1
|
He Q, He W, Dong H, Guo Y, Yuan G, Shi X, Wang D, Lu F. Role of liver sinusoidal endothelial cell in metabolic dysfunction-associated fatty liver disease. Cell Commun Signal 2024; 22:346. [PMID: 38943171 PMCID: PMC11214243 DOI: 10.1186/s12964-024-01720-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 06/20/2024] [Indexed: 07/01/2024] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) are highly specialized endothelial cells that represent the interface between blood cells on one side and hepatocytes on the other side. LSECs not only form a barrier within the hepatic sinus, but also play important physiological functions such as regulating hepatic vascular pressure, anti-inflammatory and anti-fibrotic. Pathologically, pathogenic factors can induce LSECs capillarization, that is, loss of fenestra and dysfunction, which are conducive to early steatosis, lay the foundation for the progression of metabolic dysfunction-associated fatty liver disease (MAFLD), and accelerate metabolic dysfunction-associated steatohepatitis (MASH) and liver fibrosis. The unique localization, phenotype, and function of LSECs make them potential candidates for reducing liver injury, inflammation, and preventing or reversing fibrosis in the future.
Collapse
Affiliation(s)
- Qiongyao He
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wu He
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Hui Dong
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yujin Guo
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gang Yuan
- Department of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaoli Shi
- Department of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dingkun Wang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Fuer Lu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
2
|
Arvanitakis K, Papadakos SP, Vakadaris G, Chatzikalil E, Stergiou IE, Kalopitas G, Theocharis S, Germanidis G. Shedding light on the role of LAG-3 in hepatocellular carcinoma: unraveling immunomodulatory pathways. HEPATOMA RESEARCH 2024. [DOI: 10.20517/2394-5079.2024.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Hepatocellular carcinoma (HCC) stands as a primary malignant liver tumor characterized by chronic inflammation and complex alterations within the tumor microenvironment (TME). Lymphocyte activation gene 3 (LAG-3), also known as CD223, has gained prominence as a potential next-generation immune checkpoint, maintaining continuous expression in response to persistent antigen exposure within the TME, warranting our attention. In patients with HCC, LAG-3 expression on T cells, regulatory T cells (Tregs), and natural killer (NK) cells contributes to immune evasion, while high expression of LAG-3 leads to increased angiogenesis and poor prognosis. By interacting with major histocompatibility complex class II molecules, LAG-3 promotes T cell exhaustion and suppresses antitumor responses, often in collaboration with other immune checkpoints like programmed cell death protein 1 (PD-1), while on Tregs and NK cells, LAG-3 modulates their suppressive functions, indirectly facilitating tumor immune escape. LAG-3 expression may offer prognostic insights, correlating with disease progression and outcomes in HCC patients, while various preclinical studies highlight the potential of LAG-3-targeted therapies in reinvigorating immune responses against HCC, with a few combination approaches targeting LAG-3 alongside other checkpoints demonstrating synergistic effects in restoring T cell function. Therefore, harnessing LAG-3 as a therapeutic target holds promise for enhancing antitumor immunity and potentially improving HCC treatment outcomes. Our narrative review aims to delve into the full spectrum of LAG-3 signaling in HCC, with the goal of a better understanding of the pathophysiological and immunological basis of its use to arrest HCC growth and development.
Collapse
|
3
|
Fernández-Iglesias A, Gracia-Sancho J. Role of liver sinusoidal endothelial cells in the diagnosis and treatment of liver diseases. SINUSOIDAL CELLS IN LIVER DISEASES 2024:467-481. [DOI: 10.1016/b978-0-323-95262-0.00023-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
4
|
Merle P, Kudo M, Krotneva S, Ozgurdal K, Su Y, Proskorovsky I. Regorafenib versus Cabozantinib as a Second-Line Treatment for Advanced Hepatocellular Carcinoma: An Anchored Matching-Adjusted Indirect Comparison of Efficacy and Safety. Liver Cancer 2023; 12:145-155. [PMID: 37325487 PMCID: PMC10267565 DOI: 10.1159/000527403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 09/07/2022] [Indexed: 08/15/2023] Open
Abstract
INTRODUCTION The tyrosine kinase inhibitors regorafenib and cabozantinib remain the mainstay in second-line treatment of advanced hepatocellular carcinoma (HCC). There is currently no clear evidence of superiority in efficacy or safety to guide choice between the two treatments. METHODS We conducted an anchored matching-adjusted indirect comparison using individual patient data from the RESORCE trial of regorafenib and published aggregate data from the CELESTIAL trial of cabozantinib. Second-line HCC patients with prior sorafenib exposure of ≥3 months were included in the analyses. Hazard ratios (HRs) and restricted mean survival time (RMST) were estimated to quantify differences in overall survival (OS) and progression-free survival (PFS). Safety outcomes compared were rates of grade 3 or 4 adverse events (AEs), occurring in >10% of patients, and discontinuation or dose reduction due to treatment-related AEs. RESULTS After matching adjustment for differences in baseline patient characteristics, regorafenib showed a favorable OS (HR, 0.80; 95% CI: 0.54, 1.20) and ∼3-month-longer RMST over cabozantinib (RMST difference, 2.76 months; 95% CI: -1.03, 6.54), although not statistically significant. For PFS, there was no numerical difference in HR (HR, 1.00; 95% CI: 0.68, 1.49) and no clinically meaningful difference based on RMST analyses (RMST difference, -0.59 months; 95% CI: -1.83, 0.65). Regorafenib showed a significantly lower incidence of discontinuation (risk difference, -9.2%; 95% CI: -17.7%, -0.6%) and dose reductions (-15.2%; 95% CI: -29.0%, -1.5%) due to treatment-related AEs (any grade). Regorafenib was also associated with a lower incidence (not statistically significant) of grade 3 or 4 diarrhea (risk difference, -7.1%; 95% CI: -14.7%, 0.4%) and fatigue (-6.3%; 95% CI: -14.6%, 2.0%). CONCLUSION This indirect treatment comparison suggests, relative to cabozantinib, that regorafenib could be associated with favorable OS (not statistically significant), lower rates of dose reductions and discontinuation due to treatment-related AEs, and lower rates of severe diarrhea and fatigue.
Collapse
Affiliation(s)
- Philippe Merle
- Hepatology and Gastroenterology Unit, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon, France, INSERM U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | | | | | - Yun Su
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, New Jersey, USA
| | | |
Collapse
|
5
|
Rimassa L, Finn RS, Sangro B. Combination immunotherapy for hepatocellular carcinoma. J Hepatol 2023:S0168-8278(23)00178-2. [PMID: 36933770 DOI: 10.1016/j.jhep.2023.03.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/17/2023] [Accepted: 03/09/2023] [Indexed: 03/20/2023]
Abstract
Single-agent immune checkpoint inhibitors (ICIs) have been tested in patients with advanced hepatocellular carcinoma (HCC) showing an objective response rate of 15-20%, mostly without a significant overall survival (OS) benefit. Furthermore, approximately 30% of HCC shows intrinsic resistance to ICIs. In the absence of predictive biomarkers to identify patients likely to benefit most from immunotherapy, research has moved to exploring combinations with potential activity in broader patient populations. Basket trials, including cohorts of patients with HCC, and early phase studies tested the combination of ICIs with antiangiogenic agents as well as the combination of two different ICIs. The achieved promising results provided the rationale for the following phase 3 trials, which tested the combination of anti-PD-1/PD-L1 with bevacizumab, or tyrosine kinase inhibitors (TKIs), or anti-CTLA-4. Positive results from the IMbrave150 trial led to the practice-changing approval of atezolizumab-bevacizumab, the first regimen to demonstrate improved survival in the front-line setting, since the approval of sorafenib. More recently, the HIMALAYA trial demonstrated the superiority of durvalumab-tremelimumab (STRIDE regimen) over sorafenib, establishing a new first-line option. In contrast, inconsistent results have been achieved with combinations of ICIs and TKIs, with only one phase 3 trial showing an OS benefit. The rapidly evolving therapeutic landscape for patients with advanced HCC has left significant unmet needs to be addressed in future research. These include choice and sequencing of treatments, identification of biomarkers, combinations with locoregional therapies, and development of new immunotherapy agents. This review summarizes the scientific rationale and available clinical data for combination immunotherapy in advanced HCC.
Collapse
Affiliation(s)
- Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele (Milan), Italy; Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano (Milan), Italy.
| | - Richard S Finn
- Department of Medicine, Division of Hematology/ Oncology, Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Bruno Sangro
- Liver Unit and HPB Oncology Area, Clinica Universidad de Navarra and CIBEREHD, Pamplona, Spain
| |
Collapse
|
6
|
Fasolino G, Awada G, Moschetta L, Koulalis JS, Neyns B, Verhelst B, Van Elderen P, Nelis P, de Lichtbuer PC, Cools W, Ten Tusscher M. Assessment of Retinal Pigment Epithelium Alterations and Chorioretinal Vascular Network Analyses in Patients under Treatment with BRAF/MEK Inhibitor for Different Malignancies: A Pilot Study. J Clin Med 2023; 12:jcm12031214. [PMID: 36769861 PMCID: PMC9918243 DOI: 10.3390/jcm12031214] [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: 12/06/2022] [Revised: 01/26/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
In the last two decades, an increasing number of so-called molecular-targeted therapies have become available for the treatment of patients with advanced malignancies. These drugs have included inhibitors of proteins in the MAPK pathway, such as BRAF and MEK inhibitors, which are characterized by a distinct toxicity profile. The eye is particularly susceptible to adverse effects due to MEK inhibitors, and the term MEKAR (MEK-inhibitor-associated retinopathy) indicates the presence of subretinal fluid, mimicking central serous chorioretinopathy (CSC). The pathogenesis of the retinal alterations related to MAPK pathway inhibitors is still unclear, and questions are still open. The present study aims to assess the presence of retinal pigment epithelium alterations as predictive parameters for retinal toxicity, analyzing, at the same time, the chorioretinal vascular network in patients undergoing BRAF/MEK inhibitor treatment for different malignancies.
Collapse
|
7
|
Impact of Cabozantinib Exposure on Proteinuria and Muscle Toxicity in Patients with Unresectable Hepatocellular Carcinoma. Pharmaceuticals (Basel) 2022; 15:ph15121460. [PMID: 36558911 PMCID: PMC9783864 DOI: 10.3390/ph15121460] [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: 10/16/2022] [Revised: 11/15/2022] [Accepted: 11/24/2022] [Indexed: 11/27/2022] Open
Abstract
This prospective study investigated the impact of cabozantinib exposure on proteinuria and muscle toxicity, in a cohort of 14 Japanese patients with unresectable hepatocellular carcinoma (uHCC). We measured the trough concentration of cabozantinib (Ctrough) weekly for 6 weeks after starting treatment. Although the initial dose was less than 60 mg in most cases, dose interruption occurred in 79%, primarily because of proteinuria and/or malaise. The median and coefficient of variation of maximum Ctrough at 7−42 d were 929.0 ng/mL and 59.2%, respectively. The urinary protein-to-creatinine ratio (UPCR), serum creatine kinase, and serum aldolase values were all significantly elevated following treatment. Moreover, maximum changes in serum creatine kinase and aldolase were significantly associated with maximum Ctrough (r = 0.736, p < 0.01; r = 0.798, p < 0.001; respectively). Receiver operating characteristic (ROC) curve analysis showed that changes in serum creatine kinase ≥70.5 U/L and aldolase ≥6.1 U/L from baseline relatively accurately predicted inclusion in the high-maximum Ctrough (≥929.0 ng/mL) group, with an area under the ROC of 0.929 and 0.833, respectively. Measurement of serum creatine kinase and aldolase may increase the clinical usefulness of cabozantinib treatment for uHCC and help alleviate difficulties with dose adjustments.
Collapse
|
8
|
Sugimoto R, Satoh T, Ueda A, Senju T, Tanaka Y, Yamashita S, Koyanagi T, Kurashige T, Higuchi N, Nakamura T, Tanaka M, Azuma Y, Ohno A, Ooho A, Ooe M, Mutsuki T, Uchimura K, Kuniyoshi M, Tada S, Aratake Y, Yoshimoto T, Yamashita N, Harada S, Nakamuta M, Motomura K, Kohjima M. Atezolizumab plus bevacizumab treatment for unresectable hepatocellular carcinoma progressing after molecular targeted therapy: A multicenter prospective observational study. Medicine (Baltimore) 2022; 101:e30871. [PMID: 36221372 PMCID: PMC9542563 DOI: 10.1097/md.0000000000030871] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To evaluate the efficacy of atezolizumab plus bevacizumab treatment in patients with hepatocellular carcinoma (HCC) previously treated with molecular targeted agents (MTAs). Thirty-one patients treated with atezolizumab plus bevacizumab for unresectable HCC and previously treated with MTAs were enrolled in this study. The treatment lines ranged from second to sixth lines. The treatment effect on HCC differed from that during first-line treatment. The treatment effect was determined using the Response Evaluation Criteria in Solid Tumors (RECIST) and modified RECIST. The treatment response was different for each MTA immediately prior to atezolizumab + bevacizumab treatment. Tumors treated with lenvatinib followed by atezolizumab + bevacizumab showed rapid growth for a short period of time followed by shrinkage. However, patients who received ramucirumab, sorafenib, and regorafenib did not show such changes. This was likely because of differences in the mechanism of action of the MTA administered immediately beforehand. The side-effect profile differed from that observed in the IMbrave150 phase 3 study of atezolizumab plus bevacizumab, which showed more adverse events related to hepatic reserve. Patients treated with the combination of atezolizumab and bevacizumab after lenvatinib therapy may experience rapid tumor growth and subsequent shrinkage.
Collapse
Affiliation(s)
- Rie Sugimoto
- Department of Hepato-Biliary-Pancreatology, National Hospital Organization Kyushu Cancer Center, Fukuoka City, Fukuoka, Japan
- *Correspondence: Rie Sugimoto, Department of Hepato-Biliary-Pancreatology, National Hospital Organization Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka 811-1395, Japan (e-mail: )
| | - Takeaki Satoh
- Department of Center for Liver Disease, Kokura Medical Center, Kitakyushu City, Fukuoka, Japan
| | - Akihiro Ueda
- Department of Internal Medicine, Fukuoka City Hospital, Fukuoka City, Fukuoka, Japan
| | - Takeshi Senju
- Department of Hepato-Biliary-Pancreatology, National Hospital Organization Kyushu Cancer Center, Fukuoka City, Fukuoka, Japan
| | - Yuki Tanaka
- Department of Hepato-Biliary-Pancreatology, National Hospital Organization Kyushu Cancer Center, Fukuoka City, Fukuoka, Japan
| | - Shinsaku Yamashita
- Department of Center for Liver Disease, Kokura Medical Center, Kitakyushu City, Fukuoka, Japan
| | - Toshimasa Koyanagi
- Department of Internal Medicine, Fukuoka City Hospital, Fukuoka City, Fukuoka, Japan
| | - Tomoyuki Kurashige
- Department of Center for Liver Disease, Kokura Medical Center, Kitakyushu City, Fukuoka, Japan
| | - Nobito Higuchi
- Department of Internal Medicine, Fukuoka City Hospital, Fukuoka City, Fukuoka, Japan
| | - Tsukasa Nakamura
- Department of Internal Medicine, Fukuoka City Hospital, Fukuoka City, Fukuoka, Japan
| | - Masatake Tanaka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| | - Yuuki Azuma
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| | - Akari Ohno
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| | - Aritsune Ooho
- Department of Hepatology, Steel Memorial Yawata Hospital, Kitakyushu City, Fukuoka, Japan
| | - Mari Ooe
- Department of Hepatology, Steel Memorial Yawata Hospital, Kitakyushu City, Fukuoka, Japan
| | - Taiji Mutsuki
- Department of Hepatology, Steel Memorial Yawata Hospital, Kitakyushu City, Fukuoka, Japan
| | | | - Masami Kuniyoshi
- Department of Gastroenterology, Kyushu Rosai Hospital, Kitakyushu City, Fukuoka, Japan
| | - Seiya Tada
- Department of Gastroenterology and Hepatology, Fukuokahigashi Medical Center, Koga City, Fukuoka, Japan
| | - Yoshifusa Aratake
- Department of Gastroenterology, National Hospital Organization Kyushu Medical Center, Fukuoka City, Fukuoka, Japan
| | - Tsuyoshi Yoshimoto
- Department of Gastroenterology, National Hospital Organization Kyushu Medical Center, Fukuoka City, Fukuoka, Japan
| | - Naoki Yamashita
- Department of Gastroenterology, National Hospital Organization Kyushu Medical Center, Fukuoka City, Fukuoka, Japan
| | - Shigeru Harada
- Department of Gastroenterology, Chihaya Hospital, Fukuoka, Japan
| | - Makoto Nakamuta
- Department of Gastroenterology, National Hospital Organization Kyushu Medical Center, Fukuoka City, Fukuoka, Japan
| | - Kenta Motomura
- Department of Hepatology, Aso Iizuka Hospital, Iizuka City, Fukuoka, Japan
| | - Motoyuki Kohjima
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Fukuoka, Japan
| |
Collapse
|
9
|
Necroptosis modulation by cisplatin and sunitinib in hepatocellular carcinoma cell line. Life Sci 2022; 301:120594. [PMID: 35500680 DOI: 10.1016/j.lfs.2022.120594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 11/20/2022]
Abstract
Aim Hepatocellular carcinoma (HCC) is one of the leading causes of cancer death worldwide. Systemic chemotherapy such as cisplatin and multi-targeted receptor tyrosine kinase inhibitors, including sunitinib, has marginal activity and frequent toxicity. Recently, necroptosis has been investigated as a potential target in treating cancer. Our aim is to evaluate the influence of cisplatin-sunitinib combination on HepG2 cells regarding their cytotoxicity and implicated intracellular pathways. MATERIALS AND METHODS The half-maximal inhibitory concentration (IC50) values of cisplatin, sunitinib, and their combination were determined by Sulforhodamine-B assay. Bcl-2 and Bax protein levels were assayed using western blot. ELISA technique was used to measure pRIPK3/RIPK3, pERK/ERK, caspase-9, caspase-8, malondialdehyde (MDA), glutathione (GSH), and glutathione peroxidase (GPx). KEY FINDINGS Cisplatin-sunitinib combination exhibited a superior cytotoxic effect on HepG2 cells. Low concentrations of 4 μg/ml cisplatin and 2.8 μg/ml sunitinib showed significant Bcl-2 down-regulation and Bax up-regulation. The combined treatment also lowered pRIPK3/RIPK3 by 74% (p < 0.05) compared to the control. Significant increase in pERK/ERK by 3.9 folds over the normal control was also demonstrated. Moreover, combined treatment produced a significant 4 and 4.6 folds increase in caspase-9 and -8 levels. An increase in MDA level by 1.3 folds, a decrease in the intracellular GSH level by 63%, and an increase in GPx level by 1.17 folds were demonstrated. SIGNIFICANCE Sunitinib modulated cisplatin effect on cytotoxicity, oxidative stress, apoptosis, necroptosis and MAPK pathways. Sunitinib enhanced cisplatin-induced apoptosis and increased oxidative stress, but decreased necroptosis. Combined cisplatin and sunitinib might be promising for treating advanced HCC.
Collapse
|
10
|
Liu S, Qiu J, He W, Geng C, He G, Liu C, Cai D, Liu X, Tian B, Pan H. TUG1 long non-coding RNA enlists the USF1 transcription factor to overexpress ROMO1 leading to hepatocellular carcinoma growth and metastasis. MedComm (Beijing) 2021; 1:386-399. [PMID: 34766130 PMCID: PMC8491240 DOI: 10.1002/mco2.38] [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: 06/17/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent and highly aggressive cancer. Long non‐coding RNAs (lncRNAs) are recognized as potential molecular targets for HCC and are currently under increased research focus. Here, we investigate the regulatory processes underlying the axis of the lncRNA taurine upregulated gene 1 (TUG1), Upstream Transcription Factor 1 (USF1), and reactive oxygen species modulator 1 (ROMO1) in the propagation and metastasis of HCC cells. Distribution of lncRNA TUG1 was found to be prominent in HCC cell cytoplasm and nuclei. LncRNA TUG1 conscripted the USF1 transcription factor to enhance the promoter function of ROMO1. Enlisting the USF1 transcription factor to increase ROMO1 expression following upregulation of TUG1 lncRNA enhanced HCC Huh7 cell proliferation, motility, and metastasis. Rapid tumor proliferation in nude mice provided in vivo verification. The importance of the lncRNA TUG1/USF1/ROMO1 complex as a target for HCC therapy is a key result of this investigation which is exemplified by its role in regulating the proliferation, motility, and metastasis of HCC cells.
Collapse
Affiliation(s)
- Shihai Liu
- Medical Animal Laboratory The Affiliated Hospital of Qingdao University Qingdao China
| | - Jing Qiu
- Department of stomatology Qingdao Municipal Hospital Qingdao China
| | - Weitai He
- School of Biological Science and Technology University of Jinan Jinan China
| | - Chao Geng
- Department of Clinical Laboratory The Affiliated Hospital of Qingdao University Qingdao China
| | - Guifang He
- Medical Animal Laboratory The Affiliated Hospital of Qingdao University Qingdao China
| | - Changchang Liu
- Medical Animal Laboratory The Affiliated Hospital of Qingdao University Qingdao China
| | - Duo Cai
- Medical Animal Laboratory The Affiliated Hospital of Qingdao University Qingdao China
| | - Xiangping Liu
- Medical Research Center The Affiliated Hospital of Qingdao University Qingdao China
| | - Ben Tian
- Department of Neurosurgery Intensive Medicine The First Affiliated Hospital of Baotou Medical College Baotou China
| | - Huazheng Pan
- Department of Clinical Laboratory The Affiliated Hospital of Qingdao University Qingdao China
| |
Collapse
|
11
|
Kim MY, Lee H, Ji SY, Kim SY, Hwangbo H, Park SH, Kim GY, Park C, Leem SH, Hong SH, Choi YH. Induction of Apoptosis by Isoalantolactone in Human Hepatocellular Carcinoma Hep3B Cells through Activation of the ROS-Dependent JNK Signaling Pathway. Pharmaceutics 2021; 13:pharmaceutics13101627. [PMID: 34683920 PMCID: PMC8540929 DOI: 10.3390/pharmaceutics13101627] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 12/16/2022] Open
Abstract
Isoalantolactone (IALT) is one of the isomeric sesquiterpene lactones isolated from the roots of Inula helenium L. IALT is known to possess various biological and pharmacological activities, but its anti-cancer mechanisms are not well understood. The aim of the present study was to investigate the anti-proliferative effects of IALT in human hepatocellular carcinoma (HCC) cells and to evaluate the potential anti-cancer mechanisms. Our results demonstrated that IALT treatment concentration-dependently suppressed the cell survival of HCC Hep3B cells, which was associated with the induction of apoptosis. IALT increased the expression of death-receptor-related proteins, activated caspases, and induced Bid truncation, subsequently leading to cleavage of poly (ADP-ribose) polymerase. In addition, IALT contributed to the cytosolic release of cytochrome c by destroying mitochondrial integrity, following an increase in the Bax/Bcl-2 expression ratio. However, IALT-mediated growth inhibition and apoptosis were significantly attenuated in the presence of a pan-caspase inhibitor, suggesting that IALT induced caspase-dependent apoptosis in Hep3B cells. Moreover, IALT activated the mitogen-activated protein kinases signaling pathway, and the anti-cancer effect of IALT was significantly diminished in the presence of a potent c-Jun N-terminal kinase (JNK) inhibitor. IALT also improved the generation of intracellular reactive oxygen species (ROS), whereas the ROS inhibitor significantly abrogated IALT-induced growth reduction, apoptosis, and JNK activation. Furthermore, ROS-dependent apoptosis was revealed as a mechanism involved in the anti-cancer activity of IALT in a 3D multicellular tumor spheroid model of Hep3B cells. Taken together, our findings indicate that IALT exhibited anti-cancer activity in HCC Hep3B cells by inducing ROS-dependent activation of the JNK signaling pathway.
Collapse
Affiliation(s)
- Min Yeong Kim
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea; (M.Y.K.); (H.L.); (S.Y.J.); (S.Y.K.); (H.H.)
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea
| | - Hyesook Lee
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea; (M.Y.K.); (H.L.); (S.Y.J.); (S.Y.K.); (H.H.)
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea
| | - Seon Yeong Ji
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea; (M.Y.K.); (H.L.); (S.Y.J.); (S.Y.K.); (H.H.)
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea
| | - So Young Kim
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea; (M.Y.K.); (H.L.); (S.Y.J.); (S.Y.K.); (H.H.)
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea
| | - Hyun Hwangbo
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea; (M.Y.K.); (H.L.); (S.Y.J.); (S.Y.K.); (H.H.)
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea
| | - Shin-Hyung Park
- Department of Pathology, Dong-eui University College of Korean Medicine, Busan 47227, Korea;
| | - Gi-Young Kim
- Department of Marine Life Science, College of Ocean Sciences, Jeju National University, Jeju 63243, Korea;
| | - Cheol Park
- Division of Basic Sciences, College of Liberal Studies, Dong-Eui University, Busan 47340, Korea;
| | - Sun-Hee Leem
- Department of Biomedical Sciences, College of Natural Sciences, Dong-A University, Busan 49315, Korea;
- Department of Health Sciences, The Graduated of Dong-A University, Busan 49315, Korea
| | - Su Hyun Hong
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea; (M.Y.K.); (H.L.); (S.Y.J.); (S.Y.K.); (H.H.)
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea
- Correspondence: (S.H.H.); (Y.H.C.); Tel.: +82-051-890-3334 (S.H.H.); +82-051-890-3319 (Y.H.C.)
| | - Yung Hyun Choi
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea; (M.Y.K.); (H.L.); (S.Y.J.); (S.Y.K.); (H.H.)
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea
- Correspondence: (S.H.H.); (Y.H.C.); Tel.: +82-051-890-3334 (S.H.H.); +82-051-890-3319 (Y.H.C.)
| |
Collapse
|
12
|
Rapposelli IG, Shimose S, Kumada T, Okamura S, Hiraoka A, Di Costanzo GG, Marra F, Tamburini E, Forgione A, Foschi FG, Silletta M, Lonardi S, Masi G, Scartozzi M, Nakano M, Shibata H, Kawata K, Pellino A, Vivaldi C, Lai E, Takata A, Tajiri K, Toyoda H, Tortora R, Campani C, Viola MG, Piscaglia F, Conti F, Fulgenzi CAM, Frassineti GL, Rizzato MD, Salani F, Astara G, Torimura T, Atsukawa M, Tada T, Burgio V, Rimini M, Cascinu S, Casadei-Gardini A. Identification of lenvatinib prognostic index via recursive partitioning analysis in advanced hepatocellular carcinoma. ESMO Open 2021; 6:100190. [PMID: 34144271 PMCID: PMC8219999 DOI: 10.1016/j.esmoop.2021.100190] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND After the advent of new treatment options for advanced hepatocellular carcinoma (HCC), the identification of prognostic factors is crucial for the selection of the most appropriate therapy for each patient. PATIENTS AND METHODS With the aim to fill this gap, we applied recursive partitioning analysis (RPA) to a cohort of 404 patients treated with lenvatinib. RESULTS The application of RPA resulted in a classification based on five variables that originated a new prognostic score, the lenvatinib prognostic index (LEP) index, identifying three groups: low risk [patients with prognostic nutritional index (PNI) >43.3 and previous trans-arterial chemoembolization (TACE)]; medium risk [patients with PNI >43.3 but without previous TACE and patients with PNI <43.3, albumin-bilirubin (ALBI) grade 1 and Barcelona Clinic Liver Cancer stage B (BCLC-B)]; high risk [patients with PNI <43.3 and ALBI grade 2 and patients with PNI <43.3, albumin-bilirubin (ALBI) grade 1 and Barcelona Clinic Liver Cancer stage C (BCLC-C)]. Median overall survival was 29.8 months [95% confidence interval (CI) 22.8-29.8 months] in low risk patients (n = 128), 17.0 months (95% CI 15.0-24.0 months) in medium risk (n = 162) and 8.9 months (95% CI 8.0-10.7 months) in high risk (n = 114); low risk hazard ratio (HR) 1 (reference group), medium risk HR 1.95 (95% CI 1.38-2.74), high risk HR 4.84 (95% CI 3.16-7.43); P < 0.0001. The LEP index was validated in a cohort of 127 Italian patients treated with lenvatinib. While the same classification did not show a prognostic value in a cohort of 311 patients treated with sorafenib, we also show a possible predictive role in favor of lenvatinib in the low risk group. CONCLUSIONS LEP index is a promising, easy-to-use tool that may be used to stratify patients undergoing systemic treatment of advanced HCC.
Collapse
Affiliation(s)
- I G Rapposelli
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori 'Dino Amadori'-IRST, Meldola, Italy
| | - S Shimose
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - T Kumada
- Faculty of Nursing, Gifu Kyoritsu University, Ogaki, Japan
| | - S Okamura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - A Hiraoka
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama, Japan
| | - G G Di Costanzo
- Liver Unit, Department of Transplantation, Cardarelli Hospital, Naples, Italy
| | - F Marra
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, Florence, Italy
| | - E Tamburini
- Department of Medical Oncology, Card. G. Panico Hospital of Tricase, Tricase, Italy
| | - A Forgione
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - F G Foschi
- Department of Internal Medicine, Faenza Hospital, AUSL Romagna, Faenza, Italy
| | - M Silletta
- Medical Oncology Unit, University Campus Bio-Medico, Rome, Italy
| | - S Lonardi
- Early Phase Clinical Trial Unit, Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy; Medical Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - G Masi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy; Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - M Scartozzi
- Medical Oncology, University and University Hospital of Cagliari, Italy
| | - M Nakano
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - H Shibata
- Department of Gastroenterology, Tokushima Prefectural Central Hospital, Tokushima, Japan
| | - K Kawata
- Hepatology Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - A Pellino
- Medical Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - C Vivaldi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy; Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - E Lai
- Medical Oncology, University and University Hospital of Cagliari, Italy
| | - A Takata
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - K Tajiri
- Department of Gastroenterology, Toyama University Hospital, Toyama, Japan
| | - H Toyoda
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki, Japan
| | - R Tortora
- Liver Unit, Department of Transplantation, Cardarelli Hospital, Naples, Italy
| | - C Campani
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, Florence, Italy
| | - M G Viola
- Department of Surgery, Card. G. Panico Hospital of Tricase, Tricase, Italy
| | - F Piscaglia
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - F Conti
- Department of Internal Medicine, Faenza Hospital, AUSL Romagna, Faenza, Italy
| | - C A M Fulgenzi
- Medical Oncology Unit, University Campus Bio-Medico, Rome, Italy
| | - G L Frassineti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori 'Dino Amadori'-IRST, Meldola, Italy
| | - M D Rizzato
- Medical Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - F Salani
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - G Astara
- Medical Oncology, University and University Hospital of Cagliari, Italy
| | - T Torimura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - M Atsukawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - T Tada
- Department of Internal Medicine, Japanese Red Cross Himeji Hospital, Himeji, Japan
| | - V Burgio
- Unit of Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M Rimini
- Department of Oncology and Hematology, Division of Oncology, University Hospital of Modena, Modena, Italy
| | - S Cascinu
- Unit of Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy; School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - A Casadei-Gardini
- Unit of Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy; School of Medicine, Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
13
|
Abstract
Liver sinusoidal endothelial cells (LSECs) form the wall of the hepatic sinusoids. Unlike other capillaries, they lack an organized basement membrane and have cytoplasm that is penetrated by open fenestrae, making the hepatic microvascular endothelium discontinuous. LSECs have essential roles in the maintenance of hepatic homeostasis, including regulation of the vascular tone, inflammation and thrombosis, and they are essential for control of the hepatic immune response. On a background of acute or chronic liver injury, LSECs modify their phenotype and negatively affect neighbouring cells and liver disease pathophysiology. This Review describes the main functions and phenotypic dysregulations of LSECs in liver diseases, specifically in the context of acute injury (ischaemia-reperfusion injury, drug-induced liver injury and bacterial and viral infection), chronic liver disease (metabolism-associated liver disease, alcoholic steatohepatitis and chronic hepatotoxic injury) and hepatocellular carcinoma, and provides a comprehensive update of the role of LSECs as therapeutic targets for liver disease. Finally, we discuss the open questions in the field of LSEC pathobiology and future avenues of research.
Collapse
|
14
|
Kuznietsova H, Byelinska I, Dziubenko N, Lynchak O, Milokhov D, Khilya O, Finiuk N, Klyuchivska O, Stoika R, Rybalchenko V. Suppression of systemic inflammation and signs of acute and chronic cholangitis by multi-kinase inhibitor 1-(4-Cl-benzyl)-3-chloro-4-(CF3-phenylamino)-1H-pyrrole-2,5-dione. Mol Cell Biochem 2021; 476:3021-3035. [PMID: 33792809 DOI: 10.1007/s11010-021-04144-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 03/23/2021] [Indexed: 12/12/2022]
Abstract
An aberrant activity of growth factor receptors followed by excessive cell proliferation plays a significant role in pathogenesis of cholangitis. Therefore, inhibition of these processes could be a fruitful therapeutic strategy. The effects of multi-kinase inhibitor 1-(4-Cl-benzyl)-3-chloro-4-(CF3-phenylamino)-1H-pyrrole-2,5-dione (MI-1) on the hepatic and systemic manifestations of acute and chronic cholangitis in rats were addressed. MI-1 (2.7 mg/kg per day) was applied to male rats that experienced α-naphthylisothiocyanate-induced acute (3 days) or chronic (28 days) cholangitis. Liver autopsy samples, blood serum markers, and leukograms were studied. MI-1 localization in liver cells and its impact on viability of HepG2 (human hepatoma), HL60 (human leukemia), and NIH3T3 (normal murine fibroblasts) cell lines and lymphocytes of human peripheral blood (MTT, DNA fragmentation, DNA comet assays, Propidium Iodide staining) were assessed. Under both acute and chronic cholangitis, MI-1 substantially reduced liver injury, fibrosis, and inflammatory scores (by 46-86%) and normalized blood serum markers and leukograms. Moreover, these effects were preserved after a 28-day recovery period (without any treatment). MI-1 inhibited the HL60, HepG2 cells, and human lymphocytes viability (IC50 0.6, 9.5 and 8.3 µg/ml, respectively), while NIH3T3 cells were resistant to that. Additionally, HepG2 cells and lymphocytes being incubated with MI-1 demonstrated insignificant pro-apoptotic and pro-necrotic changes and DNA single-strand breaks, suggesting that MI-1 effects in liver might be partly caused by its cytotoxic action towards liver cells and lymphocytes. In conclusion, MI-1 attenuated the systemic inflammation and signs of acute and chronic cholangitis partly through cytotoxicity towards cells of hepatic and leukocytic origin.
Collapse
Affiliation(s)
- Halyna Kuznietsova
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine.
| | - Iryna Byelinska
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Natalia Dziubenko
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Oksana Lynchak
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Demyd Milokhov
- Chemistry Department, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Olga Khilya
- Chemistry Department, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Nataliya Finiuk
- Institute of Cell Biology, National Academy of Sciences of Ukraine, Lviv, Ukraine
- Ivan Franko National University of Lviv, Lviv, Ukraine
| | - Olga Klyuchivska
- Institute of Cell Biology, National Academy of Sciences of Ukraine, Lviv, Ukraine
| | - Rostyslav Stoika
- Institute of High Technologies, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
- Institute of Cell Biology, National Academy of Sciences of Ukraine, Lviv, Ukraine
- Ivan Franko National University of Lviv, Lviv, Ukraine
| | - Volodymyr Rybalchenko
- Institute of High Technologies, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| |
Collapse
|
15
|
Su CM, Wang HC, Hsu FT, Lu CH, Lai CK, Chung JG, Kuo YC. Astragaloside IV Induces Apoptosis, G 1-Phase Arrest and Inhibits Anti-apoptotic Signaling in Hepatocellular Carcinoma. In Vivo 2020; 34:631-638. [PMID: 32111763 DOI: 10.21873/invivo.11817] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIM Hepatocellular carcinoma (HCC) is a primary malignancy of the liver and the third leading cause of cancer death worldwide. Although multiple chemotherapies options are available for HCC, chemo-induced toxicity is inevitable during clinical treatment. Therefore, identifying possible adjuvant agents with both liver-protective and antitumor effects is critical. Herbal medicines have chemopreventive and anti-HCC effect, such as Juzen taiho-to and Sho-saiko-to. Astragaloside IV is a compound extracted from the Chinese medical herb Astragalus membranaceus (Fisch.) Bge. with liver protection potential. However, whether astragaloside IV may also possess tumor-inhibitory capability and its underlying mechanism is remaining unknown. MATERIALS AND METHODS Viability analysis, cell-cycle analysis, apoptosis analysis, western blotting analysis and invasion trans-well assay were performed to identify tumor-inhibitory potential of astragaloside IV on HCC cells (SK-Hep1 and Hep3B cells). RESULTS We found that astragaloside IV may induce cytotoxicity and extrinsic/intrinsic apoptosis effect, but also trigger G1 arrest in HCC cells. The expression of anti-apoptotic proteins of HCC were all reduced by astragaloside IV. Additionally, astragaloside IV also suppressed HCC cell invasion ability. CONCLUSION Astragaloside IV effectively suppressed HCC cell proliferation, invasion and anti-apoptosis in vitro.
Collapse
Affiliation(s)
- Chun-Min Su
- Department of Surgery, Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C
| | - Hsiao-Chia Wang
- Emergency Department, Cathay General Hospital, Taipei, Taiwan, R.O.C.,School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, R.O.C
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C
| | - Chun-Hui Lu
- Division of Pharmacy, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan, R.O.C.,Department of Business Management, National Sun Yat-sen University, Kaohsiung, Taiwan, R.O.C
| | - Chien-Kai Lai
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C
| | - Yu-Cheng Kuo
- School of Medicine, China Medical University, Taichung, Taiwan, R.O.C. .,Department of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan, R.O.C
| |
Collapse
|
16
|
Garcia-Lezana T, Lopez-Canovas JL, Villanueva A. Signaling pathways in hepatocellular carcinoma. Adv Cancer Res 2020; 149:63-101. [PMID: 33579428 DOI: 10.1016/bs.acr.2020.10.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite the recent introduction of new effective systemic agents, the survival of patients with hepatocellular carcinoma (HCC) at advanced stages remains dismal. This underscores the need for new therapies, which has spurred extensive research on the identification of the main drivers of pathway de-regulation as a source of novel therapeutic targets. Frequently altered pathways in HCC involve growth factor receptors (e.g., VEGFR, FGFR, TGFA, EGFR, IGFR) and/or its cytoplasmic intermediates (e.g., PI3K-AKT-mTOR, RAF/ERK/MAPK) as well as key pathways in cell differentiation (e.g., Wnt/β-catenin, JAK/STAT, Hippo, Hedgehog, Notch). Somatic mutations, chromosomal aberrations and epigenetic changes are common mechanisms for pathway deregulation in HCC. Aberrant pathway activation has also been explored as a biomarker to predict response to specific therapies, but currently, these strategies are not implemented when deciding systemic therapies in HCC patients. Beyond the well-established molecular cascades, there are numerous emerging signaling pathways also deregulated in HCC (e.g., tumor microenvironment, non-coding RNA, intestinal microbiota), which have opened new avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Teresa Garcia-Lezana
- Division of Liver Diseases, Liver Cancer Program, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Juan Luis Lopez-Canovas
- Department of Cell Biology, Physiology and Immunology, Maimonides Institute of Biomedical Research of Córdoba (IMIBIC), University of Córdoba, Córdoba, Spain
| | - Augusto Villanueva
- Division of Liver Diseases, Liver Cancer Program, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
17
|
Luo Y, Song L, Wang X, Huang Y, Liu Y, Wang Q, Hong M, Yuan Z. Uncovering the Mechanisms of Cryptotanshinone as a Therapeutic Agent Against Hepatocellular Carcinoma. Front Pharmacol 2020; 11:1264. [PMID: 32903546 PMCID: PMC7438559 DOI: 10.3389/fphar.2020.01264] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/30/2020] [Indexed: 12/20/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a fatal and dominant form of liver cancer that currently has no effective treatment or positive prognosis. In this study, we explored the antitumor effects of cryptotanshinone (CPT) against HCC and the molecular mechanisms underlying these effects using a systems pharmacology and experimental validation approach. First, we identified a total of 296 CPT targets, 239 of which were also HCC-related targets. We elucidated the mechanisms by which CPT affects HCC through multiple network analysis, including CPT-target network analysis, protein-protein interaction network analysis, target-function network analysis, and pathway enrichment analysis. In addition, we found that CPT induced apoptosis in Huh7 and MHCC97-H ells due to increased levels of cleaved PARP, Bax, and cleaved caspase-3 and decreased Bcl-2 expression. CPT also induced autophagy in HCC cells by increasing LC3-II conversion and the expression of Beclin1 and ATG5, while decreasing the expression of p62/SQSTM1. Autophagy inhibitors (3-methyladenine and chloroquine) enhanced CPT-induced proliferation and apoptosis, suggesting that CPT-induced autophagy may protect HCC cells against cell death. Furthermore, CPT was found to inhibit the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway. Interestingly, activation of PI3K by insulin-like growth factor-I inhibited CPT-induced apoptosis and autophagy, suggesting that the PI3K/AKT/mTOR signaling pathway is involved in both CPT-induced apoptosis and autophagy. Finally, CPT was found to inhibit the growth of Huh7 xenograft tumors. In conclusion, we first demonstrated the antitumor effects of CPT in Huh7 and MHCC97-H cells, both in vitro and in vivo. We elucidated the potential antitumor mechanism of CPT, which involved inducing apoptosis and autophagy by inhibiting the PI3K/Akt/mTOR signaling pathway. Our findings may provide valuable insights into the clinical application of CPT, serving as a potential candidate therapeutic agent for HCC treatment.
Collapse
Affiliation(s)
- Yi Luo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Song
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyu Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yujie Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongqiang Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ming Hong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongyu Yuan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|