1
|
Donia T, Ali EMM, Kalantan AA, Alzahrani FA, Eid TM, Khamis AA. Synergistic anticancer efficacy of polydatin and sorafenib against the MCF-7 breast cancer cell line via inhibiting of PI3K/AKT/mTOR pathway and reducing resistance to treatment. Biochem Biophys Res Commun 2024; 739:150972. [PMID: 39541924 DOI: 10.1016/j.bbrc.2024.150972] [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: 08/14/2024] [Revised: 10/25/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
Polydatin (PD), a glucoside derivative of resveratrol, has been investigated for its potential to mitigate sorafenib (SOF) side effects and combat multidrug resistance in cancer treatment. The study evaluated its mechanism of action for inhibiting the protein kinase B/mTOR pathway in promoting breast cancer proliferation. The combined PD and SOF have synergistic effects with a combination index (CI) < 1 in the liver (HepG2) and breast (MCF-7) cancer cell lines. Molecular docking studies were conducted to analyze interactions of PD& SOF with protein kinases as well as apoptotic and multidrug resistance proteins, including AKT1, PI3K, mTOR, Apaf-1, and ABCB1 in MCF-7 cells. Experimental validation through real-time PCR confirmed. PD has a strong binding affinity, particularly with AKT1 (-56 kcal/mol) and ABCB1 (-27.16 kcal/mol), a gene associated with multidrug resistance. These interactions were linked to anti-proliferative anti-angiogenic effects and reduced resistance to treatment, demonstrating PD has potential therapeutic benefits. Furthermore, PD combined with SOF induced apoptosis, inhibited cell growth, and arrested MCF-7 cells in the sub-G1 phase with increased intracellular ROS. This was accompanied by reduced expression of AKT1 and ABCB1 genes, reinforcing the anticancer efficacy of PD/SOF combination therapy. In conclusion, the findings suggest that PD/SOF could serve as a promising anticancer treatment strategy, warranting further investigation for potential clinical applications and mechanistic studies in vivo.
Collapse
Affiliation(s)
- Thoria Donia
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt.
| | - Ehab M M Ali
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt; Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, 21589 Jeddah, Saudi Arabia.
| | - Abdulaziz A Kalantan
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, 21589 Jeddah, Saudi Arabia.
| | - Faisal Ay Alzahrani
- Department of Chemistry, College of Sciences & Arts, King Abdulaziz University, 21911 Rabigh, Saudi Arabia.
| | - Thamir M Eid
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, 21589 Jeddah, Saudi Arabia.
| | - Abeer A Khamis
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt.
| |
Collapse
|
2
|
Franco-Juárez EX, González-Villasana V, Camacho-Moll ME, Rendón-Garlant L, Ramírez-Flores PN, Silva-Ramírez B, Peñuelas-Urquides K, Cabello-Ruiz ED, Castorena-Torres F, Bermúdez de León M. Mechanistic Insights about Sorafenib-, Valproic Acid- and Metformin-Induced Cell Death in Hepatocellular Carcinoma. Int J Mol Sci 2024; 25:1760. [PMID: 38339037 PMCID: PMC10855535 DOI: 10.3390/ijms25031760] [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: 10/09/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 02/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is among the main causes of death by cancer worldwide, representing about 80-90% of all liver cancers. Treatments available for advanced HCC include atezolizumab, bevacizumab, sorafenib, among others. Atezolizumab and bevacizumab are immunological options recently incorporated into first-line treatments, along with sorafenib, for which great treatment achievements have been reached. However, sorafenib resistance is developed in most patients, and therapeutical combinations targeting cancer hallmark mechanisms and intracellular signaling have been proposed. In this review, we compiled evidence of the mechanisms of cell death caused by sorafenib administered alone or in combination with valproic acid and metformin and discussed them from a molecular perspective.
Collapse
Affiliation(s)
- Edgar Xchel Franco-Juárez
- Departamento de Biología Molecular, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey 64720, Nuevo Leon, Mexico; (E.X.F.-J.); (M.E.C.-M.); (P.N.R.-F.); (K.P.-U.)
- Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66451, Nuevo Leon, Mexico; (V.G.-V.); (L.R.-G.); (E.D.C.-R.)
| | - Vianey González-Villasana
- Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66451, Nuevo Leon, Mexico; (V.G.-V.); (L.R.-G.); (E.D.C.-R.)
| | - María Elena Camacho-Moll
- Departamento de Biología Molecular, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey 64720, Nuevo Leon, Mexico; (E.X.F.-J.); (M.E.C.-M.); (P.N.R.-F.); (K.P.-U.)
| | - Luisa Rendón-Garlant
- Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66451, Nuevo Leon, Mexico; (V.G.-V.); (L.R.-G.); (E.D.C.-R.)
| | - Patricia Nefertari Ramírez-Flores
- Departamento de Biología Molecular, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey 64720, Nuevo Leon, Mexico; (E.X.F.-J.); (M.E.C.-M.); (P.N.R.-F.); (K.P.-U.)
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64710, Nuevo Leon, Mexico;
| | - Beatriz Silva-Ramírez
- Departamento de Inmunogenética, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey 64720, Nuevo Leon, Mexico;
| | - Katia Peñuelas-Urquides
- Departamento de Biología Molecular, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey 64720, Nuevo Leon, Mexico; (E.X.F.-J.); (M.E.C.-M.); (P.N.R.-F.); (K.P.-U.)
| | - Ethel Daniela Cabello-Ruiz
- Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66451, Nuevo Leon, Mexico; (V.G.-V.); (L.R.-G.); (E.D.C.-R.)
| | - Fabiola Castorena-Torres
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64710, Nuevo Leon, Mexico;
| | - Mario Bermúdez de León
- Departamento de Biología Molecular, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey 64720, Nuevo Leon, Mexico; (E.X.F.-J.); (M.E.C.-M.); (P.N.R.-F.); (K.P.-U.)
| |
Collapse
|
3
|
Ng WH, Soo KC, Huynh H. Vinorelbine Improves the Efficacy of Sorafenib against Hepatocellular Carcinoma: A Promising Therapeutic Approach. Int J Mol Sci 2024; 25:1563. [PMID: 38338842 PMCID: PMC10855313 DOI: 10.3390/ijms25031563] [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: 12/22/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading global cause of cancer-related mortality. Despite the widespread adoption of sorafenib as the standard HCC treatment, its efficacy is constrained, frequently encountering resistance. To augment the effectiveness of sorafenib, this study investigated the synergy of sorafenib and vinorelbine using 22 HCC patient-derived xenograft (PDX) models. In this study, mice bearing HCC tumors were treated with the vehicle, sorafenib (15 mg/kg), vinorelbine (3 mg/kg), and sorafenib-vinorelbine combination (Sora/Vino). Rigorous monitoring of the tumor growth and side effects coupled with comprehensive histological and molecular analyses was conducted. The overall survival (OS) of mice bearing HCC orthotopic tumors was also assessed. Our data showed a notable 86.4% response rate to Sora/Vino, surpassing rates of 31.8% for sorafenib and 9.1% for vinorelbine monotherapies. Sora/Vino significantly inhibited tumor growth, prolonged OS of mice bearing HCC orthotopic tumors (p < 0.01), attenuated tumor cell proliferation and angiogenesis, and enhanced necrosis and apoptosis. The combination therapy effectively suppressed the focal adhesion kinase (FAK) pathway, which is a pivotal player in cell proliferation, tumor angiogenesis, survival, and metastasis. The noteworthy antitumor activity in 22 HCC PDX models positions Sora/Vino as a promising candidate for early-phase clinical trials, leveraging the established use of sorafenib and vinorelbine in HCC and other cancers.
Collapse
Affiliation(s)
- Wai Har Ng
- Laboratory of Molecular Endocrinology, National Cancer Centre Singapore, Singapore 168583, Singapore;
| | - Khee Chee Soo
- Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, Singapore 168583, Singapore
| | - Hung Huynh
- Laboratory of Molecular Endocrinology, National Cancer Centre Singapore, Singapore 168583, Singapore;
| |
Collapse
|
4
|
Huynh H, Ng WH, Soo KC. Everolimus Acts in Synergy with Vinorelbine to Suppress the Growth of Hepatocellular Carcinoma. Int J Mol Sci 2023; 25:17. [PMID: 38203186 PMCID: PMC10779360 DOI: 10.3390/ijms25010017] [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: 11/15/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a challenging cancer to treat, as traditional chemotherapies have shown limited effectiveness. The mammalian target of rapamycin/sirolimus (mTOR) and microtubules are prominent druggable targets for HCC. In this study, we demonstrated that co-targeting mTOR using mTOR inhibitors (everolimus and sirolimus) along with the microtubule inhibitor vinorelbine yielded results superior to those of the monotherapies in HCC PDX models. Our research showed that the vinorelbine arrests cells at the mitotic phase, induces apoptosis, and normalizes tumor blood vessels but upregulates survivin and activates the mTOR/p70S6K/4EBP1 pathway. The addition of the everolimus significantly improved the tumor response to the vinorelbine, leading to improved overall survival (OS) in most tested orthotopic HCC PDX models. The mechanistic investigation revealed that this marked antitumor effect was accompanied by the downregulations of mTOR targets (p-p70S6K, p-4EBP1, and p-S6K); several key cell-cycle regulators; and the antiapoptotic protein survivin. These effects did not compromise the normalization of the blood vessels observed in response to the vinorelbine in the vinorelbine-sensitive PDX models or to the everolimus in the everolimus-sensitive PDX models. The combination of the everolimus and vinorelbine (everolimus/vinorelbine) also promoted apoptosis with minimal toxicity. Given the cost-effectiveness and established effectiveness of everolimus, and especially sirolimus, this strategy warrants further investigation in early-phase clinical trials.
Collapse
Affiliation(s)
- Hung Huynh
- Laboratory of Molecular Endocrinology, National Cancer Centre Singapore, Singapore 168583, Singapore;
| | - Wai Har Ng
- Laboratory of Molecular Endocrinology, National Cancer Centre Singapore, Singapore 168583, Singapore;
| | - Khee Chee Soo
- Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, Singapore 168583, Singapore
| |
Collapse
|
5
|
Yeh DW, Liu C, Hernandez JC, Tahara SM, Tsukamoto H, Machida K. Polycomb repressive complex 2 binds and stabilizes NANOG to suppress differentiation-related genes to promote self-renewal. iScience 2023; 26:107035. [PMID: 37448562 PMCID: PMC10336160 DOI: 10.1016/j.isci.2023.107035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/20/2023] [Accepted: 05/31/2023] [Indexed: 07/15/2023] Open
Abstract
The synergistic effect of alcohol and HCV mediated through TLR4 signaling transactivates NANOG, a pluripotency transcription factor important for the stemness of tumor-initiating stem-like cells (TICs). NANOG together with the PRC2 complex suppresses expression of oxidative phosphorylation (OXPHOS) genes to generate TICs. The phosphodegron sequence PEST domain of NANOG binds EED to stabilize NANOG protein by blocking E3 ligase recruitment and proteasome-dependent degradation, while the tryptophan-rich domain of NANOG binds EZH2 and SUZ12. Human ARID1A gene loss results in the resistance to combined FAO and PRC2 inhibition therapies due to reduction of mitochondrial ROS levels. CRISPR-Cas9-mediated ARID1A knockout and/or constitutively active CTNNB1 driver mutations promoted tumor development in humanized FRG HCC mouse models, in which use of an interface inhibitor antagonizing PRC2-NANOG binding and/or FAO inhibitor blocked tumor growth. Together, the PRC2-NANOG interaction becomes a new drug target for HCC via inducing differentiation-related genes, destabilizing NANOG protein, and suppressing NANOG activity.
Collapse
Affiliation(s)
- Da-Wei Yeh
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Cheng Liu
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Juan Carlos Hernandez
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Stanley M. Tahara
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Hidekazu Tsukamoto
- Department of Pathology; University of Southern California, Los Angeles, CA 90033, USA
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA 90033, USA
| | - Keigo Machida
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA 90033, USA
| |
Collapse
|
6
|
Hernandez JC, Chen CL, Machida T, Uthaya Kumar DB, Tahara SM, Montana J, Sher L, Liang J, Jung JU, Tsukamoto H, Machida K. LIN28 and histone H3K4 methylase induce TLR4 to generate tumor-initiating stem-like cells. iScience 2023; 26:106254. [PMID: 36949755 PMCID: PMC10025994 DOI: 10.1016/j.isci.2023.106254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/09/2022] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Chemoresistance and plasticity of tumor-initiating stem-like cells (TICs) promote tumor recurrence and metastasis. The gut-originating endotoxin-TLR4-NANOG oncogenic axis is responsible for the genesis of TICs. This study investigated mechanisms as to how TICs arise through transcriptional, epigenetic, and post-transcriptional activation of oncogenic TLR4 pathways. Here, we expressed constitutively active TLR4 (caTLR4) in mice carrying pLAP-tTA or pAlb-tTA, under a tetracycline withdrawal-inducible system. Liver progenitor cell induction accelerated liver tumor development in caTLR4-expressing mice. Lentiviral shRNA library screening identified histone H3K4 methylase SETD7 as central to activation of TLR4. SETD7 combined with hypoxia induced TLR4 through HIF2 and NOTCH. LIN28 post-transcriptionally stabilized TLR4 mRNA via de-repression of let-7 microRNA. These results supported a LIN28-TLR4 pathway for the development of HCCs in a hypoxic microenvironment. These findings not only advance our understanding of molecular mechanisms responsible for TIC generation in HCC, but also represent new therapeutic targets for the treatment of HCC.
Collapse
Affiliation(s)
- Juan Carlos Hernandez
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
- MS Biotechnology Program, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Chia-Lin Chen
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
- Department of Life Sciences & Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 110, Taiwan
| | - Tatsuya Machida
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Dinesh Babu Uthaya Kumar
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Stanley M. Tahara
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Jared Montana
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Linda Sher
- Department of Surgery, University of Southern California, Los Angeles, CA 90033, USA
| | | | - Jae U. Jung
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Hidekazu Tsukamoto
- Department of Pathology, University of Southern California, Los Angeles, CA 90033, USA
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA
- Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Keigo Machida
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA
| |
Collapse
|
7
|
Wu Y, Tan HWS, Lin JY, Shen HM, Wang H, Lu G. Molecular mechanisms of autophagy and implications in liver diseases. LIVER RESEARCH 2023; 7:56-70. [PMID: 39959698 PMCID: PMC11792062 DOI: 10.1016/j.livres.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/03/2022] [Accepted: 02/15/2023] [Indexed: 02/22/2023]
Abstract
Autophagy is a highly conserved process in which cytosolic contents are degraded by the lysosome, which plays an important role in energy and nutrient balance, and protein or organelle quality control. The liver is the most important organ for metabolism. Studies to date have revealed a significant role of autophagy in the maintenance of liver homeostasis under basal and stressed conditions, and the impairment of autophagy has been closely linked to various liver diseases. Therefore, a comprehensive understanding of the roles of autophagy in liver diseases may help in the development of therapeutic strategies via targeting autophagy. In this review, we will summarize the latest understanding of the molecular mechanisms of autophagy and systematically discuss its implications in various liver diseases, including alcohol-related liver disease, non-alcoholic fatty liver disease, viral hepatitis, hepatocellular carcinoma, and acetaminophen-induced liver injury.
Collapse
Affiliation(s)
- Yuankai Wu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hayden Weng Siong Tan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jin-Yi Lin
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Han-Ming Shen
- Department of Biomedical Sciences, Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Haihe Wang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Guang Lu
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
8
|
Ghamry HI. Impending Chemotherapeutic Impact of Arthrospira platensis Nanoparticles and/or Sorafenib against Hepatocellular Carcinoma through Modulation of Antioxidant Status, Tumor Marker Genes, and Anti-Inflammatory Signaling Pathways. TOXICS 2023; 11:107. [PMID: 36850982 PMCID: PMC9964820 DOI: 10.3390/toxics11020107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/19/2023] [Accepted: 01/19/2023] [Indexed: 06/18/2023]
Abstract
This study investigated Arthrospira platensis nanoparticles (NSP) to overcome sorafenib resistance in diethyl nitrosamine-induced hepatocellular carcinoma (HCC) in rats. This study used sixty Wistar male rats randomly grouped into two main groups, the normal control group, and the HCC model. For the normal control group (n = 12), animals were injected i.p. with PBS two times/week for 16 weeks. The remaining 48 rats were injected i.p. with using a single dose of diethyl nitrosamine (DENA) (200 mg/kg, ip), followed by phenobarbital sodium (0.05%) in drinking water for 16 weeks. At the end of the 16th week, rats were allocated into four groups (11 rats/each), one group was left without treatment (DENA group), and the other three groups were treated with either sorafenib (30 mg/kg; p.o.) or Arthrospira platensis Nanoparticles (NSP) (0.5 mg/kg body weight) once daily orally with the aid of gastric gavage or their combination for another four weeks. Blood and tissue samples were collected for further biochemical, histological, immunohistochemical, and gene expression analysis. Our result revealed that DENA-treated rats showed a marked elevation of hepatic enzyme markers with an increase in the total protein and globulin and decreases in the hepatic SOD. Catalase and GSH, with significantly increased MDA levels, subsequently increased the tumor biomarkers (AFP and CEA). On the molecular level, the DENA-treated rats showed significant up-regulation of Cyp19 mRNA and the inflammatory cytokines (TNF-α, iNOS, and TGF-1β) as well as the Ki-67 gene expression (p < 0.05) with down-regulation of the PPAR-γ and FOXO-1. In addition, the HCC group showed a loss of hepatic architecture, as well as atypia, swelling, macrosteatosis of hepatocytes, and fibrosis, besides increased vascularization. The immunohistochemical findings show increased expression of both GPC-3 and Hep Par 1 in the HCC group. SOR, NSP, or a combination of NSP and SOR.NSP treatment significantly overturned the DENA's harmful effect near the normal levels and restored all cancer biomarkers and antioxidant activities, indicating the chemotherapeutic impact of NSP. The present study provides evidence that NSP exerts a major anticancer effect on DENA-induced HCC. SOR/NSP is a promising combination for tumor suppression and overcoming sorafenib resistance in HCC by modulating antioxidants, anti-inflammatory signals, and tumor markers.
Collapse
Affiliation(s)
- Heba I Ghamry
- Department of Home Economics, College of Home Economics, King Khalid University, P.O. Box 960, Abha 61421, Saudi Arabia
| |
Collapse
|
9
|
Regorafenib and Ruthenium Complex Combination Inhibit Cancer Cell Growth by Targeting PI3K/AKT/ERK Signalling in Colorectal Cancer Cells. Int J Mol Sci 2022; 24:ijms24010686. [PMID: 36614133 PMCID: PMC9820863 DOI: 10.3390/ijms24010686] [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] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 01/03/2023] Open
Abstract
Cancer is one of the leading cause of lethality worldwide, CRC being the third most common cancer reported worldwide, with 1.85 million cases and 850,000 deaths annually. As in all other cancers, kinases are one of the major enzymes that play an essential role in the incidence and progression of CRC. Thus, using multi-kinase inhibitors is one of the therapeutic strategies used to counter advanced-stage CRC. Regorafenib is an FDA-approved drug in the third-line therapy of refractory metastatic colorectal cancer. Acquired resistance to cancers and higher toxicity of these drugs are disadvantages to the patients. To counter this, combination therapy is used as a strategy where a minimal dose of drugs can be used to get a higher efficacy and reduce drug resistance development. Ruthenium-based compounds are observed to be a potential alternative to platinum-based drugs due to their significant safety and effectiveness. Formerly, our lab reported Ru-1, a ruthenium-based compound, for its anticancer activity against multiple cancer cells, such as HepG2, HCT116, and MCF7. This study evaluates Ru-1's activity against regorafenib-resistant HCT116 cells and as a combination therapeutic with regorafenib. Meanwhile, the mechanism of the effect of Ru-1 alone and with regorafenib as a combination is still unknown. In this study, we tested a drug combination (Ru-1 and regorafenib) against a panel of HT29, HCT116, and regorafenib-resistant HCT116 cells. The combination showed a synergistic inhibitory activity. Several mechanisms underlying these numerous synergistic activities, such as anti-proliferative efficacy, indicated that the combination exhibited potent cytotoxicity and enhanced apoptosis induction. Disruption of mitochondrial membrane potential increased intracellular ROS levels and decreased migratory cell properties were observed. The combination exhibited its activity by regulating PI3K/Akt and p38 MAP kinase signalling. This indicates that the combination of REG/Ru-1 targets cancer cells by modulating the PI3K/Akt and ERK signalling.
Collapse
|
10
|
Pan B, Wei X, Xu X. Patient-derived xenograft models in hepatopancreatobiliary cancer. Cancer Cell Int 2022; 22:41. [PMID: 35090441 PMCID: PMC8796540 DOI: 10.1186/s12935-022-02454-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 01/04/2022] [Indexed: 12/20/2022] Open
Abstract
Animal models are crucial tools for evaluating the biological progress of human cancers and for the preclinical investigation of anticancer drugs and cancer prevention. Various animals are widely used in hepatopancreatobiliary cancer research, and mouse models are the most popular. Generally, genetic tools, graft transplantation, and chemical and physical measures are adopted to generate sundry mouse models of hepatopancreatobiliary cancer. Graft transplantation is commonly used to study tumour progression. Over the past few decades, subcutaneous or orthotopic cell-derived tumour xenograft models (CDX models) have been developed to simulate distinct tumours in patients. However, two major limitations exist in CDX models. One model poorly simulates the microenvironment of tumours in humans, such as the vascular, lymphatic and immune environments. The other model loses genetic heterogeneity compared with the corresponding primary tumour. Increased efforts have focused on developing better models for hepatopancreatobiliary cancer research. Hepatopancreatobiliary cancer is considered a tumour with high molecular heterogeneity, making precision medicine challenging in cancer treatment. Developing a new animal model that can better mimic tumour tissue and more accurately predict the efficacy of anticancer treatments is urgent. For the past several years, the patient-derived xenograft model (PDX model) has emerged as a promising tool for translational research. It can retain the genetic and histological stability of their originating tumour at limited passages and shed light on precision cancer medicine. In this review, we summarize the methodology, advantages/disadvantages and applications of PDX models in hepatopancreatobiliary cancer research.
Collapse
|
11
|
Zhao Y, Liu Y, Zhou L, Du GS, He Q. Trends of rapamycin in survival benefits of liver transplantation for hepatocellular carcinoma. World J Gastrointest Surg 2021; 13:953-966. [PMID: 34621472 PMCID: PMC8462078 DOI: 10.4240/wjgs.v13.i9.953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/17/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
The proportion of liver transplantation (LT) for hepatocellular carcinoma (HCC) has kept on increasing over the past years and account for 20%-40% of all LT. Post-transplant HCC recurrence is considered the most important factor affecting the long-term survival of patients. The use of different types of immunosuppressive agents after LT is closely associated with an increased risk for HCC recurrence. The most commonly used conventional immunosuppressive drugs include the calcineurin inhibitors tacrolimus (FK506) and mammalian target of rapamycin inhibitor rapamycin (RAPA). Compared with tacrolimus, RAPA may carry an advantage in survival benefit because of its anti-tumor effects. However, no sufficient evidence to date has proven that RAPA could increase long-term recurrence-free survival and its anti-tumor mechanism of combined therapy remains incompletely clear. In this review, we will focus on recent advances in clinical application experience and basic research results of RAPA in patients undergoing LT for HCC to further guide the clinical practice.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yu Liu
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Lin Zhou
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Guo-Sheng Du
- Department of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Qiang He
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
12
|
Gao S, Ni Q, Wu X, Cao T. GHR knockdown enhances the sensitivity of HCC cells to sorafenib. Aging (Albany NY) 2020; 12:18127-18136. [PMID: 32970612 PMCID: PMC7585089 DOI: 10.18632/aging.103625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/04/2020] [Indexed: 01/24/2023]
Abstract
Sorafenib is approved for treatment of advanced hepatocellular carcinoma (HCC) by the Drug Administration. However, the efficacy of sorafenib has become very limited because most tumors have developed resistance to this drug. In this study, we found that sorafenib stimulated GHR expression in HCC cell lines. Thus, GHR might be linked to sorafenib resistance. To verify this hypothesis, we researched the roles of GHR knockdown and sorafenib combination in cell viability, apoptosis, cycle, and migration. The results showed that GHR blockage enhanced sorafenib blocking of cell cycle progression, leading to inhibition of this drug on HCC cell viability, and the improved promoting ability of sorafenib on cell apoptosis. In addition, it was found that GHR knockdown enhanced sorafenib inhibition of cell migration. The synergistic antitumor effects of sorafenib and GHR knockdown combination may be attributed to inhibition of PI3K/AKT/ERK1/2 signaling pathway. In conclusion, the findings suggest that GHR knockdown enhances the sensitivity of HCC cells to sorafenib. and the inactivation of PI3K/AKT/ERK1/2 signaling pathway may be the underlying mechanisms. This highlights the absence of GHR as a promising way to enhance sorafenib efficacy in HCC.
Collapse
Affiliation(s)
- Shuang Gao
- Department of Gastroenterology, The Third Affiliated Hospital of Naval Military Medical University, Shanghai 201800, China
| | - Qianwen Ni
- Department of Gastroenterology, Zhongshan Qingpu Hospital Fudan University, Shanghai 201799, China
| | - Xiuli Wu
- Department of Gastroenterology, Luoyang First People's Hospital, Luoyang 471000, China
| | - Tieliu Cao
- Department of Traditional Chinese Medicine, Minhang Branch, Shanghai Cancer Center, Fudan University, Shanghai 200240, China
| |
Collapse
|
13
|
Abd-Elbaset M, Mansour AM, Ahmed OM, Abo-Youssef AM. The potential chemotherapeutic effect of β-ionone and/or sorafenib against hepatocellular carcinoma via its antioxidant effect, PPAR-γ, FOXO-1, Ki-67, Bax, and Bcl-2 signaling pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:1611-1624. [PMID: 32270258 DOI: 10.1007/s00210-020-01863-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/24/2020] [Indexed: 12/20/2022]
Abstract
Proliferation and apoptosis are two primary driving forces behind the pathogenesis of hepatocellular carcinoma (HCC). HCC is associated with Ki-67 and Bcl-2 overexpression, reduced Bax expression inducing disturbance of equilibrium between cellular proliferation and apoptosis, and exacerbated by reduced expression of PPAR-γ and FOXO-1. Our objective was to examine the mechanism by which the cyclic isoprenoid, β-ionone (βI), attenuated hepatocarcinogenesis and compare its possible anticancer activity with sorafenib (SF) as standard HCC treatment. HCC induction was achieved by supplying Wistar rats with 0.01% diethylnitrosamine (DENA) for 8 consecutive weeks by free access of drinking water. The effects of βI (160 mg/kg/day) administered orally were evaluated by biochemical, oxidative stress, macroscopical, and histopathological analysis. In addition, immunohistochemical assay for localization and expression of Bax and Bcl-2 and RT-PCR for expression levels of PPAR-γ, FOXO-1, and Ki-67 mRNA were performed. βI treatment significantly reduced the incidence, total number, and multiplicity of visible hepatocyte nodules, attenuated LPO, near-normal restoration of all cancer biomarkers, and antioxidant activities, indicating the chemotherapeutic impact of βI. Histopathological analysis of the liver confirmed that further. βI also induced pro-apoptotic protein Bax expression and reduced anti-apoptotic expression of Bcl-2 protein. Moreover, βI induced mRNA expression of tumor suppressor genes (PPAR-γ and FOXO-1) and decreased proliferative marker Ki-67 mRNA expression. For the first time, the present study provides evidence that βI exerts a major anticancer effect on DENA-induced HCC, at least in part, through inhibition of cell proliferation, oxidative stress, and apoptogenic signal induction mediated by downregulation of Bcl-2 and upregulation of Bax, PPAR-γ, and FOXO-1 expressions.
Collapse
Affiliation(s)
- Mohamed Abd-Elbaset
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, P.O. Box 62514, Beni-Suef, Egypt.
| | - Ahmed M Mansour
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Boys), Al-Azhar University, P.O. Box 11884, Cairo, Egypt
| | - Osama M Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Amira M Abo-Youssef
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, P.O. Box 62514, Beni-Suef, Egypt
| |
Collapse
|
14
|
Chen CY, Fang JY, Chen CC, Chuang WY, Leu YL, Ueng SH, Wei LS, Cheng SF, Hsueh C, Wang TH. 2-O-Methylmagnolol, a Magnolol Derivative, Suppresses Hepatocellular Carcinoma Progression via Inhibiting Class I Histone Deacetylase Expression. Front Oncol 2020; 10:1319. [PMID: 32850418 PMCID: PMC7431949 DOI: 10.3389/fonc.2020.01319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/24/2020] [Indexed: 12/24/2022] Open
Abstract
Magnolia officinalis is widely used in Southeast Asian countries for the treatment of fever, headache, diarrhea, and stroke. Magnolol is a phenolic compound extracted from M. officinalis, with proven antibacterial, antioxidant, anti-inflammatory, and anticancer activities. In this study, we modified magnolol to synthesize a methoxylated derivative, 2-O-methylmagnolol (MM1), and investigated the use of MM1, and magnolol in the treatment of liver cancer. We found that both magnolol and MM1 exhibited inhibitory effects on the growth, migration, and invasion of hepatocellular carcinoma (HCC) cell lines and halted the cell cycle at the G1 phase. MM1 also demonstrated a substantially better tumor-suppressive effect than magnolol. Further analysis suggested that by inhibiting class I histone deacetylase expression in HCC cell lines, magnolol and MM1 induced p21 expression and p53 activation, thereby causing cell cycle arrest and inhibiting HCC cell growth, migration, and invasion. Subsequently, we verified the significant tumor-suppressive effects of magnolol and MM1 in an animal model. Collectively, these findings demonstrate the anti-HCC activities of magnolol and MM1 and their potential for clinical use.
Collapse
Affiliation(s)
- Chi-Yuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan
| | - Jia-You Fang
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan City, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Chin-Chuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Natural Products, Chang Gung University, Taoyuan City, Taiwan
| | - Wen-Yu Chuang
- Department of Anatomic Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan City, Taiwan
| | - Yann-Lii Leu
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan City, Taiwan.,Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan.,Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Shir-Hwa Ueng
- Department of Anatomic Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan City, Taiwan
| | - Li-Shan Wei
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Shu-Fang Cheng
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Natural Products, Chang Gung University, Taoyuan City, Taiwan
| | - Chuen Hsueh
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Department of Anatomic Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan City, Taiwan
| | - Tong-Hong Wang
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan.,Department of Hepato-Gastroenterology, Liver Research Center, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| |
Collapse
|
15
|
Rodríguez-Hernández MA, de la Cruz-Ojeda P, Gallego P, Navarro-Villarán E, Staňková P, Del Campo JA, Kučera O, Elkalaf M, Maseko TE, Červinková Z, Muntané J. Dose-dependent regulation of mitochondrial function and cell death pathway by sorafenib in liver cancer cells. Biochem Pharmacol 2020; 176:113902. [PMID: 32156660 DOI: 10.1016/j.bcp.2020.113902] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/03/2020] [Indexed: 01/14/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and the fourth most frequent cause of cancer-related death worldwide. Sorafenib is the first line recommended therapy for patients with locally advanced/metastatic HCC. The low response rate is attributed to intrinsic resistance of HCC cells to Sorafenib. The potential resistance to Sorafenib-induced cell death is multifactorial and involves all hallmarks of cancer. However, the presence of sub-therapeutic dose can negatively influence the antitumoral properties of the drug. In this sense, the present study showed that the sub-optimal Sorafenib concentration (10 nM) was associated with activation of caspase-9, AMP-activated protein kinase (AMPK), sustained autophagy, peroxisome proliferator-activated receptor-coactivator 1α (PGC-1α) and mitochondrial function in HepG2 cells. The increased mitochondrial respiration by Sorafenib (10 nM) was also observed in permeabilized HepG2 cells, but not in isolated rat mitochondria, which suggests the involvement of an upstream component in this regulatory mechanism. The basal glycolysis was dose dependently increased at early time point studied (6 h). Interestingly, Sorafenib increased nitric oxide (NO) generation that played an inhibitory role in mitochondrial respiration in sub-therapeutic dose of Sorafenib. The administration of sustained therapeutic dose of Sorafenib (10 µM, 24 h) induced mitochondrial dysfunction and dropped basal glycolysis derived acidification, as well as increased oxidative stress and apoptosis in HepG2. In conclusion, the accurate control of the administered dose of Sorafenib is relevant for the potential prosurvival or proapoptotic properties induced by the drug in liver cancer cells.
Collapse
Affiliation(s)
- María A Rodríguez-Hernández
- Institute of Biomedicine of Seville (IBIS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain
| | - Patricia de la Cruz-Ojeda
- Institute of Biomedicine of Seville (IBIS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Paloma Gallego
- Institute of Biomedicine of Seville (IBIS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Elena Navarro-Villarán
- Institute of Biomedicine of Seville (IBIS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain
| | - Pavla Staňková
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic; COST-European Cooperation in Science & Technology, Mitoeagle Action number: CA15203, Brussels, Belgium
| | - José A Del Campo
- Institute of Biomedicine of Seville (IBIS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain
| | - Otto Kučera
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic; COST-European Cooperation in Science & Technology, Mitoeagle Action number: CA15203, Brussels, Belgium
| | - Moustafa Elkalaf
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Tumisang E Maseko
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Zuzana Červinková
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic; COST-European Cooperation in Science & Technology, Mitoeagle Action number: CA15203, Brussels, Belgium
| | - Jordi Muntané
- Institute of Biomedicine of Seville (IBIS), IBiS/Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain; CENTRO DE INVESTIGACIÓN BIOMÉDICA EN RED de Enfermedades Hepáticas y Digestivas (CIBERehd), Spain; COST-European Cooperation in Science & Technology, Mitoeagle Action number: CA15203, Brussels, Belgium; Department of General Surgery, "Virgen del Rocío" University Hospital/IBiS/CSIC/University of Seville, Seville, Spain.
| |
Collapse
|
16
|
The Synergistic Anti-Cancer Effects of NVP-BEZ235 and Regorafenib in Hepatocellular Carcinoma. Molecules 2020; 25:molecules25102454. [PMID: 32466169 PMCID: PMC7287658 DOI: 10.3390/molecules25102454] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/18/2020] [Accepted: 05/24/2020] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver cancer worldwide. Regorafenib is a multi-kinase inhibitor and the second-line treatment for HCC. Since the PI3K/Akt/mTOR signaling pathway is dysregulated in HCC, we evaluated the therapeutic effects of regorafenib combined with a dual PI3K/mTOR inhibitor BEZ235 in the human HCC cell lines (n = 3). The combined treatment with BEZ235 and regorafenib enhanced the inhibition of cell proliferation and increased the expression of cleaved caspase-3 and cleaved PARP in HCC cells. Moreover, the combined treatment suppressed HCC cell migration and invasion in the transwell assay. Further, the Western blot analyses confirmed the involvement of epithelial-mesenchymal transition (EMT)-related genes such as slug, vimentin, and matrix metalloproteinase (MMP)-9/-2. Additionally, the proteinase activity of MMP-9/-2 was analyzed using gelatin zymography. Furthermore, the inhibition of phosphorylation of the Akt, mTOR, p70S6K, and 4EBP1 after combined treatment was validated using Western blot analysis. Therefore, these results suggest that the combined treatment with BEZ235 and regorafenib benefits patients with HCC.
Collapse
|
17
|
Rodríguez-Hernández MA, Chapresto-Garzón R, Cadenas M, Navarro-Villarán E, Negrete M, Gómez-Bravo MA, Victor VM, Padillo FJ, Muntané J. Differential effectiveness of tyrosine kinase inhibitors in 2D/3D culture according to cell differentiation, p53 status and mitochondrial respiration in liver cancer cells. Cell Death Dis 2020; 11:339. [PMID: 32382022 PMCID: PMC7206079 DOI: 10.1038/s41419-020-2558-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/24/2022]
Abstract
Sorafenib and Regorafenib are the recommended first- and second-line therapies in patients with advanced hepatocellular carcinoma (HCC). Lenvatinib and Cabozantinib have shown non-inferior antitumoral activities compared with the corresponding recommended therapies. The clinical trials have established recommended doses for each treatment that lead different blood concentrations in patients for Sorafenib (10 µM), Regorafenib (1 µM), Lenvatinib (0.1 µM), and Cabozantinib (1 µM). However, very low response rates are observed in patients attributed to intrinsic resistances or upregulation of survival signaling. The aim of the study was the comparative dose-response analysis of the drugs (0-100 µM) in well-differentiated (HepG2, Hep3B, and Huh7), moderately (SNU423), and poorly (SNU449) differentiated liver cancer cells in 2D/3D cultures. Cells harbors wild-type p53 (HepG2), non-sense p53 mutation (Hep3B), inframe p53 gene deletion (SNU423), and p53 point mutation (Huh7 and SNU449). The administration of regular used in vitro dose (10 µM) in 3D and 2D cultures, as well as the dose-response analysis in 2D cultures showed Sorafenib and Regorafenib were increasingly effective in reducing cell proliferation, and inducing apoptosis in well-differentiated and expressing wild-type p53 in HCC cells. Lenvatinib and Cabozantinib were particularly effective in moderately to poorly differentiated cells with mutated or lacking p53 that have lower basal oxygen consumption rate (OCR), ATP, and maximal respiration capacity than observed in differentiated HCC cells. Sorafenib and Regorafenib downregulated, and Lenvatinib and Cabozantinib upregulated epidermal growth factor receptor (EGFR) and mesenchymal-epithelial transition factor receptor (c-Met) in HepG2 cells. Conclusions: Sorafenib and Regorafenib were especially active in well-differentiated cells, with wild-type p53 and increased mitochondrial respiration. By contrast, Lenvatinib and Cabozantinib appeared more effective in moderately to poorly differentiated cells with mutated p53 and low mitochondrial respiration. The development of strategies that allow us to deliver increased doses in tumors might potentially enhance the effectiveness of the treatments.
Collapse
Affiliation(s)
- María A Rodríguez-Hernández
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Spanish Network for Biomedical Research in Hepatic and Digestive diseases (CIBERehd), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Raquel Chapresto-Garzón
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Miryam Cadenas
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Elena Navarro-Villarán
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Spanish Network for Biomedical Research in Hepatic and Digestive diseases (CIBERehd), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - María Negrete
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Miguel A Gómez-Bravo
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Spanish Network for Biomedical Research in Hepatic and Digestive diseases (CIBERehd), Institute of Health Carlos III (ISCIII), Madrid, Spain
- Department of General Surgery, Hospital University "Virgen del Rocío"/CSIC/University of Seville/IBIS, Seville, Spain
| | - Victor M Victor
- Spanish Network for Biomedical Research in Hepatic and Digestive diseases (CIBERehd), Institute of Health Carlos III (ISCIII), Madrid, Spain
- Service of Endocrinology, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
- Department of Physiology, University of Valencia, Valencia, Spain
| | - Francisco J Padillo
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Spanish Network for Biomedical Research in Hepatic and Digestive diseases (CIBERehd), Institute of Health Carlos III (ISCIII), Madrid, Spain
- Department of General Surgery, Hospital University "Virgen del Rocío"/CSIC/University of Seville/IBIS, Seville, Spain
| | - Jordi Muntané
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain.
- Spanish Network for Biomedical Research in Hepatic and Digestive diseases (CIBERehd), Institute of Health Carlos III (ISCIII), Madrid, Spain.
- Department of General Surgery, Hospital University "Virgen del Rocío"/CSIC/University of Seville/IBIS, Seville, Spain.
| |
Collapse
|
18
|
Downregulation of thioredoxin-1-dependent CD95 S-nitrosation by Sorafenib reduces liver cancer. Redox Biol 2020; 34:101528. [PMID: 32388267 PMCID: PMC7210585 DOI: 10.1016/j.redox.2020.101528] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) represents 80% of the primary hepatic neoplasms. It is the sixth most frequent neoplasm, the fourth cause of cancer-related death, and 7% of registered malignancies. Sorafenib is the first line molecular targeted therapy for patients in advanced stage of HCC. The present study shows that Sorafenib exerts free radical scavenging properties associated with the downregulation of nuclear factor E2-related factor 2 (Nrf2)-regulated thioredoxin 1 (Trx1) expression in liver cancer cells. The experimental downregulation and/or overexpression strategies showed that Trx1 induced activation of nitric oxide synthase (NOS) type 3 (NOS3) and S-nitrosation (SNO) of CD95 receptor leading to an increase of caspase-8 activity and cell proliferation, as well as reduction of caspase-3 activity in liver cancer cells. In addition, Sorafenib transiently increased mRNA expression and activity of S-nitrosoglutathione reductase (GSNOR) in HepG2 cells. Different experimental models of hepatocarcinogenesis based on the subcutaneous implantation of HepG2 cells in nude mice, as well as the induction of HCC by diethylnitrosamine (DEN) confirmed the relevance of Trx1 downregulation during the proapoptotic and antiproliferative properties induced by Sorafenib. In conclusion, the induction of apoptosis and antiproliferative properties by Sorafenib were related to Trx1 downregulation that appeared to play a relevant role on SNO of NOS3 and CD95 in HepG2 cells. The transient increase of GSNOR might also participate in the deactivation of CD95-dependent proliferative signaling in liver cancer cells. Sorafenib induces mitochondrial ROS generation, but also acts as nucleophilic scavenger. Sorafenib reduces Nrf2-depenent Trx1 expression, and SNO–NOS3 and SNO-CD95 ratios. Sorafenib-related antitumoral in vivo activity involves diminution of Trx1 and SNO-CD95.
Collapse
|
19
|
MEK inhibition by cobimetinib suppresses hepatocellular carcinoma and angiogenesis in vitro and in vivo. Biochem Biophys Res Commun 2019; 523:147-152. [PMID: 31836141 DOI: 10.1016/j.bbrc.2019.12.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 12/05/2019] [Indexed: 01/10/2023]
Abstract
Hepatocellular carcinoma which is featured with the extensive vascularization is the third most frequent cause of cancer-related deaths with limited therapeutic options, particularly for advanced disease. Cobimetinib, a MEK inhibitor, has been approved for the treatment of melanomas with a BRAF mutation. In this work, we investigated the efficacy of cobimetinib in sensitive and resistant HCC cells. Using a panel of HCC cell lines and normal hepatocellular cells as control, we showed that cobimetinib is active against HCC cells and spare normal hepatocellular cells. Cobimetinib at nanomolar concentration inhibited proliferation and induced apoptosis in sorafenib-resistant HCC cells (Hep3B-r), suggesting its ability to overcome HCC resistance to standard of care. This was further demonstrated by our results that cobimetinib significantly augmented the inhibitory effects of sorafenib and doxorubicin in HCC cells. Notably, cobimetinib dose-dependently inhibited tumor angiogenesis by inhibiting HCC endothelial cell (HCCEC) growth, survival and capillary network work formation. Cobimetinib suppressed ERK/RSK without affecting JNK or p38 signaling pathways in Hep3B-r and HCCEC cells. In addition, cobimetinib negatively influenced the apoptosis pathways by increasing pro-apoptotic protein Bim and decreasing anti-apoptotic proteins Mcl-1 and Bcl-2. In addition, we validated the in vitro findings in HCC xenograft mouse model and demonstrated that cobimetinib inhibited ERK signaling, promoted apoptosis, and was active against resistant HCC growth and angiogenesis in vivo, without causing significant toxicity in mice. Our findings support the clinical trials of cobimetinib for HCC treatment and highlight the therapeutic value of inhibiting MEK/ERK/RSK to overcome HCC resistance.
Collapse
|
20
|
Lin CH, Elkholy KH, Wani NA, Li D, Hu P, Barajas JM, Yu L, Zhang X, Jacob ST, Khan WN, Bai XF, Noonan AM, Ghoshal K. Ibrutinib Potentiates Antihepatocarcinogenic Efficacy of Sorafenib by Targeting EGFR in Tumor Cells and BTK in Immune Cells in the Stroma. Mol Cancer Ther 2019; 19:384-396. [PMID: 31582534 DOI: 10.1158/1535-7163.mct-19-0135] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 08/12/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC), the most prevalent primary liver cancer, is a leading cause of cancer-related death worldwide because of rising incidence and limited therapy. Although treatment with sorafenib or lenvatinib is the standard of care in patients with advanced-stage HCC, the survival benefit from sorafenib is limited due to low response rate and drug resistance. Ibrutinib, an irreversible tyrosine kinase inhibitor (TKI) of the TEC (e.g., BTK) and ErbB (e.g., EGFR) families, is an approved treatment for B-cell malignancies. Here, we demonstrate that ibrutinib inhibits proliferation, spheroid formation, and clonogenic survival of HCC cells, including sorafenib-resistant cells. Mechanistically, ibrutinib inactivated EGFR and its downstream Akt and ERK signaling in HCC cells, and downregulated a set of critical genes involved in cell proliferation, migration, survival, and stemness, and upregulated genes promoting differentiation. Moreover, ibrutinib showed synergy with sorafenib or regorafenib, a sorafenib congener, by inducing apoptosis of HCC cells. In vivo, this TKI combination significantly inhibited HCC growth and prolonged survival of immune-deficient mice bearing human HCCLM3 xenograft tumors and immune-competent mice bearing orthotopic mouse Hepa tumors at a dose that did not exhibit systemic toxicity. In immune-competent mice, the ibrutinib-sorafenib combination reduced the numbers of BTK+ immune cells in the tumor microenvironment. Importantly, we found that the BTK+ immune cells were also enriched in the tumor microenvironment in a subset of primary human HCCs. Collectively, our findings implicate BTK signaling in hepatocarcinogenesis and support clinical trials of the sorafenib-ibrutinib combination for this deadly disease.
Collapse
Affiliation(s)
- Cho-Hao Lin
- Department of Pathology, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Khadija H Elkholy
- Department of Pathology, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Nissar A Wani
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
| | - Ding Li
- Department of Pathology, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Peng Hu
- Department of Pathology, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Juan M Barajas
- Department of Pathology, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Lianbo Yu
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Xiaoli Zhang
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Samson T Jacob
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
| | - Wasif N Khan
- Department of Microbiology & Immunology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Xue-Feng Bai
- Department of Pathology, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Anne M Noonan
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Kalpana Ghoshal
- Department of Pathology, The Ohio State University, Columbus, Ohio. .,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
21
|
Zheng JF, He S, Zeng Z, Gu X, Cai L, Qi G. PMPCB Silencing Sensitizes HCC Tumor Cells to Sorafenib Therapy. Mol Ther 2019; 27:1784-1795. [PMID: 31337603 PMCID: PMC6822227 DOI: 10.1016/j.ymthe.2019.06.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/10/2019] [Accepted: 06/19/2019] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) tumors invariably develop resistance to cytotoxic and targeted agents, resulting in failed treatment and tumor recurrence. Previous in vivo short hairpin RNA (shRNA) screening evidence revealed mitochondrial-processing peptidase (PMPC) as a leading gene contributing to tumor cell resistance against sorafenib, a multikinase inhibitor used to treat advanced HCC. Here, we investigated the contributory role of the β subunit of PMPC (PMPCB) in sorafenib resistance. Silencing PMPCB increased HCC tumor cell susceptibility to sorafenib therapy, decreased liver tumor burden, and improved survival of tumor-bearing mice receiving sorafenib. Moreover, sorafenib + PMPCB shRNA combination therapy led to attenuated liver tumor burden and improved survival outcome for tumor-bearing mice, and it reduced colony formation in murine and human HCC cell lines in vitro. Additionally, PMPCB silencing enhanced PINK1-Parkin signaling and downregulated the anti-apoptotic protein MCL-1 in sorafenib-treated HCC cells, which is indicative of a healthier pro-apoptotic phenotype. Higher pre-treatment MCL-1 expression was associated with inferior survival outcomes in sorafenib-treated HCC patients. Elevated MCL-1 expression was present in sorafenib-resistant murine HCC cells, while MCL-1 knockdown sensitized these cells to sorafenib. In conclusion, our findings advocate combination regimens employing sorafenib with PMPCB knockdown or MCL-1 knockdown to circumvent sorafenib resistance in HCC patients.
Collapse
Affiliation(s)
- Jian-Feng Zheng
- Department of Laboratory Medicine, Baoan Central Hospital of Shenzhen, The Fifth Affiliated Hospital of Shenzhen University, Shenzhen 518102, Guangdong, P.R. China.
| | - Shaozhong He
- Department of Oncology, Baoan Central Hospital of Shenzhen, The Fifth Affiliated Hospital of Shenzhen University, Shenzhen 518102, Guangdong, P.R. China
| | - Zongyue Zeng
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Xinqi Gu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, P.R. China
| | - Lei Cai
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, P.R. China
| | - Guangying Qi
- Department of Pathology and Physiopathology, Guilin Medical University, Guilin 541004, Guangxi, P.R. China.
| |
Collapse
|
22
|
Preclinical efficacy of a novel dual PI3K/mTOR inhibitor, CMG002, alone and in combination with sorafenib in hepatocellular carcinoma. Cancer Chemother Pharmacol 2019; 84:809-817. [PMID: 31385002 DOI: 10.1007/s00280-019-03918-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/29/2019] [Indexed: 12/23/2022]
Abstract
PURPOSE Sorafenib has been the only first systemic drug that improves survival of patients with advanced hepatocellular carcinoma (HCC). However, because the response rate of sorafenib is relatively low, novel therapeutic strategies are needed to improve survival in patients with HCC. This study investigated the effect of CMG002 alone and in combination with sorafenib on HCC in vitro and vivo. METHODS The effect of a newly developed dual PI3K/mTOR inhibitor, CMG002, on the proliferation of Huh-7 and HepG2 HCC cells was investigated using the MTT assay. Western blotting was performed to assess phosphorylation of the key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. HepG2 cells were inoculated into mice, which were treated with vehicle, sorafenib, CMG002, and their combinations. Tumor cell proliferation and tumor angiogenesis were evaluated by immunohistochemical analysis of Ki-67 and CD31, respectively. Tumor cell apoptosis was detected by the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Levels of key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways were evaluated by western blot analysis. RESULTS The combination of sorafenib and CMG002 additively inhibited Huh-7 and HepG2 cell proliferation compared to single-agent treatment. Sorafenib and CMG002 as single agents differentially inhibited or activated key enzymes in the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. The combination of sorafenib and CMG002 inhibited all key enzymes in the two pathways. Treatment with CMG002 for 4 weeks alone and in combination with sorafenib strongly inhibited tumor growth. CMG002 inhibited HCC cell proliferation, induced apoptosis, and decreased tumor angiogenesis. Furthermore, these effects were enhanced when CMG002 was combined with sorafenib. CONCLUSIONS The combination of CMG002 and sorafenib significantly inhibited HCC cell proliferation and tumorigenesis by inhibiting the Ras/Raf/MAPK and PI3K/AKT/mTOR pathways. These findings suggest that CMG002 to be a potential novel candidate treatment for HCC.
Collapse
|
23
|
Pan J, Lu C, Jun W, Wu Y, Shi X, Ding Y. The up-regulation of P62 levels is associated with resistance of sorafenib in hepatocarcinoma cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:2622-2630. [PMID: 31934090 PMCID: PMC6949556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 05/22/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Sorafenib is one of the most commonly used systemic therapies for hepatocellular carcinoma (HCC), but the acquired resistance towards sorafenib found in HCC patients usually led to failure of treatment and poor prognosis. Therefore, there is an urgent need to study the molecular mechanism caused by the acquired resistance. Previous studies demonstrated that P62 plays an important role in tumor cell resistance towards systemic therapies including chemotherapy and targeted therapy. However, the role of P62 in acquired resistance to sorafenib in HCC has not been clearly investigated. MATERIALS AND METHODS In this study we screened the most sensitive HCC cell lines towards sorafenib using CCK8. Then on this cell line, we analyzed the relationship between P62 expression level and the sensitivity towards sorafenib by western blot and CCK8. After knockdown and overexpression of P62 in HCC cells, cells were then treated with sorafenib. After that, we detect changes of sensitivity towards sorafenib. HCC samples were used to investigate the expression of P62 and their survival time. RESULTS Among four HCC cell lines in our lab, HepG2 cell line with the highest sensitivity to sorafenib was screened and selected. After treatment with sorafenib, the expression of P62 was significantly increased. In HCC cells, we found that significant up-regulation of P62 was correlated with the reduction of sorafenib sensitivity. In HCC samples, we found that the expression of P62 was associated with sorafenib resistance and a shorter survival time. CONCLUSION The up-regulation of P62 could reduce the sensitivity of HCC towards sorafenib. Thus, P62 could be therapeutic target to overcome sorafenib acquired resistance in the future.
Collapse
Affiliation(s)
- Junping Pan
- Department of Hepatobiliary Surgery, The Drum Tower Clinical Medical College, Nanjing Medical UniversityNanjing, Jiangsu Province, China
| | - Chenglin Lu
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical SchoolNanjing, Jiangsu Province, China
| | - Wang Jun
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical SchoolNanjing, Jiangsu Province, China
| | - Yafu Wu
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical SchoolNanjing, Jiangsu Province, China
| | - Xiaolei Shi
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical SchoolNanjing, Jiangsu Province, China
| | - Yitao Ding
- Department of Hepatobiliary Surgery, The Drum Tower Clinical Medical College, Nanjing Medical UniversityNanjing, Jiangsu Province, China
| |
Collapse
|
24
|
6PGD inhibition sensitizes hepatocellular carcinoma to chemotherapy via AMPK activation and metabolic reprogramming. Biomed Pharmacother 2019; 111:1353-1358. [PMID: 30841449 DOI: 10.1016/j.biopha.2019.01.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 12/25/2022] Open
Abstract
Better understanding of the molecular mechanism involved in hepatocellular carcinoma (HCC) progression is essential for the development of therapeutic strategies to overcome chemoresistance in HCC patients. In this work, we show that 6-phosphogluconate dehydrogenase (6PGD), a key enzyme of the oxidative pentose phosphate pathway, is important for HCC growth and survival. Compared to normal liver tissues, we demonstrate that 6PGD expression is upregulated in HCC tissues. 6PGD overexpression increases 6PGD activity and promotes growth in normal liver cells. In contrast, targeting 6PGD using both genetic and pharmacological approaches inhibits HCC growth and survival. Combination of chemotherapeutic agents with 6PGD inhibition achieves greater efficacy in inhibiting HCC growth and survival than chemotherapeutic agent alone. We further show that inhibition of 6PGD activates AMP-activated protein kinase (AMPK) and acetyl-coenzyme A carboxylase 1 (ACC1), and decreases level of NADPH/NAD + and NADH in HCC, leading to SIRT1 activity reduction and oxidative stress. Conversely, AMPK depletion significantly abolishes the effects of physcion (a selective small-molecule 6PGD inhibitor) in decreasing NADPH/NAD + ratio, growth and survival, confirming the role of AMPK as the relevant upstream activator with 6PGD inhibition in HCC cells. Our work is the first to demonstrate the upregulation of 6PGD and its critical involvement in growth and survival in HCC. Our findings suggest 6PGD as a promising therapeutic target to overcome chemoresistance in HCC.
Collapse
|
25
|
Kim YS, Lee YM, Oh TI, Shin DH, Kim GH, Kan SY, Kang H, Kim JH, Kim BM, Yim WJ, Lim JH. Emodin Sensitizes Hepatocellular Carcinoma Cells to the Anti-Cancer Effect of Sorafenib through Suppression of Cholesterol Metabolism. Int J Mol Sci 2018; 19:ijms19103127. [PMID: 30321984 PMCID: PMC6213641 DOI: 10.3390/ijms19103127] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/01/2018] [Accepted: 10/10/2018] [Indexed: 12/12/2022] Open
Abstract
Reduced therapeutic efficacy of sorafenib, a first-generation multikinase inhibitor, is often observed during the treatment of advanced hepatocellular carcinoma (HCC). Emodin is an active component of Chinese herbs, and is effective against leukemia, lung cancer, colon cancer, pancreatic cancer, and HCC; however, the sensitizing effect of emodin on sorafenib-based HCC therapy has not been evaluated. Here, we demonstrate that emodin significantly improved the anti-cancer effect of sorafenib in HCC cells, such as HepG2, Hep3B, Huh7, SK-HEP-1, and PLC/PRF5. Mechanistically, emodin inhibits sterol regulatory element-binding protein-2 (SREBP-2) transcriptional activity, which suppresses cholesterol biosynthesis and oncogenic protein kinase B (AKT) signaling. Additionally, attenuated cholesterol synthesis and oncogenic AKT signaling inactivated signal transducer and activator of transcription 3 (STAT3), an oncogenic transcription factor. Furthermore, emodin synergistically increased cell cycle arrest in the G1 phase and apoptotic cells in the presence of sorafenib. Animal models xenografted with HepG2 or SK-HEP-1 cells also showed that the combination of emodin and sorafenib was sufficient to inhibit tumor growth. Overall, these results suggested that the combination of emodin and sorafenib may offer a potential therapy for patients with advanced HCC.
Collapse
Affiliation(s)
- Young-Seon Kim
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Yoon-Mi Lee
- Department of Food Bioscience, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Taek-In Oh
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Dong Hoon Shin
- Research Institute, National Cancer Center, Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang 10408, Korea.
| | - Geon-Hee Kim
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Sang-Yeon Kan
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Hyeji Kang
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
| | - Ji Hyung Kim
- College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea.
| | - Byeong Mo Kim
- Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases (SIRIC), Yonsei University College of Medicine, Seodaemun-gu, Seoul 03722, Korea.
| | - Woo Jong Yim
- Jung-Ang Microbe Research Institute (JM), 398, Jikji-daero, Heungdeok-gu, Cheongju 28576, Chungbuk, Korea.
| | - Ji-Hong Lim
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Chungbuk, Korea.
- Diabetes and Bio-Research Center, Konkuk University, Chungju 27478, Chungbuk, Korea.
| |
Collapse
|
26
|
Thompson EM, Landi D, Ashley D, Keir ST, Bigner D. Bevacizumab, irinotecan, temozolomide, tyrosine kinase inhibition, and MEK inhibition are effective against pleomorphic xanthoastrocytoma regardless of V600E status. J Neurooncol 2018; 140:261-268. [DOI: 10.1007/s11060-018-2975-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 08/09/2018] [Indexed: 02/04/2023]
|
27
|
Jung DH, Tak E, Hwang S, Song GW, Ahn CS, Kim KH, Moon DB, Ha TY, Park GC, Ryoo BY, Lee KJ, Kim N, Kwon JH, Jwa EK, Lee SG. Antitumor effect of sorafenib and mammalian target of rapamycin inhibitor in liver transplantation recipients with hepatocellular carcinoma recurrence. Liver Transpl 2018; 24:932-945. [PMID: 29710388 DOI: 10.1002/lt.25191] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/23/2017] [Accepted: 04/13/2018] [Indexed: 12/12/2022]
Abstract
Both sorafenib and mammalian target of rapamycin inhibitor (mTORi) have antitumor effects. This study aimed to evaluate their antitumor effects in liver transplantation (LT) recipients with hepatocellular carcinoma (HCC) recurrence. We performed a laboratory study using sorafenib and mTORi and subsequently validated their survival benefit in a clinical LT setting. In the laboratory study, the HepG2.2.15 liver tumor cell line and 5 patient-derived graft HCC cell lines were used for in vitro cytotoxic studies. After treatment with everolimus and sorafenib, cell viability and apoptosis assays revealed noticeable cytotoxic effects with individual agents and augmented effects by combination therapy. An in vivo mouse study also demonstrated similar cytotoxic outcomes. In the clinical study including 232 LT recipients with HCC recurrence, the 3-month medication drop-out rate was 35.6% for sorafenib administration and 23.5% for mTORi administration. Postrecurrence survival rates were not different according to sorafenib administration (P = 0.17) but were significantly improved following mTORi administration (P < 0.001). In mTORi subgroups with and without sorafenib, there was no difference in the overall postrecurrence patient survival period (P = 0.26), indicating an absence of synergistic or additional antitumor effect from sorafenib. The median progression-free and overall survival period was 6.4 and 11.8 months, respectively, after sorafenib administration. Time of tumor recurrence and use of mTORi were independent risk factors. In conclusion, our laboratory study demonstrated synergistic antitumor effects of sorafenib and mTORi, but this was not reproduced in our clinical LT study. Our clinical result of mTORi administration showed improved postrecurrence survival, thus administering mTORi in LT recipients with HCC recurrence appears worthwhile. However, the antitumor effect of sorafenib on posttransplant recurrence was not determined in this retrospective study, thus requiring further studies with early start of sorafenib administration. Liver Transplantation 24 932-945 2018. © 2018 AASLD.
Collapse
Affiliation(s)
- Dong-Hwan Jung
- Division of Hepatobiliary Surgery and Liver Transplantation, Departments of Surgery
| | - Eunyoung Tak
- Asan Institute of Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Shin Hwang
- Division of Hepatobiliary Surgery and Liver Transplantation, Departments of Surgery
| | - Gi-Won Song
- Division of Hepatobiliary Surgery and Liver Transplantation, Departments of Surgery
| | - Chul-Soo Ahn
- Division of Hepatobiliary Surgery and Liver Transplantation, Departments of Surgery
| | - Ki-Hun Kim
- Division of Hepatobiliary Surgery and Liver Transplantation, Departments of Surgery
| | - Deok-Bog Moon
- Division of Hepatobiliary Surgery and Liver Transplantation, Departments of Surgery
| | - Tae-Yong Ha
- Division of Hepatobiliary Surgery and Liver Transplantation, Departments of Surgery
| | - Gil-Chun Park
- Division of Hepatobiliary Surgery and Liver Transplantation, Departments of Surgery
| | | | - Kyung Jin Lee
- Asan Institute of Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Nayoung Kim
- Asan Institute of Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae-Hyeon Kwon
- Division of Hepatobiliary Surgery and Liver Transplantation, Departments of Surgery
| | - Eun-Kyoung Jwa
- Division of Hepatobiliary Surgery and Liver Transplantation, Departments of Surgery
| | - Sung-Gyu Lee
- Division of Hepatobiliary Surgery and Liver Transplantation, Departments of Surgery
| |
Collapse
|
28
|
Yin X, Xiao Y, Han L, Zhang B, Wang T, Su Z, Zhang N. Ceramide-Fabricated Co-Loaded Liposomes for the Synergistic Treatment of Hepatocellular Carcinoma. AAPS PharmSciTech 2018; 19:2133-2143. [PMID: 29714002 DOI: 10.1208/s12249-018-1005-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/24/2018] [Indexed: 02/06/2023] Open
Abstract
Combination therapy is one of the important methods to improve therapeutic effect on the treatment of hepatocellular carcinoma (HCC). Sorafenib (SF) is a canonical US Food and Drug Administration-approved multikinase molecule inhibitor against HCC. However, therapeutic benefit with Sorafenib alone was usually unsatisfactory. Ceramide (CE) is an endogenous bioactive sphingolipid, which has a strong potential to suppress various tumors. The combination of SF and CE was hoping to exert maximum synergistic antitumor effect through different tumor-suppressible mechanisms. In this respect, SF and CE co-loaded liposomes (SF/CE-liposomes) were developed to verify synergistic antitumor efficacy. The optimal molar ratio of SF and CE was determined through combination index. SF/CE-liposomes were prepared by thin-film hydration method, which exhibited spherical or ellipsoidal shape. Particle size of SF/CE-liposomes was 174 ± 4 nm with homogeneous distribution. Release profile of SF demonstrated that addition of CE imposed no significant impact on the release of SF. SF/CE-liposomes exhibited acceptable stability in different media and desirable storage stability over 30 days at 4°C. In vitro cellular uptake confirmed that SF/CE-liposomes could be efficiently internalized into HepG2 cells. In vitro cytotoxicity evaluation indicated that SF/CE-liposomes exhibited higher cytotoxicity on HepG2 cells. IC50 value of SF/CE-liposomes was 11.5 ± 0.44 μM, which was significantly lower than that of SF-liposomes (**p < 0.01). Evaluation of in vivo synergistic effect on H22-bearing mice verified that SF/CE-liposomes achieved robust antitumor activity in preventing tumor growth. All results suggested that SF/CE-liposomes might be served as an efficient co-delivery system for improving therapeutic efficacy of HCC.
Collapse
|
29
|
Refolo MG, Lippolis C, Carella N, Cavallini A, Messa C, D'Alessandro R. Chlorogenic Acid Improves the Regorafenib Effects in Human Hepatocellular Carcinoma Cells. Int J Mol Sci 2018; 19:ijms19051518. [PMID: 29783729 PMCID: PMC5983789 DOI: 10.3390/ijms19051518] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/14/2018] [Accepted: 05/17/2018] [Indexed: 12/24/2022] Open
Abstract
Chlorogenic acid (CGA) is a polyphenol present in many human dietary foods. Several studies indicated a beneficial role of CGA in the prevention of cancer and an enhancement of chemotherapy when combined with CGA in the treatment of human hepatocarcinoma (HCC). Drug toxicity, resistance and subsequent disease progression represent a problem in HCC management, although treatment with the multikinase inhibitor Regorafenib improved overall survival. This study focused on the evaluation of the effects of combined treatment using both low Regorafenib concentrations and CGA as natural compound in HCC cells. The analysis of cell proliferation by Ki67 staining and cell cycle progression showed that CGA enhanced Regorafenib-mediated cell growth inhibition. Moreover, CGA potentiated the apoptotic effect of Regorafenib by the activation of the pro-apoptotic Annexin V, Bax and Caspase 3/7 and the inhibition of anti-apoptotic Bcl2 and Bcl-xL. Combined treatments were also effective in inhibiting cell motility. The mechanisms underlying the positive effects of combining CGA and Regorafenib were also addressed and an increased inhibition of MAPK (mitogen-activated protein kinase)and PI3K/Akt/mTORC (phosphatidylinositol-3-kinase (PI3K)/Akt and the mammalian target of rapamycin (mTOR) signaling was observed. Overall, these data demonstrated that co-treatment with Regorafenib and CGA enhanced Regorafenib action, reducing its cytotoxicity in HCC cells. In conclusion, this drug combination could be considered as a safe and more effective approach in HCC therapy.
Collapse
Affiliation(s)
- Maria Grazia Refolo
- Laboratory of Cellular and Molecular Biology, Department of Clinical Pathology, National Institute for Digestive Diseases, IRCCS "Saverio de Bellis", Via Turi 27, 70013 Castellana Grotte, BA, Italy.
| | - Catia Lippolis
- Laboratory of Cellular and Molecular Biology, Department of Clinical Pathology, National Institute for Digestive Diseases, IRCCS "Saverio de Bellis", Via Turi 27, 70013 Castellana Grotte, BA, Italy.
| | - Nicola Carella
- Laboratory of Cellular and Molecular Biology, Department of Clinical Pathology, National Institute for Digestive Diseases, IRCCS "Saverio de Bellis", Via Turi 27, 70013 Castellana Grotte, BA, Italy.
| | - Aldo Cavallini
- Laboratory of Cellular and Molecular Biology, Department of Clinical Pathology, National Institute for Digestive Diseases, IRCCS "Saverio de Bellis", Via Turi 27, 70013 Castellana Grotte, BA, Italy.
| | - Caterina Messa
- Laboratory of Cellular and Molecular Biology, Department of Clinical Pathology, National Institute for Digestive Diseases, IRCCS "Saverio de Bellis", Via Turi 27, 70013 Castellana Grotte, BA, Italy.
| | - Rosalba D'Alessandro
- Laboratory of Cellular and Molecular Biology, Department of Clinical Pathology, National Institute for Digestive Diseases, IRCCS "Saverio de Bellis", Via Turi 27, 70013 Castellana Grotte, BA, Italy.
| |
Collapse
|
30
|
Machida K. Pluripotency Transcription Factors and Metabolic Reprogramming of Mitochondria in Tumor-Initiating Stem-like Cells. Antioxid Redox Signal 2018; 28:1080-1089. [PMID: 29256636 PMCID: PMC5865250 DOI: 10.1089/ars.2017.7241] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 12/15/2017] [Accepted: 12/16/2017] [Indexed: 12/26/2022]
Abstract
Significance: Neoplasms contain tumor-initiating stem-like cells (TICs) that drive malignant progression and tumor growth with drug resistance. TICs proliferate through a self-renewal process in which the two daughter cells differ in their proliferative potential, with one retaining the self-renewing phenotype and another displaying the differentiated phenotype. Recent Advances: Cancer traits (hepatocellular carcinoma) are triggered by alcoholism, obesity, and hepatitis B or C virus (HBV and HCV), including genetic changes, angiogenesis, defective tumor immunity, immortalization, metabolic reprogramming, excessive and prolonged inflammation, migration/invasion/metastasis, evasion of cell cycle arrest, anticell death, and compensatory regeneration/proliferation. Critical Issues: This review describes how metabolic reprogramming in mitochondria promotes self-renewal and oncogenicity of TICs. Pluripotency transcription factors (TFs), NANOG, OCT4, MYC, and SOX2, contribute to cancer progression by mitochondrial reprogramming, leading to the genesis of TICs and cancer. For example, oxidative phosphorylation (OXPHOS) and fatty acid metabolism are identified as major pathways contributing to pluripotency TF-mediated oncogenesis. Future Directions: Identification of novel metabolic pathways provides potential drug targets for neutralizing the activity of highly malignant TICs found in cancer patients. Antioxid. Redox Signal. 28, 1080-1089.
Collapse
Affiliation(s)
- Keigo Machida
- Department of Molecular Microbiology and Immunology, Southern California Research Center for ALPD and Cirrhosis, University of Southern California Keck School of Medicine, Los Angeles, California
| |
Collapse
|
31
|
Hwang S, Ahn CS, Kim KH, Moon DB, Ha TY, Song GW, Jung DH, Park GC, Lee SG. A cross-sectional analysis of long-term immunosuppressive regimens after liver transplantation at Asan Medical Center: Increased preference for mycophenolate mofetil. Ann Hepatobiliary Pancreat Surg 2018. [PMID: 29536052 PMCID: PMC5845607 DOI: 10.14701/ahbps.2018.22.1.19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Backgrounds/Aims Long-term immunosuppression regimens after liver transplantation (LT) are rarely reported in detail. We aimed to provide information on actual long-term immunosuppression regimens through this cross-sectional study. Methods Our institutional LT database was searched for adult patients who underwent primary LT operation from 2000 to 2016. We identified 3620 live recipients with actual information on immunosuppressive agent use for 1-17 years. Results The study cohort was divided into 7 groups according to posttransplantation period. The immunosuppressive agents used at the cross-sectional review period were tacrolimus in 2884 (79.7%), cyclosporine in 445 (12.3%), mycophenolate mofetil in 2007 (55.4%), and everolimus in 138 (3.8%) recipients. There was no marked difference in immunosuppressive agent use according to pretransplantation liver malignancy or type of LT operation. Tacrolimus, cyclosporine, mycophenolate mofetil, and everolimus were used in 97.4%, 1.8%, 60.9%, and 9.2%, respectively, in the year 2 group; 94.1%, 3.9%, 51.6%, and 8.3%, respectively, in the year 3 group; 87.3%, 8.4%, 68.9%, and 4.8%, respectively, in the year 4-5 group; 78.2%, 12.9%, 64.6%, and 3.0%, respectively, in the year 6-7 group; 76.9%, 10.8%, 58.8%, and 2.4%, respectively, in the year 8-10 group; 66.7%, 22.4%, 43.4%, and 1.5%, respectively, in the year 11-15 group; and 73.8%, 15.4%, 32.9%, and 1.7%, respectively, in the year ≥15 group. Conclusions Tacrolimus and mycophenolate mofetil are the primary immunosuppressive agents after LT, and the indications for everolimus have started to increase at our institution. We believe our results will help establish tailored long-term immunosuppression regimens.
Collapse
Affiliation(s)
- Shin Hwang
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chul-Soo Ahn
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ki-Hun Kim
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Deok-Bog Moon
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Tae-Yong Ha
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Gi-Won Song
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dong-Hwan Jung
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Gil-Chun Park
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung-Gyu Lee
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
32
|
Fong ELS, Toh TB, Lin QXX, Liu Z, Hooi L, Mohd Abdul Rashid MB, Benoukraf T, Chow EKH, Huynh TH, Yu H. Generation of matched patient-derived xenograft in vitro-in vivo models using 3D macroporous hydrogels for the study of liver cancer. Biomaterials 2018; 159:229-240. [PMID: 29353739 DOI: 10.1016/j.biomaterials.2017.12.026] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 12/21/2017] [Accepted: 12/31/2017] [Indexed: 12/28/2022]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer death worldwide, often manifesting at the advanced stage when cure is no longer possible. The discrepancy between preclinical findings and clinical outcome in HCC is well-recognized. So far, sorafenib is the only targeted therapy approved as first-line therapy for patients with advanced HCC. There is an urgent need for improved preclinical models for the development of HCC-targeted therapies. Patient-derived xenograft (PDX) tumor models have been shown to closely recapitulate human tumor biology and predict patient drug response. However, the use of PDX models is currently limited by high costs and low throughput. In this study, we engineered in vitro conditions conducive for the culture of HCC-PDX organoids derived from a panel of 14 different HCC-PDX lines through the use of a three-dimensional macroporous cellulosic sponge system. To validate the in vitro HCC-PDX models, both in vivo and in vitro HCC-PDX models were subjected to whole exome sequencing and RNA-sequencing. Correlative studies indicate strong concordance in genomic and transcriptomic profiles as well as intra-tumoral heterogeneity between each matched in vitro-in vivo HCC-PDX pairs. Furthermore, we demonstrate the feasibility of using these in vitro HCC-PDX models for drug testing, paving the way for more efficient preclinical studies in HCC drug development.
Collapse
Affiliation(s)
- Eliza Li Shan Fong
- Department of Biomedical Engineering, National University of Singapore, Singapore.
| | - Tan Boon Toh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Quy Xiao Xuan Lin
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Zheng Liu
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Lissa Hooi
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Touati Benoukraf
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | | | - Hanry Yu
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Mechanobiology Institute, National University of Singapore, Singapore; BioSyM, Singapore-MIT Alliance for Research and Technology, Singapore; Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China; NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, Singapore
| |
Collapse
|
33
|
Rodríguez-Hernández MA, González R, de la Rosa ÁJ, Gallego P, Ordóñez R, Navarro-Villarán E, Contreras L, Rodríguez-Arribas M, González-Gallego J, Álamo-Martínez JM, Marín-Gómez LM, Del Campo JA, Quiles JL, Fuentes JM, de la Cruz J, Mauriz JL, Padillo FJ, Muntané J. Molecular characterization of autophagic and apoptotic signaling induced by sorafenib in liver cancer cells. J Cell Physiol 2018; 234:692-708. [PMID: 30132846 DOI: 10.1002/jcp.26855] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/10/2018] [Indexed: 12/14/2022]
Abstract
Sorafenib is the unique accepted molecular targeted drug for the treatment of patients in advanced stage of hepatocellular carcinoma. The current study evaluated cell signaling regulation of endoplasmic reticulum (ER) stress, c-Jun-N-terminal kinase (JNK), Akt, and 5'AMP-activated protein kinase (AMPK) leading to autophagy and apoptosis induced by sorafenib. Sorafenib induced early (3-12 hr) ER stress characterized by an increase of Ser51 P-eIF2α/eIF2α, C/EBP homologous protein (CHOP), IRE1α, and sXBP1, but a decrease of activating transcription factor 6 expression, overall temporally associated with the increase of Thr183,Tyr185 P-JNK1/2/JNK1/2, Thr172 P-AMPKα, Ser413 P-Foxo3a, Thr308 P-AKt/AKt and Thr32 P-Foxo3a/Foxo3a ratios, and reduction of Ser2481 P-mammalian target of rapamycin (mTOR)/mTOR and protein translation. This pattern was related to a transient increase of tBid, Bim EL , Beclin-1, Bcl-xL, Bcl-2, autophagy markers, and reduction of myeloid cell leukemia-1 (Mcl-1) expression. The progressive increase of CHOP expression, and reduction of Thr308 P-AKt/AKt and Ser473 P-AKt/AKt ratios were associated with the reduction of autophagic flux and an additional upregulation of Bim EL expression and caspase-3 activity (24 hr). Small interfering-RNA (si-RNA) assays showed that Bim, but not Bak and Bax, was involved in the induction of caspase-3 in sorafenib-treated HepG2 cells. Sorafenib increased autophagic and apoptotic markers in tumor-derived xenograft model. In conclusion, the early sorafenib-induced ER stress and regulation of JNK and AMPK-dependent signaling were related to the induction of survival autophagic process. The sustained drug treatment induced a progressive increase of ER stress and PERK-CHOP-dependent rise of Bim EL , which was associated with the shift from autophagy to apoptosis. The kinetic of Bim EL expression profile might also be related to the tight balance between AKt- and AMPK-related signaling leading to Foxo3a-dependent BIM EL upregulation.
Collapse
Affiliation(s)
- María A Rodríguez-Hernández
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Raúl González
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Ángel J de la Rosa
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Paloma Gallego
- Unit for the Clinical Management of Digestive Diseases, Hospital University "Nuestra Señora de Valme", Seville, Spain
| | - Raquel Ordóñez
- Institute of Biomedicine (IBIOMED), Department of Biomedical Sciences, University of León, León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Elena Navarro-Villarán
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Laura Contreras
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Department of Genetics, University of Seville, Seville, Spain
| | - Mario Rodríguez-Arribas
- Department of Biochemistry, Molecular Biology and Genetics, Faculty of Nursery and Occupational Therapy, University of Extremadura, Cáceres, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Javier González-Gallego
- Institute of Biomedicine (IBIOMED), Department of Biomedical Sciences, University of León, León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - José M Álamo-Martínez
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Department of General Surgery, Hospital University "Virgen del Rocío"/CSIC/University of Seville/IBiS/CSIC/University of Seville, Spain
| | - Luís M Marín-Gómez
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Department of General Surgery, Hospital University "Virgen del Rocío"/CSIC/University of Seville/IBiS/CSIC/University of Seville, Spain
| | - José A Del Campo
- Unit for the Clinical Management of Digestive Diseases, Hospital University "Nuestra Señora de Valme", Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - José L Quiles
- Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, Department of Physiology, University of Granada, Granada, Spain
| | - José M Fuentes
- Department of Biochemistry, Molecular Biology and Genetics, Faculty of Nursery and Occupational Therapy, University of Extremadura, Cáceres, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jesús de la Cruz
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Department of Genetics, University of Seville, Seville, Spain
| | - José L Mauriz
- Institute of Biomedicine (IBIOMED), Department of Biomedical Sciences, University of León, León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Francisco J Padillo
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Department of General Surgery, Hospital University "Virgen del Rocío"/CSIC/University of Seville/IBiS/CSIC/University of Seville, Spain
| | - Jordi Muntané
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Department of General Surgery, Hospital University "Virgen del Rocío"/CSIC/University of Seville/IBiS/CSIC/University of Seville, Spain
| |
Collapse
|
34
|
Tan J, Ye J, Song M, Zhou M, Hu Y. Ribavirin augments doxorubicin's efficacy in human hepatocellular carcinoma through inhibiting doxorubicin-induced eIF4E activation. J Biochem Mol Toxicol 2017; 32. [PMID: 29112301 DOI: 10.1002/jbt.22007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/02/2017] [Accepted: 10/13/2017] [Indexed: 02/06/2023]
Abstract
Activation of eukaryotic translation initiation factor 4E (eIF4E) is a cellular survival mechanism in response to chemotherapy in cancers. In this work, we demonstrate that targeting eIF4E by ribavirin sensitizes hepatocellular carcinoma (HCC) cell response to doxorubicin. Ribavirin inhibits growth and survival of HCC cells, and to a greater extent than in normal liver cells. Its combination with doxorubicin achieves greater efficacy than single drug in vitro and in vivo. Ribavirin suppresses phosphorylation of molecules involved in Akt/mTOR/eIF4E pathway. Overexpression of the phosphomimetic form (S209D) but not the nonphosphorylatable form (S209A) eIF4E significantly reverses the inhibitory effects of ribavirin. Interestingly, doxorubicin significantly increases p-eIF4E(S209) level in a dose- and time-dependent manner, suggesting that doxorubicin induces eIF4E activation in HCC cells. In addition, eIF4E activation induced by doxorubicin in HCC cells is inhibited by ribavirin. Our work demonstrates the greater efficacy of ribavirin and doxorubicin combination and its underlying mechanisms.
Collapse
Affiliation(s)
- Jun Tan
- Department of Hepatology, Ningbo No. 2 Hospital, Ningbo 315010, People's Republic of China
| | - Jingfen Ye
- Department of Hepatology, Ningbo No. 2 Hospital, Ningbo 315010, People's Republic of China
| | - Meijun Song
- Department of Respiratory Medicine, Ningbo Medical Treatment Center Li Huili Hospital, Ningbo 315041, People's Republic of China
| | - Mi Zhou
- School of Medicine, Ningbo University, Ningbo 315211, People's Republic of China
| | - Yaoren Hu
- Department of Hepatology, Ningbo No. 2 Hospital, Ningbo 315010, People's Republic of China
| |
Collapse
|
35
|
Ravaud A, Gomez-Roca C, Picat MQ, Digue L, Chevreau C, Gimbert A, Chauzit E, Sitta R, Cornelis F, Asselineau J, Aziza R, Daste A, Quemener C, Baud J, Bikfalvi A, Pedenon–Périchout D, Doussau A, Molimard M, Delord JP. Phase I study of axitinib and everolimus in metastatic solid tumours and extension to metastatic renal cell carcinoma: Results of EVAX study. Eur J Cancer 2017; 85:39-48. [DOI: 10.1016/j.ejca.2017.07.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/19/2017] [Indexed: 11/25/2022]
|
36
|
Swamy SG, Kameshwar VH, Shubha PB, Looi CY, Shanmugam MK, Arfuso F, Dharmarajan A, Sethi G, Shivananju NS, Bishayee A. Targeting multiple oncogenic pathways for the treatment of hepatocellular carcinoma. Target Oncol 2017; 12:1-10. [PMID: 27510230 DOI: 10.1007/s11523-016-0452-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common forms of liver cancer diagnosed worldwide. HCC occurs due to chronic liver disease and is often diagnosed at advanced stages. Chemotherapeutic agents such as doxorubicin are currently used as first-line agents for HCC therapy, but these are non-selective cytotoxic molecules with significant side effects. Sorafenib, a multi-targeted tyrosine kinase inhibitor, is the only approved targeted drug for HCC patients. However, due to adverse side effects and limited efficacy, there is a need for the identification of novel pharmacological drugs beyond sorafenib. Several agents that target and inhibit various signaling pathways involved in HCC are currently being assessed for HCC treatment. In the present review article, we summarize the diverse signal transduction pathways responsible for initiation as well as progression of HCC and also the potential anticancer effects of selected targeted therapies that can be employed for HCC therapy.
Collapse
Affiliation(s)
- Supritha G Swamy
- Department of Biotechnology, JSS Science and Technology University, JSS Technical Institutions Campus, Mysore, Karnataka, 570006, India
| | - Vivek H Kameshwar
- Department of Biotechnology, JSS Science and Technology University, JSS Technical Institutions Campus, Mysore, Karnataka, 570006, India
| | - Priya B Shubha
- Department of Studies in Chemistry, University of Mysore, Manasagangotri, Mysore, 570 006, Karnataka, India
| | - Chung Yeng Looi
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Frank Arfuso
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Biosciences Research Precinct, Curtin University, Bentley, Western Australia, 6009, Australia
| | - Arunasalam Dharmarajan
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Biosciences Research Precinct, Curtin University, Bentley, Western Australia, 6009, Australia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Biosciences Research Precinct, Curtin University, Bentley, Western Australia, 6009, Australia
| | - Nanjunda Swamy Shivananju
- Department of Biotechnology, JSS Science and Technology University, JSS Technical Institutions Campus, Mysore, Karnataka, 570006, India.
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, 18301 N. Miami Avenue, Miami, FL, 33169, USA.
| |
Collapse
|
37
|
Cao JY, Yin HS, Li HS, Yu XQ, Han X. Interleukin-27 augments the inhibitory effects of sorafenib on bladder cancer cells. ACTA ACUST UNITED AC 2017; 50:e6207. [PMID: 28746469 PMCID: PMC5520222 DOI: 10.1590/1414-431x20176207] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/22/2017] [Indexed: 01/08/2023]
Abstract
Both sorafenib and interleukin-27 (IL-27) are antineoplastic drugs. This study aimed to investigate the synergistic effect of these two drugs on bladder cancer cells. HTB-9 and T24 cells were stimulated with IL-27 (50 ng/mL), sorafenib (2 μM) or the synergistic action of these two drugs. The cells without treatment acted as control. Cell proliferation, apoptosis and invasion were measured by bromodeoxyuridine assay, flow cytometry and modified Boyden chamber, respectively. Simultaneously, both modified Boyden chamber and scratch assay were used to assess cell migration. Finally, the phosphorylation levels of key kinases in the Akt/mechanistic target of rapamycin (mTOR)/mitogen-activated protein kinase (MAPK) pathway, and expression levels of matrix metalloproteinase (MMP)-2 and MMP-9 were detected by western blot analysis. Stimulation with IL-27 or sorafenib repressed proliferation, migration and invasion but promoted apoptosis, and the effects were all enhanced by the combination of these two drugs in HTB-9 cells. The effect of the combined treatment on bladder cancer cells was verified in T24 cells. Additionally, the phosphorylation levels of AKT, mTOR and MAPK as well as the expression levels of MMP-2 and MMP-9 were all decreased by a single treatment of IL-27 or sorafenib, and further decreased by the combined treatment of these two drugs. The combination of IL-27 and sorafenib inhibited proliferation, migration and invasion and promoted apoptosis of bladder cancer cells compared with mono-drug treatment. Additionally, the AKT/mTOR/MAPK pathway might be implicated in the functional effects by down-regulations of MMP-2 and MMP-9.
Collapse
Affiliation(s)
- J Y Cao
- Department of Urology, Binzhou Medical University Hospital, Binzhou, China
| | - H S Yin
- Department of Urology, Binzhou Medical University Hospital, Binzhou, China
| | - H S Li
- Department of Gastrointestinal Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - X Q Yu
- Department of Gastrointestinal Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - X Han
- Department of Gastrointestinal Surgery, Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|
38
|
Nakamoto Y. Promising new strategies for hepatocellular carcinoma. Hepatol Res 2017; 47:251-265. [PMID: 27558453 DOI: 10.1111/hepr.12795] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/16/2016] [Accepted: 08/19/2016] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common causes of cancer death worldwide. It usually arises based on a background of chronic liver diseases, defined as the hypercarcinogenic state. The current treatment options for HCC ranging from locoregional treatments to chemotherapies, including sorafenib, effectively regulate the limited sizes and numbers of the nodules. However, these treatments remain unsatisfactory because they have insufficient antitumor effects on the large and numerous nodules associated with HCC and because of a high recurrence rate in the surrounding inflamed liver. To develop novel and promising therapies with higher antitumor effects, recent progress in identifying molecular targets and developing immunological procedures for HCC are reviewed. The molecular targets discussed include the intracellular signaling pathways of protein kinase B/mammalian target of rapamycin and RAS/RAF/mitogen-activated protein kinase, Wnt/β-catenin and glutamine synthetase, insulin-like growth factor, signal transducer and activator of transcription 3, nuclear factor-κB and telomerase reverse transcriptase, and c-MET. Immunological studies have focused mainly on target identification, T cells, natural killer cells, dendritic cells, natural killer T cells, and vaccine development.
Collapse
Affiliation(s)
- Yasunari Nakamoto
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| |
Collapse
|
39
|
Tan J, Song M, Zhou M, Hu Y. Antibiotic tigecycline enhances cisplatin activity against human hepatocellular carcinoma through inducing mitochondrial dysfunction and oxidative damage. Biochem Biophys Res Commun 2017; 483:17-23. [PMID: 28069382 DOI: 10.1016/j.bbrc.2017.01.021] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 01/05/2017] [Indexed: 01/08/2023]
Abstract
Targeting mitochondrial metabolism has been recently demonstrated to be a promising therapeutic strategy for the treatment of various cancer. In this work, we demonstrate that antibiotic tigecycline is selectively against hepatocellular carcinoma (HCC) through inducing mitochondrial dysfunction and oxidative damage. Tigecycline is more effective in inhibiting proliferation and inducing apoptosis of HCC than normal liver cells. Importantly, tigecycline significantly enhances the inhibitory effects of chemotherapeutic drug cisplatin in HCC in vitro and in vivo. Mechanistically, tigecycline specifically inhibits mitochondrial translation as shown by the decreased protein levels of Cox-1 and -2 but not Cox-4 or Grp78, and increased mRNA levels of Cox-1 and -2 but not Cox-4 in HCC cells exposed to tigecycline. In addition, tigecycline significantly induces mitochondrial dysfunction in HCC cells via decreasing mitochondrial membrane potential, complex I and IV activities, mitochondrial respiration and ATP levels. Tigecycline also increases levels of mitochondrial superoxide, hydrogen peroxide and ROS levels. Consistent with oxidative stress, oxidative damage on DNA, protein and lipid are also observed in tigecycline-treated cells. Importantly, antioxidant N-acetyl-l-cysteine (NAC) reverses the effects of tigecycline, suggesting that oxidative stress is required for the action of tigecycline in HCC cells. We further show that HCC cells have higher level of mitochondrial biogenesis than normal liver cells which might explain the different sensitivity to tigecycline between HCC and normal liver cells. Our work is the first to demonstrate that tigecycline is a promising candidate for HCC treatment and highlight the therapeutic value of targeting mitochondrial metabolism in HCC.
Collapse
Affiliation(s)
- Jun Tan
- Department of Hepatology, Ningbo No. 2 Hospital, Ningbo, 315010, China
| | - Meijun Song
- Department of Respiratory Medicine, Ningbo Medical Treatment Center Li Huili Hospital, Ningbo, 315041, China
| | - Mi Zhou
- School of Medicine, Ningbo University, Ningbo, 315211, China.
| | - Yaoren Hu
- Department of Hepatology, Ningbo No. 2 Hospital, Ningbo, 315010, China.
| |
Collapse
|
40
|
Eritja N, Chen BJ, Rodríguez-Barrueco R, Santacana M, Gatius S, Vidal A, Martí MD, Ponce J, Bergadà L, Yeramian A, Encinas M, Ribera J, Reventós J, Boyd J, Villanueva A, Matias-Guiu X, Dolcet X, Llobet-Navàs D. Autophagy orchestrates adaptive responses to targeted therapy in endometrial cancer. Autophagy 2017; 13:608-624. [PMID: 28055301 PMCID: PMC5361596 DOI: 10.1080/15548627.2016.1271512] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Targeted therapies in endometrial cancer (EC) using kinase inhibitors rarely result in complete tumor remission and are frequently challenged by the appearance of refractory cell clones, eventually resulting in disease relapse. Dissecting adaptive mechanisms is of vital importance to circumvent clinical drug resistance and improve the efficacy of targeted agents in EC. Sorafenib is an FDA-approved multitarget tyrosine and serine/threonine kinase inhibitor currently used to treat hepatocellular carcinoma, advanced renal carcinoma and radioactive iodine-resistant thyroid carcinoma. Unfortunately, sorafenib showed very modest effects in a multi-institutional phase II trial in advanced uterine carcinoma patients. Here, by leveraging RNA-sequencing data from the Cancer Cell Line Encyclopedia and cell survival studies from compound-based high-throughput screenings we have identified the lysosomal pathway as a potential compartment involved in the resistance to sorafenib. By performing additional functional biology studies we have demonstrated that this resistance could be related to macroautophagy/autophagy. Specifically, our results indicate that sorafenib triggers a mechanistic MAPK/JNK-dependent early protective autophagic response in EC cells, providing an adaptive response to therapeutic stress. By generating in vivo subcutaneous EC cell line tumors, lung metastatic assays and primary EC orthoxenografts experiments, we demonstrate that targeting autophagy enhances sorafenib cytotoxicity and suppresses tumor growth and pulmonary metastasis progression. In conclusion, sorafenib induces the activation of a protective autophagic response in EC cells. These results provide insights into the unopposed resistance of advanced EC to sorafenib and highlight a new strategy for therapeutic intervention in recurrent EC.
Collapse
Affiliation(s)
- Núria Eritja
- a Department of Basic Sciences , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.4 , Lleida , Spain.,b Department of Pathology , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida/Hospital Universitari Arnau de Vilanova , Lleida , Spain
| | | | | | - Maria Santacana
- a Department of Basic Sciences , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.4 , Lleida , Spain.,b Department of Pathology , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida/Hospital Universitari Arnau de Vilanova , Lleida , Spain
| | - Sònia Gatius
- a Department of Basic Sciences , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.4 , Lleida , Spain.,b Department of Pathology , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida/Hospital Universitari Arnau de Vilanova , Lleida , Spain
| | - August Vidal
- e Department of Pathology , University Hospital of Bellvitge, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Catalonia , Spain
| | - Maria Dolores Martí
- f Department of Gynecology , University Hospital of Bellvitge, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Catalonia , Spain
| | - Jordi Ponce
- f Department of Gynecology , University Hospital of Bellvitge, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Catalonia , Spain
| | - Laura Bergadà
- a Department of Basic Sciences , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.4 , Lleida , Spain.,b Department of Pathology , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida/Hospital Universitari Arnau de Vilanova , Lleida , Spain
| | - Andree Yeramian
- a Department of Basic Sciences , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.4 , Lleida , Spain.,b Department of Pathology , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida/Hospital Universitari Arnau de Vilanova , Lleida , Spain
| | - Mario Encinas
- g Department of Experimental Medicine , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8 , Lleida , Spain
| | - Joan Ribera
- g Department of Experimental Medicine , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8 , Lleida , Spain
| | - Jaume Reventós
- e Department of Pathology , University Hospital of Bellvitge, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Catalonia , Spain.,f Department of Gynecology , University Hospital of Bellvitge, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Catalonia , Spain
| | - Jeff Boyd
- h Department of Human and Molecular Genetics , Herbert Wertheim College of Medicine, Florida International University , Miami , FL , USA
| | - Alberto Villanueva
- i Chemoresistance and Predictive Factors Group, Program Against Cancer Therapeutic Resistance (ProCURE) , Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Catalonia , Spain
| | - Xavier Matias-Guiu
- a Department of Basic Sciences , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.4 , Lleida , Spain.,b Department of Pathology , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida/Hospital Universitari Arnau de Vilanova , Lleida , Spain.,e Department of Pathology , University Hospital of Bellvitge, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Catalonia , Spain.,f Department of Gynecology , University Hospital of Bellvitge, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat , Barcelona , Catalonia , Spain
| | - Xavier Dolcet
- a Department of Basic Sciences , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.4 , Lleida , Spain.,b Department of Pathology , Universitat de Lleida/Institut de Recerca Biomèdica de Lleida/Hospital Universitari Arnau de Vilanova , Lleida , Spain
| | - David Llobet-Navàs
- d Institute of Genetic Medicine, Newcastle University , Newcastle-Upon-Tyne , UK
| |
Collapse
|
41
|
Abstract
Fully grown oocytes arrest meiosis at prophase I and deposit maternal RNAs. A subset of maternal transcripts is stored in a dormant state in the oocyte, and the timely driven translation of specific mRNAs guides meiotic progression, the oocyte-embryo transition, and early embryo development. In the absence of transcription, the regulation of gene expression in oocytes is controlled almost exclusively at the level of transcriptome and proteome stabilization and at the level of protein synthesis.This chapter focuses on the recent findings on RNA distribution related to the temporal and spatial translational control of the meiotic cycle progression in mammalian oocytes. We discuss the most relevant mechanisms involved in the organization of the oocyte's maternal transcriptome storage and localization, and the regulation of translation, in correlation with the regulation of oocyte meiotic progression.
Collapse
|
42
|
Fritsche-Guenther R, Witzel F, Kempa S, Brummer T, Sers C, Blüthgen N. Effects of RAF inhibitors on PI3K/AKT signalling depend on mutational status of the RAS/RAF signalling axis. Oncotarget 2016; 7:7960-9. [PMID: 26799289 PMCID: PMC4884967 DOI: 10.18632/oncotarget.6959] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/25/2015] [Indexed: 01/20/2023] Open
Abstract
Targeted therapies within the RAS/RAF/MEK/ERK signalling axis become increasingly popular, yet cross-talk and feedbacks in the signalling network lead to unexpected effects. Here we look systematically into how inhibiting RAF and MEK with clinically relevant inhibitors result in changes in PI3K/AKT activation. We measure the signalling response using a bead-based ELISA, and use a panel of three cell lines, and isogenic cell lines that express mutant forms of the oncogenes KRAS and BRAF to interrogate the effects of the MEK and RAF inhibitors on signalling. We find that treatment with the RAF inhibitors have opposing effects on AKT phosphorylation depending on the mutational status of two important oncogenes, KRAS and BRAF. If these two genes are in wildtype configuration, RAF inhibitors reduce AKT phosphorylation. In contrast, if BRAF or KRAS are mutant, RAF inhibitors will leave AKT phosphorylation unaffected or lead to an increase of AKT phosphorylation. Down-regulation of phospho-AKT by RAF inhibitors also extends to downstream transcription factors, and correlates with apoptosis induction. Our results show that oncogenes rewire signalling such that targeted therapies can have opposing effects on parallel pathways, which depend on the mutational status of the cell.
Collapse
Affiliation(s)
- Raphaela Fritsche-Guenther
- Max-Delbrück-Center for Molecular Medicin (MDC) Berlin Buch, The Berlin Institute for Medical Systems Biology (BIMSB), 13125 Berlin, Germany
| | - Franziska Witzel
- Institute of Pathology, Molecular Tumor Pathology, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany.,Institute for Theoretical Biology, Charité - Universitätsmedizin Berlin, 10115 Berlin, Germany
| | - Stefan Kempa
- Max-Delbrück-Center for Molecular Medicin (MDC) Berlin Buch, The Berlin Institute for Medical Systems Biology (BIMSB), 13125 Berlin, Germany
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research and Centre for Biological Signalling Studies BIOSS, Albert-Ludwigs University Freiburg, 79104 Freiburg, Germany
| | - Christine Sers
- Institute of Pathology, Molecular Tumor Pathology, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Nils Blüthgen
- Institute of Pathology, Molecular Tumor Pathology, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany.,Institute for Theoretical Biology, Charité - Universitätsmedizin Berlin, 10115 Berlin, Germany.,Integrative Research Institute for the Life Sciences, Humboldt University Berlin, 10099 Berlin, Germany
| |
Collapse
|
43
|
Liu Y, Sun L, Su X, Guo S. Inhibition of eukaryotic initiation factor 4E phosphorylation by cercosporamide selectively suppresses angiogenesis, growth and survival of human hepatocellular carcinoma. Biomed Pharmacother 2016; 84:237-243. [DOI: 10.1016/j.biopha.2016.09.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 06/01/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023] Open
|
44
|
Navarro-Villarán E, Tinoco J, Jiménez G, Pereira S, Wang J, Aliseda S, Rodríguez-Hernández MA, González R, Marín-Gómez LM, Gómez-Bravo MA, Padillo FJ, Álamo-Martínez JM, Muntané J. Differential Antitumoral Properties and Renal-Associated Tissue Damage Induced by Tacrolimus and Mammalian Target of Rapamycin Inhibitors in Hepatocarcinoma: In Vitro and In Vivo Studies. PLoS One 2016; 11:e0160979. [PMID: 27518575 PMCID: PMC4982663 DOI: 10.1371/journal.pone.0160979] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 07/27/2016] [Indexed: 12/30/2022] Open
Abstract
Orthotopic liver transplantation (OLT) is the recommended treatment for patients at early stages of hepatocarcinoma (HCC) with potential portal hypertension and/or bilirubinemia, but without vascular-associated diseases. The patients are receiving immunosuppressive therapy to reduce graft rejection, but differential side effects have been related to calcineurin and mTOR inhibitor administration regarding tumor recurrence and nephrotoxicity. The in vitro studies showed that Tacrolimus exerted a more potent pro-apoptotic effect than Everolimus (Huh 7>Hep 3B>HepG2), being sirolimus only active in Hep3B cell line. Tacrolimus and Everolimus exerted potent antiproliferative properties in Huh 7 and Hep3B in which cells Sirolimus was inactive. Interestingly, Tacrolimus- and Everolimus-dependent G0/G1 cell accumulation occurred as a consequence of drastic reduction in S, as well as in S and G2+M phases, respectively. The in vivo studies support data on the more effective antitumoral properties of Everolimus, eventual risk of pro-angiogenic tumoral properties and nephrotoxicity of Tacrolimus, and pro-proliferative properties of Sirolimus in tumors developed in nude mice.
Collapse
Affiliation(s)
- Elena Navarro-Villarán
- Institute of Biomedicine of Seville (IBIS), “Virgen del Rocío”-“Virgen Macarena” University Hospital/CSIC/University of Seville, Seville, Spain
| | - José Tinoco
- Department of General Surgery, “Virgen del Rocío”-“Virgen Macarena” University Hospital/CSIC/University of Seville/IBIS/CSIC/University of Seville, Seville, Spain
| | - Granada Jiménez
- Department of General Surgery, “Virgen del Rocío”-“Virgen Macarena” University Hospital/CSIC/University of Seville/IBIS/CSIC/University of Seville, Seville, Spain
| | - Sheila Pereira
- Institute of Biomedicine of Seville (IBIS), “Virgen del Rocío”-“Virgen Macarena” University Hospital/CSIC/University of Seville, Seville, Spain
| | - Jize Wang
- Department of General Surgery, “Virgen del Rocío”-“Virgen Macarena” University Hospital/CSIC/University of Seville/IBIS/CSIC/University of Seville, Seville, Spain
| | - Sara Aliseda
- Institute of Biomedicine of Seville (IBIS), “Virgen del Rocío”-“Virgen Macarena” University Hospital/CSIC/University of Seville, Seville, Spain
| | - María A. Rodríguez-Hernández
- Institute of Biomedicine of Seville (IBIS), “Virgen del Rocío”-“Virgen Macarena” University Hospital/CSIC/University of Seville, Seville, Spain
| | - Raúl González
- Institute of Biomedicine of Seville (IBIS), “Virgen del Rocío”-“Virgen Macarena” University Hospital/CSIC/University of Seville, Seville, Spain
| | - Luís M. Marín-Gómez
- Department of General Surgery, “Virgen del Rocío”-“Virgen Macarena” University Hospital/CSIC/University of Seville/IBIS/CSIC/University of Seville, Seville, Spain
| | - Miguel A. Gómez-Bravo
- Department of General Surgery, “Virgen del Rocío”-“Virgen Macarena” University Hospital/CSIC/University of Seville/IBIS/CSIC/University of Seville, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Francisco J. Padillo
- Department of General Surgery, “Virgen del Rocío”-“Virgen Macarena” University Hospital/CSIC/University of Seville/IBIS/CSIC/University of Seville, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - José M. Álamo-Martínez
- Department of General Surgery, “Virgen del Rocío”-“Virgen Macarena” University Hospital/CSIC/University of Seville/IBIS/CSIC/University of Seville, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Jordi Muntané
- Department of General Surgery, “Virgen del Rocío”-“Virgen Macarena” University Hospital/CSIC/University of Seville/IBIS/CSIC/University of Seville, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- * E-mail:
| |
Collapse
|
45
|
Singh AR, Joshi S, Burgoyne AM, Sicklick JK, Ikeda S, Kono Y, Garlich JR, Morales GA, Durden DL. Single Agent and Synergistic Activity of the "First-in-Class" Dual PI3K/BRD4 Inhibitor SF1126 with Sorafenib in Hepatocellular Carcinoma. Mol Cancer Ther 2016; 15:2553-2562. [PMID: 27496136 DOI: 10.1158/1535-7163.mct-15-0976] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 07/08/2016] [Indexed: 01/30/2023]
Abstract
Deregulated PI3K/AKT/mTOR, Ras/Raf/MAPK, and c-Myc signaling pathways are of prognostic significance in hepatocellular carcinoma (HCC). Sorafenib, the only drug clinically approved for patients with advanced HCC, blocks the Ras/Raf/MAPK pathway but it does not inhibit the PI3K/AKT/mTOR pathway or c-Myc activation. Hence, there is an unmet medical need to identify potent PI3K/BRD4 inhibitors, which can be used either alone or in combination with sorafenib to treat patients with advanced HCC. Herein, we show that SF1126 (pan PI3K/BRD4 inhibitor) as single agent or in combination with sorafenib inhibited proliferation, cell cycle, apoptosis, and multiple key enzymes in PI3K/AKT/mTOR and Ras/Raf/MAPK pathway in Hep3B, HepG2, SK-Hep1, and Huh7 HCC cell lines. We demonstrate that the active moiety of the SF1126 prodrug LY294002 binds to and blocks BRD4 interaction with the acetylated histone-H4 chromatin mark protein and displaced BRD4 coactivator protein from the transcriptional start site of MYC in Huh7 and SK-Hep1 HCC cell lines. Moreover, SF1126 blocked expression levels of c-Myc in HCC cells. Treatment of SF1126 either alone or in combination with sorafenib showed significant antitumor activity in vivo Our results establish that SF1126 is a dual PI3K/BRD4 inhibitor. This agent has completed a phase I clinical trial in humans with good safety profile. Our data support the potential future consideration of a phase II clinical trial of SF1126, a clinically relevant dual "first-in-class" PI3K/BRD4 inhibitor in advanced HCC, and a potential combination with sorafenib. Mol Cancer Ther; 15(11); 2553-62. ©2016 AACR.
Collapse
Affiliation(s)
- Alok R Singh
- Department of Pediatrics, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Shweta Joshi
- Department of Pediatrics, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Adam M Burgoyne
- Division of Hematology-Oncology, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Jason K Sicklick
- Division of Surgical Oncology, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Sadakatsu Ikeda
- Division of Hematology-Oncology, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Yuko Kono
- Division of Hepatology, Department of Medicine, University of California San Diego, La Jolla, California
| | | | | | - Donald L Durden
- Department of Pediatrics, Moores Cancer Center, University of California San Diego, La Jolla, California.
- SignalRx Pharmaceuticals, San Diego, California
- Division of Pediatric Hematology-Oncology, UCSD Rady Children's Hospital, University of California San Diego Health System, La Jolla, California
| |
Collapse
|
46
|
Liu C, Yang Z, Wang L, Lu Y, Tang B, Miao H, Xu Q, Chen X. Combination of sorafenib and gadolinium chloride (GdCl3) attenuates dimethylnitrosamine(DMN)-induced liver fibrosis in rats. BMC Gastroenterol 2015; 15:159. [PMID: 26572488 PMCID: PMC4647665 DOI: 10.1186/s12876-015-0380-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 10/19/2015] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND/AIMS Liver sinusoidal endothelial cells (SECs), hepatic stellate cells (HSCs) and Kupffer cells (KCs) are involved in the development of liver fibrosis and represent a potential therapeutic target. The therapeutic effects on liver fibrosis of sorafenib, a multiple tyrosine kinase inhibitor, and gadolinium chloride (GdCl3), which depletes KCs, were evaluated in rats. METHODS Liver fibrosis was induced in rats with dimethylnitrosamine, and the effects of sorafenib and/or GdCl3 in these rats were monitored. Interactions among ECs, HSCs and KCs were assessed by laser confocal microscopy. RESULTS The combination of sorafenib and GdCl3, but not each agent alone, attenuated liver fibrosis and significantly reduced liver function and hydroxyproline (Hyp). Sorafenib significantly inhibited the expression of angiogenesis-associated cell markers and cytokines, including CD31, von Willebrand factor (vWF), and vascular endothelial growth factor, whereas GdCl3 suppressed macrophage-related cell markers and cytokines, including CD68, tumor necrosis factor-α, interleukin-1β, and CCL2. Laser confocal microscopy showed that sorafenib inhibited vWF expression and GdCl3 reduced CD68 staining. Sorafenib plus GdCl3 suppressed the interactions of HSCs, ECs and KCs. CONCLUSION Sorafenib plus GdCl3 can suppress collagen accumulation, suggesting that this combination may be a potential therapeutic strategy in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Cheng Liu
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
- Laboratory of Molecular Pathology, Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Zongguo Yang
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
| | - Lei Wang
- Department of Hepatology, Affiliated hospital of Shandong University of Trasitional Chinese Medicine, 250014, Jinan, China.
| | - Yunfei Lu
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
| | - Bozong Tang
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
| | - Hui Miao
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
| | - Qingnian Xu
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
| | - Xiaorong Chen
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center, Shanghai, 201508, China.
| |
Collapse
|
47
|
Gao JJ, Shi ZY, Xia JF, Inagaki Y, Tang W. Sorafenib-based combined molecule targeting in treatment of hepatocellular carcinoma. World J Gastroenterol 2015; 21:12059-12070. [PMID: 26576091 PMCID: PMC4641124 DOI: 10.3748/wjg.v21.i42.12059] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/28/2015] [Accepted: 09/14/2015] [Indexed: 02/06/2023] Open
Abstract
Sorafenib is the only and standard systematic chemotherapy drug for treatment of advanced hepatocellular carcinoma (HCC) at the current stage. Although sorafenib showed survival benefits in large randomized phase III studies, its clinical benefits remain modest and most often consist of temporary tumor stabilization, indicating that more effective first-line treatment regimens or second-line salvage therapies are required. The molecular pathogenesis of HCC is very complex, involving hyperactivated signal transduction pathways such as RAS/RAF/MEK/ERK and PI3K/AKT/mTOR and aberrant expression of molecules such as receptor tyrosine kinases and histone deacetylases. Simultaneous or sequential abrogation of these critical pathways or the functions of these key molecules involved in angiogenesis, proliferation, and apoptosis may yield major improvements in the management of HCC. In this review, we summarize the emerging sorafenib-based combined molecule targeting for HCC treatment and analyze the rationales of these combinations.
Collapse
|
48
|
He C, Dong X, Zhai B, Jiang X, Dong D, Li B, Jiang H, Xu S, Sun X. MiR-21 mediates sorafenib resistance of hepatocellular carcinoma cells by inhibiting autophagy via the PTEN/Akt pathway. Oncotarget 2015; 6:28867-28881. [PMID: 26311740 PMCID: PMC4745697 DOI: 10.18632/oncotarget.4814] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 07/17/2015] [Indexed: 02/06/2023] Open
Abstract
Sorafenib resistance remains a major obstacle for the effective treatments of hepatocellular carcinoma (HCC). Recent studies indicate that activated Akt contributes to the acquired resistance to sorafenib, and miR-21 dysregulates phosphatase and tensin homolog (PTEN), which inhibits Akt activation. Sorafenib-resistant HCC cells were shown to be refractory to sorafenib-induced growth inhibition and apoptosis. Akt and its downstream factors were highly activated and/or upregulated in sorafenib-resistant cells. Inhibition of autophagy decreased the sensitivity of sorafenib-resistant cells to sorafenib, while its induction had the opposite effect. Differential screening of miRNAs showed higher levels of miR-21 in sorafenib-resistant HCC cells. Exposure of HCC cells to sorafenib led to an increase in miR-21 expression, a decrease in PTEN expression and sequential Akt activation. Transfection of miR-21 mimics in HCC cells restored sorafenib resistance by inhibiting autophagy. Anti-miR-21 oligonucleotides re-sensitized sorafenib-resistant cells by promoting autophagy. Inhibition of miR-21 enhances the efficacy of sorafenib in treating sorafenib-resistant HCC tumors in vivo. We conclude that miR-21 participates in the acquired resistance of sorafenib by suppresing autophagy through the Akt/PTEN pathway. MiR-21 could serve as a therapeutic target for overcoming sorafenib resistance in the treatment of HCC.
Collapse
Affiliation(s)
- Changjun He
- Department of Surgery, the Affiliated Cancer Hospital of Harbin Medical University, Harbin, China
- Department of Pharmacology, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin Medical University, Harbin, China
- The Hepatosplenic Surgery Center, Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuesong Dong
- The Hepatosplenic Surgery Center, Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Zhai
- The Hepatosplenic Surgery Center, Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xian Jiang
- The Hepatosplenic Surgery Center, Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Deli Dong
- Department of Pharmacology, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin Medical University, Harbin, China
| | - Baoxin Li
- Department of Pharmacology, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin Medical University, Harbin, China
| | - Hongchi Jiang
- The Hepatosplenic Surgery Center, Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shidong Xu
- Department of Surgery, the Affiliated Cancer Hospital of Harbin Medical University, Harbin, China
| | - Xueying Sun
- The Hepatosplenic Surgery Center, Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
49
|
Zhai B, Hu F, Yan H, Zhao D, Jin X, Fang T, Pan S, Sun X, Xu L. Bufalin Reverses Resistance to Sorafenib by Inhibiting Akt Activation in Hepatocellular Carcinoma: The Role of Endoplasmic Reticulum Stress. PLoS One 2015; 10:e0138485. [PMID: 26381511 PMCID: PMC4575108 DOI: 10.1371/journal.pone.0138485] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 08/31/2015] [Indexed: 12/17/2022] Open
Abstract
Sorafenib is the standard first-line therapeutic treatment for patients with advanced hepatocellular carcinoma (HCC), but its use is hampered by the development of drug resistance. The activation of Akt by sorafenib is thought to be responsible for this resistance. Bufalin is the major active ingredient of the traditional Chinese medicine Chan su, which inhibits Akt activation; therefore, Chan su is currently used in the clinic to treat cancer. The present study aimed to investigate the ability of bufalin to reverse both inherent and acquired resistance to sorafenib. Bufalin synergized with sorafenib to inhibit tumor cell proliferation and induce apoptosis. This effect was at least partially due to the ability of bufalin to inhibit Akt activation by sorafenib. Moreover, the ability of bufalin to inactivate Akt depended on endoplasmic reticulum (ER) stress mediated by inositol-requiring enzyme 1 (IRE1). Silencing IRE1 with siRNA blocked the bufalin-induced Akt inactivation, but silencing eukaryotic initiation factor 2 (eIF2) or C/EBP-homologous protein (CHOP) did not have the same effect. Additionally, silencing Akt did not influence IRE1, CHOP or phosphorylated eIF2α expression. Two sorafenib-resistant HCC cell lines, which were established from human HCC HepG2 and Huh7 cells, were refractory to sorafenib-induced growth inhibition but were sensitive to bufalin. Thus, Bufalin reversed acquired resistance to sorafenib by downregulating phosphorylated Akt in an ER-stress-dependent manner via the IRE1 pathway. These findings warrant further studies to examine the utility of bufalin alone or in combination with sorafenib as a first- or second-line treatment after sorafenib failure for advanced HCC.
Collapse
Affiliation(s)
- Bo Zhai
- Department of General Surgery, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Fengli Hu
- Department of Gastroenterology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Haijiang Yan
- Department of General Surgery, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Dali Zhao
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xin Jin
- Department of General Surgery, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Taishi Fang
- Department of General Surgery, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shangha Pan
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xueying Sun
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lishan Xu
- Department of General Surgery, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
50
|
Kanda T, Yokosuka O. The androgen receptor as an emerging target in hepatocellular carcinoma. J Hepatocell Carcinoma 2015; 2:91-9. [PMID: 27508198 PMCID: PMC4918288 DOI: 10.2147/jhc.s48956] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the male-dominant liver diseases with poor prognosis, although treatments for HCC have been progressing in the past decades. Androgen receptor (AR) is a member of the nuclear receptor superfamily. Previous studies reported that AR was expressed in human HCC and non-HCC tissues. AR is activated both ligand-dependently and ligand-independently. The latter is associated with a mitogen-activated protein kinase–, v-akt murine thymoma viral oncogene homolog 1–, or signal-transducer and activator of transcription–signaling pathway, which has been implicated in the development of HCC. It has been reported that more than 200 RNA expression levels are altered by androgen treatment. In the liver, androgen-responsive genes are cytochrome P450s, transforming growth factor β, vascular endothelial growth factor, and glucose-regulated protein 78 kDa, which are also associated with human hepatocarcinogenesis. Recent studies also revealed that AR plays a role in cell migration and metastasis. It is possible that cross-talk among AR-signaling, endoplasmic reticulum stress, and innate immune response is important for human hepatocarcinogenesis and HCC development. This review shows that AR could play a potential role in human HCC and represent one of the important target molecules for the treatment of HCC.
Collapse
Affiliation(s)
- Tatsuo Kanda
- Department of Gastroenterology and Nephrology, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Osamu Yokosuka
- Department of Gastroenterology and Nephrology, Chiba University, Graduate School of Medicine, Chiba, Japan
| |
Collapse
|