and in vivo results support the feasibility of investigating the synergism regimen in clinical tests.
Collapse
Affiliation(s)
- Jianli Zhang
- Department of Respiratory Diseases, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | | | | |
Collapse
|
28
|
Mikhail S, Cosgrove D, Zeidan A. Hepatocellular carcinoma: systemic therapies and future perspectives. Expert Rev Anticancer Ther 2014; 14:1205-18. [PMID: 25199765 DOI: 10.1586/14737140.2014.949246] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma is (HCC) the most common primary malignancy of the liver in adults. It is also the fifth most common solid cancer worldwide and the third leading cause of cancer-related deaths. Treatment options for HCC include liver transplantation, surgical resection, locoregional therapies and chemotherapy. The median survival time of patients following the diagnosis of unresectable disease is approximately 6-20 months, whereas the 5-year survival is less than 5%. Given the projected increase in incidence of HCC due to hepatitis C virus infection and obesity related cirrhosis, there is an urgent need for more intensive research in this cancer. In this article, we review the systemic options available for patients with HCC, its molecular pathogenesis and future therapeutic directions with special emphasis on immune-based and molecularly-targeted therapy.
Collapse
Affiliation(s)
- Sameh Mikhail
- Wexner Medical Center, Ohio State University, 320 W.10th Street, Columbus, Ohio, 43210, USA
| | | | | |
Collapse
|
29
|
|
30
|
Liu F, He Y, Liang Y, Wen L, Zhu Y, Wu Y, Zhao L, Li Y, Mao X, Liu H. PI3-kinase inhibition synergistically promoted the anti-tumor effect of lupeol in hepatocellular carcinoma. Cancer Cell Int 2013; 13:108. [PMID: 24176221 PMCID: PMC3833314 DOI: 10.1186/1475-2867-13-108] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 10/28/2013] [Indexed: 01/06/2023] Open
Abstract
Background Lup-20(29)-en-3H-ol (Lupeol), a dietary triterpene, has been shown to possess multiple pharmacological activities including anti-tumor effects Methods In the current study, we noted that low doses of lupeol (<40 μM) promoted the growth of hepatocellular carcinoma (HCC) cells with a significant activation of the PI3-kinase/Akt signaling pathway. We further investigated the combined anti-tumor effect of lupeol and S14161, a newly identified PI3-Kinase inhibitor in vitro and in vivo Results The results demonstrated that lupeol and S14161 could exert a synergistic antitumor effect resulting in chemo-sensitization of HCC to low doses of lupeol. Using an in vivo HCC model, we further demonstrated that lupeol and S14161 synergistically inhibited tumor growth without any adverse effects on body weight Conclusion Our studies showed that the activation of PI3-kinase/Akt pathway resulted in the tumor-promoting effect with low doses of lupeol. Combining PI3-kinase inhibitor with lupeol could synergistically augment the anti-tumor effect of lupeol and might be an applicable strategy for HCC therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Haiyan Liu
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, P, R, China.
| |
Collapse
|
31
|
Liang SR, Hu GR, Fang LJ, Huang SJ, Li JS, Zhao MY, Meng MJ. CpG oligodeoxynucleotides enhance chemosensitivity of 5-fluorouracil in HepG2 human hepatoma cells via downregulation of the antiapoptotic factors survivin and livin. Cancer Cell Int 2013; 13:106. [PMID: 24161202 PMCID: PMC4176997 DOI: 10.1186/1475-2867-13-106] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 10/23/2013] [Indexed: 12/21/2022] Open
Abstract
Background Recent studies indicated that a synthetic oligonucleotide containing un-methylated CpG oligodeoxynucleotides (CpG-ODN) has a potential function for cancer therapy. In this study, we evaluated the chemosensitizing effects of CpG-ODN in 5-fluorouracil (5-FU)-treated HepG2 human hepatoma cells. Methods Cell viability assay were utilized to evaluate the direct cytotoxicity of CpG-ODN in the presence or absence of 5-FU in HepG2 cells, and apoptosis as well as cell-cycle was examined by flow cytometry analysis. The mRNA expression of Bcl-2, Livin and Survivin within HepG2 cells treated with CpG-ODN and/or 5-FU were analyzed by Real Time PCR assay in vitro. Results CpG-ODN in combination with 5-FU could decrease cell viability, increase apoptosis and further induce HepG2 cells cycle arrest at S phase when compared with CpG-ODN or 5-FU. CpG-ODN or 5-FU could down-regulate the mRNA expression of Bcl-2 within HepG2 cells. The mRNA expression of Livin and Survivin decreased in cells treated with CpG-ODN alone but increased in cells treated with 5-FU alone. However, CpG-ODN in combination with 5-FU could down-regulate the mRNA expression of Livin and Survivin within HepG2 cells. Conclusions Our finding demonstrated that CpG-ODN enhanced the chemosentivity of 5-FU in HepG2 human hepatoma cells at least in part by down-regulating the expression of Livin and Survivin, leading to apoptosis and further inducing cell cycle arrest at S phase. Therefore, CpG-ODN may be a potential candidate as chemosensitizer for human hepatocellular carcinoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Min-Jie Meng
- School of Life Science and Biopharmaceutical, Guangdong Pharmaceutical University, Guangzhou 510006, P, R, China.
| |
Collapse
|
32
|
Krawczyk J, Keane N, Swords R, O'Dwyer M, Freeman CL, Giles FJ. Perifosine--a new option in treatment of acute myeloid leukemia? Expert Opin Investig Drugs 2013; 22:1315-27. [PMID: 23931614 DOI: 10.1517/13543784.2013.826648] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Perifosine is a novel targeted oral Akt inhibitor. In preclinical leukemia models, perifosine has an independent cytotoxic potential but also synergizes well with other rationally selected targeted agents. The evidence from clinical trials supporting the use of perifosine in the therapy of leukemias is limited. The optimal dose and schedule have yet to be defined. However, given its favorable toxicity profile and mechanism of action, the therapeutic potential of perifosine should be evaluated in well-designed clinical trials. AREAS COVERED The role of the phosphatidylinositol-3 kinase (PI3K)/Akt zpathway in normal cells, cancer and leukemias is discussed. The mechanism of action of perifosine and the basic information on the development and chemical properties are summarized. The evidence from in vivo and in vitro studies is presented. The efficacy and side effect profile are summarized. EXPERT OPINION The safety and tolerability profile of perifosine are satisfactory. The evidence from clinical trials in patients with leukemias is very limited. The preclinical data are encouraging. Perifosine has the potential to play a role in the treatment of leukemias in the future. Its role needs to be confirmed in clinical trials.
Collapse
|
33
|
Shen J, Liang L, Wang C. Perifosine inhibits lipopolysaccharide (LPS)-induced tumor necrosis factor (TNF)-α production via regulation multiple signaling pathways: new implication for Kawasaki disease (KD) treatment. Biochem Biophys Res Commun 2013; 437:250-5. [PMID: 23806687 DOI: 10.1016/j.bbrc.2013.06.055] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 06/14/2013] [Indexed: 11/16/2022]
Abstract
Kawasaki disease (KD) is a multisystem vasculitis of unknown etiology, with coronary artery aneurysms occurring in majority of untreated cases. Tumor necrosis factor (TNF)-α is the pleiotropic inflammatory cytokine elevated during the acute phase of KD, which induces damage to vascular endothelial cells to cause systemic vasculitis. We here investigated the potential role of perifosine, a novel Akt inhibitor, on TNFα expression in LPS-stimulated macrophages and in ex-vivo cultured peripheral blood mononuclear cells (PBMCs) of acute KD patients. Here, we found that perifosine inhibited LPS-induced TNFα expression and production in mouse macrophages (RAW 264.7 cells and bone marrow-derived macrophages (BMDMs)). Meanwhile, perifosine administration down-regulated TNFα production in PBMCs isolated from acute KD patients. For the mechanism study, we found that perifosine significantly inhibited Akt and ERK/mitogen-activated protein kinases (MAPK) signaling, while activating AMP-activated protein kinase (AMPK) signaling in both patients' PBMCs and LPS-stimulated macrophages. Interestingly, although perifosine is generally known as an Akt inhibitor, our data suggested that ERK inhibition and AMPK activation, but not Akt inactivation were possibly involved in perifosine-mediated inhibition against TNFα production in monocytes. In conclusion, our data suggested that perifosine significantly inhibited TNFα production via regulation multiple signaling pathways. The results of this study should have significant translational relevance in managing this devastating disease.
Collapse
Affiliation(s)
- Jie Shen
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang University School of Medicine, China
| | | | | |
Collapse
|
34
|
Jin YL, Wang ZQ, Qu H, Wang HX, Ibrahim M, Zhang J, Huang YH, Wu J, Bai LL, Wang XY, Meng JY, Tang JW. Annexin A7 gene is an important factor in the lymphatic metastasis of tumors. Biomed Pharmacother 2013; 67:251-9. [DOI: 10.1016/j.biopha.2012.11.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 11/23/2012] [Indexed: 12/16/2022] Open
|
35
|
Gomide A, Thomé C, dos Santos G, Ferreira G, Faça V, Rego E, Greene L, Stabeli R, Ciancaglini P, Itri R. Disrupting membrane raft domains by alkylphospholipids. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:1384-9. [DOI: 10.1016/j.bbamem.2013.01.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 01/17/2013] [Accepted: 01/22/2013] [Indexed: 11/28/2022]
|
36
|
Yao C, Wei JJ, Wang ZY, Ding HM, Li D, Yan SC, Yang YJ, Gu ZP. Perifosine induces cell apoptosis in human osteosarcoma cells: new implication for osteosarcoma therapy? Cell Biochem Biophys 2013; 65:217-27. [PMID: 23015227 DOI: 10.1007/s12013-012-9423-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Despite the advances of adjuvant chemotherapy and significant improvement of survival, the prognosis for patients with osteosarcoma is generally poor. The search for more effective anti-osteosarcoma agents is necessary and urgent. Here we report that perifosine induces cell apoptosis and growth inhibition in cultured human osteosarcoma cells. Perifosine blocks Akt/mTOR complex 1 (mTORC1) signaling, while promoting caspase-3, c-Jun N-terminal kinases (JNK), and p53 activation. Further, perifosine inhibits survivin expression probably by disrupting its association with heat shock protein-90 (HSP-90). These signaling changes together were responsible for a marked increase of osteosarcoma cell apoptosis and growth inhibition. Finally, we found that a low dose of perifosine enhanced etoposide- or doxorubicin-induced anti-OS cells activity. The results together suggest that perifosine might be used as a novel and effective anti-osteosarcoma agent.
Collapse
Affiliation(s)
- Chen Yao
- Department of Orthopedics, BenQ Medical Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Pal I, Mandal M. PI3K and Akt as molecular targets for cancer therapy: current clinical outcomes. Acta Pharmacol Sin 2012; 33:1441-58. [PMID: 22983389 DOI: 10.1038/aps.2012.72] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The PI3K-Akt pathway is a vital regulator of cell proliferation and survival. Alterations in the PIK3CA gene that lead to enhanced PI3K kinase activity have been reported in many human cancer types, including cancers of the colon, breast, brain, liver, stomach and lung. Deregulation of PI3K causes aberrant Akt activity. Therefore targeting this pathway could have implications for cancer treatment. The first generation PI3K-Akt inhibitors were proven to be highly effective with a low IC(50), but later, they were shown to have toxic side effects and poor pharmacological properties and selectivity. Thus, these inhibitors were only effective in preclinical models. However, derivatives of these first generation inhibitors are much more selective and are quite effective in targeting the PI3K-Akt pathway, either alone or in combination. These second-generation inhibitors are essentially a specific chemical moiety that helps to form a strong hydrogen bond interaction with the PI3K/Akt molecule. The goal of this review is to delineate the current efforts that have been undertaken to inhibit the various components of the PI3K and Akt pathway in different types of cancer both in vitro and in vivo. Our focus here is on these novel therapies and their inhibitory effects that depend upon their chemical nature, as well as their development towards clinical trials.
Collapse
|
38
|
Thomé CH, dos Santos GA, Ferreira GA, Scheucher PS, Izumi C, Leopoldino AM, Simão AM, Ciancaglini P, de Oliveira KT, Chin A, Hanash SM, Falcão RP, Rego EM, Greene LJ, Faça VM. Linker for activation of T-cell family member2 (LAT2) a lipid raft adaptor protein for AKT signaling, is an early mediator of alkylphospholipid anti-leukemic activity. Mol Cell Proteomics 2012; 11:1898-912. [PMID: 23001822 DOI: 10.1074/mcp.m112.019661] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lipid rafts are highly ordered membrane domains rich in cholesterol and sphingolipids that provide a scaffold for signal transduction proteins; altered raft structure has also been implicated in cancer progression. We have shown that 25 μm 10-(octyloxy) decyl-2-(trimethylammonium) ethyl phosphate (ODPC), an alkylphospholipid, targets high cholesterol domains in model membranes and induces apoptosis in leukemia cells but spares normal hematopoietic and epithelial cells under the same conditions. We performed a quantitative (SILAC) proteomic screening of ODPC targets in a lipid-raft-enriched fraction of leukemic cells to identify early events prior to the initiation of apoptosis. Six proteins, three with demonstrated palmitoylation sites, were reduced in abundance. One, the linker for activation of T-cell family member 2 (LAT2), is an adaptor protein associated with lipid rafts in its palmitoylated form and is specifically expressed in B lymphocytes and myeloid cells. Interestingly, LAT2 is not expressed in K562, a cell line more resistant to ODPC-induced apoptosis. There was an early loss of LAT2 in the lipid-raft-enriched fraction of NB4 cells within 3 h following treatment with 25 μm ODPC. Subsequent degradation of LAT2 by proteasomes was observed. Twenty-five μm ODPC inhibited AKT activation via myeloid growth factors, and LAT2 knockdown in NB4 cells by shRNA reproduced this effect. LAT2 knockdown in NB4 cells also decreased cell proliferation and increased cell sensitivity to ODPC (7.5×), perifosine (3×), and arsenic trioxide (8.5×). Taken together, these data indicate that LAT2 is an early mediator of the anti-leukemic activity of alkylphospholipids and arsenic trioxide. Thus, LAT2 may be used as a target for the design of drugs for cancer therapy.
Collapse
Affiliation(s)
- Carolina H Thomé
- Instituto Nacional de Ciência e Tecnologia em Células-Tronco e Terapia Celular, Fundação Hemocentro de Ribeirão Preto, 14051-140, Ribeirão Preto, SP, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sun W, Modak S. Emerging treatment options for the treatment of neuroblastoma: potential role of perifosine. Onco Targets Ther 2012; 5:21-9. [PMID: 22419878 PMCID: PMC3299554 DOI: 10.2147/ott.s14578] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Achieving a cure for high-risk neuroblastoma, the most common extracranial solid tumor in children, remains a formidable task despite the recent addition of antibody-mediated anti-GD2 immunotherapy to established multimodality therapy. The PI3K/Akt pathway is a pivotal signaling pathway utilized by a plethora of receptor tyrosine kinases that contribute to the aggressive phenotype of high-risk neuroblastoma. Akt is aberrantly activated in high-risk neuroblastoma and is therefore an attractive therapeutic target. Perifosine is the best-characterized Akt inhibitor in preclinical studies and in clinical trials in adults, although safety in children is not yet confirmed. It is a synthetic third-generation alkylphospholipid with good oral bioavailability and modest side effects. Perifosine targets the lipid-binding PH domain of Akt and inhibits the translocation of Akt to the cell membrane, an essential step for Akt activation. It decreases Akt phosphorylation and increases caspase-dependent apoptosis in neuroblastoma cell lines, inhibits growth of neuroblastoma xenografts, and overcomes RTK/ligand-mediated chemoresistance. It is currently being studied in two Phase I clinical trials in children with recurrent or refractory solid tumors including neuroblastoma. In the single agent trial (ClinicalTrials.gov identifier NCT00776867), maximum tolerated dose has not yet been reached and pharmacokinetic data has been accrued. In the second study (ClinicalTrials.gov identifier NCT01049841), patients are treated with a combination of perifosine and the mTOR-inhibitor temsirolimus based on preclinical data showing synergy of the two agents, and the premise that direct Akt inhibition may overcome Akt activation secondary to mTOR inhibition. Based on results from adult trials, it is unlikely that perifosine alone will produce dramatic therapeutic effects against high-risk neuroblastoma. However, given the recent encouraging early-phase combination therapy results in adults with multiple myeloma and colorectal carcinoma, rational perifosine-containing combination regimens hold promise for neuroblastoma therapy. These will be explored after safety in children is established in Phase I studies.
Collapse
Affiliation(s)
- Weili Sun
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | | |
Collapse
|
40
|
PI3K/AKT/PTEN Signaling as a Molecular Target in Leukemia Angiogenesis. Adv Hematol 2012; 2012:843085. [PMID: 22505939 PMCID: PMC3299269 DOI: 10.1155/2012/843085] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 12/11/2011] [Indexed: 11/17/2022] Open
Abstract
PI3K/AKT/PTEN pathway is important in the regulation of angiogenesis mediated by vascular endothelial growth factor in many tumors including leukemia. The signaling pathway is activated in leukemia patients as well as leukemia cell lines together with a decrease in the expression of PTEN gene. The mechanism by which the signaling pathway regulates angiogenesis remains to be further elucidated. However, it has become an attractive target for drug therapy against leukemia, because angiogenesis is a key process in malignant cell growth. In this paper, we will focus on the roles and mechanisms of PI3K/AKT/PTEN pathway in regulating angiogenesis.
Collapse
|
41
|
A triterpenoid from Thalictrum fortunei induces apoptosis in BEL-7402 cells through the P53-induced apoptosis pathway. Molecules 2011; 16:9505-19. [PMID: 22086402 PMCID: PMC6264704 DOI: 10.3390/molecules16119505] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 11/04/2011] [Accepted: 11/08/2011] [Indexed: 11/16/2022] Open
Abstract
Thalictrum fortunei S. Moore, a perennial plant distributed in the southeastern part of China, has been used in Traditional Chinese Medicine for thousands of years for its antitumor, antibacterial and immunoregulatory effects. In order to investigate the active components and the mechanism of the anti-tumor effects of Thalictrum fortunei, the growth inhibitory effects of eight triterpenoids isolated from the aerial parts of the plant on tumor cell lines were examined by 3-(4,5)-dimethylthiazoy1-3,5-diphenyltetrazolium bromide (MTT) assay. The MTT-assay results showed that the inhibitory activity of 3-O-β-d-glucopyranosyl-(1→4)-β-d-fucopyranosyl(22S,24Z)-cycloart-24-en-3β,22,26-triol 26-O-β-d-glucopyranoside (1) was stronger than that of the other seven tested triterpenoids on human hepatoma Bel-7402 cell line (Bel-7402), human colon lovo cells (LoVo), human non-small cells lung cancer NCIH-460 cells (NCIH-460) and human gastric carcinoma SGC-7901 cells (SGC-7901) after 48 h treatment in vitro, with the IC50 values of 66.4, 84.8, 73.5, 89.6 μM, respectively. Moreover, the antitumor mechanism of compound 1 on Bel-7402 cell was explored through nucleus dyeing, fluorescence assay, flow cytometry and western blot. The flow cytometric analysis results revealed that compound 1 caused apoptosis and mitochondrial membrane potential (MMP) loss in Bel-7402 cells. A fluorescence assay indicated that intracellular reactive oxygen species (ROS) were markedly provoked by compound 1 treatment compared to control cells. Immunoblot results showed that compound 1 significantly increased the expression levels of cleaved caspase-3, P53 and Bax protein, and decreased the expression level of Bcl-2 protein. These findings indicate that compound 1 inhibits the growth activity of tumor cells, probably through the P53 protein-induced apoptosis pathway.
Collapse
|
42
|
Perifosine as potential anti-cancer agent inhibits proliferation, migration, and tube formation of human umbilical vein endothelial cells. Mol Cell Biochem 2011; 368:1-8. [PMID: 21769450 DOI: 10.1007/s11010-011-0986-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 07/06/2011] [Indexed: 10/18/2022]
Abstract
Targeting angiogenesis is considered an effective strategy for treating the expansion and metastasis of tumors. The aim of this study is to assess the effects of perifosine, an inhibitor of Akt, on cell proliferation, apoptosis, angiogenesis, and VEGF-induced cell migration in cultured human umbilical vein endothelial cells (HUVECs) in vitro. MTT and cell cycle analysis results indicated that perifosine inhibited the growth of HUVECs in a dose-dependent manner, arrested cell cycle progression at the G(2) phase with regulation the expression of p21 and cyclinB1. Apoptosis induced by the higher concentrations of perifosine in HUVECs was also observed. In addition, tube formation of HUVECs and VEGF-induced cell migration were markedly inhibited by perifosine. Western blotting analysis of cell signaling molecules indicated that perifosine inhibited ERK and p38 phosphorylation in HUVECs. These results suggest that perifosine exerts anti-angiogenic activity in HUVECs and is a promising agent for treatment of angiogenesis related-diseases.
Collapse
|
43
|
Krajarng A, Nakamura Y, Suksamrarn S, Watanapokasin R. α-Mangostin induces apoptosis in human chondrosarcoma cells through downregulation of ERK/JNK and Akt signaling pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:5746-5754. [PMID: 21446759 DOI: 10.1021/jf200620n] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Chondrosarcoma is a malignant primary bone tumor that is resistant to chemotherapy and radiation therapy. α-Mangostin, a component of Garcinia mangostana Linn, is a xanthone derivative shown to have antioxidant and antitumor properties. This study is the first to investigate anticancer effects of α-mangostin in the human chondrosarcoma cell line SW1353. We showed that α-mangostin inhibited cell proliferation of SW1353 cells in a time- and dose-dependent manner by using the trypan blue exclusion method. Hoechst 33342 nuclear staining and nucleosomal DNA-gel electrophoresis revealed that α-mangostin could induce nuclear condensation and fragmentation, typically seen in apoptosis. Flow cytometry using Annexin V/PI double staining assessed apoptosis, necrosis and viability. α-Mangostin activated caspase-3, -8, -9 expression, decreased Bcl-2 and increased Bax. This promotes mitochondrial dysfunction, leading to the release of cytochrome c from the mitochondria to the cytoplasm. In addition, total and phosphorylated ERK and JNK were downregulated in α-mangostin-treated SW1353 cells but no changes in p38. α-Mangostin also decreased phosphorylated Akt without altering total Akt. These results suggest that α-mangostin inhinbited cell proliferation and induced apoptosis through downregulation of ERK, JNK and Akt signaling pathway in human chondrosarcoma SW1353 cells.
Collapse
Affiliation(s)
- Aungkana Krajarng
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
| | | | | | | |
Collapse
|