1
|
Wei J, Zhao X, Long F, Tian K, Wu L. Lianhua Qingwen exerts anti-liver cancer effects and synergistic efficacy with sorafenib through PI3K/AKT pathway: Integrating network pharmacology, molecular docking, and experimental validation. Gene 2024; 912:148383. [PMID: 38493972 DOI: 10.1016/j.gene.2024.148383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Liver cancer is one of the most lethal malignancies and sorafenib resistance is the main treatment obstacle for patients with advanced liver cancer. Developing drugs that sensitize liver cancer patients to sorafenib is of great importance. Lianhua Qingwen (LHQW), a sort of Traditional Chinese Medicine (TCM) approved by the Chinese Food and Drug Administration (CFDA), is reported to exert synergistic effects with oseltamivir against Influenza virus. However, whether LHQW could exhibit anti-liver cancer effects and enhance the efficacy of sorafenib against liver cancer have not been reported. In the present study, the potential anti-liver cancer effects of LHQW and its synergistic effects with sorafenib were investigated via applying network pharmacology, molecular docking, and in vitro experiments. An "ingredient-compound- target-liver cancer" network was constructed which included 12 ingredients, 164 compounds, and 402 targets. AKT1 was identified as the most hub gene and the PI3K/AKT pathway was revealed as the most enriched pathway. Subsequently, the molecular docking results showed that kaempferol, luteolin, and quercetin were screened as the top 3 compounds which showed the tightest binding to AKT1. Further, the in vitro experiments verified that LHQW significantly inhibited liver cancer cell proliferation and induced apoptosis. Western blot assays confirmed that LHQW could attenuate the PI3K/AKT pathway. Interestingly, LHQW showed a synergistic effect with sorafenib against liver cancer via reducing cell viability, inducing apoptosis, and down- regulating PI3K/AKT pathway. This study broadens the potential application of LHQW and provides insights for liver cancer treatment.
Collapse
Affiliation(s)
- Jinrui Wei
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530200, Guangxi, China
| | - Xuqi Zhao
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, 7 Guangxi 530004, China
| | - Fuli Long
- Department of Hepatology, the First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530200, Guangxi, China
| | - Kunpeng Tian
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, 7 Guangxi 530004, China; Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha 410007, China.
| | - Lichuan Wu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, 7 Guangxi 530004, China.
| |
Collapse
|
2
|
Zhang F, Wang B, Zhang W, Xu Y, Zhang C, Xue X. Transcription Factor MAZ Potentiates the Upregulated NEIL3-mediated Aerobic Glycolysis, thereby Promoting Angiogenesis in Hepatocellular Carcinoma. Curr Cancer Drug Targets 2024; 24:1235-1249. [PMID: 38347781 DOI: 10.2174/0115680096265896231226062212] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 11/06/2023] [Accepted: 11/17/2023] [Indexed: 09/25/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is characterized by high vascularity and notable abnormality of blood vessels, where angiogenesis is a key process in tumorigenesis and metastasis. The main functions of Nei Like DNA Glycosylase 3 (NEIL3) include DNA alcoholization repair, immune response regulation, nervous system development and function, and DNA damage signal transduction. However, the underlying mechanism of high expression NEIL3 in the development and progression of HCC and whether the absence or silencing of NEIL3 inhibits the development of cancer remain unclear. Therefore, a deeper understanding of the mechanisms by which increased NEIL3 expression promotes cancer development is needed. METHODS Expression of NEIL3 and its upstream transcription factor MAZ in HCC tumor tissues was analyzed in bioinformatics efforts, while validation was done by qRT-PCR and western blot in HCC cell lines. The migration and tube formation capacity of HUVEC cells were analyzed by Transwell and tube formation assays. Glycolytic capacity was analyzed by extracellular acidification rate, glucose uptake, and lactate production levels. Chromatin immunoprecipitation (ChIP) and dual-luciferase reporter gene assays were utilized to investigate specific interactions between MAZ and NEIL3. RESULTS NEIL3 and MAZ were substantially upregulated in HCC tissues and cells. NEIL3 was involved in modulating the glycolysis pathway, suppression of which reversed the stimulative impact of NEIL3 overexpression on migration and angiogenesis in HUVEC cells. MAZ bound to the promoter of NEIL3 to facilitate NEIL3 transcription. Silencing MAZ reduced NEIL3 expression and suppressed the glycolysis pathway, HUVEC cell migration, and angiogenesis. CONCLUSION MAZ potentiated the upregulated NEIL3-mediated glycolysis pathway and HCC angiogenesis. This study provided a rationale for the MAZ/NEIL3/glycolysis pathway as a possible option for anti-angiogenesis therapy in HCC.
Collapse
Affiliation(s)
- Fabiao Zhang
- Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Binfeng Wang
- Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Wenlong Zhang
- Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Yongfu Xu
- Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Caiming Zhang
- Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Xiangyang Xue
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| |
Collapse
|
3
|
Xu FQ, Dong MM, Wang ZF, Cao LD. Metabolic rearrangements and intratumoral heterogeneity for immune response in hepatocellular carcinoma. Front Immunol 2023; 14:1083069. [PMID: 36776894 PMCID: PMC9908004 DOI: 10.3389/fimmu.2023.1083069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Liver cancer is one of the most common malignant tumors globally. Not only is it difficult to diagnose, but treatments are scarce and the prognosis is generally poor. Hepatocellular carcinoma (HCC) is the most common type of liver cancer. Aggressive cancer cells, such as those found in HCC, undergo extensive metabolic rewiring as tumorigenesis, the unique feature, ultimately causes adaptation to the neoplastic microenvironment. Intratumoral heterogeneity (ITH) is defined as the presence of distinct genetic features and different phenotypes in the same tumoral region. ITH, a property unique to malignant cancers, results in differences in many different features of tumors, including, but not limited to, tumor growth and resistance to chemotherapy, which in turn is partly responsible for metabolic reprogramming. Moreover, the different metabolic phenotypes might also activate the immune response to varying degrees and help tumor cells escape detection by the immune system. In this review, we summarize the reprogramming of glucose metabolism and tumoral heterogeneity and their associations that occur in HCC, to obtain a better understanding of the mechanisms of HCC oncogenesis.
Collapse
Affiliation(s)
- Fei-Qi Xu
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.,The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meng-Meng Dong
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Zhi-Fei Wang
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Li-Dong Cao
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Chen X, Wang Y, Liu H, Zhang J, Wang J, Jin X, Ma Y. CSP I-plus modified rEndostatin inhibits hepatocellular carcinoma metastasis via down-regulation of VEGFA and integrinβ1. BMC Cancer 2022; 22:1200. [PMID: 36419008 PMCID: PMC9682839 DOI: 10.1186/s12885-022-10318-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/15/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND In our previous study, N end of the Circumsporozoite protein (CSP I-plus) modified recombinant human Endostatin (rEndostatin, endostar) (rES-CSP) was constructed, which had antiangiogenic capability and bound to hepatocellular carcinoma in vivo and in vitro. In this study, the inhibition of rES-CSP on hepatocellular carcinoma metastasis was verified in vivo and in vitro, and its possible mechanism was explored. METHODS Firstly, the impact of rES-CSP on the migration, adhesion of hepatoma cell HCCLM3 was identified by wound healing, transwell, and on metastasis of orthotopic xenograft model was identified in nude mouse. Then the expression of metastasis-associated molecules (MMP2, E-cadherin, integrinβ1) and angiogenesis-related factors (VEGFA) in vitro and in vivo were detected by real-time PCR, western blotting, immunohistochemistry. RESULTS Finally, we found that rES-CSP could inhibit the migration and invasion of HCCLM3, and decrease tumor metastasis and growth in nude mouse orthotopic xenograft models. The tumor inhibiting rates of rES-CSP and Endostar were 42.46 ± 5.39% and 11.1 ± 1.88%. The lung metastasis rates of the control, Endostar and rES-CSP were 71, 50, and 42.8%, respectively. Compared with Endostar, rES-CSP significantly down-regulated the expression of VEGFA and integrinβ1. Heparin, a competitive inhibitor of CSP I-plus, which can be bind to the highly-sulfated heparan sulfate proteoglycans (HSPGs) over-expressed in liver and hepatocellular carcinoma, alleviated the down-regulation of VEGFA and integrinβ1. CONCLUSIONS These indicate that rES-CSP may play a role in inhibiting tumor growth and metastasis by down-regulating the angiogenic factor VEGF and the metastasis-related molecules or by interfering with HSPGs-mediated tumor metastasis.
Collapse
Affiliation(s)
- Xueqin Chen
- grid.411847.f0000 0004 1804 4300Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Higher Education Mega Center, Guangzhou, 510006 China
| | - Yan Wang
- grid.411847.f0000 0004 1804 4300Zhongshan Campus Laboratory Center, Guangdong Pharmaceutical University, Guangzhou, 510006 China
| | - Hancong Liu
- grid.411847.f0000 0004 1804 4300Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Higher Education Mega Center, Guangzhou, 510006 China
| | - Jingjing Zhang
- grid.411847.f0000 0004 1804 4300Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Higher Education Mega Center, Guangzhou, 510006 China
| | - Jie Wang
- grid.411847.f0000 0004 1804 4300Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Higher Education Mega Center, Guangzhou, 510006 China
| | - Xiaobao Jin
- grid.411847.f0000 0004 1804 4300Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Higher Education Mega Center, Guangzhou, 510006 China
| | - Yan Ma
- grid.411847.f0000 0004 1804 4300Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Higher Education Mega Center, Guangzhou, 510006 China
| |
Collapse
|
5
|
Bhosale PB, Abusaliya A, Kim HH, Ha SE, Park MY, Jeong SH, Vetrivel P, Heo JD, Kim JA, Won CK, Kim HW, Kim GS. Apigetrin Promotes TNFα-Induced Apoptosis, Necroptosis, G2/M Phase Cell Cycle Arrest, and ROS Generation through Inhibition of NF-κB Pathway in Hep3B Liver Cancer Cells. Cells 2022; 11:cells11172734. [PMID: 36078142 PMCID: PMC9454891 DOI: 10.3390/cells11172734] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/26/2022] [Accepted: 08/31/2022] [Indexed: 11/25/2022] Open
Abstract
Apigetrin (7-(β-D-glucopyranosyloxy)-4′,5-dihydroxyflavone), a glycoside bioactive dietary flavonoid derived from Taraxacum officinale and Teucrium gnaphalodes, is known to possess anticancer, antioxidant, and anti-inflammatory effects on numerous cancers. In the present study, we examined the effect of apigetrin in Hep3B hepatocellular cancer cell line (HCC). Apigetrin inhibited cell growth and proliferation of Hep3B cells, as confirmed by MTT and colony formation assay. We used apigetrin at concentrations of 0, 50, and 100 µM for later experiments. Of these concentrations, 100 µM of apigetrin showed a significant effect on cell inhibition. In apigetrin-treated Hep3B cells, cell cycle arrest occurred at the G2/M phase. Apoptosis and necroptosis of Hep3B cells treated with apigetrin were confirmed by Annexin V/propidium iodide (PI) staining and flow cytometry results. Morphological observation through 4′,6-diamidino-2-phenylindole (DAPI) staining showed intense blue fluorescence representing chromatin condensation. Hematoxylin staining showed necroptotic features such as formation of vacuoles and swelling of organelles. Apigetrin increased reactive oxygen species (ROS) levels in cells, based on fluorescence imaging. Furthermore, the underlying mechanism involved in the apoptosis and necroptosis was elucidated through western blotting. Apigetrin up-regulated TNFα, but down-regulated phosphorylation of p-p65, and IκB. Apigetrin inhibited the expression of Bcl-xl but increased Bax levels. Up-regulation of cleaved PARP and cleaved caspase 3 confirmed the induction of apoptosis in apigetrin-treated Hep3B cells. Additionally, necroptosis markers RIP3, p-RIP3, and p-MLKL were significantly elevated by apigetrin dose-dependently, suggesting necroptotic cell death. Taken together, our findings strongly imply that apigetrin can induce apoptosis and necroptosis of Hep3B hepatocellular cancer cells. Thus, apigetrin as a natural compound might have potential for treating liver cancer.
Collapse
Affiliation(s)
- Pritam Bhagwan Bhosale
- Department of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
| | - Abuyaseer Abusaliya
- Department of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
| | - Hun Hwan Kim
- Department of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
| | - Sang Eun Ha
- Department of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
- Biological Resources Research Group, Bioenvironmental Science & Toxicology Division, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju 52834, Korea
| | - Min Yeong Park
- Department of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
| | - Se Hyo Jeong
- Department of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
| | - Preethi Vetrivel
- Department of Pharmacy, National University of Singapore, Singapore 117643, Singapore
| | - Jeong Doo Heo
- Department of Pharmacy, National University of Singapore, Singapore 117643, Singapore
| | - Jin-A Kim
- Department of Physical Therapy, International University of Korea, Jinju 52833, Korea
| | - Chung kil Won
- Department of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
| | - Hyun-Wook Kim
- Division of Animal Bioscience & Intergrated Biotechnology, Jinju 52725, Korea
| | - Gon Sup Kim
- Department of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea
- Correspondence: ; Tel.: +82-55-772-2346
| |
Collapse
|
6
|
MYBL1 induces transcriptional activation of ANGPT2 to promote tumor angiogenesis and confer sorafenib resistance in human hepatocellular carcinoma. Cell Death Dis 2022; 13:727. [PMID: 35987690 PMCID: PMC9392790 DOI: 10.1038/s41419-022-05180-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 02/06/2023]
Abstract
Angiogenesis is considered as an important process in tumor growth, metastasis of hepatocellular carcinoma (HCC) and associated with cancer progression, suggesting that an important research and development field of clinical molecular targeted drugs for HCC. However, the molecular mechanisms underlying tumor angiogenesis in HCC remains elusive. In the current study, we demonstrate that upregulation of AMYB proto-oncogene-like 1 (MYBL1) was associated with high endothelial vessel (EV) density and contributed to poor prognosis of HCC patient. Functionally, MYBL1 overexpressing enhanced the capacity of HCC cells to induce tube formation, migration of HUVECs, neovascularization in CAMs, finally, enhanced HCC cells metastasis, while silencing MYBL1 had the converse effect. Furthermore, HCC cells with high MYBL1 expression were more resistance to sorafenib treatment. We observed that CD31 staining was significantly increased in tumors formed by MYBL1-overexpressing cells but decreased in MYBL1-silenced tumors. Mechanistically, MYBL1 binds to the ANGPT2 promoter and transcriptionally upregulate ANGPT2 mRNA expression. Strikingly, treatment with monoclonal antibody against ANGPT2 significantly inhibited the growth of MYBL1-overexpressing tumors and efficiently impaired angiogenesis. Furthermore, the histone post-translational factors: protein arginine methyltransferase 5 (PRMT5), MEP50, and WDR5 were required for MYBL1-mediated ANGPT2 upregulation. Importantly, we confirmed the correlation between MYBL1 and ANGPT2 expression in a large cohort of clinical HCC samples and several published datasets in pancreatic cancer, esophageal carcinoma, stomach adenocarcinoma, and colon cancer. Our results demonstrate that MYBL1 upregulated the ANGPT2 expression, then induced angiogenesis and confer sorafenib resistance to HCC cells, and MYBL1 may represent a novel prognostic biomarker and therapeutic target for patients with HCC.
Collapse
|
7
|
Zhao M, Wang Y, Zhang Y, Li X, Mi J, Wang Q, Geng Z, Zuo L, Song X, Ge S, Zhang Z, Tang M, Li H, Wang Z, Jiang C, Su F. The upregulation of stromal antigen 3 expression suppresses the phenotypic hallmarks of hepatocellular carcinoma through the Smad3-CDK4/CDK6-cyclin D1 and CXCR4/RhoA pathways. BMC Gastroenterol 2022; 22:378. [PMID: 35941537 PMCID: PMC9361574 DOI: 10.1186/s12876-022-02400-z] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 06/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The stromal antigen 3 (STAG3) gene encodes an adhesion complex subunit that can regulate sister chromatid cohesion during cell division. Chromosome instability caused by STAG3 gene mutation may potentially promote tumor progression, but the effect of STAG3 on hepatocellular carcinoma (HCC) and the related molecular mechanism are not reported in the literature. The mechanism of the occurrence and development of HCC is not adequately understood. Therefore, the biological role of STAG3 in HCC remains to be studied, and whether STAG3 might be a sensitive therapeutic target in HCC remains to be determined. METHODS The expression and clinical significance of STAG3 in HCC tissues and cell lines were determined by RT-qPCR and immunohistochemistry analyses. The biological functions of STAG3 in HCC were determined through in vitro and in vivo cell function tests. The molecular mechanism of STAG3 in HCC cells was then investigated by western blot assay. RESULTS The mRNA expression of STAG3 was lower in most HCC cells than in normal cells. Subsequently, an immunohistochemical analysis of STAG3 was performed with 126 samples, and lower STAG3 expression was associated with worse overall survival in HCC patients. Moreover, cytofunctional tests revealed that the lentivirus-mediated overexpression of STAG3 in HCC cells inhibited cell proliferation, migration, and invasion; promoted apoptosis; induced G1/S phase arrest in vitro; and inhibited tumor growth in vivo. Furthermore, studies of the molecular mechanism suggested that the overexpression of STAG3 increased Smad3 expression and decreased CDK4, CDK6, cyclin D1, CXCR4 and RhoA expression. CONCLUSION STAG3 exhibits anticancer effects against HCC, and these effects involve the Smad3-CDK4/CDK6-cyclin D1 and CXCR4/RhoA pathways. STAG3 is a tumor-suppressor gene that may serve as a potential target for molecular therapy, which provides a new idea for the treatment of HCC.
Collapse
Affiliation(s)
- Menglin Zhao
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233030, Anhui, China
| | - Yanyan Wang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233030, Anhui, China
| | - Yue Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233030, Anhui, China
| | - Xinwei Li
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233030, Anhui, China
| | - Jiaqi Mi
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233030, Anhui, China
| | - Qiang Wang
- Department of Network Information Center, Bengbu Medical College, No. 2600 Donghai Road, Bengbu, 233030, Anhui, China
| | - Zhijun Geng
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Lugen Zuo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233030, Anhui, China
| | - Xue Song
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Sitang Ge
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233030, Anhui, China
| | - Zining Zhang
- Department of Clinical Medicine Science, Bengbu Medical College, No. 2600 Donghai Road, Bengbu, 233030, Anhui, China
| | - Mingyue Tang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233030, Anhui, China
| | - Huiyuan Li
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233030, Anhui, China
| | - Zishu Wang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233030, Anhui, China
| | - Chenchen Jiang
- Cancer Neurobiology Group, School of Medicine & Public Health, The University of Newcastle, Callaghan, NSW, 2308, Australia.
| | - Fang Su
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233030, Anhui, China.
| |
Collapse
|
8
|
Yang D, Su Y, Zhao F, Chen C, Zhao K, Xiong X, Ding Y. A Practical Nomogram and Risk Stratification System Predicting the Cancer-Specific Survival for Patients With Advanced Hepatocellular Carcinoma. Front Oncol 2022; 12:914192. [PMID: 35903694 PMCID: PMC9316183 DOI: 10.3389/fonc.2022.914192] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) has the highest cancer-related mortality rate. This study aims to create a nomogram to predict the cancer-specific survival (CSS) in patients with advanced hepatocellular carcinoma. Methods Patients diagnosed with advanced HCC (AJCC stage III and IV) during 1975 to 2018 were obtained from the Surveillance, Epidemiology, and End Results (SEER) database. Qualified patents were randomized into training cohort and validation cohort at a ratio of 7:3. The results of univariate and multivariate Cox regression analyses were used to construct the nomogram. Consistency index (C-index), area under the time-dependent receiver operating characteristic (ROC) curve [time-dependent area under the curve (AUC)], and calibration plots were used to identify and calibrate the nomogram. The net reclassification index (NRI), integrated discrimination improvement (IDI), and C-index, and decision curve analysis DCA were adopted to compare the nomogram’s clinical utility with the AJCC criteria. Results The 3,103 patients with advanced hepatocellular carcinoma were selected (the training cohort: 2,175 patients and the validation cohort: 928 patients). The C-index in both training cohort and validation cohort were greater than 0.7. The AUC for ROC in the training cohort was 0.781, 0.771, and 0.791 at 1, 2, and 3 years CSS, respectively. Calibration plots showed good consistency between actual observations and the 1-, 2-, and 3-year CSS predicted by the nomogram. The 1-, 2-, and 3-year NRI were 0.77, 0.46, and 0.48, respectively. The 1-, 2-, and 3-year IDI values were 0.16, 0.15, and 0.12 (P < 0.001), respectively. DCA curves in both the training and validation cohorts demonstrated that the nomogram showed better predicted 1-, 2-, and 3-year CSS probabilities than AJCC criteria. Conclusions This study established a practical nomogram for predicting CSS in patients with advanced HCC and a risk stratification system that provided an applicable tool for clinical management.
Collapse
Affiliation(s)
- Dashuai Yang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Su
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fangrui Zhao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chen Chen
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Youming Ding, ; Chen Chen,
| | - Kailiang Zhao
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangyun Xiong
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Youming Ding
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Youming Ding, ; Chen Chen,
| |
Collapse
|
9
|
Jiang J, Zheng Y, Chen F, Dong L, Guo X. Activation of YAP1 by STK25 contributes to the progression of hepatocellular carcinoma. Tissue Cell 2022; 76:101797. [DOI: 10.1016/j.tice.2022.101797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 04/01/2022] [Accepted: 04/03/2022] [Indexed: 12/24/2022]
|
10
|
Xu W, Hu B, Cheng Y, Guo Y, Yao W, Qian H. Material basis research for Echinacea purpurea (L.) Moench against hepatocellular carcinoma in a mouse model through integration of metabonomics and molecular docking. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 98:153948. [PMID: 35152087 DOI: 10.1016/j.phymed.2022.153948] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/09/2022] [Accepted: 01/15/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Echinacea purpurea (L.) Moench (EP), a well-known "immunostimulant" in the West, is one of the most popular botanicals for patients with cancer. It has been proved to be effective against hepatocellular carcinoma (HCC), while the active ingredients remains unclear. PURPOSE This study aimed to investigate the inhibitory effect and interpret the material basis of EP against HCC through metabolomics and molecular docking. METHODS Tumor growth, biochemical analysis and pathological changes were detected in HCC-induced mice to evaluate the efficacy of EP. An integrative method combining molecular docking and LC-MS-based metabolomics was performed to evaluate the inhibitory role and screen the material basis of EP against HCC. RESULTS EP significantly suppressed tumor growth and decreased the levels of AFP. Histological analysis showed that wide areas of necrosis in the EP-treated tumors that were almost absent in those in model group. Serum metabolomics results revealed EP could significantly improve 12 serum different metabolites induced by HCC, which were involved into phenylalanine, tyrosine and tryptophan biosynthesis and phenylalanine metabolism. Then, 5 related genes were selected out to be the key targets of EP against HCC based on Metscape. 22 identified compounds were docked through Sybyl-X. The herb-compound-gene-metabolic pathways network (HCGMN) was constructed to reveal the associations between EP and HCC. Finally, 19 compounds were screened as promising active ingredients of EP against HCC. CONCLUSION The results showed that the approach integrating of metabonomics and molecular docking is a powerful strategy to obtain the active ingredients from plants.
Collapse
Affiliation(s)
- Wenqian Xu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, No. 1800, Lihu Ave,, Wuxi, Jiangsu 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Bin Hu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, No. 1800, Lihu Ave,, Wuxi, Jiangsu 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Yuliang Cheng
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, No. 1800, Lihu Ave,, Wuxi, Jiangsu 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Yahui Guo
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, No. 1800, Lihu Ave,, Wuxi, Jiangsu 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Weirong Yao
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - He Qian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, No. 1800, Lihu Ave,, Wuxi, Jiangsu 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
11
|
Shi M, Lv X, Zhu M, Dong Y, Hu L, Qian Y, Fan C, Tian N. HMGA1 promotes hepatocellular carcinoma proliferation, migration, and regulates cell cycle via miR-195-5p. Anticancer Drugs 2022; 33:e273-e285. [PMID: 34407055 DOI: 10.1097/cad.0000000000001201] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
HMGA1 has been reported to be aberrantly expressed and correlate with the poor prognosis of many carcinomas. This study aimed to investigate the clinical significance and molecular mechanism of HMGA1 as a tumor-suppressing gene in hepatocellular carcinoma (HCC). Analysis of TCGA dataset by TANRIC website and R2 platform, we found that HMGA1 expression was significantly higher in HCC tissues compared to that in normal liver tissues and was associated with Edmondson grade. Patients with highly expressed HMGA1 had worse overall survival. Gene ontology enrichment analysis and Kyoto Encyclopedia of Genes and Genomes analysis showed the potential relationships between HMGA1 and other genes in HCC. We also demonstrated that the downregulation of HMGA1 dramatically suppressed the proliferation and migration of HCC cells. Furthermore, ectopic expression of HMGA1 blocked G0/G1 to S transition. Subsequent investigation characterized HMGA1 as a direct target of miR-195-5p, and miR-195-5p downregulation abrogated the effect of HMGA1 on HCC proliferation, migration, and cell cycle arrest. In addition, we also demonstrated that miR-195-5p downregulation abrogated the effect of HMGA1 on HCC growth in vivo. Taken together, our data provide strong evidence that HMGA1 promotes HCC and is negatively regulated by the tumor-suppressor, miR-195-5p.
Collapse
Affiliation(s)
- Minyang Shi
- Department of Cell Biology, Institute of Molecular Medicine, Life Science College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | | | | | | | | | | | | | | |
Collapse
|
12
|
The Unfolded Protein Response Is Associated with Cancer Proliferation and Worse Survival in Hepatocellular Carcinoma. Cancers (Basel) 2021; 13:cancers13174443. [PMID: 34503253 PMCID: PMC8430652 DOI: 10.3390/cancers13174443] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary We studied the association between the unfolded protein response (UPR) and carcinogenesis, cancer progression, and survival in hepatocellular carcinoma (HCC). We studied 655 HCC patients from 4 independent cohorts using an UPR score. The UPR was enhanced as normal liver became cancerous and as HCC advanced in stage. The UPR was correlated with cancer cell proliferation that was confirmed by multiple parameters. Significantly, a high UPR score was associated with worse patient survival. Interestingly, though UPR was associated with a high mutational load, it was not associated with immune response, immune cell infiltration, or angiogenesis. To our knowledge, this is the first study to investigate the clinical relevance of the unfolded protein response in HCC. Abstract Hepatocellular carcinoma is a leading cause of cancer death worldwide. The unfolded protein response (UPR) has been revealed to confer tumorigenic capacity in cancer cells. We hypothesized that a quantifiable score representative of the UPR could be used as a biomarker for cancer progression in HCC. In this study, a total of 655 HCC patients from 4 independent HCC cohorts were studied to examine the relationships between enhancement of the UPR and cancer biology and patient survival in HCC utilizing an UPR score. The UPR correlated with carcinogenic sequence and progression of HCC consistently in two cohorts. Enhanced UPR was associated with the clinical parameters of HCC progression, such as cancer stage and multiple parameters of cell proliferation, including histological grade, mKI67 gene expression, and enrichment of cell proliferation-related gene sets. The UPR was significantly associated with increased mutational load, but not with immune cell infiltration or angiogeneis across independent cohorts. The UPR was consistently associated with worse survival across independent cohorts of HCC. In conclusion, the UPR score may be useful as a biomarker to predict prognosis and to better understand HCC.
Collapse
|
13
|
Younossi ZM, Henry L. Epidemiology of non-alcoholic fatty liver disease and hepatocellular carcinoma. JHEP Rep 2021; 3:100305. [PMID: 34189448 PMCID: PMC8215299 DOI: 10.1016/j.jhepr.2021.100305] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/07/2021] [Accepted: 05/01/2021] [Indexed: 02/07/2023] Open
Abstract
The prevalence of hepatocellular carcinoma (HCC) is increasing worldwide, whereas that of most other cancers is decreasing. Non-alcoholic fatty liver disease (NAFLD), which has increased with the epidemics of obesity and type 2 diabetes, increases the risk of HCC. Interestingly, NAFLD-associated HCC can develop in patients with or without cirrhosis. A lack of awareness about NAFLD-related HCC has led to delays in diagnosis. Therefore, a large number of patients with HCC are diagnosed with advanced-stage HCC with low 5-year survival. In this context, increasing awareness of NAFLD and NAFLD-related HCC may lead to earlier diagnosis and more effective interventions.
Collapse
Key Words
- ALD, alcohol-related liver disease
- CVD, cardiovascular disease
- ELF, enhanced liver fibrosis
- FIB-4, fibrosis-4
- HCC, hepatocellular carcinoma
- NAFLD, non-alcoholic fatty liver disease
- NASH, non-alcoholic steatohepatitis
- PDGF, platelet-derived growth factor
- STAT3, signal transducer and activator of transcription 3
- TNF, tumour necrosis factor-α
- VEGF, vascular endothelial growth factor
- awareness
- cirrhosis
- natural history
- non-cirrhosis
- surveillance
Collapse
Affiliation(s)
- Zobair M. Younossi
- Center for Liver Disease and Department of Medicine, Inova Fairfax Medical Campus, Falls Church, VA, United States
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, United States
- Medical Service Line. Inova Health Systems, Falls Church, VA, United States
| | - Linda Henry
- Center for Outcomes Research in Liver Diseases, Washington DC, United States
| |
Collapse
|
14
|
Vijayan K, Wei L, Glennon EKK, Mattocks C, Bourgeois N, Staker B, Kaushansky A. Host-targeted Interventions as an Exciting Opportunity to Combat Malaria. Chem Rev 2021; 121:10452-10468. [PMID: 34197083 DOI: 10.1021/acs.chemrev.1c00062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Terminal and benign diseases alike in adults, children, pregnant women, and others are successfully treated by pharmacological inhibitors that target human enzymes. Despite extensive global efforts to fight malaria, the disease continues to be a massive worldwide health burden, and new interventional strategies are needed. Current drugs and vector control strategies have contributed to the reduction in malaria deaths over the past 10 years, but progress toward eradication has waned in recent years. Resistance to antimalarial drugs is a substantial and growing problem. Moreover, targeting dormant forms of the malaria parasite Plasmodium vivax is only possible with two approved drugs, which are both contraindicated for individuals with glucose-6-phosphate dehydrogenase deficiency and in pregnant women. Plasmodium parasites are obligate intracellular parasites and thus have specific and absolute requirements of their hosts. Growing evidence has described these host necessities, paving the way for opportunities to pharmacologically target host factors to eliminate Plasmodium infection. Here, we describe progress in malaria research and adjacent fields and discuss key challenges that remain in implementing host-directed therapy against malaria.
Collapse
Affiliation(s)
| | - Ling Wei
- Seattle Children's Research Institute, Seattle, Washington 98109, United States
| | | | - Christa Mattocks
- Department of Global Health, University of Washington, Seattle, Washington 98195, United States
| | - Natasha Bourgeois
- Seattle Children's Research Institute, Seattle, Washington 98109, United States.,Department of Global Health, University of Washington, Seattle, Washington 98195, United States
| | - Bart Staker
- Seattle Children's Research Institute, Seattle, Washington 98109, United States
| | - Alexis Kaushansky
- Seattle Children's Research Institute, Seattle, Washington 98109, United States.,Department of Global Health, University of Washington, Seattle, Washington 98195, United States.,Department of Pediatrics, University of Washington, Seattle, Washington 98105, United States.,Brotman Baty Institute for Precision Medicine, Seattle, Washington 98195, United States
| |
Collapse
|
15
|
Shao M, Shi R, Gao ZX, Gao SS, Li JF, Li H, Cui SZ, Hu WM, Chen TY, Wu GR, Zhang J, Xu J, Sy MS, Li C. Crizotinib and Doxorubicin Cooperatively Reduces Drug Resistance by Mitigating MDR1 to Increase Hepatocellular Carcinoma Cells Death. Front Oncol 2021; 11:650052. [PMID: 34094940 PMCID: PMC8170002 DOI: 10.3389/fonc.2021.650052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/16/2021] [Indexed: 11/24/2022] Open
Abstract
As the sixth most lethal cancers worldwide, hepatocellular carcinoma (HCC) has been treated with doxorubicin (Dox) for decades. However, chemotherapy resistance, especially for Dox is an even more prominent problem due to its high cardiotoxicity. To find a regimen to reduce Dox resistance, and identify the mechanisms behind it, we tried to identify combination of drugs that can overcome drug resistance by screening tyrosine kinase inhibitor(s) with Dox with various HCC cell lines in vitro and in vivo. We report here that combination of Crizo and Dox has a synergistic effect on inducing HCC cell death. Accordingly, Crizo plus Dox increases Dox accumulation in nucleus 3-16 times compared to Dox only; HCC cell death enhanced at least 50% in vitro and tumor weights reduced ranging from 35 to 65%. Combining these two drugs reduces multiple drug resistance 1 (MDR1) protein as a result of activation of protein kinase RNA-like endoplasmic reticulum kinase (PERK), which phosphorylates eIF2α, leading to protein translational repression. Additionally, PERK stimulation activates C-Jun terminal kinase (JNK), resulting in accumulation of unfused autophagosome to enhance autophagic cell death via Poly-ADP-ribosyltransferase (PARP-1) cleavage. When the activity of PERK or JNK is blocked, unfused autophagosome is diminished, cleaved PARP-1 is reduced, and cell death is abated. Therefore, Crizo plus Dox sensitize HCC drug resistance by engaging PERK-p- eIF2α-MDR1, and kill HCC cells by engaging PERK-JNK- autophagic cell death pathways. These newly discovered mechanisms of Crizo plus Dox not only provide a potential treatment for HCC but also point to an approach to overcome MDR1 related drug resistance in other cancers.
Collapse
Affiliation(s)
- Ming Shao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Run Shi
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Zhen-Xing Gao
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Shan-Shan Gao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jing-Feng Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Huan Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Shu-Zhong Cui
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.,Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Wei-Min Hu
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Tian-Yun Chen
- Department of Stomatology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gui-Ru Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jie Zhang
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jiang Xu
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Man-Sun Sy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Chaoyang Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
16
|
Tian P, Wang Y, Du W. Ultrasound-targeted microbubble destruction enhances the anti-tumor action of miR-4284 inhibitor in non-small cell lung cancer cells. Exp Ther Med 2021; 21:551. [PMID: 33850523 PMCID: PMC8027739 DOI: 10.3892/etm.2021.9983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) are known to be involved in various human cancer types. Ultrasound-targeted microbubble destruction (UTMD) may improve the transfection efficiency of exogenous genes into target tissues and organs, thereby improving cancer treatment. In the present study, the role of miR-4284 in non-small cell lung cancer (NSCLC) was investigated and the effect of UTMD-mediated inhibition of miR-4284 on tumor progression was further analyzed. The expression of miR-4284 in NSCLC cells and tissues was detected by reverse transcription-quantitative PCR. UTMD-mediated inhibition of miR-4284 was achieved by co-transfection of microvesicles and miR-4284 inhibitors into NSCLC cells. A Cell Counting Kit-8 assay was used to determine NSCLC cell proliferation, and the migration and invasion of NSCLC cells were examined by Transwell assays. Compared with that in the control group, the expression of miR-4284 was increased in NSCLC tissues and cells. Knockdown of miR-4284 in NSCLC cells inhibited cell proliferation, migration and invasion. UTMD improved the transfection efficiency of miR-4284 inhibitors in NSCLC cells, resulting in more significant inhibition of tumor cell proliferation, migration and invasion. In conclusion, the results indicated that the expression of miR-4284 was increased in clinical samples and cell lines of NSCLC and that knockdown of miR-4284 inhibited the proliferation, migration and invasion of tumor cells. UTMD-mediated miR-4284 inhibition further promoted this effect, indicating that this technique may represent a novel strategy for the treatment of NSCLC.
Collapse
Affiliation(s)
- Peng Tian
- Department of Ultrasonics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Yanzhen Wang
- Department of Ultrasonics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Wenyan Du
- Department of Science and Education, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| |
Collapse
|
17
|
Notartomaso S, Scarselli P, Mascio G, Liberatore F, Mazzon E, Mammana S, Gugliandolo A, Cruccu G, Bruno V, Nicoletti F, Battaglia G. N-Acetylcysteine causes analgesia in a mouse model of painful diabetic neuropathy. Mol Pain 2021; 16:1744806920904292. [PMID: 32009537 PMCID: PMC6997966 DOI: 10.1177/1744806920904292] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
N-Acetylcysteine, one of the most prescribed antioxidant drugs, enhances pain
threshold in rodents and humans by activating mGlu2 metabotropic glutamate
receptors. Here, we assessed the analgesic activity of N-acetylcysteine in the
streptozotocin model of painful diabetic neuropathy and examined the effect of
N-acetylcysteine on proteins that are involved in mechanisms of nociceptive
sensitization. Mice with blood glucose levels ≥250 mg/dl in response to a single
intraperitoneal (i.p.) injection of streptozotocin (200 mg/kg) were used for the
assessment of mechanical pain thresholds. Systemic treatment with
N-acetylcysteine (100 mg/kg, i.p., either single injection or daily injections
for seven days) caused analgesia in diabetic mice. N-acetylcysteine-induced
analgesia was abrogated by the Sxc− inhibitors, sulfasalazine (8 mg/kg, i.p.), erastin (30 mg/kg,
i.p.), and sorafenib (10 mg/kg, i.p.), or by the mGlu2/3 receptor antagonist,
LY341495 (1 mg/kg, i.p.). Repeated administrations of N-acetylcysteine in
diabetic mice reduced ERK1/2 phosphorylation in the dorsal region of the lumbar
spinal cord. The analgesic activity of N-acetylcysteine was occluded by the MEK
inhibitor, PD0325901 (25 mg/kg, i.p.), the TRPV1 channel blocker, capsazepine
(40 mg/kg, i.p.), or by a cocktail of NMDA and mGlu5 metabotropic glutamate
receptor antagonists (memantine, 25 mg/kg, plus MTEP, 5 mg/kg,
both i.p.). These findings offer the first demonstration that N-acetylcysteine
relieves pain associated with diabetic neuropathy and holds promise for the use
of N-acetylcysteine as an add-on drug in diabetic patients.
Collapse
Affiliation(s)
| | - Pamela Scarselli
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | - Giada Mascio
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | | | | | - Santa Mammana
- IRCCS Centro Neurolesi "Bonino-Pulejo", Messina, Italy
| | | | - Giorgio Cruccu
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Valeria Bruno
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy.,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Ferdinando Nicoletti
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy.,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Giuseppe Battaglia
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy.,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| |
Collapse
|
18
|
Oshi M, Kim TH, Tokumaru Y, Yan L, Matsuyama R, Endo I, Cherkassky L, Takabe K. Enhanced DNA Repair Pathway is Associated with Cell Proliferation and Worse Survival in Hepatocellular Carcinoma (HCC). Cancers (Basel) 2021; 13:cancers13020323. [PMID: 33477315 PMCID: PMC7830462 DOI: 10.3390/cancers13020323] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary We studied the relationship between enhancement of DNA repair and cancer aggressiveness, tumor immune microenvironment, and patient survival in 749 hepatocellular carcinoma (HCC) patients from 5 cohorts using a DNA repair pathway score. We show that the DNA repair pathway was enhanced by the stepwise carcinogenic process of HCC, notably in grade 3 compared to grade 1 or 2 HCC. DNA repair high HCC was associated with worse survival, elevated intratumor heterogeneity, and mutation load, but not with the fraction of immune cell infiltration nor cytolytic activity. The expression of proliferation- and other cancer aggressiveness-related gene sets was also increased. Interestingly, these features were more pronounced in low-grade compared to high-grade HCC. In conclusion, the DNA repair score may be used to understand the role of DNA repair pathways in patient prognosis and treatment sensitivity and be used to improve patient outcome. To our knowledge, this is the first study using DNA repair pathway-related gene set expression data to examine and validate the clinical relevance of DNA repair pathway activity in HCC. Abstract Hepatocellular carcinoma (HCC) is one of the most common malignancies and a leading cause of cancer-related deaths worldwide. In this study, a total of 749 HCC patients from 5 cohorts were studied to examine the relationships between enhancement of DNA repair and cancer aggressiveness, tumor immune microenvironment, and patient survival in HCC, utilizing a DNA repair pathway score. Our findings suggest that the DNA repair pathway was not only enhanced by the stepwise carcinogenic process of HCC, but also significantly enhanced in grade 3 HCC compared with grade 1 and 2 tumors. DNA repair high HCC was associated with worse survival, elevated intratumor heterogeneity, and mutation load, but not with the fraction of immune cell infiltration nor immune response. HCC tumors with a DNA repair high score enriched the cell proliferation- and other cancer aggressiveness-related gene sets. Interestingly, these features were more pronounced in grade 1 and 2 HCC compared to grade 3 HCC. To our knowledge, this is the first study to use DNA repair pathway-related gene set expression data to examine and validate the clinical relevance of DNA repair pathway activity in HCC. The DNA repair score may be used to better understand and predict prognosis in HCC.
Collapse
Affiliation(s)
- Masanori Oshi
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (M.O.); (T.H.K.); (Y.T.); (L.C.)
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan; (R.M.); (I.E.)
| | - Tae Hee Kim
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (M.O.); (T.H.K.); (Y.T.); (L.C.)
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14263, USA
| | - Yoshihisa Tokumaru
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (M.O.); (T.H.K.); (Y.T.); (L.C.)
- Department of Surgical Oncology, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Li Yan
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Ryusei Matsuyama
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan; (R.M.); (I.E.)
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan; (R.M.); (I.E.)
| | - Leonid Cherkassky
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (M.O.); (T.H.K.); (Y.T.); (L.C.)
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (M.O.); (T.H.K.); (Y.T.); (L.C.)
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan; (R.M.); (I.E.)
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14263, USA
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8520, Japan
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan
- Correspondence: ; Tel.: +1-716-8455540; Fax: +1-716-8451668
| |
Collapse
|
19
|
Xu J, Wan Z, Tang M, Lin Z, Jiang S, Ji L, Gorshkov K, Mao Q, Xia S, Cen D, Zheng J, Liang X, Cai X. N 6-methyladenosine-modified CircRNA-SORE sustains sorafenib resistance in hepatocellular carcinoma by regulating β-catenin signaling. Mol Cancer 2020; 19:163. [PMID: 33222692 PMCID: PMC7681956 DOI: 10.1186/s12943-020-01281-8] [Citation(s) in RCA: 210] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 11/12/2020] [Indexed: 12/22/2022] Open
Abstract
Background and aims Accumulating evidence suggests that the primary and acquired resistance of hepatocellular carcinoma (HCC) to sorafenib is mediated by multiple molecular, cellular, and microenvironmental mechanisms. Understanding these mechanisms will enhance the likelihood of effective sorafenib therapy. Methods In vitro and in vivo experiments were performed and clinical samples and online databases were acquired for clinical investigation. Results In this study, we found that a circular RNA, circRNA-SORE, which is up-regulated in sorafenib-resistant HCC cells, was necessary for the maintenance of sorafenib resistance, and that silencing circRNA-SORE substantially increased the efficacy of sorafenib-induced apoptosis. Mechanistic studies determined that circRNA-SORE sequestered miR-103a-2-5p and miR-660-3p by acting as a microRNA sponge, thereby competitively activating the Wnt/β-catenin pathway and inducing sorafenib resistance. The increased level of circRNA-SORE in sorafenib-resistant cells resulted from increased RNA stability. This was caused by an increased level of N6-methyladenosine (m6A) at a specific adenosine in circRNA-SORE. In vivo delivery of circRNA-SORE interfering RNA by local short hairpin RNA lentivirus injection substantially enhanced sorafenib efficacy in animal models. Conclusions This work indicates a novel mechanism for maintaining sorafenib resistance and is a proof-of-concept study for targeting circRNA-SORE in sorafenib-treated HCC patients as a novel pharmaceutical intervention for advanced HCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-020-01281-8.
Collapse
Affiliation(s)
- Junjie Xu
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.,Key Laboratory of Laparoscopic Technology of Zhejiang Province, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou, China.,Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China.,Zhejiang University Cancer Center, Hangzhou, China
| | - Zhe Wan
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.,Key Laboratory of Laparoscopic Technology of Zhejiang Province, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou, China.,Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China.,Zhejiang University Cancer Center, Hangzhou, China
| | - Minyue Tang
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Zhongjie Lin
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.,Key Laboratory of Laparoscopic Technology of Zhejiang Province, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou, China.,Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China.,Zhejiang University Cancer Center, Hangzhou, China
| | - Shi Jiang
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.,Key Laboratory of Laparoscopic Technology of Zhejiang Province, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou, China.,Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China.,Zhejiang University Cancer Center, Hangzhou, China
| | - Lin Ji
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.,Key Laboratory of Laparoscopic Technology of Zhejiang Province, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou, China.,Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China.,Zhejiang University Cancer Center, Hangzhou, China
| | - Kirill Gorshkov
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Qijiang Mao
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.,Key Laboratory of Laparoscopic Technology of Zhejiang Province, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou, China.,Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China.,Zhejiang University Cancer Center, Hangzhou, China
| | - Shunjie Xia
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.,Key Laboratory of Laparoscopic Technology of Zhejiang Province, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou, China.,Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China.,Zhejiang University Cancer Center, Hangzhou, China
| | - Dong Cen
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.,Key Laboratory of Laparoscopic Technology of Zhejiang Province, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou, China.,Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China.,Zhejiang University Cancer Center, Hangzhou, China
| | - Junhao Zheng
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.,Key Laboratory of Laparoscopic Technology of Zhejiang Province, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou, China.,Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China.,Zhejiang University Cancer Center, Hangzhou, China
| | - Xiao Liang
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China. .,Key Laboratory of Laparoscopic Technology of Zhejiang Province, Hangzhou, China. .,Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou, China. .,Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China. .,Zhejiang University Cancer Center, Hangzhou, China.
| | - Xiujun Cai
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China. .,Key Laboratory of Laparoscopic Technology of Zhejiang Province, Hangzhou, China. .,Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Hangzhou, China. .,Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, China. .,Zhejiang University Cancer Center, Hangzhou, China.
| |
Collapse
|
20
|
Luo D, Hou D, Wen T, Feng M, Zhang H. Efficacy and safety of Brucea javanica oil emulsion for liver cancer: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e23197. [PMID: 33217831 PMCID: PMC7676520 DOI: 10.1097/md.0000000000023197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/11/2020] [Accepted: 10/17/2020] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Brucea javanica oil emulsion (BJOE), extracted from the Chinese herb Bruceae Fructus (Yadanzi), is a broad-spectrum anti-tumor drug and has been widely used for the treatment of liver cancer in China. The aim of this study is to systematically investigate the efficacy and safety of BJOE for the treatment of liver cancer. METHODS Seven electronic databases including the Cochrane Library, PubMed, Excerpt Medica Database, Chinese Biomedical Literature Database, China National Knowledge Infrastructure, China Scientific Journal Database, and Wanfang Database will be systematically retrieved for data extraction from their inceptions to September 2020. Cochrane Risk of Bias tool will be used to assess the risk of bias of included studies. The RevMan 5.4 and Stata 16.0 software will be applied for statistical analyses. Statistical heterogeneity will be computed by I tests. Sensitivity analysis will be conducted to evaluate the stability of the results. The publication bias will be evaluated by funnel plots and Egger test. The quality of evidence will be assessed by the GRADE system. RESULTS The results of our research will be published in a peer-reviewed journal or presenting the findings at a relevant conference. CONCLUSION The conclusion of this study will provide helpful evidence of the effect and safety of BJOE for the treatment of liver cancer in clinical practice. OSF REGISTRATION NUMBER 10.17605/OSF.IO/UC8XQ.
Collapse
Affiliation(s)
| | - Daorui Hou
- Department of Oncology
- Department of Traditional Chinese Medicine Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan Province, China
| | | | | | | |
Collapse
|
21
|
Wu J, Chai H, Li F, Ren Q, Gu Y. SETD1A augments sorafenib primary resistance via activating YAP in hepatocellular carcinoma. Life Sci 2020; 260:118406. [PMID: 32918976 DOI: 10.1016/j.lfs.2020.118406] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/24/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022]
Abstract
AIMS Sorafenib, the approved first-line chemotherapy drug for HCC (Hepatocellular Carcinoma), remains the key treatment agent which effectively improves the survival rate of advanced HCC patients. However, the sorafenib primary resistance limits the application of sorafenib for HCC treatment. The aims of current study are to explore the role and mechanism of SETD1A (Histone Lysine Methyltransferase SET Domain Containing 1A) in sorafenib primary resistance. MAIN METHODS The SETD1A expression in HCC was analyzed by Gene Expression Profiling Interactive Analysis. The survival of HCC patients was analyzed by Kaplan-Meier Plotter. Western Blot and Real-time qPCR were performed to measure the protein and mRNA levels, respectively. Cell counting kit-8 assay and colony formation assay were performed to determine cell viability and proliferation. Propidium Iodide and Trypan Blue staining assays were performed to investigate cell death. KEY FINDINGS Here, we showed that the expression of SETD1A was markedly upregulated in both HCC cell lines and tumor tissues compared to normal hepatocytes and corresponding non-tumor liver tissues, respectively. Regardless of whether treated with sorafenib, the patients who had higher level of SETD1A underwent lower survival rate of overall. In addition, SETD1A expression was positively correlated with the IC50 of sorafenib treated HCC cell lines. Furthermore, we indicated that knockdown of SETD1 augmented proliferation inhibition and cell death induced by sorafenib. SETD1A deficiency impaired YAP (Yes-associated protein) phosphorylation and activation. YAP activation contributed to SETD1A mediated sorafenib primary resistance. SIGNIFICANCE The current study demonstrated that SETD1A enhanced YAP activation to induce sorafenib primary resistance in HCC.
Collapse
Affiliation(s)
- Jugang Wu
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Hongjuan Chai
- Department of Gynecology and Obstetrics, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Feng Li
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Qing Ren
- Department of Gynecology and Obstetrics, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Department of Gynecology and Obstetrics, Hainan West Central Hospital (Shanghai Ninth People's Hospital, Hainan Branch), Shanghai JiaoTong University School of Medicine, Hainan, China.
| | - Yan Gu
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| |
Collapse
|
22
|
Hou D, Xiong J, Li Y, Peng Y, Xiong L. Efficacy and safety of Xiaoaiping injection for liver cancer: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e21993. [PMID: 32871951 PMCID: PMC7458193 DOI: 10.1097/md.0000000000021993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Xiaoaiping injection, extracted from the Chinese herb Marsdenia tenacissima (Roxb.) Wight et Arn., is a broad-spectrum anti-tumor drug and has been widely used for the treatment of liver cancer in China. The aim of this study is to systematically investigate the efficacy and safety of Xiaoaiping injection for the treatment of liver cancer. METHODS AND ANALYSIS Seven electronic databases including the Cochrane Library, PubMed, Excerpt Medica Database, Chinese Biomedical Literature Database, China National Knowledge Infrastructure, China Scientific Journal Database, and Wanfang Database will be systematically retrieved for data extraction from their inceptions to August 2020. Cochrane Risk of Bias tool will be used to assess the risk of bias of included studies. The RevMan 5.4 and Stata 16.0 software will be applied for statistical analyses. Statistical heterogeneity will be computed by I tests. Sensitivity analysis will be conducted to evaluate the stability of the results. The publication bias will be evaluated by funnel plots and Eggers test. The quality of evidence will be assessed by the GRADE system. RESULTS The results of our research will be published in a peer-reviewed journal. CONCLUSION The conclusion of this study will provide helpful evidence of the effect and safety of Xiaoaiping injection for the treatment of liver cancer in clinical practice. OSF REGISTRATION NUMBER 10.17605/OSF.IO/9BD6A.
Collapse
Affiliation(s)
- Daorui Hou
- Department of Traditional Chinese Medicine Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan Province
| | - Jian Xiong
- Department of Oncology, Guang’anmen Hospital, Beijing
| | - Ya Li
- Hangzhou Lin’an TCM Hospital, Hangzhou, Zhejiang Province
| | | | - Lu Xiong
- Department of Oncology, Guang’anmen Hospital, Beijing
| |
Collapse
|
23
|
Mao X, Guo Y, Lu Z, Wen F, Liang H, Sun W. Enhanced CT Textures Derived From Computer Mathematic Distribution Analysis Enables Arterial Enhancement Fraction Being an Imaging Biomarker Option of Hepatocellular Carcinoma. Front Oncol 2020; 10:1337. [PMID: 32850426 PMCID: PMC7431458 DOI: 10.3389/fonc.2020.01337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose: This study aims to explore the imaging–clinic relationship and an optional imaging biomarker of hepatocellular carcinoma (HCC) by using texture analysis on arterial enhancement fraction (AEF). Materials and Methods: The HCC patients treated in No. 2 Interventional Ward, ShengJing Hospital of China Medical University from June 2018 to June 2019 were enrolled, for whom tri-phasic enhanced CT scans were acquired. Perfusion analysis and texture analysis were then performed on the tri-phasic enhanced CT images. After the region of interest (ROI) of viable HCC was drawn, 13 AEF textures describing the values distribution were conducted. A between-groups comparison of AEF textures was made where the cases had grouping properties, a correlation analysis was made between AEF textures and alpha-fetoprotein (AFP) as well as other clinical data which were digital, and regression analysis was made when a significant correlation was found. SPSS 19.0 (IBM) was utilized for statistical analysis; a significant difference was considered when P < 0.05. Results: Twenty-five HCC patients were enrolled. Several AEF textures were found to have a correlation with clinical features, including previous surgery history, age, glutamic oxaloacetylase, indirect bilirubin, creatinine, and AFP. The majority of AEF textures (up to 9/13) were found to have a correlation with AFP (SD, variance, uniformity, energy, entropy, inertia, correlation, inverse difference moment, and cluster prominence), while six or seven textures have a linear or cubic relationship with AFP (SD, variance, uniformity, inertia, correlation, cluster prominence, plus inverse difference moment). Conclusion: The AEF textures of HCC are strongly correlated with and are impacted by AFP, which may enable AEF to act as an optional imaging biomarker of HCC.
Collapse
Affiliation(s)
- Xiaonan Mao
- Department of Radiology, ShengJing Hospital of China Medical University, Shenyang, China
| | - Yan Guo
- GE Healthcare, Shanghai, China
| | - Zaiming Lu
- Department of Radiology, ShengJing Hospital of China Medical University, Shenyang, China
| | - Feng Wen
- Department of Radiology, ShengJing Hospital of China Medical University, Shenyang, China
| | - Hongyuan Liang
- Department of Radiology, ShengJing Hospital of China Medical University, Shenyang, China
| | - Wei Sun
- Department of Radiology, ShengJing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
24
|
Feng J, Li J, Wu L, Yu Q, Ji J, Wu J, Dai W, Guo C. Emerging roles and the regulation of aerobic glycolysis in hepatocellular carcinoma. J Exp Clin Cancer Res 2020; 39:126. [PMID: 32631382 PMCID: PMC7336654 DOI: 10.1186/s13046-020-01629-4] [Citation(s) in RCA: 403] [Impact Index Per Article: 80.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
Liver cancer has become the sixth most diagnosed cancer and the fourth leading cause of cancer death worldwide. Hepatocellular carcinoma (HCC) is responsible for up to 75-85% of primary liver cancers, and sorafenib is the first targeted drug for advanced HCC treatment. However, sorafenib resistance is common because of the resultant enhancement of aerobic glycolysis and other molecular mechanisms. Aerobic glycolysis was firstly found in HCC, acts as a hallmark of liver cancer and is responsible for the regulation of proliferation, immune evasion, invasion, metastasis, angiogenesis, and drug resistance in HCC. The three rate-limiting enzymes in the glycolytic pathway, including hexokinase 2 (HK2), phosphofructokinase 1 (PFK1), and pyruvate kinases type M2 (PKM2) play an important role in the regulation of aerobic glycolysis in HCC and can be regulated by many mechanisms, such as the AMPK, PI3K/Akt pathway, HIF-1α, c-Myc and noncoding RNAs. Because of the importance of aerobic glycolysis in the progression of HCC, targeting key factors in its pathway such as the inhibition of HK2, PFK or PKM2, represent potential new therapeutic approaches for the treatment of HCC.
Collapse
Affiliation(s)
- Jiao Feng
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jingjing Li
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Qiang Yu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jie Ji
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China.
| | - Weiqi Dai
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China.
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China.
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan University, Shanghai, 200032, China.
- Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200336, China.
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China.
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China.
| |
Collapse
|
25
|
Lin H, Zhang R, Wu W, Lei L. Comprehensive network analysis of the molecular mechanisms associated with sorafenib resistance in hepatocellular carcinoma. Cancer Genet 2020; 245:27-34. [PMID: 32559715 DOI: 10.1016/j.cancergen.2020.04.076] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/28/2020] [Accepted: 04/23/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) is an intractable disease because patients with HCC frequently develop sorafenib resistance after long-term chemotherapy. Although studies has demonstrated the availability of cumulative information on drug-resistant patients, little is known about the strategies and molecular mechanisms to reverse sorafenib resistance. Here, the present study identified critical mRNAs and transcription factors (TFs) associated with sorafenib resistance of HCC and evaluated the significance correlation between drug-resistant genes and TFs in comprehensive network for HCC xenografts mice. METHODS The expression profiles of mRNAs were compared between sorafenib-acquired resistant tissue and sorafenib sensitive tissue utilizing RNA-Seq data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). Gene Ontology and KEGG pathway analysis were performed to investigate the biological function of significantly dysregulated mRNA. Furthermore, the Kaplan-Meier survival analyses were performed to evaluate the effect of mRNA on over survival. Subsequently, TFs were predicted using TRANSFAC and TF-mRNA regulatory networks were visualized using cytoscape software. RESULTS A total of 827 mRNAs were found to be differentially expressed in sorafenib-acquired resistant tissue compared with control. Thereafter, the results of functional enrichment analysis showed the dysregulated mRNAs involved in drug-resistant signaling pathway, including MAPK, JAK-STAT, TGF-β and drug-metabolism cytochrome P450 signaling pathway. CDK1, CDKN1A and TAPBP might serve as prognostic signature of resistance of HCC to sorafenib according to the survival analysis. Furthermore, TF-mRNA networks were constructed. There were 18 TFs were predicted to regulate differentially expressed mRNAs, which play an essential role in the regulation of dysfunctional gene networks. NFKB1 was presented in the TF-mRNA networks as the node with the highest degree and MYC was predicted as prognostic TF in drug resistance of HCC CONCLUSIONS: Taken together, our findings showed that novel mRNAs and TFs, which served as critical biomarkers to predict survival and therapeutic targets of resistance to sorafenib in HCC. Furthermore, we constructed the TF-mRNA networks, which provides valuable theoretical references to further evaluate the molecular mechanisms of resistance to sorafenib in HCC.
Collapse
Affiliation(s)
- Haoming Lin
- Department of Pancreto-biliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China.
| | - Rui Zhang
- Department of Pancreto-biliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China.
| | - Wenrui Wu
- Department of Pancreto-biliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China.
| | - Liming Lei
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Laboratory of South China Structural Heart Disease, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
26
|
Galle PR, Foerster F, Kudo M, Chan SL, Llovet JM, Qin S, Schelman WR, Chintharlapalli S, Abada PB, Sherman M, Zhu AX. Biology and significance of alpha-fetoprotein in hepatocellular carcinoma. Liver Int 2019; 39:2214-2229. [PMID: 31436873 DOI: 10.1111/liv.14223] [Citation(s) in RCA: 391] [Impact Index Per Article: 65.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/19/2019] [Accepted: 08/03/2019] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common causes of cancer-related deaths globally due, in part, to the majority of patients being diagnosed with intermediate or advanced stage disease. Our increased understanding of the heterogeneous molecular pathogenesis of HCC has led to significant developments in novel targeted therapies. Despite these advances, there remains a high unmet need for new treatment options. HCC is a complex disease with multiple pathogenic mechanisms caused by a variety of risk factors, making it difficult to characterize with a single biomarker. In fact, numerous biomarkers have been studied in HCC, but alpha-fetoprotein (AFP) remains the most widely used and accepted serum marker since its discovery over 60 years ago. This review summarizes the most relevant studies associated with the regulation of AFP at the gene and protein levels; the pathophysiology of AFP as a pro-proliferative protein; and the correlation of AFP with molecular HCC subclasses, the vascular endothelial growth factor pathway and angiogenesis. Also described are the historical and current uses of AFP for screening and surveillance, diagnosis, its utility as a prognostic and predictive biomarker and its role as a tumour antigen in HCC. Taken together, these data demonstrate the relevance of AFP for patients with HCC and identify several remaining questions that will benefit from future research.
Collapse
Affiliation(s)
- Peter R Galle
- Department of Internal Medicine I, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Friedrich Foerster
- Department of Internal Medicine I, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | | | - Josep M Llovet
- Translational Research in Hepatic Oncology, Liver Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain.,Mount Sinai Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Shukui Qin
- Cancer Center of Bayi Hospital, Nanjing Chinese Medicine University, Nanjing, China
| | | | | | | | | | - Andrew X Zhu
- Massachusetts General Hospital Cancer Center, Harvard Medical Center, Boston, MA, USA
| |
Collapse
|
27
|
Hilmi M, Neuzillet C, Calderaro J, Lafdil F, Pawlotsky JM, Rousseau B. Angiogenesis and immune checkpoint inhibitors as therapies for hepatocellular carcinoma: current knowledge and future research directions. J Immunother Cancer 2019; 7:333. [PMID: 31783782 PMCID: PMC6884868 DOI: 10.1186/s40425-019-0824-5] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 11/13/2019] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the second deadliest cancer worldwide, due to its high incidence and poor prognosis. Frequent initial presentation at advanced stages along with impaired liver function limit the use of a broad therapeutic arsenal in patients with HCC. Although main HCC oncogenic drivers have been deciphered in recent years (TERT, TP53, CTNNB1 mutations, miR122 and CDKN2A silencing), therapeutic applications derived from this molecular knowledge are still limited. Given its high vascularization and immunogenicity, antiangiogenics and immune checkpoint inhibitors (ICI), respectively, are two therapeutic approaches that have shown efficacy in HCC. Depending on HCC immune profile, combinations of these therapies aim to modify the protumoral/antitumoral immune balance, and to reactivate and favor the intratumoral trafficking of cytotoxic T cells. Combination therapies involving antiangiogenics and ICI may be synergistic, because vascular endothelial growth factor A inhibition increases intratumoral infiltration and survival of cytotoxic T lymphocytes and decreases regulatory T lymphocyte recruitment, resulting in a more favorable immune microenvironment for ICI antitumoral activity. First results from clinical trials evaluating combinations of these therapies are encouraging with response rates never observed before in patients with HCC. A better understanding of the balance and interactions between protumoral and antitumoral immune cells will help to ensure the success of future therapeutic trials. Here, we present an overview of the current state of clinical development of antitumoral therapies in HCC and the biological rationale for their use. Moreover, translational studies on tumor tissue and blood, prior to and during treatment, will help to identify biomarkers and immune signatures with predictive value for both clinical outcome and response to combination therapies.
Collapse
MESH Headings
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/adverse effects
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/antagonists & inhibitors
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/pathology
- Clinical Trials as Topic
- Combined Modality Therapy
- Disease Susceptibility
- Humans
- Immunomodulation/drug effects
- Liver Neoplasms/drug therapy
- Liver Neoplasms/etiology
- Liver Neoplasms/pathology
- Molecular Targeted Therapy
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/metabolism
- Treatment Outcome
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Marc Hilmi
- Department of Medical Oncology, Curie Institute, University of Versailles Saint-Quentin, Paris, France
| | - Cindy Neuzillet
- Department of Medical Oncology, Curie Institute, University of Versailles Saint-Quentin, Paris, France
| | - Julien Calderaro
- Department of Pathology, Henri Mondor Hospital, Créteil, France
- IMRB-INSERM U955 Team 18, Créteil, France
- Université Paris-Est-Créteil, Créteil, France
| | - Fouad Lafdil
- IMRB-INSERM U955 Team 18, Créteil, France
- Université Paris-Est-Créteil, Créteil, France
- Institut Universitaire de France, Paris, France
| | - Jean-Michel Pawlotsky
- IMRB-INSERM U955 Team 18, Créteil, France
- Université Paris-Est-Créteil, Créteil, France
- National Reference Center for Viral Hepatitis B, C and D, Department of Virology, Henri Mondor Hospital, Créteil, France
| | - Benoit Rousseau
- IMRB-INSERM U955 Team 18, Créteil, France.
- Department of Medicine, Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
28
|
Yu Ping Feng San Exert Anti-Angiogenesis Effects through the Inhibition of TSLP-STAT3 Signaling Pathways in Hepatocellular Carcinoma. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:1947156. [PMID: 31885639 PMCID: PMC6925680 DOI: 10.1155/2019/1947156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 08/09/2019] [Accepted: 09/17/2019] [Indexed: 12/14/2022]
Abstract
Background Clinically, Yu ping feng san (YPFS) has been extensively used as a medication for treating immune deficiency, and YPFS is combined with chemotherapy drugs to treat cancer, including hepatocellular carcinoma (HCC), lung cancer, and pancreatic cancer. Previous research has shown that YPFS has a therapeutic effect on HCC by improving the immunosuppressive state of the liver cancer microenvironment. The present study aimed to investigate the effect of YPFS on angiogenesis of HCC. Methods High-performance liquid chromatography (HPLC) was used to certify the composition of YPFS. An orthotopic transplanted model of murine HCC was entrenched. Immunohistochemistry was used to observe the changes of the microvessel density (MVD). The MTT assay was used to detect the cell viability. ELISA was performed to analyze the expression of related factors. Western blot was used to analyze the protein expression. Tube formation assay was used to analyze the anti-angiogenic efficiency. Results YPFS significantly reduced the tumor volume and weight, thus exerted the growth inhibitory effect. The level of MVD and VEGF was obviously decreased in YPFS-treated HCC-bearing mice, and the YPFS treatment also reduced the VEGF level in Hepa1-6 cells. Further study revealed that the expression of TSLP/TSLPR and p-STAT3/STAT3 was decreased by YPFS. The level of MVD and VEGF and the expression of TSLP/TSLPR and p-STAT3/STAT3 in tumor tissue and Hepa1-6 cells were suppressed by incubation with the anti-TSLP antibody, whereas treatment with the anti-TSLP antibody in YPFS-treated cells did not cause further significant inhibition compared with the cells treated only with YPFS. More importantly, YPFS inhibited proliferation, expression of p-STAT3/STAT3, and tube formation of HUVECs induced by TSLP. Conclusions These results indicated that YPFS attenuated the activation of the TSLP-STAT3 signaling pathway by inhibiting the immune-related factor-TSLP, thereby inhibiting the formation of hepatic microvessels and exerting an anti-HCC effect.
Collapse
|
29
|
Rasheduzzaman M, Yin H, Park SY. Cardiac glycoside sensitized hepatocellular carcinoma cells to TRAIL via ROS generation, p38MAPK, mitochondrial transition, and autophagy mediation. Mol Carcinog 2019; 58:2040-2051. [PMID: 31392779 DOI: 10.1002/mc.23096] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/17/2019] [Accepted: 07/26/2019] [Indexed: 12/18/2022]
Abstract
A major concern in the clinical application of tumor necrosis factor related apoptosis-inducing ligand (TRAIL) in tumors is the development of resistance. Therefore, agents that can potentially restore TRAIL sensitivity are important therapeutic targets for cancer treatment. Herein, we evaluated lanatoside c and digoxin, both of which are widely used cardiac glycosides (CGs), for their ability to sensitize human hepatocellular carcinoma cells (Huh-7 and HepG2) through TRAIL-induced apoptosis. CGs functionalize TRAIL as shown by its effect on intracellular reactive oxygen species (ROS) generation, which damages mitochondrial integrity and thereby confers intrinsic apoptotic caspase cascade during combined treatment. Caspase activation is dependent on ROS as shown by the ability of CGs to generate ROS and the ROS-N-acetylcysteine (NAC) relationship, which inhibits apoptosis during cotreatment by preventing the formation of caspase-8 and -3. Furthermore, CGs triggered p38MAPK phosphorylation and NAC pre-exposure blocked p38MAPK phosphorylation, which demonstrated that p38MAPK was dependent upon ROS generation. Additionally, CGs were found to be potent inducers of AMPK-mediated protective autophagy as pharmacological and genetic autophagy inhibition reached the higher threshold of TRAIL-mediated apoptosis. Finally, CGs downregulated the expression of the antiapoptotic protein Bcl-2 and increased the translocation of proapoptotic protein cytochrome c, thereby inducing apoptosis. Collectively, these results indicate that CGs potentiate the enhanced cytotoxic capacity to TRAIL through ROS generation, p38MAPK phosphorylation, cell survival protein downregulation, and protective autophagy inhibition.
Collapse
Affiliation(s)
- Mohammad Rasheduzzaman
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Brisbane, Queensland, Australia.,Biosafety Research Institute, Department of Veterinary Medicine, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, South Korea
| | - Honghua Yin
- Biosafety Research Institute, Department of Veterinary Medicine, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, South Korea
| | - Sang-Youel Park
- Biosafety Research Institute, Department of Veterinary Medicine, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, South Korea
| |
Collapse
|
30
|
Méndez-Blanco C, Fondevila F, García-Palomo A, González-Gallego J, Mauriz JL. Sorafenib resistance in hepatocarcinoma: role of hypoxia-inducible factors. Exp Mol Med 2018; 50:1-9. [PMID: 30315182 PMCID: PMC6185986 DOI: 10.1038/s12276-018-0159-1] [Citation(s) in RCA: 235] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/05/2018] [Accepted: 06/18/2018] [Indexed: 12/14/2022] Open
Abstract
Sorafenib, a multikinase inhibitor with antiproliferative, antiangiogenic, and proapoptotic properties, constitutes the only effective first-line drug approved for the treatment of advanced hepatocellular carcinoma (HCC). Despite its capacity to increase survival in HCC patients, its success is quite low in the long term owing to the development of resistant cells through several mechanisms. Among these mechanisms, the antiangiogenic effects of sustained sorafenib treatment induce a reduction of microvessel density, promoting intratumoral hypoxia and hypoxia-inducible factors (HIFs)-mediated cellular responses that favor the selection of resistant cells adapted to the hypoxic microenvironment. Clinical data have demonstrated that overexpressed HIF-1α and HIF-2α in HCC patients are reliable markers of a poor prognosis. Thus, the combination of current sorafenib treatment with gene therapy or inhibitors against HIFs have been documented as promising approaches to overcome sorafenib resistance both in vitro and in vivo. Because the depletion of one HIF-α subunit elevates the expression of the other HIF-α isoform through a compensatory loop, targeting both HIF-1α and HIF-2α would be a more interesting strategy than therapies that discriminate among HIF-α isoforms. In conclusion, there is a marked correlation between the hypoxic microenvironment and sorafenib resistance, suggesting that targeting HIFs is a promising way to increase the efficiency of treatment. Targeting hypoxia-inducible factors (HIFs), regulatory proteins induced by low oxygen levels, could increase the effectiveness of sorafenib, the only systemic therapy approved for advanced liver cancer. Long-term treatment with sorafenib starves tumors of oxygen, which can lead to the proliferation of cancer cells that are able to survive low oxygen levels. HIFs regulate genes involved in this adaptation and HIF levels are increased in sorafenib-resistant cells. José Mauriz at the University of León, Spain, and colleagues review recent studies on the effects of HIF inhibition on sorafenib efficacy. They conclude that HIF-1α and HIF-2α are predictive markers of sorafenib resistance and that using inhibitors of both these factors as an add-on therapy could improve patient survival. This strategy may be applicable to other types of cancer in which reduced oxygen conditions lead to drug resistance.
Collapse
Affiliation(s)
- Carolina Méndez-Blanco
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Flavia Fondevila
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Andrés García-Palomo
- Institute of Biomedicine, University of León, León, Spain.,Service of Oncology, Complejo Asistencial Universitario de León, León, Spain
| | - Javier González-Gallego
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - José L Mauriz
- Institute of Biomedicine, University of León, León, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain.
| |
Collapse
|
31
|
Morse MA, Sun W, Kim R, He AR, Abada PB, Mynderse M, Finn RS. The Role of Angiogenesis in Hepatocellular Carcinoma. Clin Cancer Res 2018; 25:912-920. [PMID: 30274981 DOI: 10.1158/1078-0432.ccr-18-1254] [Citation(s) in RCA: 385] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/17/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) accounts for about 90% of all primary liver cancers and is the second leading cause of cancer-related deaths worldwide. The hypervascular nature of most HCC tumors underlines the importance of angiogenesis in the pathobiology of these tumors. Several angiogenic pathways have been identified as being dysregulated in HCC, suggesting they may be involved in the development and pathogenesis of HCC. These data provide practical targets for systemic treatments such as those targeting the vascular endothelial growth factor receptor and its ligand. However, the clinical relevance of other more recently identified angiogenic pathways in HCC pathogenesis or treatment remains unclear. Research into molecular profiles and validation of prognostic or predictive biomarkers will be required to identify the patient subsets most likely to experience meaningful benefit from this important class of agents.
Collapse
Affiliation(s)
- Michael A Morse
- Department of Medicine, Division of Medical Oncology, Duke University Health System, Durham, North Carolina.
| | - Weijing Sun
- Division of Medical Oncology, University of Kansas School of Medicine, Kansas City, Kansas
| | - Richard Kim
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Aiwu Ruth He
- Department of Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | | | | | - Richard S Finn
- Department of Medicine, Division of Hematology/Oncology, Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
32
|
Zhang J, Fang C, Qu M, Wu H, Wang X, Zhang H, Ma H, Zhang Z, Huang Y, Shi L, Liang S, Gao Z, Song W, Wang X. CD13 Inhibition Enhances Cytotoxic Effect of Chemotherapy Agents. Front Pharmacol 2018; 9:1042. [PMID: 30258365 PMCID: PMC6144529 DOI: 10.3389/fphar.2018.01042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 08/27/2018] [Indexed: 12/20/2022] Open
Abstract
Multidrug resistance (MDR) of hepatocellular carcinoma is a serious problem. Although CD13 is a biomarker in human liver cancer stem cells, the relationship between CD13 and MDR remains uncertain. This study uses liver cancer cell model to understand the role of CD13 in enhancing the cytotoxic effect of chemotherapy agents. Cytotoxic agents can induce CD13 expression. CD13 inhibitor, bestatin, enhances the antitumor effect of cytotoxic agents. Meanwhile, CD13-targeting siRNA and neutralizing antibody can enhance the cytotoxic effect of 5-fluorouracil (5FU). CD13 overexpression increases cell survival upon cytotoxic agents treatment, while the knockdown of CD13 causes hypersensitivity of cells to cytotoxic agents treatment. Mechanistically, the inhibition of CD13 leads to the increase of cellular reactive oxygen species (ROS). BC-02 is a novel mutual prodrug (hybrid drug) of bestatin and 5FU. Notably, BC-02 can inhibit cellular activity in both parental and drug-resistant cells, accompanied with significantly increased ROS level. Moreover, the survival time of Kunming mice bearing H22 cells under BC-02 treatment is comparable to the capecitabine treatment at maximum dosage. These data implicate a therapeutic method to reverse MDR by targeting CD13, and indicate that BC-02 is a potent antitumor compound.
Collapse
Affiliation(s)
- Jian Zhang
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Chunyan Fang
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Meihua Qu
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Huina Wu
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Xuejuan Wang
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Hongan Zhang
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Hui Ma
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Zhaolin Zhang
- Weifang Bochuang International Biological Medicinal Institute, Weifang, China
| | - Yongxue Huang
- Weifang Bochuang International Biological Medicinal Institute, Weifang, China
| | - Lihong Shi
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Shujuan Liang
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Zhiqin Gao
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Weiguo Song
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Xuejian Wang
- School of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
33
|
Mass spectral analysis of the multikinase inhibitor BZG and its metabolites and analysis of their binding to vascular endothelial growth factor receptor-2. Oncotarget 2018; 8:29951-29962. [PMID: 28415783 PMCID: PMC5444716 DOI: 10.18632/oncotarget.16264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/08/2017] [Indexed: 12/19/2022] Open
Abstract
We previously showed that BZG is a novel multitarget kinase inhibitor, which inhibited hepatocellular carcinoma in vivo and in vitro. In the present study, we used ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC/Q-TOF MS) to characterize BZG and its metabolites generated in vivo. The probable metabolic mechanism was further confirmed by analysis of Phase I and Phase II metabolism in liver microsomes and with recombinant enzymes. In addition, the binding affinities of BZG metabolites to vascular endothelial growth factor receptor 2 (VEGFR2) were predicted using electronic high throughput screening (eHiTS). The results showed that BZG underwent phase I and phase II metabolism. We detected 11 BZG metabolites and identified hydroxylation, glucuronation, acetylation, sulfonation and degradation as the major metabolic processes in vivo and in vitro. Five of the eleven metabolites showed highly favorable eHiTS energy scores that were lower than sorafenib. Knowledge of the in vivo metabolic pathways of BZG and its binding affinities to VEGFR2 will be beneficial for further clinical development of BZG.
Collapse
|
34
|
Liang DY, Huang W, Chang Q, Hou YQ. ShDcR3 sensitizes TRAIL-resistant HCC cells by inducing caspase-dependent apoptosis while suppressing NF-κB dependent cFLIPL expression. PLoS One 2018; 13:e0191545. [PMID: 29444104 PMCID: PMC5812574 DOI: 10.1371/journal.pone.0191545] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 01/08/2018] [Indexed: 02/06/2023] Open
Abstract
Evidence has shown that most hepatocellular carcinoma (HCC) cells are resistant to tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis. However, the molecular mechanisms underlying TRAIL-mediated apoptosis resistance are not well understood. In this study, we reported that downregulation of Decoy receptor 3 (DcR3) expression by lentiviral vectors carrying shRNA against DcR3 (LV-ShDcR3, shDcR3) in Huh7 both greatly enhanced TRAIL-mediated apoptosis and reduced cell proliferation capability. In addition, silencing DcR3 resulted in upregulation of the cell apoptotic regulators including Bid, caspase-3, and caspase-8. Caspase inhibitors inhibited shDcR3-mediated cell death, which indicated that downregulation of DcR3 expression in Huh7 cells increased TRAIL-induced caspase-dependent apoptotic cell death. Furthermore, although the knockdown of DcR3 altered the expression of some Bcl-2- and IAP-family proteins, this change was inhibited by pretreatment with a pancaspase inhibitor, which indicated the cytotoxic effect of shDcR3 was not due to the expression of these proteins. In contrast, shDcR3 significantly inhibited TRAIL-induced transcription factor nuclear κB (NF-κB) activation through the IκB kinase (IKK) pathway, as well as inhibited TRAIL-induced increases in FLICE-inhibitory protein long form (cFLIPL) expression at the transcriptional level. Silencing cFLIPL expression mimicked the cytotoxic effect of shDcR3 on TRAIL-mediated cell apoptosis. Moreover, overexpression of cFLIPL effectively prevented the increase in cell apoptosis in Huh7 cells co-treated with TRAIL and shDcR3. Taken together, our findings indicated that silencing DcR3 sensitizes TRAIL-mediated apoptosis in HCC cells by inhibiting NF-κB.
Collapse
Affiliation(s)
- Dong-Yu Liang
- Department of Central Laboratory, Songjiang Hospital Affiliated First People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
- Department of Central Laboratory, Jiading District Central Hospital Affiliated Shanghai University of Medicine&Health Sciences, Shanghai, China
| | - Wei Huang
- Department of Blood Bank, Jiading District Central Hospital Affiliated Shanghai University of Medicine&Health Sciences, Shanghai, China
| | - Qing Chang
- Department of Central Laboratory, Jiading District Central Hospital Affiliated Shanghai University of Medicine&Health Sciences, Shanghai, China
- * E-mail: (YH); (QC)
| | - Yan-Qiang Hou
- Department of Central Laboratory, Songjiang Hospital Affiliated First People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
- * E-mail: (YH); (QC)
| |
Collapse
|
35
|
Akinyemiju T, Abera S, Ahmed M, Alam N, Alemayohu MA, Allen C, Al-Raddadi R, Alvis-Guzman N, Amoako Y, Artaman A, Ayele TA, Barac A, Bensenor I, Berhane A, Bhutta Z, Castillo-Rivas J, Chitheer A, Choi JY, Cowie B, Dandona L, Dandona R, Dey S, Dicker D, Phuc H, Ekwueme DU, Zaki MES, Fischer F, Fürst T, Hancock J, Hay SI, Hotez P, Jee SH, Kasaeian A, Khader Y, Khang YH, Kumar A, Kutz M, Larson H, Lopez A, Lunevicius R, Malekzadeh R, McAlinden C, Meier T, Mendoza W, Mokdad A, Moradi-Lakeh M, Nagel G, Nguyen Q, Nguyen G, Ogbo F, Patton G, Pereira DM, Pourmalek F, Qorbani M, Radfar A, Roshandel G, Salomon JA, Sanabria J, Sartorius B, Satpathy M, Sawhney M, Sepanlou S, Shackelford K, Shore H, Sun J, Mengistu DT, Topór-Mądry R, Tran B, Ukwaja KN, Vlassov V, Vollset SE, Vos T, Wakayo T, Weiderpass E, Werdecker A, Yonemoto N, Younis M, Yu C, Zaidi Z, Zhu L, Murray CJL, Naghavi M, Fitzmaurice C. The Burden of Primary Liver Cancer and Underlying Etiologies From 1990 to 2015 at the Global, Regional, and National Level: Results From the Global Burden of Disease Study 2015. JAMA Oncol 2017; 3:1683-1691. [PMID: 28983565 PMCID: PMC5824275 DOI: 10.1001/jamaoncol.2017.3055] [Citation(s) in RCA: 1472] [Impact Index Per Article: 184.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/30/2017] [Indexed: 12/12/2022]
Abstract
IMPORTANCE Liver cancer is among the leading causes of cancer deaths globally. The most common causes for liver cancer include hepatitis B virus (HBV) and hepatitis C virus (HCV) infection and alcohol use. OBJECTIVE To report results of the Global Burden of Disease (GBD) 2015 study on primary liver cancer incidence, mortality, and disability-adjusted life-years (DALYs) for 195 countries or territories from 1990 to 2015, and present global, regional, and national estimates on the burden of liver cancer attributable to HBV, HCV, alcohol, and an “other” group that encompasses residual causes. DESIGN, SETTINGS, AND PARTICIPANTS Mortality was estimated using vital registration and cancer registry data in an ensemble modeling approach. Single-cause mortality estimates were adjusted for all-cause mortality. Incidence was derived from mortality estimates and the mortality-to-incidence ratio. Through a systematic literature review, data on the proportions of liver cancer due to HBV, HCV, alcohol, and other causes were identified. Years of life lost were calculated by multiplying each death by a standard life expectancy. Prevalence was estimated using mortality-to-incidence ratio as surrogate for survival. Total prevalence was divided into 4 sequelae that were multiplied by disability weights to derive years lived with disability (YLDs). DALYs were the sum of years of life lost and YLDs. MAIN OUTCOMES AND MEASURES Liver cancer mortality, incidence, YLDs, years of life lost, DALYs by etiology, age, sex, country, and year. RESULTS There were 854 000 incident cases of liver cancer and 810 000 deaths globally in 2015, contributing to 20 578 000 DALYs. Cases of incident liver cancer increased by 75% between 1990 and 2015, of which 47% can be explained by changing population age structures, 35% by population growth, and −8% to changing age-specific incidence rates. The male-to-female ratio for age-standardized liver cancer mortality was 2.8. Globally, HBV accounted for 265 000 liver cancer deaths (33%), alcohol for 245 000 (30%), HCV for 167 000 (21%), and other causes for 133 000 (16%) deaths, with substantial variation between countries in the underlying etiologies. CONCLUSIONS AND RELEVANCE Liver cancer is among the leading causes of cancer deaths in many countries. Causes of liver cancer differ widely among populations. Our results show that most cases of liver cancer can be prevented through vaccination, antiviral treatment, safe blood transfusion and injection practices, as well as interventions to reduce excessive alcohol use. In line with the Sustainable Development Goals, the identification and elimination of risk factors for liver cancer will be required to achieve a sustained reduction in liver cancer burden. The GBD study can be used to guide these prevention efforts.
Collapse
Affiliation(s)
- Tomi Akinyemiju
- School of Public Health, Birmingham, University of Alabama at Birmingham
| | - Semaw Abera
- Mekelle University, School of Public Health, College of Health Sciences, Mekelle, Tigray, Ethiopia
- University of Hohenheim, Institute of Biological Chemistry and Nutrition, Stuttgart, Baden Württemberg, Germany
| | - Muktar Ahmed
- Jimma University Institute of Health, Department of Epidemiology, Jimma, Oromiya, Ethiopia
| | - Noore Alam
- Department of Health, Queensland Government, Herston, QLD, Australia
- University of Queensland, School of Public Health, Herston, QLD, Australia
| | | | - Christine Allen
- University of Washington, Institute for Health Metrics and Evaluation, Seattle
| | | | - Nelson Alvis-Guzman
- Universidad de Cartagena, Grupo de Investigación en Economía de la Salud, Cartagena, Bolivar, Colombia
| | - Yaw Amoako
- Komfo Anokye Teaching Hospital, Department of Medicine, Bantama, Ghana
| | - Al Artaman
- University of Manitoba, Community Health Sciences, Winnipeg, Manitoba, Canada
| | | | - Aleksandra Barac
- Clinical Center of Serbia, Clinic for Infectious and Tropic Diseases, Belgrade, Serbia
| | - Isabela Bensenor
- Hospital Universitário, University of São Paulo Division of Internal Medicine, São Paulo, São Paulo, Brazil
| | - Adugnaw Berhane
- Debre Berhan University, College of Health Sciences, Debre Berhan, Amhara, Ethiopia
| | - Zulfiqar Bhutta
- Aga Khan University, Centre of Excellence in Women & Child, Karachi, Sindh, Pakistan
- The Hospital for Sick Children, Centre for Global Child Health, Toronto, Ontario, Canada
| | - Jacqueline Castillo-Rivas
- Caja Costarricense de Seguro Social, Dirección Actuarial y Economica, San Jose, San Jose, Costa Rica
| | | | - Jee-Young Choi
- Seoul National University, College of Medicine Medical Library, Seoul, South Korea
| | - Benjamin Cowie
- Doherty Institute, WHO Collaborating Centre for Viral Hepatitis, Melbourne, Victoria, Australia
| | - Lalit Dandona
- University of Washington, Institute for Health Metrics and Evaluation, Seattle
- Public Health Foundation of India, Research, Gurgaon, NCR, India
| | - Rakhi Dandona
- University of Washington, Institute for Health Metrics and Evaluation, Seattle
- Public Health Foundation of India, Research, Gurgaon, NCR, India
| | - Subhojit Dey
- Indian Institute of Public Health-Delhi, Environmental and Occupational Health, Gurgaon, Haryana, India
| | - Daniel Dicker
- University of Washington, Institute for Health Metrics and Evaluation, Seattle
| | - Huyen Phuc
- Duy Tan University, Institute for Global Health Innovations, Da Nang, Vietnam
| | - Donatus U. Ekwueme
- Division of Cancer Prevention and Control, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | - Florian Fischer
- Bielefeld University, School of Public Health, Bielefeld, Germany
| | - Thomas Fürst
- Swiss Tropical and Public Health Institute, Epidemiology and Public Health, Basel, Switzerland
- University of Basel, Switzerland
- Imperial College London, School of Public Health, London, England
| | - Jamie Hancock
- University of Washington, Institute for Health Metrics and Evaluation, Seattle
| | - Simon I. Hay
- University of Washington, Institute for Health Metrics and Evaluation, Seattle
| | - Peter Hotez
- Baylor College of Medicine, National School of Tropical Medicine, Houston, Texas
- Sabin Vaccine Institute & Texas Children's Hospital Center for Vaccine Development, Houston
| | - Sun Ha Jee
- Graduate School of Public Health, Yonsei University, Epidemiology and Health Promotion, Seoul, South Korea
| | - Amir Kasaeian
- Tehran University of Medical Sciences, Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran, Tehran, Iran
| | - Yousef Khader
- Jordan University of Science and Technology, Public Health, Irbid, Irbid, Jordan
| | - Young-Ho Khang
- Seoul National University College of Medicine, Institute of Health Policy and Management, Seoul, Seoul Metropolitan City, South Korea
| | - Anil Kumar
- Public Health Foundation of India Research, Gurgaon (NCR), Haryana, India
| | - Michael Kutz
- University of Washington, Institute for Health Metrics and Evaluation, Seattle
| | - Heidi Larson
- Department Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, England
| | - Alan Lopez
- University of Washington, Institute for Health Metrics and Evaluation, Seattle
- University of Melbourne, Melbourne School of Population and Global Health, Melbourne, VIC, Australia
| | - Raimundas Lunevicius
- Aintree University Hospital NHS Foundation Trust, General Surgery Department, Liverpool, England
- School of Medicine, University of Liverpool, Liverpool, England
| | - Reza Malekzadeh
- Tehran University of Medical Sciences, Digestive Diseases Research Institute, Tehran, Tehran, Iran
| | - Colm McAlinden
- University Hospitals Bristol, Department of Medicine, Bristol, England
| | - Toni Meier
- Martin Luther University Halle-Wittenberg, Institute for Agricultural and Nutritional Sciences, Halle (Saale), Germany
| | | | - Ali Mokdad
- University of Washington, Institute for Health Metrics and Evaluation, Seattle
| | - Maziar Moradi-Lakeh
- Iran University of Medical Sciences, Gastrointestinal and Liver Disease Research Center, Tehran, Tehran, Iran
- Iran University of Medical Sciences, Preventive Medicine and Public Health Research Center, Tehran, Tehran, Iran
| | - Gabriele Nagel
- Ulm University, Institute of Epidemiology and Medical Biometry, Ulm, Germany
| | - Quyen Nguyen
- Duy Tan University, Institute for Global Health Innovations, Da Nang, Vietnam
| | - Grant Nguyen
- University of Washington, Institute for Health Metrics and Evaluation, Seattle
| | - Felix Ogbo
- Western Sydney University, Centre for Health Research, School of Medicine, Penrith, NSW, Australia
- Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
| | - George Patton
- University of Melbourne, Paediatrics, Melbourne, Victoria, Australia
| | - David M. Pereira
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Farshad Pourmalek
- Department of Urology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mostafa Qorbani
- Alborz University of Medical Sciences, Noncommunicable Diseases Research Center, Karaj, Alborz, Iran
| | - Amir Radfar
- A. T. Still University, College of Graduate Health Studies, Mesa, Arizona
| | - Gholamreza Roshandel
- Golestan University of Medical Sciences, Golestan Research Center of Gastroenterology and Hepatology, Gorgan, Iran
| | - Joshua A Salomon
- Harvard T.H. Chan School of Public Health, Department of Global Health and Population, Boston, Massachusetts
| | - Juan Sanabria
- Marshall University School of Medicine, Surgery, Huntington, West Virginia
- Case Western Reserve University, Nutrition and Preventive Medicine, Ohio
| | - Benn Sartorius
- University of KwaZulu-Natal, Public Health Medicine, Durban, KwaZulu-Natal, South Africa
| | - Maheswar Satpathy
- Utkal University, Centre for Advanced Study in Psychology, Bhubaneswar, Odisha, India
- AIIMS New Delhi, JPN Apex Trauma Centre, New Delhi, Delhi, India
| | - Monika Sawhney
- Marshall University Public Health, Huntington, West Virginia
| | - Sadaf Sepanlou
- Tehran University of Medical Sciences, Digestive Diseases Research Institute, Tehran, Tehran, Iran
| | - Katya Shackelford
- University of Washington, Institute for Health Metrics and Evaluation, Seattle
| | - Hirbo Shore
- Haramaya University School of Public Health, Harari, Harari, Ethiopia
| | - Jiandong Sun
- Queensland University of Technology, School of Public Health and Social Work, Brisbane, Queensland, Australia
| | | | - Roman Topór-Mądry
- Faculty of Health Sciences Jagiellonian University Medical College, Institute of Public Health, Kraków, Poland
- Faculty of Health Sciences Wroclaw Medical University, Wroclaw, Poland
| | - Bach Tran
- Hanoi Medical University, Institute for Preventive Medicine and Public Health, Hanoi, Vietnam
- Johns Hopkins University, Bloomberg School of Public Health, Baltimore, Maryland
| | | | - Vasiliy Vlassov
- Department of Health Care Administration and Economy, National Research University Higher School of Economics, Moscow, Russia
| | - Stein Emil Vollset
- Norwegian Institute of Public Health, Centre for Disease Burden, Bergen, Norway
- University of Bergen, Department of Global Public Health and Primary Care, Bergen, Norway
| | - Theo Vos
- University of Washington, Institute for Health Metrics and Evaluation, Seattle
| | - Tolassa Wakayo
- Jimma University, Population and Family Health, Oromia, Ethiopia
| | - Elisabete Weiderpass
- Cancer Registry of Norway, Institute of Population Based Cancer Research, Oslo, Norway
- University of Tromsø, The Arctic University of Norway, Department of Community Medicine, Faculty of Health Sciences, Tromsø, Norway
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Andrea Werdecker
- Federal Institute for Population Research, Competence Center Mortality-Follow-Up of the National Cohort, Wiesbaden, Hesse, Germany
| | - Naohiro Yonemoto
- Kyoto University, School of Public Health Biostatistics, Sakyo, Kyoto, Japan
| | - Mustafa Younis
- Jackson State University, Health Policy & Management, Jackson, Mississippi
- Harvard Asia Aging Center, Harvard Medical School, Boston, Massachuetts
| | - Chuanhua Yu
- Department of Epidemiology and Biostatistics, Wuhan University, Wuhan, Hubei Province, China
| | - Zoubida Zaidi
- Department of Epidemiology, University Hospital of Setif, Setif, Algeria
- University Ferhat Abbas, Faculty of Medicine, Setif, Algeria
| | - Liguo Zhu
- Jiangsu Provincial Center for Disease Control and Prevention, Major Project Execution Office, Nanjing, Jiangsu, China
| | | | - Mohsen Naghavi
- University of Washington, Institute for Health Metrics and Evaluation, Seattle
| | - Christina Fitzmaurice
- University of Washington, Institute for Health Metrics and Evaluation, Seattle
- Division of Hematology, Department of Medicine, University of Washington, Seattle
| |
Collapse
|
36
|
Kohorst MA, Warad DM, Matsumoto JM, Heimbach JK, El-Youssef M, Arndt CAS, Rodriguez V, Nageswara Rao AA. Management of pediatric hepatocellular carcinoma: A multimodal approach. Pediatr Transplant 2017. [PMID: 28631359 DOI: 10.1111/petr.13007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
HCC is rare in the pediatric population, but is the second most common liver malignancy in children. Survival rates for primary unresectable HCC have been dismal. The objective of this study was to describe our experience with a multimodal approach for the management of unresectable HCC in two adolescent patients and to review the literature. Both patients are currently alive with no recurrence at 51 and 29 months post-transplant. Multimodality treatment involving chemotherapy with doxorubicin, cisplatin, and sorafenib; TACE; timely liver transplantation; and post-transplant therapy with sorafenib and mTOR inhibitors may help improve outcomes and prolong survival in pediatric patients with unresectable HCC.
Collapse
Affiliation(s)
- Mira A Kohorst
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Hematology and Oncology, Mayo Clinic, Rochester, MN, USA
| | - Deepti M Warad
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Hematology and Oncology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Mounif El-Youssef
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Gastroenterology, Mayo Clinic, Rochester, MN, USA
| | - Carola A S Arndt
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Hematology and Oncology, Mayo Clinic, Rochester, MN, USA
| | - Vilmarie Rodriguez
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Hematology and Oncology, Mayo Clinic, Rochester, MN, USA
| | - Amulya A Nageswara Rao
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Hematology and Oncology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
37
|
Liu X, Xu J, Wang S, Yu X, Kou B, Chai M, Zang Y, Chen D. Synergistic inhibitory effects on hepatocellular carcinoma with recombinant human adenovirus Aspp2 and oxaliplatin via p53-independent pathway in vitro and in vivo. Int J Oncol 2017; 51:1291-1299. [PMID: 28902369 DOI: 10.3892/ijo.2017.4105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/26/2017] [Indexed: 11/06/2022] Open
Abstract
The present study was designed to investigate the synergistic inhibitory effects on hepatocellular carcinoma with recombinant human adenovirus Aspp2 (Aspp2-ad) and oxaliplatin via p53-independent pathway in vitro and in vivo. After being treated with Aspp2-ad and/or oxaliplatin for 24-48 h, HepG2P53-/- and Hep3B cells showed a significant growth inhibition compared with vehicle control. Combination group showed a synergetic effect, the inhibitory rates were all above 80% at 48 h point in HepG2P53-/- and Hep3B cells. The apoptotic cell numbers of Aspp2-ad and/or oxaliplatin treatment groups were increased remarkably, especially for the combined therapy group in the liver cancer cells. The Hep3B xenograft experiment also showed similar inhibition of Aspp2-ad and/or oxaliplatin to the in vitro experiment. H&E results showed that combination group had the least mitotic indexes and the most necrosis. The immunohistochemistry results showed that PCNA, CD31 expression decreased greatly in treatment groups. These results suggested that Aspp2-ad might inhibit proliferation and vascular growth of hepatocarcinoma. Aspp2 induced apoptosis protein expression in Aspp2-ad and combination groups, the Aspp2, Bax and activation of caspase-3 expression increased greatly both in vitro and in vivo. But interestingly, the autophagy proteins showed different responses not only in HepG2P53-/- and Hep3B cells but also in vitro and in vivo. We found that Aspp2-ad downregulated the p-ERK, p-STAT3 expression, the synergistic effects were observed in combination group, while there was not response of mTOR to Aspp2-ad. In conclusion, Aspp2-ad, in P53-independent manner, regulated ERK and STAT3 signal moleculars to inhibit hepatocarcinoma in coordination with oxaliplatin by influencing the protein expression of proliferation, apoptosis, autophagy and vascular growth. Aspp2-ad has the potential to be developed in gene therapy for HCC, especially for P53 deletion or mutation in HCC.
Collapse
Affiliation(s)
- Xiaoni Liu
- Beijing Institute of Hepatology and Beijing YouAn Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Jianji Xu
- Beijing Institute of Hepatology and Beijing YouAn Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Shuang Wang
- Beijing Institute of Hepatology and Beijing YouAn Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Xiaoxiao Yu
- Beijing Institute of Hepatology and Beijing YouAn Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Boxin Kou
- Beijing Institute of Hepatology and Beijing YouAn Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Mengyin Chai
- Beijing Institute of Hepatology and Beijing YouAn Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Yunjin Zang
- Beijing Institute of Hepatology and Beijing YouAn Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Dexi Chen
- Beijing Institute of Hepatology and Beijing YouAn Hospital, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
38
|
Takahashi K, Putchakayala KG, Safwan M, Kim DY. Extrahepatic metastasis of hepatocellular carcinoma to the paravertebral muscle: A case report. World J Hepatol 2017; 9:973-978. [PMID: 28839518 PMCID: PMC5550763 DOI: 10.4254/wjh.v9.i22.973] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/29/2017] [Accepted: 05/18/2017] [Indexed: 02/06/2023] Open
Abstract
Identification of extrahepatic metastases (EHM) of hepatocellular carcinoma (HCC) has been paradoxically increasing due to an increase in the survival of HCC patients. However, metastasis of HCC to the skeletal muscle tissue is extremely rare. We describe a unique case of HCC metastasizing to the paravertebral muscle. A 55-year-old man with a history of hepatitis B cirrhosis underwent partial liver resection with complete removal of HCC. Three months later, a computed tomography (CT) scan showed intrahepatic recurrence. The tumors were treated with yttrium-90 microspheres, trans-catheter arterial chemoembolization, and sorafenib. Six months later, a CT scan showed an enhancing lesion of the left paravertebral muscle that on biopsy were consistent with metastatic HCC. The tumor was treated with stereotactic hypo-fractionated image-guided radiation therapy (SHFRT). A follow-up scan 3 mo post-radiotherapy revealed a stable appearance of the paravertebral muscle metastasis. Because of the progression in the intrahepatic tumors, the patient was treated with capecitabine, which was changed to dasatinib 6 mo later. The patient passed away three years after the primary surgical resection. Management of EHM poses an extreme challenge. This is the first case of HCC with EHM to the paravertebral muscle in which stability of disease was achieved using SHFRT. This case highlights the importance of early detection of hepatitis B viral infection and initiation of anti-viral therapy to decrease recurrence of HCC and prevent EHM.
Collapse
Affiliation(s)
- Kazuhiro Takahashi
- Transplant and Hepatobiliary Surgery, Henry Ford Hospital, Detroit, MI 48202, United States
| | - Krishna G Putchakayala
- Transplant and Hepatobiliary Surgery, Henry Ford Hospital, Detroit, MI 48202, United States
| | - Mohamed Safwan
- Transplant and Hepatobiliary Surgery, Henry Ford Hospital, Detroit, MI 48202, United States
| | - Dean Y Kim
- Transplant and Hepatobiliary Surgery, Henry Ford Hospital, Detroit, MI 48202, United States.
| |
Collapse
|
39
|
Zhou DY, Qin J, Huang J, Wang F, Xu GP, Lv YT, Zhang JB, Shen LM. Zoledronic acid inhibits infiltration of tumor-associated macrophages and angiogenesis following transcatheter arterial chemoembolization in rat hepatocellular carcinoma models. Oncol Lett 2017; 14:4078-4084. [PMID: 28943915 PMCID: PMC5592878 DOI: 10.3892/ol.2017.6717] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 02/03/2017] [Indexed: 12/13/2022] Open
Abstract
Hepatic transcatheter arterial chemoembolization (TACE), a minimally invasive procedure to block the blood supply of tumors and release of cytotoxic agents, is preferentially applied to patients with hepatocellular carcinoma (HCC) who are not able to receive radical treatments. However, the long-term effects of TACE are unsatisfactory, as the microenvironment following procedure stimulates tumor angiogenesis, which promotes recurrence and metastasis of residual tumors. Tumor associated macrophages (TAMs) have been revealed to stimulate tumor growth and angiogenesis associated with poor prognosis in HCC. The present study focused on the changes in TAMs following TACE, and explored the effects of TACE in combination with the TAM inhibitor zoledronic acid (ZA) in rat HCC models. Orthotropic HCC rats were divided into three groups: Sham TACE, TACE alone and TACE combined with ZA treatment. At 7 or 14 days following TACE, tumor growth was evaluated by magnetic resonance imaging (MRI). Infiltration of TAMs was assessed by histological analysis and flow cytometry. Tumor angiogenesis was measured as the mean vessel density, and initial slope was calculated from dynamic contrast enhancement MRI. Local and systemic levels of vascular endothelial growth factor (VEGF) were determined by western blotting or an ELISA, respectively. The results revealed that TACE inhibited tumor growth at 7 days following the procedure, but this inhibition was attenuated at 14 days following the procedure compared with the sham TACE control. If combined with ZA treatment, TACE exhibited a stable inhibition effect on tumor growth until the end of observation. Investigation of the underlying mechanisms demonstrated that TACE combined with ZA treatment inhibited infiltration of F4/80 positive TAMs and tumor angiogenesis compared with the TACE alone group at 14 days following the procedure. Additionally, the combination treatment significantly inhibited secretion of VEGF in the present models. In conclusion, ZA treatment enhanced the effects of TACE through inhibiting TAM infiltration and tumor angiogenesis in rat HCC models.
Collapse
Affiliation(s)
- Da-Yong Zhou
- Department of Interventional Radiology and Vascular Surgery, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Jie Qin
- Department of Anatomy, Histology and Embryology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Jian Huang
- Department of Interventional Radiology and Vascular Surgery, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Feng Wang
- Department of Pathology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Guo-Peng Xu
- Department of Respiration, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Yan-Tian Lv
- Department of Respiration, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Ji-Bin Zhang
- Department of Radiology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Li-Ming Shen
- Department of Interventional Radiology and Vascular Surgery, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| |
Collapse
|
40
|
Neuzillet C, de Mestier L, Rousseau B, Mir O, Hebbar M, Kocher HM, Ruszniewski P, Tournigand C. Unravelling the pharmacologic opportunities and future directions for targeted therapies in gastro-intestinal cancers part 2: Neuroendocrine tumours, hepatocellular carcinoma, and gastro-intestinal stromal tumours. Pharmacol Ther 2017; 181:49-75. [PMID: 28723416 DOI: 10.1016/j.pharmthera.2017.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Until the 1990s, cytotoxic chemotherapy has been the cornerstone of medical therapy for gastrointestinal (GI) cancers. Better understanding of the cancer cell molecular biology has led to the therapeutic revolution of targeted therapies, i.e. monoclonal antibodies or small molecule inhibitors directed against proteins that are specifically overexpressed or mutated in cancer cells. These agents, being more specific to cancer cells, were expected to be less toxic than conventional cytotoxic agents. However, their effects have sometimes been disappointing, due to intrinsic or acquired resistance mechanisms, or to an activity restricted to some tumour settings, illustrating the importance of patient selection and early identification of predictive biomarkers of response to these therapies. Targeted agents have provided clinical benefit in many GI cancer types. Particularly, some GI tumours are considered chemoresistant and targeted therapies have offered a new therapeutic base for their management. Hence, somatostatin receptor-directed strategies, sorafenib, and imatinib have revolutioned the management of neuroendocrine tumours (NET), hepatocellular carcinoma (HCC), and gastrointestinal stromal tumours (GIST), respectively, and are now used as first-line treatment in many patients affected by these tumours. However, these agents face problems of resistances and identification of predictive biomarkers from imaging and/or biology. We propose a comprehensive two-part review providing a panoramic approach of the successes and failures of targeted agents in GI cancers to unravel the pharmacologic opportunities and future directions for these agents in GI oncology. In this second part, we will focus on NET, HCC, and GIST, whose treatment relies primarily on targeted therapies.
Collapse
Affiliation(s)
- Cindy Neuzillet
- INSERM UMR1149, Beaujon University Hospital (Assistance Publique-Hôpitaux de Paris, AP-HP), Paris 7 Diderot University, 100 Boulevard du Général Leclerc, 92110 Clichy, France; Department of Medical Oncology, Henri Mondor University Hospital (AP-HP), Paris Est Créteil University (UPEC), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France; Tumour Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom; Barts and The London HPB Centre, The Royal London Hospital, Whitechapel, London E1 1BB, United Kingdom.
| | - Louis de Mestier
- INSERM UMR1149, Beaujon University Hospital (Assistance Publique-Hôpitaux de Paris, AP-HP), Paris 7 Diderot University, 100 Boulevard du Général Leclerc, 92110 Clichy, France; Department of Gastroenterology and Pancreatology, Beaujon University Hospital (AP-HP), Paris 7 Diderot University, 100 Boulevard du Général Leclerc, 92110 Clichy, France
| | - Benoît Rousseau
- Department of Medical Oncology, Henri Mondor University Hospital (AP-HP), Paris Est Créteil University (UPEC), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France; Institut Mondor de Recherche Biomédicale, INSERM UMR955 Team 18, Paris Est Créteil University (UPEC), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Olivier Mir
- Department of Cancer Medicine - Sarcoma Group, Department of Early Drug Development (DITEP) - Phase 1 Unit, Gustave Roussy Cancer Campus, University of Paris Sud, 114, Rue Edouard Vaillant, 94800 Villejuif, France
| | - Mohamed Hebbar
- Department of Medical Oncology, Lille University Hospital, 1, Rue Polonovski, 59037 Lille, France
| | - Hemant M Kocher
- Tumour Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom; Barts and The London HPB Centre, The Royal London Hospital, Whitechapel, London E1 1BB, United Kingdom
| | - Philippe Ruszniewski
- INSERM UMR1149, Beaujon University Hospital (Assistance Publique-Hôpitaux de Paris, AP-HP), Paris 7 Diderot University, 100 Boulevard du Général Leclerc, 92110 Clichy, France
| | - Christophe Tournigand
- Department of Medical Oncology, Henri Mondor University Hospital (AP-HP), Paris Est Créteil University (UPEC), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| |
Collapse
|
41
|
Dahlmanns M, Yakubov E, Chen D, Sehm T, Rauh M, Savaskan N, Wrosch JK. Chemotherapeutic xCT inhibitors sorafenib and erastin unraveled with the synaptic optogenetic function analysis tool. Cell Death Discov 2017; 3:17030. [PMID: 28835855 PMCID: PMC5541984 DOI: 10.1038/cddiscovery.2017.30] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 04/23/2017] [Indexed: 01/19/2023] Open
Abstract
In the search for new potential chemotherapeutics, the compounds’ toxicity to healthy cells is an important factor. The brain with its functional units, the neurons, is especially endangered during the radio- and chemotherapeutic treatment of brain tumors. The effect of the potential compounds not only on neuronal survival but also neuronal function needs to be taken into account. Therefore, in this study we aimed to comprehend the biological effects of chemotherapeutic xCT inhibition on healthy neuronal cells with our synaptic optogenetic function analysis tool (SOFA). We combined common approaches, such as investigation of morphological markers, neuronal function and cell metabolism. The glutamate-cystine exchanger xCT (SLC7A11, system Xc−) is the main glutamate exporter in malignant brain tumors and as such a relevant drug target for treating deadly glioblastomas (WHO grades III and IV). Recently, two small molecules termed sorafenib (Nexavar) and erastin have been found to efficiently block xCT function. We investigated neuronal morphology, metabolic secretome profiles, synaptic function and cell metabolism of primary hippocampal cultures (containing neurons and glial cells) treated with sorafenib and erastin in clinically relevant concentrations. We found that sorafenib severely damaged neurons already after 24 h of treatment. Noteworthy, also at a lower concentration, where no morphological damage or metabolic disturbance was monitored, sorafenib still interfered with synaptic and metabolic homeostasis. In contrast, erastin-treated neurons displayed mostly inconspicuous morphology and metabolic rates. Key parameters of proper neuronal function, such as synaptic vesicle pool sizes, were however disrupted following erastin application. In conclusion, our data revealed that while sorafenib and erastin effectively inhibited xCT function they also interfered with essential neuronal (synaptic) function. These findings highlight the particular importance of investigating the effects of potential neurooncological and general cancer chemotherapeutics also on healthy neuronal cells and their function as revealed by the SOFA tool.
Collapse
Affiliation(s)
- Marc Dahlmanns
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Eduard Yakubov
- Translational Neurooncology Laboratory, Department of Neurosurgery, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany.,Paracelsus Medical University, Nuremberg, Germany
| | - Daishi Chen
- Translational Neurooncology Laboratory, Department of Neurosurgery, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Tina Sehm
- Translational Neurooncology Laboratory, Department of Neurosurgery, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Nicolai Savaskan
- Translational Neurooncology Laboratory, Department of Neurosurgery, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany.,BiMECON Ent., Berlin, Germany
| | - Jana Katharina Wrosch
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
42
|
Gao L, Wang L, Sun Z, Li H, Wang Q, Yi C, Wang X. Morusin shows potent antitumor activity for human hepatocellular carcinoma in vitro and in vivo through apoptosis induction and angiogenesis inhibition. Drug Des Devel Ther 2017; 11:1789-1802. [PMID: 28670112 PMCID: PMC5481341 DOI: 10.2147/dddt.s138320] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most aggressive cancers with high mortality worldwide. Research and development of novel agents for HCC therapy is in demand, urgently. Morusin has been reported to exhibit potential cytotoxic activity in several cancer cell lines. However, whether it has potential antiangiogenic activity especially in HCC remains unclear. In the current study, we found that morusin exerted growth inhibition effects on human HCC cells (HepG2 and Hep3B) in vitro and human HCC cell (HepG2) xenografts in vivo. Moreover, apoptosis induction was observed in a dose-dependent manner after morusin treatment along with an increase in the expression of active caspase-3 and the Bax/Bcl-2 expression ratio. More importantly, morusin inhibited proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs) in vitro and downregulated angiogenic proteins in HCC cells and HUVECs. In vivo, tumor angiogenesis was also attenuated after morusin treatment. In addition, morusin suppressed constitutive as well as IL-6-induced STAT3 phosphorylation in HCC cells and corresponding tumor tissues. Overall, morusin has a potential anticancer effect on human HCC cells in vitro and in vivo by inducing apoptosis and inhibiting anti-angiogenesis. The corresponding mechanism might be associated with the attenuation of the IL-6/STAT3 signaling pathway. Morusin might serve as a promising novel anticancer agent in HCC therapy, and requires further study.
Collapse
Affiliation(s)
| | - Li Wang
- Laboratory of Lung Cancer, Lung Cancer Center
| | - Zhen Sun
- Laboratory of Experimental Oncology, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, People's Republic of China
| | - Haiyan Li
- Laboratory of Experimental Oncology, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, People's Republic of China
| | - Qiaoping Wang
- Laboratory of Experimental Oncology, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, People's Republic of China
| | | | - Xiujie Wang
- Laboratory of Experimental Oncology, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
43
|
TIP30 regulates lipid metabolism in hepatocellular carcinoma by regulating SREBP1 through the Akt/mTOR signaling pathway. Oncogenesis 2017; 6:e347. [PMID: 28604762 PMCID: PMC5519197 DOI: 10.1038/oncsis.2017.49] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 03/18/2017] [Accepted: 05/02/2017] [Indexed: 12/24/2022] Open
Abstract
Lipid reprogramming has been considered as a crucial characteristic in hepatocellular carcinoma (HCC) initiation and progression. However, detailed molecular mechanisms have yet to be clearly defined. Here, we examined the effects of tumor suppressor TIP30 on the regulation of HCC lipid metabolism. We found that decreased TIP30 expression leads to elevated fatty acid synthesis and enhanced levels of lipogenic enzymes SCD and FASN in HCC cells. Moreover, SREBP1 is one of the key transcription factors regulating liver lipid metabolism, and TIP30 deficiency significantly increased SREBP1 expression and nuclear accumulation. Small interfering RNAs targeting SREBP1 could reverse fatty acid synthesis induced by TIP30 deficiency. Furthermore, downregulating TIP30 activated the Akt/mTOR signaling pathway to upregulate SREBP1 expression, which promoted lipid metabolism by activating gene transcription of lipogenesis, including fasn and scd. We also showed that TIP30 deficiency-regulated lipid metabolism promoted proliferation of HCC cells. Clinically, our data revealed that TIP30 expression significantly correlated with SREBP1 in patients with HCC and that a combination of TIP30 and SREBP1 is a powerful predictor of HCC prognosis. Together, our data suggested a novel function of TIP30 in HCC progression and indicate that TIP30 regulation of SREBP1 may represent a novel target for HCC treatment.
Collapse
|
44
|
Quantification of hepatocellular carcinoma heterogeneity with multiparametric magnetic resonance imaging. Sci Rep 2017; 7:2452. [PMID: 28550313 PMCID: PMC5446396 DOI: 10.1038/s41598-017-02706-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022] Open
Abstract
Tumour heterogeneity poses a significant challenge for treatment stratification. The goals of this study were to quantify heterogeneity in hepatocellular carcinoma (HCC) using multiparametric magnetic resonance imaging (mpMRI), and to report preliminary data correlating quantitative MRI parameters with advanced histopathology and gene expression in a patient subset. Thirty-two HCC patients with 39 HCC lesions underwent mpMRI including diffusion-weighted imaging (DWI), blood-oxygenation-level-dependent (BOLD), tissue-oxygenation-level-dependent (TOLD) and dynamic contrast-enhanced (DCE)-MRI. Histogram characteristics [central tendency (mean, median) and heterogeneity (standard deviation, kurtosis, skewness) MRI parameters] in HCC and liver parenchyma were compared using Wilcoxon signed-rank tests. Histogram data was correlated between MRI methods in all patients and with histopathology and gene expression in 14 patients. HCCs exhibited significantly higher intra-tissue heterogeneity vs. liver with all MRI methods (P < 0.030). Although central tendency parameters showed significant correlations between MRI methods and with each of histopathology and gene expression, heterogeneity parameters exhibited additional complementary correlations between BOLD and DCE-MRI and with histopathologic hypoxia marker HIF1α and gene expression of Wnt target GLUL, pharmacological target FGFR4, stemness markers EPCAM and KRT19 and immune checkpoint PDCD1. Histogram analysis combining central tendency and heterogeneity mpMRI features is promising for non-invasive HCC characterization on the imaging, histologic and genomics levels.
Collapse
|
45
|
Abstract
Hepatocellular carcinoma (HCC), also called malignant hepatoma, is one of the deadliest cancers due to its complexities, reoccurrence after surgical resection, metastasis and heterogeneity. Incidence and mortality of HCC are increasing in Western countries and are expected to rise as a consequence of the obesity epidemic. Multiple factors trigger the initiation and progression of HCC including chronic alcohol consumption, viral hepatitis B and C infection, metabolic disorders and age. Although Sorafenib is the only FDA approved drug for the treatment of HCC, numerous treatment modalities such as transcatheter arterial chemoembolization/transarterial chemoembolization (TACE), radiotherapy, locoregional therapy and chemotherapy have been tested in the clinics. Polymeric nanoparticles, liposomes, and micelles carrying small molecules, proteins, peptides and nucleic acids have attracted great attention for the treatment of various cancers including HCC. Herein, we discuss the pathogenesis of HCC in relation to its various recent treatment methodologies using nanodelivery of monoclonal antibodies (mAbs), small molecules, miRNAs and peptides. Synopsis of recent clinical trials of mAbs and peptide drugs has been presented with a broad overview of the pathogenesis of the disease and treatment efficacy.
Collapse
Affiliation(s)
- Rinku Dutta
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States.
| |
Collapse
|
46
|
Liu Y, Gao F, Song W. Periostin contributes to arsenic trioxide resistance in hepatocellular carcinoma cells under hypoxia. Biomed Pharmacother 2017; 88:342-348. [PMID: 28119236 DOI: 10.1016/j.biopha.2017.01.052] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 01/09/2017] [Accepted: 01/09/2017] [Indexed: 01/23/2023] Open
Abstract
Hypoxia has been suggested to induce chemoresistance in tumor cells. In this study, we aimed to test the hypothesis that hypoxia-inducible factor-1alpha (HIF-1α)/periostin axis might promote arsenic trioxide resistance in hepatocellular carcinoma (HCC) cells under hypoxia. HCC cells were exposed to hypoxia and measured for periostin expression. Loss-of-function studies were done to assess the role of periostin in arsenic trioxide resistance. In vivo xenograft mouse studies were performed to determine the effect of periostin silencing on HCC susceptibility to arsenic trioxide. It was found that periostin expression was significantly increased in SMMC7721 and Hep3B HCC cells after hypoxic treatment. Depletion of HIF-1α blocked the upregulation of periostin induced by hypoxia. HCC cells under hypoxia displayed more resistant to arsenic trioxide than those under normoxia. Interestingly, downregulation of periostin re-sensitized hypoxic SMMC7721 and Hep3B cells to arsenic trioxide, which was accompanied by increased apoptosis. Luciferase reporter assay revealed that periostin overexpression enhanced HIF-1α-dependent transcriptional activity and induced the expression of vascular endothelial growth factor, Mcl-1, and Bcl-xL in SMMC7721 cells. Administration of arsenic trioxide resulted in a significant inhibition of SMMC7721 tumor growth. Notably, downregulation of periostin significantly enhanced the anticancer effect of arsenic trioxide against SMMC7721 tumors and reduced the percentage of Ki-67-positive proliferating cells. Taken together, periostin contributes to arsenic trioxide resistance in HCC under hypoxic microenvironment, which is likely associated with promotion of HIF-1α-dependent activation of survival genes. Targeting periostin may represent a promising strategy to improve arsenic trioxide-based anticancer therapy against HCC.
Collapse
Affiliation(s)
- Yujin Liu
- Department of Interventional Radiology, Yueyang Hospital of Integrated Traditional Chinese & Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Feng Gao
- Department of Interventional Radiology, Yueyang Hospital of Integrated Traditional Chinese & Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weixiang Song
- Department of Interventional Radiology, Yueyang Hospital of Integrated Traditional Chinese & Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
47
|
Pan JX, Wang F, Ye LY. Doxorubicin-induced epithelial–mesenchymal transition through SEMA 4A in hepatocellular carcinoma. Biochem Biophys Res Commun 2016; 479:610-614. [DOI: 10.1016/j.bbrc.2016.09.167] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 09/30/2016] [Indexed: 12/22/2022]
|