1
|
Buzatu IM, Tataranu LG, Duta C, Stoian I, Alexandru O, Dricu A. A Review of FDA-Approved Multi-Target Angiogenesis Drugs for Brain Tumor Therapy. Int J Mol Sci 2025; 26:2192. [PMID: 40076810 PMCID: PMC11899917 DOI: 10.3390/ijms26052192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/16/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Neovascularization is an important process in brain tumor development, invasion and metastasis. Several research studies have indicated that the VEGF signaling target has potential for reducing angiogenesis in brain tumors. However, targeting VEGF signaling has not met the expected efficacy, despite initial enthusiasm. This is partly because tumors cleverly use alternative growth factor pathways, other than VEGF signaling, to restore angiogenesis. Multi-target inhibitors have been developed to inhibit several receptor kinases that play a role in the development of angiogenesis. By simultaneously affecting various receptor kinases, these treatments can potentially obstruct various angiogenic pathways that are involved in brain cancer advancement, often offering a more holistic strategy than treatments focusing on just one kinase. Since 2009, the FDA has approved a number of multi-kinase inhibitors that target angiogenic growth factor receptors (e.g., VEGFR, PDGFR, FGFR, RET, c-KIT, MET, AXL and others) for treatment of malignant diseases, including brain cancer. Here, we present some recent results from the literature regarding the preclinical and clinical effects of these inhibitors on brain tumors.
Collapse
Affiliation(s)
- Iuliana Mihaela Buzatu
- Department of Microbiology, “Fundeni” Clinical Institute, Șoseaua Fundeni 258, 022328 Bucharest, Romania;
| | - Ligia Gabriela Tataranu
- Department of Neurosurgery, Clinical Emergency Hospital “Bagdasar-Arseni”, Soseaua Berceni 12, 041915 Bucharest, Romania;
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Carmen Duta
- Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania; (C.D.); (I.S.); (A.D.)
| | - Irina Stoian
- Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania; (C.D.); (I.S.); (A.D.)
| | - Oana Alexandru
- Department of Neurology, University of Medicine and Pharmacy of Craiova, Petru Rares 2, 200349 Craiova, Romania
| | - Anica Dricu
- Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania; (C.D.); (I.S.); (A.D.)
| |
Collapse
|
2
|
Shi L, Zhang S, Liu G, Nie Z, Ding P, Chang W, Dai Y, Ma X. Toxin protein LukS-PV targeting complement receptor C5aR1 inhibits cell proliferation in hepatocellular carcinoma via the HDAC7-Wnt/β-catenin axis. J Biol Chem 2025; 301:108148. [PMID: 39736396 PMCID: PMC11910327 DOI: 10.1016/j.jbc.2024.108148] [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: 07/18/2024] [Revised: 10/30/2024] [Accepted: 12/20/2024] [Indexed: 01/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the common malignant tumors. Complement system has become a new focus of cancer research by changing the biological behavior of cancer cells to influence the growth of cancer. Recent studies reported that the complement C5a-C5aR1 axis can promote the malignant phenotype of multiple tumors through various signaling pathways. LukS-PV (Panton-Valentine), the S component of Staphylococcus aureus-secreted PV leucocidin, can also bind C5aR1 specifically. This project aims to investigate the role of LukS-PV on HCC cell proliferation and explore underlying molecular mechanisms. Our findings revealed that LukS-PV targeting C5aR1 inhibited HCC cell proliferation in vitro and in vivo. Interestingly, we discovered that LukS-PV inhibited the proliferation of HCC cells by upregulating the acetylation level of β-catenin to promote its protein degradation. In addition, histone deacetylase (HDAC)7 identified as a regulator mediates the deacetylation of β-catenin. Furthermore, our results showed that LukS-PV inhibited proliferation in HCC cells by downregulating HDAC7 to promote the degradation of β-catenin through ubiquitin-proteasome system. Collectively, our findings revealed that LukS-PV targeting C5aR1 inhibits HCC cell proliferation through the HDAC7-Wnt/β-catenin axis. These results revealing a novel mechanism that LukS-PV as a bacterial toxin inhibits HCC cell proliferation through epigenetic remodeling by targeting complement receptor C5aR1, suggest the strong potential of LukS-PV as a promising candidate for HCC treatment.
Collapse
Affiliation(s)
- Lan Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shanshan Zhang
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Gan Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhengchao Nie
- Department of Clinical Laboratory, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Pengsheng Ding
- Department of Clinical Laboratory, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wenjiao Chang
- Department of Clinical Laboratory, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yuanyuan Dai
- Department of Clinical Laboratory, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiaoling Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
3
|
Zhang H, Pei S, Li J, Zhu J, Li H, Wu G, Weng R, Chen R, Fang Z, Sun J, Chen K. Insights about exosomal circular RNAs as novel biomarkers and therapeutic targets for hepatocellular carcinoma. Front Pharmacol 2024; 15:1466424. [PMID: 39444611 PMCID: PMC11496148 DOI: 10.3389/fphar.2024.1466424] [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: 07/18/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024] Open
Abstract
One of the most prevalent pathological types of Primary Liver Cancer (PLC) is the Hepatocellular Carcinoma (HCC) poses a global health issue. The high recurrence and metastasis rate of HCC, coupled with a low 5-year survival rate, result in a bleak prognosis. Exosomes, small extracellular vesicles released by various cells, contain diverse non-coding RNA molecules, including circular RNAs (circRNAs), which play a significant role in intercellular communication and can impact HCC progression. Studies have revealed the potential clinical applications of exosomal circRNAs as biomarkers and therapeutic targets for HCC. These circRNAs can be transferred via exosomes to nearby non-cancerous cells, thereby regulating HCC progression and influencing malignant phenotypes, such as cell proliferation, invasion, metastasis, and drug resistance. This review provides a comprehensive overview of the identified exosomal circRNAs, highlighting their potential as non-invasive biomarkers for HCC, and suggesting new perspectives for HCC diagnosis and treatment. The circRNA from exosomal organelles promotes metastasis and immune scape because of their unique chirality which is different from the Biomolecular Homochirality.
Collapse
Affiliation(s)
- Haiyan Zhang
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- Zhejiang Chinese Medical University, Shuren College, Hangzhou, China
| | - Shanshan Pei
- School of Pharmacy, Beihua University, Jilin, China
| | - Jiaxuan Li
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jiajie Zhu
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Hongyu Li
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Guangshang Wu
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Ruiqi Weng
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Ruyi Chen
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Zhongbiao Fang
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jingbo Sun
- School of Pharmacy, Beihua University, Jilin, China
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| |
Collapse
|
4
|
Gong J, Han G, Chen Z, Zhang Y, Xu B, Xu C, Gao W, Wu J. CircDCAF8 promotes the progression of hepatocellular carcinoma through miR-217/NAP1L1 Axis, and induces angiogenesis and regorafenib resistance via exosome-mediated transfer. J Transl Med 2024; 22:517. [PMID: 38816735 PMCID: PMC11137954 DOI: 10.1186/s12967-024-05233-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/23/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Circular RNAs (circRNAs), which are a new type of single-stranded circular RNA, have significant involvement in progression of many diseases, including tumors. Currently, multiple circRNAs have been identified in hepatocellular carcinoma (HCC). Our study aims to investigate the function and mechanism of circDCAF8 in HCC. METHODS The expression of circDCAF8 (hsa_circ_0014879) in HCC and para-carcinoma tissue samples was determined using quantitative real-time polymerase chain reaction (qRT-PCR). The biological function of circDCAF8 in HCC was confirmed by experiments conducted both in vitro and in vivo. And the relationship between circDCAF8, miR-217 and NAP1L1 was predicted by database and verified using qRT-PCR, RNA-binding protein immunoprecipitation (RIP) and dual-luciferase reporter assays. Exosomes isolated from HCC cells were utilized to assess the connection of exosomal circDCAF8 with HCC angiogenesis and regorafenib resistance. RESULTS CircDCAF8 is upregulated in HCC tissues and cell lines, and is linked to an unfavourable prognosis for HCC patients. Functionally, circDCAF8 was proved to facilitate proliferation, migration, invasion and Epithelial-Mesenchymal Transformation (EMT) in HCC cells. Animal examinations also validated the tumor-promoting characteristics of circDCAF8 on HCC. Besides, exosomal circDCAF8 promoted angiogenesis in HUVECs. Mechanistically, circDCAF8 interacted with miR-217 and NAP1L1 was a downstream protein of miR-217. CircDCAF8 promoted NAP1L1 expression by sponging miR-217. In addition, exosomes may transfer circDCAF8 from regorafenib-resistant HCC cells to sensitive cells, where it would confer a resistant phenotype. CONCLUSION CircDCAF8 facilitates HCC proliferation and metastasis via the miR-217/NAP1L1 axis. Meanwhile, circDCAF8 can promote angiogenesis and drive resistance to regorafenib, making it a viable therapeutic target for HCC patients.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Exosomes/metabolism
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Drug Resistance, Neoplasm/genetics
- Neovascularization, Pathologic/genetics
- Disease Progression
- Animals
- Phenylurea Compounds/pharmacology
- Phenylurea Compounds/therapeutic use
- Cell Line, Tumor
- Pyridines/pharmacology
- Mice, Nude
- Gene Expression Regulation, Neoplastic
- Male
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- Mice
- Mice, Inbred BALB C
- Female
- Base Sequence
- Human Umbilical Vein Endothelial Cells/metabolism
- Middle Aged
- Angiogenesis
Collapse
Affiliation(s)
- Jiahao Gong
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu Province, China
| | - Guoyong Han
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu Province, China
| | - Zhiqiang Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu Province, China
| | - Yinqi Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu Province, China
| | - Bin Xu
- Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong Province, China
| | - Chao Xu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu Province, China
| | - Wen Gao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China.
| | - Jindao Wu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu Province, China.
| |
Collapse
|
5
|
Prasad YR, Anakha J, Pande AH. Treating liver cancer through arginine depletion. Drug Discov Today 2024; 29:103940. [PMID: 38452923 DOI: 10.1016/j.drudis.2024.103940] [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: 11/10/2023] [Revised: 02/16/2024] [Accepted: 02/29/2024] [Indexed: 03/09/2024]
Abstract
Liver cancer, the sixth most common cancer globally and the second-leading cause of cancer-related deaths, presents a critical public health threat. Diagnosis often occurs in advanced stages of the disease, aligning incidence with fatality rates. Given that established treatments, such as stereotactic body radiation therapy and transarterial radioembolization, face accessibility and affordability challenges, the emerging focus on cancer cell metabolism, particularly arginine (Arg) depletion, offers a promising research avenue. Arg-depleting enzymes show efficacy against Arg-auxotrophic cancers, including hepatocellular carcinoma (HCC). Thus, in this review, we explore the limitations of current therapies and highlight the potential of Arg depletion, emphasizing various Arg-hydrolyzing enzymes in clinical development.
Collapse
Affiliation(s)
- Yenisetti Rajendra Prasad
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - J Anakha
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India.
| |
Collapse
|
6
|
Long J, Liu L, Yang X, Lu X, Qin L. Impact of combining Lenvatinib with Transarterial chemoembolization for unresectable hepatocellular carcinoma. Pak J Med Sci 2023; 39:1847-1852. [PMID: 37936761 PMCID: PMC10626123 DOI: 10.12669/pjms.39.6.7944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/01/2023] [Accepted: 08/08/2023] [Indexed: 11/09/2023] Open
Abstract
Objectives To investigate the impact of combining lenvatinib with transarterial chemoembolization (TACE) for unresectable hepatocellular carcinoma (HCC). Methods This was a retrospective observational study which reviewed the medical records of 103 unresectable HCC patients from January 2017 to June 2020 in The First Affiliated Hospital of Soochow University. It included 46 patients who received TACE plus lenvatinib and 57 patients who received TACE alone. The levels of serum indicators, clinical effect, adverse events, overall survival (OS), and progression-free survival (PFS) were compared between the two groups. Results AFP and VEGF levels in the TACE+lenvatinib group post-treatment were significantly lower than the TACE group (P<0.05). The clinical efficacy in the TACE+lenvatinib group (69.57%) was higher than that in the TACE group (40.35%) post-treatment (P<0.05). There were significant differences in hypertension, diarrhea, and bleeding (gingiva) between the two groups (P<0.05). There were no significant differences in one or two year PFS rate or one year OS between groups (P>0.05), while the two years survival rate in the TACE+lenvatinib group was significantly higher than that in the TACE group (P<0.05). Conclusions TACE combined with lenvatinib have a high clinical effective rate, with reduced AFP and VEGF levels, higher two year survival rate, and acceptable incidence of adverse events.
Collapse
Affiliation(s)
- Jianwu Long
- Jianwu Long, Department of General Surgery, The First Affiliated Hospital of Soochow University, Soochow, Jiangsu province, P.R. China
| | - Longfei Liu
- Longfei Liu, Department of Hepatobiliary Surgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan province, P.R. China
| | - Xuefeng Yang
- Xuefeng Yang, Department of Hepatobiliary Surgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan province, P.R. China
| | - Xianzhou Lu
- Xianzhou Lu Department of General Surgery, Hengyang County People’s Hospital, Hengyang, Hunan province, P.R. China
| | - Lei Qin
- Lei Qin, Department of General Surgery, The First Affiliated Hospital of Soochow University, Soochow, Jiangsu province, P.R. China
| |
Collapse
|
7
|
Xia B, Liu Y, Wang J, Lu Q, Lv X, Deng K, Yang J. Emerging role of exosome-shuttled noncoding RNAs in gastrointestinal cancers: From intercellular crosstalk to clinical utility. Pharmacol Res 2023; 195:106880. [PMID: 37543095 DOI: 10.1016/j.phrs.2023.106880] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023]
Abstract
Gastrointestinal cancer remains a significant global health burden. The pursuit of advancing the comprehension of tumorigenesis, along with the identification of reliable biomarkers and the development of precise therapeutic strategies, represents imperative objectives in this field. Exosomes, small membranous vesicles released by most cells, commonly carry functional biomolecules, including noncoding RNAs (ncRNAs), which are specifically sorted and encapsulated by exosomes. Exosome-mediated communication involves the release of exosomes from tumor or stromal cells and the uptake by nearby or remote recipient cells. The bioactive cargoes contained within these exosomes exert profound effects on the recipient cells, resulting in significant modifications in the tumor microenvironment (TME) and distinct alterations in gastrointestinal tumor behaviors. Due to the feasibility of isolating exosomes from various bodily fluids, exosomal ncRNAs have shown great potential as liquid biopsy-based indicators for different gastrointestinal cancers, using blood, ascites, saliva, or bile samples. Moreover, exosomes are increasingly recognized as natural delivery vehicles for ncRNA-based therapeutic interventions. In this review, we elucidate the processes of ncRNA-enriched exosome biogenesis and uptake, examine the regulatory and functional roles of exosomal ncRNA-mediated intercellular crosstalk in gastrointestinal TME and tumor behaviors, and explore their potential clinical utility in diagnostics, prognostics, and therapeutics.
Collapse
Affiliation(s)
- Bihan Xia
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Yuzhi Liu
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Jin Wang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Qing Lu
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Xiuhe Lv
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Kai Deng
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China.
| | - Jinlin Yang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China.
| |
Collapse
|
8
|
Zhang D, Jiang C, Zheng X, Lin Z, Zhuang Q, Xie H, Liang Y, Xu Y, Cui L, Liu X, Zeng Y. Normalization of Tumor Vessels by Lenvatinib-Based Metallo-Nanodrugs Alleviates Hypoxia and Enhances Calreticulin-Mediated Immune Responses in Orthotopic HCC and Organoids. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207786. [PMID: 37052507 DOI: 10.1002/smll.202207786] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/22/2023] [Indexed: 06/19/2023]
Abstract
Immunocheckpoint inhibitors combined with Lenvatinib is the first line treatment for hepatocellular carcinoma (HCC), but their potency is hampered by the low response rate and adverse events. Herein, a targeted therapeutic strategy through the coassembly of Lenvatinib, Adriamycin, Fe3+ ion, and a natural polyphenol (metallo-nanodrugs) is presented by coordination effect for potentiating tumor vascular normalization and systematic chemo-immunotherapy to effectively inhibit the progression of HCC in both orthotopic model and patients-derived organoids. In mice with orthotopic HCC, the obtained metallo-nanodrugs efficiently increase the drug accumulation in orthotopic tumors and can respond to acidic tumor environment. The promotion of tumor vascular normalization by metallo-nanodrugs is observed, which enhances the infiltrating T lymphocytes in tumor, and reinforces the calreticulin-mediated antitumor immunity through alleviating hypoxia, reducing regulatory T cells, and down-regulating PDL1 expression of tumors. The excellent therapeutic efficiency with complete remission of orthotopic tumors (3/6) and long-term survival of mice (4/6, 42 days) are also achieved. Furthermore, the excellent therapeutic effect of metallo-nanodrugs is also validated in 5 patient-derived organoids, and hence can provide a marvelous systemic chemo-immunotherapy strategy for enhancing HCC treatment.
Collapse
Affiliation(s)
- Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Chenwei Jiang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Xiaoyuan Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Zhiwen Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, P. R. China
| | - Qiuyu Zhuang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Huanzhang Xie
- Fujian Key Laboratory of Functional Marine Sensing Materials, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, 350108, P. R. China
| | - Yuzhi Liang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Yu Xu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Linsheng Cui
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, P. R. China
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, P. R. China
| | - Yongyi Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, P. R. China
| |
Collapse
|
9
|
Zhang Y, Wu X, Zhu H, Cong Y. Development and in functional study of a bi-specific sustained release drug-loaded nano-liposomes for hepatocellular carcinoma. J Biomater Appl 2023:8853282231179313. [PMID: 37243614 DOI: 10.1177/08853282231179313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
BACKGROUND Lenvatinib (LEN) is a first-line therapy for patients with hepatocellular carcinoma (HCC), but has a larger adverse effect profile. In this study, we developed a liposome with drug-carrying function and magnetic resonance imaging (MRI) imaging function to investigate the targeted drug-carrying function and MRI tracing ability of liposome for HCC. METHODS Magnetic nano-liposomes (MNL) with dual targeting function of epithelial cell adhesion molecule (EpCAM) and vimentin and capable of encapsulating LEN drugs were prepared. The characterization performance, drug loading efficiency and cytotoxicity of EpCAM/vimentin-LEN-MNL were tested, and the dual-targeting slow release drug loading function and MRI tracing ability were investigated in cellular and animal models. RESULTS EpCAM/vimentin-LEN-MNL has a mean particle size of 218.37 ± 5.13 nm and a mean potential of 32.86 ± 4.62 mV, and is spherical in shape and can be uniformly dispersed in solution. The encapsulation rate was 92.66 ± 0.73% and the drug loading rate was 9.35 ± 0.16%. It has low cytotoxicity, can effectively inhibit HCC cell proliferation and promote HCC cell apoptosis, and has specific targeting function and MRI tracing ability for HCC cells. CONCLUSIONS In this study, an HCC-specific dual-targeted sustained-release drug delivery liposome with dual-targeted recognition and sensitive MRI tracer was successfully prepared, which provides an important scientific basis for maximizing the multiple effects of nano-carriers in tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Yufei Zhang
- Shanghai Seventh People's Hospital, Shanghai, China
| | - Xiaoxiong Wu
- Shanghai Seventh People's Hospital, Shanghai, China
| | - Hongfan Zhu
- Shanghai Seventh People's Hospital, Shanghai, China
| | - Yun Cong
- Shanghai Seventh People's Hospital, Shanghai, China
| |
Collapse
|
10
|
Kim Y, Lee JS, Lee HW, Kim BK, Park JY, Kim DY, Ahn SH, Goh MJ, Kang W, Kim SU. Sorafenib versus nivolumab after lenvatinib treatment failure in patients with advanced hepatocellular carcinoma. Eur J Gastroenterol Hepatol 2023; 35:191-197. [PMID: 36574310 DOI: 10.1097/meg.0000000000002466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND AIM An optimal sequential anti-hepatocellular carcinoma (HCC) agent that can be used after failed lenvatinib treatment has not been established. Here, we compared the outcomes of sorafenib and nivolumab as second-line agents after failed lenvatinib treatment in patients with advanced HCC. METHODS Patients with advanced HCC who had received sorafenib or nivolumab as second-line agents after failed lenvatinib treatment were recruited from two Korean tertiary institutions between November 2018 and June 2020. RESULTS The median age of the 60 participants (52 treated with sorafenib and eight treated with nivolumab) at baseline was 56.8 years. The demographic, laboratory and tumor variables, as well as lenvatinib treatment duration, were similar between the two groups. The median durations of sorafenib and nivolumab treatment were 1.2 and 2.6 months, respectively ( P = 0.164). Twenty-four (40.0%) patients died during the follow-up period (median, 15.8 months). The median overall survival (OS) of the study population was 5.8 months. The median OS of patients treated with sorafenib was significantly longer than the median OS of patients treated with nivolumab (8.7 vs. 3.0 months; P = 0.046). Sorafenib treatment (vs. nivolumab) was independently associated with a lower risk of mortality (hazard ratio = 0.194; 95% confidence interval, 0.053-0.708; P = 0.013). Worse Eastern Cooperative Oncology Group performance status, larger maximal tumor size, lymph node metastases and higher total bilirubin levels were independently associated with increased mortality risk (all P < 0.05). CONCLUSIONS Lenvatinib-sorafenib sequential treatment resulted in significantly better survival did than lenvatinib-nivolumab sequential treatment in patients with advanced HCC. Larger studies are needed to validate our results.
Collapse
Affiliation(s)
- Yuna Kim
- Department of Internal Medicine
- Institute of Gastroenterology, Yonsei University College of Medicine
| | - Jae Seung Lee
- Department of Internal Medicine
- Institute of Gastroenterology, Yonsei University College of Medicine
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Hye Won Lee
- Department of Internal Medicine
- Institute of Gastroenterology, Yonsei University College of Medicine
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Beom Kyung Kim
- Department of Internal Medicine
- Institute of Gastroenterology, Yonsei University College of Medicine
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Jun Yong Park
- Department of Internal Medicine
- Institute of Gastroenterology, Yonsei University College of Medicine
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Do Young Kim
- Department of Internal Medicine
- Institute of Gastroenterology, Yonsei University College of Medicine
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Sang Hoon Ahn
- Department of Internal Medicine
- Institute of Gastroenterology, Yonsei University College of Medicine
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Myung Ji Goh
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Wonseok Kang
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea
| | - Seung Up Kim
- Department of Internal Medicine
- Institute of Gastroenterology, Yonsei University College of Medicine
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| |
Collapse
|
11
|
Birnbaum DJ, Picard M, Da Costa Q, Delayre T, Finetti P, Cabaud O, Agavnian E, De Rauglaudre B, Denicolaï E, Bertucci F, Mamessier E. PVRIG Expression Is an Independent Prognostic Factor and a New Potential Target for Immunotherapy in Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:cancers15020447. [PMID: 36672396 PMCID: PMC9856571 DOI: 10.3390/cancers15020447] [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: 11/28/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/13/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a frequent and deadly cancer in need of new treatments. Immunotherapy has shown promising results in several solid tumors. The TIGIT/DNAM-1 axis gathers targets for new immune checkpoint inhibitors (ICIs). Here, we aimed at highlighting the potential of this axis as a new therapeutic option for HCC. For this, we built a large transcriptomic database of 683 HCC samples, clinically annotated, and 319 normal liver tissues. We interrogated this database for the transcriptomic expression of each member of the TIGIT/DNAM-1 axis and tested their prognostic value for survival. We then focused on the most discriminant one for these criteria, i.e., PVRIG, and analyzed the clinical characteristics, the disease-free and overall survivals, and biological pathways associated with PVRIG High tumors. Among all members of the TIGIT/DNAM-1 axis, PVRIG expression was higher in tumors than in normal liver, was heterogeneous across tumors, and was the only member with independent prognostic value for better survival. PVRIG High tumors were characterized by a higher lymphocytic infiltrate and enriched for signatures associated with tertiary lymphoid structures and better anti-tumor immune response. These results suggest that patients with PVRIG High tumors might be good candidates for immune therapy involving ICIs, notably ICIs targeting the TIGIT/DNAM-1 axis. Further functional and clinical validation is urgently required.
Collapse
Affiliation(s)
- David Jeremie Birnbaum
- Predictive Oncology Laboratory, Labeled Team Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Paoli-Calmettes Institute (IPC), Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseilles, France
- General and Visceral Surgery Department, North’s Hospital, 13015 Marseilles, France
| | - Maelle Picard
- Predictive Oncology Laboratory, Labeled Team Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Paoli-Calmettes Institute (IPC), Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseilles, France
| | - Quentin Da Costa
- Predictive Oncology Laboratory, Labeled Team Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Paoli-Calmettes Institute (IPC), Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseilles, France
| | - Thomas Delayre
- Predictive Oncology Laboratory, Labeled Team Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Paoli-Calmettes Institute (IPC), Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseilles, France
- General and Visceral Surgery Department, North’s Hospital, 13015 Marseilles, France
| | - Pascal Finetti
- Predictive Oncology Laboratory, Labeled Team Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Paoli-Calmettes Institute (IPC), Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseilles, France
| | - Olivier Cabaud
- Predictive Oncology Laboratory, Labeled Team Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Paoli-Calmettes Institute (IPC), Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseilles, France
| | - Emilie Agavnian
- IPC/CRCM Experimental Pathology (ICEP), CRCM, Paoli-Calmettes Institute, 13009 Marseilles, France
| | - Bernadette De Rauglaudre
- Predictive Oncology Laboratory, Labeled Team Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Paoli-Calmettes Institute (IPC), Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseilles, France
- Department of Digestive Oncology and Gastro-Enterology, Timone Hospital, AP-HM, 13005 Marseille, France
| | - Emilie Denicolaï
- Predictive Oncology Laboratory, Labeled Team Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Paoli-Calmettes Institute (IPC), Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseilles, France
| | - François Bertucci
- Predictive Oncology Laboratory, Labeled Team Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Paoli-Calmettes Institute (IPC), Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseilles, France
- Department of Medical Oncology, Paoli-Calmettes Institute, 13009 Marseilles, France
| | - Emilie Mamessier
- Predictive Oncology Laboratory, Labeled Team Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Paoli-Calmettes Institute (IPC), Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseilles, France
- Correspondence: ; Tel.: +33-4-91-22-72-61
| |
Collapse
|
12
|
Han X, Wu J, Sha Z, Lai R, Shi J, Mi L, Yin F, Guo Z. Dicer Suppresses Hepatocellular Carcinoma via Interleukin-8 Pathway. Clin Med Insights Oncol 2023; 17:11795549231161212. [PMID: 37056297 PMCID: PMC10088407 DOI: 10.1177/11795549231161212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/15/2023] [Indexed: 04/15/2023] Open
Abstract
Background Elevated level of interleukin-8 (IL-8) promotes hepatocellular carcinoma (HCC) development and contributes to poor prognosis. Previously, we have proved that Dicer inhibits HCC progression. In this study, we evaluated the potential interaction between IL-8 and Dicer as well as their influence on HCC. Methods Hepatocellular carcinoma cells of SMMC-7721 were divided into 2 groups for subsequent analysis: pCMV-Dicer group for Dicer-overexpressing lentivirus transfected cells (pCMV-Dicer cells) and pCMV-NC group for empty lentivirus transfected cells (pCMV-NC cells). Cell Counting kit-8 (CCK8), wound healing, and transwell were used to evaluate the inhibitory effect of Dicer overexpression on proliferation, migration, and invasion of HCC cells. The level of IL-8 was measured by flow cytometry bead-based immunoassays. Male nude BALB/c mice injected with pCMV-Dicer or pCMV-NC cell suspensions was used for transplant of HCC tumor. Results We found that the secretion of IL-8 was reduced in the medium of pCMV-Dicer cells (P = .027). Recombinant human IL-8 (rhIL-8) reversed the inhibitory effect of Dicer on proliferation (P < .01), migration (P = .003), and invasion (P = .001), whereas IL-8 inhibitor of reparixin enhanced inhibitory effect of Dicer on proliferation (P < .05), migration (P = .008), and invasion (P = .000). Lenvatinib downregulated the IL-8 level of HCC cells (P = .000) as well as promote Dicer-induced inhibition for HCC cells referring to proliferation (P < .05), migration (P = .000), and invasion (P = .000). Animal experiments also demonstrated that Dicer cooperated with lenvatinib to inhibit the growth of HCC tumors (P < .05). Conclusions Dicer cooperated with lenvatinib to inhibit HCC growth via downregulating IL-8, and Dicer displayed its potential capability to enhance the anti-tumor effect of lenvatinib.
Collapse
Affiliation(s)
- Xin Han
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Jianhua Wu
- Animal Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Ziyue Sha
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Ruixue Lai
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Jianfei Shi
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Lili Mi
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Fei Yin
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Zhanjun Guo
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
- Zhanjun Guo, Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang 050011, P.R. China.
| |
Collapse
|
13
|
Chandrababu G, Sah SK, Kumar AR, M S, Nath LR. Green Synthesized Nanoparticles as a Plausible Therapeutic Strategy Against Hepatocellular Carcinoma: An Update on its Preclinical and Clinical Relevance. Recent Pat Anticancer Drug Discov 2023; 18:268-291. [PMID: 35616675 DOI: 10.2174/1574892817666220523124437] [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: 10/13/2021] [Revised: 01/29/2022] [Accepted: 02/18/2022] [Indexed: 11/22/2022]
Abstract
Green nanotechnology can offer notable advantages over the conventional drug delivery methods in terms of improved drug stability, drug-carrying capacity, site-specificity, and feasibility to apply different routes of administration with less systemic toxicities. Metal nanoparticles bio fabricated with phytoconstituents and microbial extracts have gained significant interest for the treatment of various solid tumors including hepatocellular carcinoma. Hepatocellular carcinoma (HCC) is an aggressive cancer with a very poor prognosis. The current treatments of HCC fails to provide tumor specificity, causing many systemic toxicities and poor overall survival benefits especially for patients in advanced and terminal stages. A novel therapeutic approach with maximal therapeutic effect and minimum adverse effects are urgently required for HCC patients. Green synthesized metal nanoparticles offer significant anticancer effects along with minimal systemic toxicities because of their site-specific delivery into the tumor microenvironment (TME). Green synthesized metal nanoparticles can therefore be a highly beneficial strategy for the treatment of HCC if properly validated with preclinical and clinical studies. This review focuses on the preclinical evidence of the most widely studied green metal nanoparticles such as green synthesized silver nanoparticles, gold nanoparticles and selenium nanoparticles. We have also summarised the clinical studies and the patents approved for nanoparticles against HCC.
Collapse
Affiliation(s)
- Gopika Chandrababu
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi-682041, Kerala, India
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi-682041, Kerala, India
| | - Sunil Kumar Sah
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi-682041, Kerala, India
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi-682041, Kerala, India
| | - Ayana R Kumar
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi-682041, Kerala, India
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi-682041, Kerala, India
| | - Sabitha M
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi-682041, Kerala, India
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi-682041, Kerala, India
| |
Collapse
|
14
|
Malik IA, Rajput M, Werner R, Fey D, Salehzadeh N, von Arnim CAF, Wilting J. Differential in vitro effects of targeted therapeutics in primary human liver cancer: importance for combined liver cancer. BMC Cancer 2022; 22:1193. [PMCID: PMC9675209 DOI: 10.1186/s12885-022-10247-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/29/2022] [Indexed: 11/21/2022] Open
Abstract
The incidence of primary liver tumors, hepatocellular carcinoma (HCC), intrahepatic cholangiocellular carcinoma (ICC), and combined HCC/ICC (cHCC/CC) is increasing. For ICC, targeted therapy exists only for a small subpopulation of patients, while for HCC, Sorafenib and Lenvatinib are in use. Diagnosis of cHCC/CC is a great challenge and its incidence is underestimated, bearing the risk of unintended non-treatment of ICC. Here, we investigated effects of targeted inhibitors on human ICC cell lines (HUH28, RBE, SSP25), in comparison to extrahepatic (E)CC lines (EGI1, CCC5, TFK1), and HCC/hepatoblastoma cell lines (HEP3B, HUH7, HEPG2). Cells were challenged with: AKT inhibitor MK-2206; multikinase inhibitors Sorafenib, Lenvatinib and Dasatinib; PI3-kinase inhibitors BKM-120, Wortmannin, LY294002, and CAL-101; and mTOR inhibitor Rapamycin. Dosage of the substances was based on the large number of published data of recent years. Proliferation was analyzed daily for four days. All cell lines were highly responsive to MK-2206. Thereby, MK-2206 reduced expression of phospho(p)-AKT in all ICC, ECC, and HCC lines, which mostly corresponded to reduction of p-mTOR, whereas p-ERK1/2 was upregulated in many cases. Lenvatinib showed inhibitory effects on the two HCC cell lines, but not on HEPG2, ICCs and ECCs. Sorafenib inhibited proliferation of all cells, except the ECC line CCC5. However, at reduced dosage, we observed increased cell numbers in some ICC experiments. Dasatinib was highly effective especially in ICC cell lines. Inhibitory effects were observed with all four PI3-kinase inhibitors. However, cell type-specific differences were also evident here. Rapamycin was most effective in the two HCC cell lines. Our studies show that the nine inhibitors differentially target ICC, ECC, and HCC/hepatoblastoma lines. Caution should be taken with Lenvatinib and Sorafenib administration in patients with cHCC/CC as the drugs may have no effects on, or might even stimulate, ICC.
Collapse
Affiliation(s)
- Ihtzaz Ahmed Malik
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany
| | - Mansi Rajput
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany
| | - Rieke Werner
- grid.411984.10000 0001 0482 5331Department of Anatomy and Cell Biology, University Medical Center Goettingen, Kreuzbergring 36, 37075 Goettingen, Germany
| | - Dorothea Fey
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany ,grid.411984.10000 0001 0482 5331Department of Anatomy and Cell Biology, University Medical Center Goettingen, Kreuzbergring 36, 37075 Goettingen, Germany
| | - Niloofar Salehzadeh
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany ,grid.411984.10000 0001 0482 5331Department of Anatomy and Cell Biology, University Medical Center Goettingen, Kreuzbergring 36, 37075 Goettingen, Germany
| | - Christine A. F. von Arnim
- grid.411984.10000 0001 0482 5331Department of Geriatrics, University Medical Center Goettingen, Waldweg 33, D-37073 Goettingen, Germany
| | - Jörg Wilting
- grid.411984.10000 0001 0482 5331Department of Anatomy and Cell Biology, University Medical Center Goettingen, Kreuzbergring 36, 37075 Goettingen, Germany
| |
Collapse
|
15
|
Tiu YC, Gong L, Zhang Y, Luo J, Yang Y, Tang Y, Lee WM, Guan XY. GLIPR1 promotes proliferation, metastasis and 5-fluorouracil resistance in hepatocellular carcinoma by activating the PI3K/PDK1/ROCK1 pathway. Cancer Gene Ther 2022; 29:1720-1730. [PMID: 35760898 DOI: 10.1038/s41417-022-00490-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/13/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023]
Abstract
Hepatocellular carcinoma (HCC) contributes to a heavy disease burden for its high prevalence and poor prognosis, with limited effective systemic therapies available. In the era of precision medicine, treatment efficacy might be improved by combining personalized systemic therapies. Since oncogenic activation is one of the primary driving forces in HCC, characterization of these oncogenes can provide insights for developing new targeted therapies. Based on RNA sequencing of epithelial-mesenchymal transition (EMT)-induced HCC cells, this study discovers and characterizes glioma pathogenesis-related protein 1 (GLIPR1) that robustly drives HCC progression and can potentially serve as a prognostic biomarker and therapeutic target with clinical utility. GLIPR1 serves opposing roles and involves distinct mechanisms in different cancers. However, based on integrated in-silico analysis, in vitro and in vivo functional investigations, we demonstrate that GLIPR1 plays a multi-faceted oncogenic role in HCC development via enhancing tumor proliferation, metastasis, and 5FU resistance. We also found that GLIPR1 induces EMT and is actively involved in the PI3K/PDK1/ROCK1 singling axis to exert its oncogenic effects. Thus, pre-clinical evaluation of GLIPR1 and its downstream factors in HCC patients might facilitate further discovery of therapeutic targets, as well as improve HCC chemotherapeutic outcomes and prognosis.
Collapse
Affiliation(s)
- Yuen Chak Tiu
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lanqi Gong
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China.,Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yu Zhang
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jie Luo
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China.,Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yuma Yang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China.,Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ying Tang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China.,Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Wing-Mui Lee
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China. .,State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China. .,Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China. .,State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
16
|
Mohapatra P, Chandrasekaran N. Wnt/β-catenin targeting in liver carcinoma through nanotechnology-based drug repurposing: A review. Biomed Pharmacother 2022; 155:113713. [PMID: 36126453 DOI: 10.1016/j.biopha.2022.113713] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/08/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Liver cancer is the fifth most widespread in the world, with a high fatality rate and poor prognosis.However,surgicalresction,thermal/radiofrequencyablation,chemo/radioembolization and pathway targeting to the cancer cells are all possible options for treating Liver Carcinoma. Unfortunately, once the tumour has developed and spread, diagnosis often occurs too late. The targeted therapy has demonstrated notable, albeit modest, efficacy in some patients with advanced HCC. This demonstrates the necessity of creating additional focused treatments and, in pursuit of this end, the need to find ever-more pathways as prospective targets. Despite the critical need, there are currently no Wnt signalling directed therapy on the research field, only a few methods have progressed beyond the early stage of clinical studies. In the present study, we report that repurposing of drug previously licensed for other diseases is one possible strategy inhibit malignant cell proliferation and renewal by removing individuals protein expression in the Wnt/β-catenin pathway. Particularly β-catenin complex is present in Liver cancer, where tumour necrosis factor is indispensable for the complex formation and β-catenin interactions are disrupted upon drug in nano-carrier through nanotechnology. This study findings not only highlight that repurposing drug could improve liver cancer treatment outcomes but also focused to character traits and functions of the Wnt signalling cascade's molecular targets and how they could be used to get anti-tumour results method to targeting Wnt/β-catenin in liver carcinoma.
Collapse
|
17
|
Hao X, Zhang Y, Shi X, Liu H, Zheng Z, Han G, Rong D, Zhang C, Tang W, Wang X. CircPAK1 promotes the progression of hepatocellular carcinoma via modulation of YAP nucleus localization by interacting with 14-3-3ζ. J Exp Clin Cancer Res 2022; 41:281. [PMID: 36131287 PMCID: PMC9494907 DOI: 10.1186/s13046-022-02494-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/23/2022] [Indexed: 12/24/2022] Open
Abstract
Abstract
Background
Circular RNA (circRNA), a new class of non-coding RNA, has obvious correlations with the occurrence and development of many diseases, including tumors. This study aimed to investigate the potential roles of circPAK1 in hepatocellular carcinoma (HCC).
Methods
High-throughput sequencing was performed on 3 pairs of HCC and matched normal tissues to determine the upregulated circRNAs. The expression level of circPAK1 was detected by qRT-PCR in HCC and paired with normal liver tissue samples. The effects of circPAK1 on proliferation, invasion, metastasis and apoptosis of HCC cells were evaluated by in vitro and in vivo experiments. We also constructed Chitosan/si-circPAK1 (CS/si-circPAK1) nanocomplexes using Chitosan material to evaluate its in vivo therapeutic effect on HCC. High-throughput sequencing, RNA-sequencing, RNA probe pull-down, RNA immunoprecipitation and Co-Immunoprecipitation assays were performed to explore the relationship between circPAK1, 14–3-3ζ, p-LATS1 and YAP. Exosomes isolated from lenvatinib-resistant HCC cell lines were used to evaluate the relationship between exosomal circPAK1 and lenvatinib resistance.
Results
CircPAK1, a novel circRNA, is highly expressed in HCC tumor tissues and cell lines as well as correlated with poor outcomes in HCC patients. Functionally, circPAK1 knockdown inhibited HCC cell proliferation, migration, invasion and angiogenesis while circPAK1 overexpression promoted HCC progression. The tumor-promoting phenotypes of circPAK1 on HCC were also confirmed by animal experiments. Importantly, the application of CS/si-circPAK1 nanocomplexes showed a better therapeutic effect on tumor growth and metastasis. Mechanistically, circPAK1 enhanced HCC progression by inactivating the Hippo signaling pathway, and this kind of inactivation is based on its competitively binding of 14–3-3 ζ with YAP, which weakens the recruitment and cytoplasmic fixation of 14–3-3 ζ to YAP, thus promoting YAP nucleus localization. Additionally, circPAK1 could be transported by exosomes from lenvatinib-resistant cells to sensitive cells and induce lenvatinib resistance of receipt cells.
Conclusion
CircPAK1 exerts its oncogenic function by competitively binding 14–3-3 ζ with YAP, thus promoting YAP nucleus localization, leading to the inactivation of a Hippo signaling pathway. Exosomal circPAK1 may drive resistance to lenvatinib, providing a potential therapeutic target for HCC patients.
Collapse
|
18
|
Sheng J, Wu J, Yin X, Sun Z, Wang X, Zhang J, Tang J, Ji Y, Song J, Wei X, Wang L, Zhao Y, Zhang H, Li T, Zhang Q, Bai X, Chen L, Chen D, Liang T. Synergetic treatment of oxygen microcapsules and lenvatinib for enhanced therapy of HCC by alleviating hypoxia condition and activating anti-tumor immunity. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
19
|
Chandok N, Sirpal S, Yoshida EM. Checkpoint inhibition in hepatocellular carcinoma: Outsmarting the Squid Game. CANADIAN LIVER JOURNAL 2022; 5:165-168. [PMID: 35991480 PMCID: PMC9236585 DOI: 10.3138/canlivj-2022-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 04/06/2022] [Indexed: 01/04/2025]
Affiliation(s)
- Natasha Chandok
- Division of Gastroenterology, William Osler Health System, Brampton, Ontario, Canada
| | - Sanjeev Sirpal
- Department of Emergency Medicine, Fleury Hospital, Montreal, Quebec, Canada
| | - Eric M Yoshida
- Division of Gastroenterology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
20
|
Castven D, Czauderna C, Becker D, Pereira S, Schmitt J, Weinmann A, Shah V, Hajduk J, Keggenhoff F, Binder H, Keck T, Heilmann-Heimbach S, Wörns MA, Thorgeirsson SS, Breuhahn K, Galle PR, Marquardt JU. Acquired Resistance to Antiangiogenic Therapies in Hepatocellular Carcinoma Is Mediated by Yes-Associated Protein 1 Activation and Transient Expansion of Stem-Like Cancer Cells. Hepatol Commun 2022; 6:1140-1156. [PMID: 34817932 PMCID: PMC9035566 DOI: 10.1002/hep4.1869] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/30/2021] [Accepted: 10/25/2021] [Indexed: 01/10/2023] Open
Abstract
Induction of neoangiogenesis is a hallmark feature during disease progression of hepatocellular carcinoma (HCC). Antiangiogenetic compounds represent a mainstay of therapeutic approaches; however, development of chemoresistance is observed in the majority of patients. Recent findings suggest that tumor-initiating cells (TICs) may play a key role in acquisition of resistance, but the exact relevance for HCC in this process remains to be defined. Primary and established hepatoma cell lines were exposed to long-term sorafenib treatment to model acquisition of resistance. Treatment effects on TICs were estimated by sphere-forming capacity in vitro, tumorigenicity in vivo, and flow cytometry. Adaptive molecular changes were assessed by whole transcriptome analyses. Compensatory mechanisms of resistance were identified and directly evaluated. Sustained antiproliferative effect following sorafenib treatment was observed in three of six HCC cell lines and was followed by rapid regrowth, thereby mimicking responses observed in patients. Resistant cells showed induction in sphere forming in vitro and tumor-initiating capacity in vivo as well as increased number of side population and epithelial cell adhesion molecule-positive cells. Conversely, sensitive cell lines showed consistent reduction of TIC properties. Gene sets associated with resistance and poor prognosis, including Hippo/yes-associated protein (YAP), were identified. Western blot and immunohistochemistry confirmed increased levels of YAP. Combined treatment of sorafenib and specific YAP inhibitor consistently revealed synergistic antioncogenic effects in resistant cell lines. Conclusion: Resistance to antiangiogenic therapy might be driven by transient expansion of TICs and activation of compensatory pro-oncogenic signaling pathways, including YAP. Specific targeting of TICs might be an effective therapeutic strategy to overcome resistance in HCC.
Collapse
Affiliation(s)
- Darko Castven
- Department of Medicine ILichtenberg Research Group for Molecular HepatocarcinogenesisUniversity Medical Center Schleswig HolsteinLuebeckGermany.,Department of Medicine IUniversity Medical CenterMainzGermany
| | - Carolin Czauderna
- Department of Medicine ILichtenberg Research Group for Molecular HepatocarcinogenesisUniversity Medical Center Schleswig HolsteinLuebeckGermany.,Department of Medicine IUniversity Medical CenterMainzGermany
| | - Diana Becker
- Department of Medicine IUniversity Medical CenterMainzGermany
| | - Sharon Pereira
- Department of Medicine IUniversity Medical CenterMainzGermany
| | - Jennifer Schmitt
- Institute of PathologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Arndt Weinmann
- Department of Medicine IUniversity Medical CenterMainzGermany
| | - Viral Shah
- Department of HematologyMedical Oncology, and PneumologyUniversity Medical CenterMainzGermany
| | - Jovana Hajduk
- Department of Medicine ILichtenberg Research Group for Molecular HepatocarcinogenesisUniversity Medical Center Schleswig HolsteinLuebeckGermany.,Department of Medicine IUniversity Medical CenterMainzGermany
| | | | - Harald Binder
- Institute for Medical Biometry and StatisticsFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Tobias Keck
- Clinic for SurgeryUniversity Medical Center Schleswig HolsteinLuebeckGermany
| | - Stefanie Heilmann-Heimbach
- Institute of Human GeneticsUniversity of Bonn School of MedicineUniversity of BonnBonnGermany.,Department of GenomicsLife and Brain CenterUniversity of BonnBonnGermany
| | - Marcus A Wörns
- Department of Medicine IUniversity Medical CenterMainzGermany
| | - Snorri S Thorgeirsson
- Laboratory of Human CarcinogenesisCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Kai Breuhahn
- Institute of PathologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Peter R Galle
- Department of Medicine IUniversity Medical CenterMainzGermany
| | - Jens U Marquardt
- Department of Medicine ILichtenberg Research Group for Molecular HepatocarcinogenesisUniversity Medical Center Schleswig HolsteinLuebeckGermany.,Department of Medicine IUniversity Medical CenterMainzGermany
| |
Collapse
|
21
|
Necroptosis modulation by cisplatin and sunitinib in hepatocellular carcinoma cell line. Life Sci 2022; 301:120594. [PMID: 35500680 DOI: 10.1016/j.lfs.2022.120594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 11/20/2022]
Abstract
Aim Hepatocellular carcinoma (HCC) is one of the leading causes of cancer death worldwide. Systemic chemotherapy such as cisplatin and multi-targeted receptor tyrosine kinase inhibitors, including sunitinib, has marginal activity and frequent toxicity. Recently, necroptosis has been investigated as a potential target in treating cancer. Our aim is to evaluate the influence of cisplatin-sunitinib combination on HepG2 cells regarding their cytotoxicity and implicated intracellular pathways. MATERIALS AND METHODS The half-maximal inhibitory concentration (IC50) values of cisplatin, sunitinib, and their combination were determined by Sulforhodamine-B assay. Bcl-2 and Bax protein levels were assayed using western blot. ELISA technique was used to measure pRIPK3/RIPK3, pERK/ERK, caspase-9, caspase-8, malondialdehyde (MDA), glutathione (GSH), and glutathione peroxidase (GPx). KEY FINDINGS Cisplatin-sunitinib combination exhibited a superior cytotoxic effect on HepG2 cells. Low concentrations of 4 μg/ml cisplatin and 2.8 μg/ml sunitinib showed significant Bcl-2 down-regulation and Bax up-regulation. The combined treatment also lowered pRIPK3/RIPK3 by 74% (p < 0.05) compared to the control. Significant increase in pERK/ERK by 3.9 folds over the normal control was also demonstrated. Moreover, combined treatment produced a significant 4 and 4.6 folds increase in caspase-9 and -8 levels. An increase in MDA level by 1.3 folds, a decrease in the intracellular GSH level by 63%, and an increase in GPx level by 1.17 folds were demonstrated. SIGNIFICANCE Sunitinib modulated cisplatin effect on cytotoxicity, oxidative stress, apoptosis, necroptosis and MAPK pathways. Sunitinib enhanced cisplatin-induced apoptosis and increased oxidative stress, but decreased necroptosis. Combined cisplatin and sunitinib might be promising for treating advanced HCC.
Collapse
|
22
|
Zhang P, Sun H, Wen P, Wang Y, Cui Y, Wu J. circRNA circMED27 acts as a prognostic factor and mediator to promote lenvatinib resistance of hepatocellular carcinoma. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:293-303. [PMID: 35024242 PMCID: PMC8718824 DOI: 10.1016/j.omtn.2021.12.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 12/07/2021] [Indexed: 12/24/2022]
Abstract
Circular RNAs (circRNAs) have been proven to play key roles in the development and progression of various types of cancers. However, there were no reported studies on the roles of circRNA mediator complex subunit 27 (circMED27) in tumors including hepatocellular carcinoma (HCC). In this study, we found that circMED27 was significantly increased in HCC serum and that higher levels of circMED27 were correlated with bad clinical characteristics and poor prognoses of patients with HCC. Furthermore, upregulated circMED27 promoted HCC resistance to lenvatinib. Our mechanistic investigations revealed that circMED27 functions as a competing endogenous RNA (ceRNA) for miR-655-3p to upregulate ubiquitin-specific peptidase 28 (USP28) expression. Thus, we are led to conclude that circMED27 acts as a potential therapeutic target for HCC patients receiving lenvatinib therapy and may represent a promising molecular biomarker for forecasting lenvatinib-resistant HCC.
Collapse
Affiliation(s)
- Pengfei Zhang
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Haixiang Sun
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Peihao Wen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Yilin Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yuehong Cui
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jing Wu
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
23
|
Xu X, Jiang W, Han P, Zhang J, Tong L, Sun X. MicroRNA-128-3p Mediates Lenvatinib Resistance of Hepatocellular Carcinoma Cells by Downregulating c-Met. J Hepatocell Carcinoma 2022; 9:113-126. [PMID: 35252056 PMCID: PMC8894104 DOI: 10.2147/jhc.s349369] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/12/2022] [Indexed: 01/20/2023] Open
Abstract
Objective Lenvatinib is a first-line multikinase inhibitor for advanced hepatocellular carcinoma (HCC), but resistance to the drug remains a major hurdle for its long-term anti-cancer activity. This resistance is thought to be due to overexpression of c-Met. This study aims to identify potential upstream microRNAs (miRNAs) that regulate c-Met, investigate the underlying mechanisms, and seek potential strategies that may reverse such resistance. Methods Lenvatinib-resistant HCC (LR-HCC) cells were established from human HCC Huh7 and SMMC-7721 cells. Assays of cell proliferation, cell cycle distribution, apoptosis, RT-qPCR, Western blot analysis and immunohistochemistry were employed. Potential miRNAs were screened by miRNA-target prediction tools and their regulatory effects were evaluated by luciferase reporter assays. Xenograft tumor models were used to evaluate the therapeutic effects. Results LR-HCC cells were refractory to lenvatinib-induced growth inhibition and apoptosis in vitro and in vivo. Sustained exposure of cells to lenvatinib resulted in increased expression and phosphorylation of c-Met, and c-Met inhibition enhanced the effects of lenvatinib in suppressing LR-HCC cells. Among eleven miRNA candidates, miR-128-3p displayed the most vigorous activity to negatively regulate c-Met and was downregulated in LR-HCC cells. MiR-128-3p mimics inhibited proliferation and induced apoptosis of LR-HCC cells, and enhanced the effects of lenvatinib in cell culture and animal models. MiR-128-3p and c-Met participate in the mechanisms underlying lenvatinib resistance through regulating Akt that mediates the apoptotic pathway and ERK (extracellular-signal-regulated kinase) modulating cell cycle progression. Conclusion The present results indicate that the miR-128-3p/c-Met axis may be potential therapeutic targets for circumventing lenvatinib resistance in HCC and warrant further investigation.
Collapse
Affiliation(s)
- Xin Xu
- Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People’s Republic of China
- Department of General Surgery, Daqing Oil Field General Hospital, Daqing, 163000, People’s Republic of China
| | - Wenjing Jiang
- Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People’s Republic of China
| | - Peng Han
- Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People’s Republic of China
| | - Jingyan Zhang
- Department of General Surgery, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, 163316, People’s Republic of China
| | - Liquan Tong
- Department of General Surgery, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, 163316, People’s Republic of China
- Liquan Tong, Department of General Surgery, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, 163316, People’s Republic of China, Email
| | - Xueying Sun
- Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People’s Republic of China
- Correspondence: Xueying Sun, Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People’s Republic of China, Email
| |
Collapse
|
24
|
Altaf S, Saleem F, Sher AA, Ali A. Potential therapeutic strategies to combat HCC. Curr Mol Pharmacol 2022; 15:929-942. [PMID: 34979895 DOI: 10.2174/1874467215666220103111009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/16/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022]
Abstract
Hepatocellular carcinoma (HCC) is a complex, life threatening and most common neoplasm in the world. HCC tumors are genetically and phenotypically heterogeneous and involve various molecular mechanisms and stimulation of several signaling pathways such as Vascular Endothelial Growth Factor, Epidermal Growth Factor Receptors (EGFR), Insulin growth factor, Ras/extracellular signal-stimulated kinase, mammalian goal of rapamycin (mTOR), c-mesenchymal-epithelial transition factor-1 (c-Met), Hedgehog, Wnt and apoptotic signaling. Lately, in patient's multi-kinase cascade blockers such as sorafenib, selumetinib and regorafenib have increased survival rate of progressive HCC. This development presents a step forward towards the therapy of liver cancer infection and attests that molecular systemic rehabilitations can be useful in HCC treatment. The development of these systemic therapeutic agents has further expanded the research area for surplus molecular mediators to auxiliary increase cure rate of patients. This article reviews the complete consideration of focus on cascades, current enduring clinical tests by means of HCC therapeutic mediators, and imminent prospects in the cure of HCC.
Collapse
Affiliation(s)
- Sidra Altaf
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | - Faiza Saleem
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | - Azam Ali Sher
- Department of Epidemiology, Michigan State University, Michigan, USA
| | - Ashiq Ali
- Department of Pathology, University of Agriculture, Faisalabad, Pakistan
| |
Collapse
|
25
|
Farooq A, Ahmed Z, Wert J, Jalil A, Yu J, Zaytsev V, Ahmad S. Updates on clinical trials for the management of hepatocellular carcinoma. THERANOSTICS AND PRECISION MEDICINE FOR THE MANAGEMENT OF HEPATOCELLULAR CARCINOMA, VOLUME 3 2022:259-273. [DOI: 10.1016/b978-0-323-99283-1.00013-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
26
|
Tanaka H, Horioka K, Hasebe T, Sawada K, Nakajima S, Konishi H, Isozaki S, Goto M, Fujii Y, Kamikokura Y, Ogawa K, Nishikawa Y. Treatment of hepatocellular carcinoma with autologous platelets encapsulating sorafenib or lenvatinib: A novel therapy exploiting tumor‐platelet interactions. Int J Cancer 2021; 150:1640-1653. [DOI: 10.1002/ijc.33915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 12/25/2022]
Affiliation(s)
- Hiroki Tanaka
- Division of Tumor Pathology, Department of Pathology Asahikawa Medical University Asahikawa Japan
| | - Kie Horioka
- Department of Oncology‐Pathology Karolinska Institutet Stockholm Sweden
- Department of Legal Medicine International University of Health and Welfare Narita Japan
| | - Takumu Hasebe
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology, Department of Medicine Asahikawa Medical University Asahikawa Japan
| | - Koji Sawada
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology, Department of Medicine Asahikawa Medical University Asahikawa Japan
| | - Shunsuke Nakajima
- Department of Emergency Medicine Asahikawa Medical University Asahikawa Japan
| | - Hiroaki Konishi
- Department of Gastroenterology and Advanced Medical Sciences Asahikawa Medical University Asahikawa Japan
| | - Shotaro Isozaki
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology, Department of Medicine Asahikawa Medical University Asahikawa Japan
| | - Masanori Goto
- Division of Tumor Pathology, Department of Pathology Asahikawa Medical University Asahikawa Japan
| | - Yumiko Fujii
- Division of Tumor Pathology, Department of Pathology Asahikawa Medical University Asahikawa Japan
| | - Yuki Kamikokura
- Division of Tumor Pathology, Department of Pathology Asahikawa Medical University Asahikawa Japan
| | | | - Yuji Nishikawa
- Division of Tumor Pathology, Department of Pathology Asahikawa Medical University Asahikawa Japan
| |
Collapse
|
27
|
Wang S, Wang X, Yang X, Liu F, Li J, Li W, Bai Z, Wang H, Mao J, Li T, He K, Wang H. Comprehensive kinomic study via a chemical proteomic approach reveals kinome reprogramming in hepatocellular carcinoma tissues. Proteomics 2021; 22:e2100141. [PMID: 34932872 DOI: 10.1002/pmic.202100141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 11/07/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. Kinases are attractive therapeutic targets since they are commonly altered in cancers. Here, to identify kinases of potential therapeutic interest in HCC, a quantitative kinomic study of tumour and adjacent non-tumour liver tissues was performed using a chemical proteomics approach. In total, 124 kinases were found differentially expressed and they were distributed over all nine kinase groups. Exploration of The Cancer Genome Atlas (TCGA) data showed that the dysregulation of 45 kinases was correlated with poor prognosis in HCC patients. We then tested 11 inhibitors targeting 12 crucial protein kinases alone or in combination for their ability to inhibit cell growth in Hep3B and PLC/PRF/5 cell lines. Six inhibitors significantly reduced viability in both cell lines. Combination inhibition of polo-like kinase 1 (PLK1) and casein kinase 1 epsilon (CSNK1E) significantly induced growth arrest in both cell lines synergistically. In summary, our analysis presents the most complete view of kinome reprogramming in HCC and provides novel insight into crucial kinases in HCC and potential therapeutic targets for HCC treatment. Moreover, the identification of hundreds of differentially expressed kinases forms a rich resource for novel drug targets or diagnostic biomarker discovery. Data are available via ProteomeXchange (identifier PXD023806).
Collapse
Affiliation(s)
- Shufeng Wang
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Xinzheng Wang
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Xin Yang
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Feng Liu
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Jin Li
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Weihua Li
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Zhaofang Bai
- Department of Liver Disease, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
| | - Hongbo Wang
- Department of Hepatobiliary Surgery Center, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Jie Mao
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Tingting Li
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Kun He
- National Center of Biomedical Analysis, Beijing, 100850, China
| | - Hongxia Wang
- National Center of Biomedical Analysis, Beijing, 100850, China
| |
Collapse
|
28
|
Khan AA, Liu ZK, Xu X. Recent advances in immunotherapy for hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int 2021; 20:511-520. [PMID: 34344612 DOI: 10.1016/j.hbpd.2021.06.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 06/22/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Treatment of hepatocellular carcinoma (HCC) is challenging as most patients are diagnosed at advanced stage with underlying chronic liver conditions. Conventional systemic chemotherapy has failed in HCC, and the clinical efficacy of FDA-approved molecular targeted agents such as sorafenib and lenvatinib remains unsatisfactory. DATA SOURCES Literature search was conducted in PubMed for relevant articles published before January 2021. The search aimed to identify recent developments in immune-based treatment approaches for HCC. Information of clinical trials was obtained from https://clinicaltrials.gov/. RESULTS Two immune checkpoint inhibitors (ICIs), nivolumab and pembrolizumab were approved as monotherapies, which has revolutionized HCC treatment. Besides, combination ICIs have also got accelerated FDA approval recently. Immune-based therapies have challenged targeted drugs owing to their safety, tolerability, and survival benefits. In addition to the significant success in ICIs, other immunotherapeutic strategies such as cancer vaccine, chimeric antigen receptor T-cells, natural killer cells, cytokines, and combination therapy, have also shown promising outcomes in clinical trials. Various diagnostic and prognostic biomarkers have been identified which can help in clinical decision making when starting treatment with ICIs. CONCLUSIONS Immunotherapy has emerged as one of the mainstream treatment modalities for advanced HCC in recent years. However, challenges such as low response rate and acquired resistance in previously respondent patients still exist. Further research is needed to understand the unique resistance mechanism to immunotherapy and to discover more predictive biomarkers to guide clinical decision making.
Collapse
Affiliation(s)
- Abid Ali Khan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Lab of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Ministry of Public Health, Hangzhou 310003, China
| | - Zhi-Kun Liu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Lab of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Ministry of Public Health, Hangzhou 310003, China
| | - Xiao Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Lab of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Ministry of Public Health, Hangzhou 310003, China; Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
29
|
Rizzo A, Ricci AD, Gadaleta-Caldarola G, Brandi G. First-line immune checkpoint inhibitor-based combinations in unresectable hepatocellular carcinoma: current management and future challenges. Expert Rev Gastroenterol Hepatol 2021; 15:1245-1251. [PMID: 34431725 DOI: 10.1080/17474124.2021.1973431] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Immunotherapy has recently taken on an extremely important role in medical oncology, as first- or later-line treatment in several tumor types, and recent years have seen the emerging of clinical trials assessing immune checkpoint inhibitors (ICIs) in unresectable hepatocellular carcinoma (HCC). AREAS COVERED Herein, we provide an overview of recently published studies exploring the dual immune checkpoint blockade or the combination of ICIs plus biological treatments as first-line treatment in HCC patients with advanced disease, especially focusing on the biological rationale behind these therapeutic strategies, and ongoing active and recruiting clinical trials. EXPERT OPINION Results of studies on monotherapy with ICIs have suggested that this strategy could be beneficial only in a minority of patients; conversely, the recently published IMbrave150 study has reported an overall survival benefit in HCC receiving the combination of atezolizumab plus bevacizumab compared to sorafenib as first-line treatment. A wide number of clinical trials is evaluating ICI-based combinations in advanced HCC, a strategy which is supported by robust preclinical and early-phase clinical data, and results of these studies are highly awaited.
Collapse
Affiliation(s)
- Alessandro Rizzo
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Bologna, Italy.,Department of Medical Oncology, Medical Oncology Unit, "Mons. R. Dimiccoli" Hospital, Barletta (BT), ASL BT, Italy
| | - Angela Dalia Ricci
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Bologna, Italy.,Department of Medical Oncology, Medical Oncology Unit, "Mons. R. Dimiccoli" Hospital, Barletta (BT), ASL BT, Italy
| | - Gennaro Gadaleta-Caldarola
- Department of Medical Oncology, Medical Oncology Unit, "Mons. R. Dimiccoli" Hospital, Barletta (BT), ASL BT, Italy
| | - Giovanni Brandi
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Bologna, Italy
| |
Collapse
|
30
|
Overview of lenvatinib as a targeted therapy for advanced hepatocellular carcinoma. Clin Exp Hepatol 2021; 7:249-257. [PMID: 34712825 PMCID: PMC8527338 DOI: 10.5114/ceh.2021.109312] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/06/2021] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common malignancy worldwide. Patients commonly present with advanced/unresectable HCC where several treatment options are not effective. In this review, the authors discuss the indications and usage of lenvatinib, a multikinase inhibitor, as first-line therapy for advanced/unresectable HCC, its mode of action, efficacy, drug reactions, response to treatment and adverse effects. Since its approval in 2007, sorafenib has been used as first-line therapy for unresectable HCC. In 2018, a phase III multinational REFLECT trial on subjects with unresectable HCC (Child-Pugh class A) demonstrated that lenvatinib was non-inferior compared to sorafenib for overall survival, with a controllable toxicity profile, leading to its approval. In addition, our review discusses studies that compare the safety and efficacy profile of lenvatinib especially in patients who have a decline in their liver function to Child-Pugh class B. A current real world analysis of lenvatinib approval for unresectable HCC worldwide is reported.
Collapse
|
31
|
Karabicici M, Azbazdar Y, Ozhan G, Senturk S, Firtina Karagonlar Z, Erdal E. Changes in Wnt and TGF-β Signaling Mediate the Development of Regorafenib Resistance in Hepatocellular Carcinoma Cell Line HuH7. Front Cell Dev Biol 2021; 9:639779. [PMID: 34458250 PMCID: PMC8386122 DOI: 10.3389/fcell.2021.639779] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive, chemo resistant neoplasm with poor prognosis and limited treatment options. Exploring activated pathways upon drug treatment can be used to discover more effective anticancer agents to overcome therapy resistance and enhance therapeutic outcomes for patients with advanced HCC. Human tumor-derived cell lines recapitulate HCC diversity and are widely used for studying mechanisms that drive drug resistance in HCC. In this study, we show that regorafenib treatment activates Wnt/β-catenin signaling only in hepatoblast-like HCC cell lines and induces enrichment of markers associated with hepatic stem/progenitor cells. Moreover, activation of Wnt/β-catenin signaling via Wnt3a/R-Spo1 treatment protects these cells from regorafenib induced apoptosis. On the other hand, regorafenib resistant cells established by long-term regorafenib treatment demonstrate diminished Wnt/β-catenin signaling activity while TGF-β signaling activity of these cells is significantly enhanced. Regorafenib resistant cells (RRCs) also show increased expression of several mesenchymal genes along with an induction of CD24 and CD133 cancer stem cell markers. Moreover, regorafenib resistant cells also exhibit significantly augmented in vitro and in vivo migration capacity which could be reversed by TGF-β type 1 receptor (TGFb -R1) inhibition. When combined with regorafenib treatment, TGFβ-R1 inhibition also significantly decreased colony formation ability and augmented cell death in resistant spheroids. Importantly, when we knocked down TGFβ-R1 using a lentiviral plasmid, regorafenib resistant cells entered senescence indicating that this pathway is important for their survival. Treatment of RRCs with TGFβ-R1 inhibitor and regorafenib significantly abolished pSTAT3, pSMAD2 and pERK (44/42) expression suggesting the involvement of both canonical and non-canonical pathways. In conclusion, our data suggest that HCC tumors with aberrant activation in the Wnt/β-catenin pathway, might have higher intrinsic regorafenib resistance and the inhibition of this pathway along with regorafenib administration might increase regorafenib-induced cell death in combinational therapies. However, to resolve acquired regorafenib resistance developed in HCC patients, the combined use of TGF-β pathway inhibitors and Regorafenib constitute a promising approach that can increase regorafenib sensitization and prevent tumor recurrence.
Collapse
Affiliation(s)
- Mustafa Karabicici
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Izmir, Turkey.,Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Izmir, Turkey
| | - Yagmur Azbazdar
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Izmir, Turkey.,Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Izmir, Turkey
| | - Gunes Ozhan
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Izmir, Turkey.,Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Izmir, Turkey
| | - Serif Senturk
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Izmir, Turkey.,Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Izmir, Turkey
| | | | - Esra Erdal
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Izmir, Turkey.,Department of Medical Biology and Genetics, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
32
|
Lu Q, Guo Q, Xin M, Lim C, Gamero AM, Gerhard GS, Yang L. LncRNA TP53TG1 Promotes the Growth and Migration of Hepatocellular Carcinoma Cells via Activation of ERK Signaling. Noncoding RNA 2021; 7:ncrna7030052. [PMID: 34564314 PMCID: PMC8482154 DOI: 10.3390/ncrna7030052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/27/2022] Open
Abstract
Long non-coding RNA (lncRNA) TP53 target 1 (TP53TG1) was discovered as a TP53 target gene. TP53TG1 has been reported as having dual roles by exerting tumor-suppressive and oncogenic activities that vary depending on the cancer type. Yet, the role of TP53TG1 in hepatocellular carcinoma (HCC) is not fully understood. In this study, we performed both gain- and loss-of-function studies to determine the biological role of TP53TG1 in HCC. We found that the knockdown of TP53 in HCC cells caused the upregulation of TP53TG1. Furthermore, we found that the knockdown of TP53TG1 not only suppressed HCC cell proliferation and migration, but also reduced intrinsic ERK signaling. In contrast, the overexpression of TP53TG1 increased ERK activation and enhanced HCC proliferation. In conclusion, our study reveals an oncogenic role of TP53TG1 in HCC, which provides a novel insight into the cell-type-specific function of TP53TG1 in HCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ling Yang
- Correspondence: ; Tel.: +1-215-707-3779
| |
Collapse
|
33
|
Cheng CLH, Tsang FHC, Wei L, Chen M, Chin DWC, Shen J, Law CT, Lee D, Wong CCL, Ng IOL, Wong CM. Bromodomain-containing protein BRPF1 is a therapeutic target for liver cancer. Commun Biol 2021; 4:888. [PMID: 34285329 PMCID: PMC8292510 DOI: 10.1038/s42003-021-02405-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
Epigenetic deregulation plays an essential role in hepatocellular carcinoma (HCC) progression. Bromodomains are epigenetic "readers" of histone acetylation. Recently, bromodomain inhibitors have exhibited promising therapeutic potential for cancer treatment. Using transcriptome sequencing, we identified BRPF1 (bromodomain and PHD finger containing 1) as the most significantly upregulated gene among the 43 bromodomain-containing genes in human HCC. BRPF1 upregulation was significantly associated with poor patient survival. Gene ablation or pharmacological inactivation of BRPF1 significantly attenuated HCC cell growth in vitro and in vivo. BRPF1 was involved in cell cycle progression, senescence and cancer stemness. Transcriptome sequencing revealed that BRPF1 is a master regulator controlling the expression of multiple key oncogenes, including E2F2 and EZH2. We demonstrated that BRPF1 activated E2F2 and EZH2 expression by facilitating promoter H3K14 acetylation through MOZ/MORF complex. In conclusion, BRPF1 is frequently upregulated in human HCCs. Targeting BRPF1 may be an approach for HCC treatment.
Collapse
Affiliation(s)
- Carol Lai-Hung Cheng
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Felice Hoi-Ching Tsang
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Lai Wei
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Mengnuo Chen
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Don Wai-Ching Chin
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Jialing Shen
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Cheuk-Ting Law
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Derek Lee
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Carmen Chak-Lui Wong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Irene Oi-Lin Ng
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Chun-Ming Wong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong. .,Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.
| |
Collapse
|
34
|
Vogel A, Frenette C, Sung M, Daniele B, Baron A, Chan SL, Blanc JF, Tamai T, Ren M, Lim HJ, Palmer DH, Takami Y, Kudo M. Baseline Liver Function and Subsequent Outcomes in the Phase 3 REFLECT Study of Patients with Unresectable Hepatocellular Carcinoma. Liver Cancer 2021; 10:510-521. [PMID: 34721512 PMCID: PMC8527908 DOI: 10.1159/000516490] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/13/2021] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Baseline liver function among patients starting treatment for unresectable hepatocellular carcinoma (uHCC) impacts survival and could impact efficacy outcomes and safety profiles of treatments. This post hoc analysis of the phase 3 REFLECT study examined the efficacy and safety outcomes for lenvatinib and for sorafenib in patients with uHCC, assessed by Child-Pugh score (CPS) and albumin-bilirubin (ALBI) grade. METHODS Efficacy and safety were assessed in patient cohorts from REFLECT according to study entry baseline ALBI grade and CPS. RESULTS Lenvatinib treatment generally provided survival benefits in all groups. Median overall survival (OS) among patients with an ALBI grade of 1 was consistently higher than among patients with an ALBI grade of 2 for both the lenvatinib and sorafenib arms (lenvatinib: 17.4 vs. 8.6 months; sorafenib: 14.6 vs. 7.7 months, respectively). Median OS among patients with a CPS of 5 was consistently higher than among patients with a CPS of 6 (lenvatinib: 15.3 vs. 9.4 months; sorafenib: 14.2 vs. 7.9 months, respectively). Progression-free survival and objective response rates for these ALBI grades and CPS demonstrated similar patterns. Among patients who received lenvatinib and experienced a treatment-related treatment-emergent adverse event leading to withdrawal, 6.6% had an ALBI grade of 1, while 13.3% had an ALBI grade of 2, and 7.9% had a CPS of 5, while 12.1% had a CPS of 6. CONCLUSIONS Better liver function at baseline, as measured by ALBI grade or CPS, may be prognostic for better survival outcomes in patients with uHCC undergoing treatment with lenvatinib or sorafenib.
Collapse
Affiliation(s)
- Arndt Vogel
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, Hannover, Germany,*Arndt Vogel,
| | - Catherine Frenette
- Department of Transplantation Hepatology, Scripps MD Anderson Cancer Center, La Jolla, California, USA
| | - Max Sung
- Department of Hematology-Oncology, Tisch Cancer Institute at Mount Sinai, New York, New York, USA
| | - Bruno Daniele
- Department of Oncology, Ospedale del Mare, Napoli, Italy
| | - Ari Baron
- Department of Clinical Oncology, Pacific Hematology Oncology Associates, San Francisco, California, USA
| | - Stephen L. Chan
- Department of Clinical Oncology, The State Key Laboratory of Translational Oncology, Sir YK Pao Centre for Cancer, The Chinese University of Hong Kong, Hong Kong, China
| | - Jean Frédéric Blanc
- Department of Clinical Research and Innovation, University Hospital of Bordeaux, Bordeaux, France
| | | | - Min Ren
- Eisai Inc., Biostatistics, Oncology Business Group, Woodcliff Lake, New Jersey, USA
| | - Howard J. Lim
- Department of Medicine, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Daniel H. Palmer
- Department of Medical Oncology, Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Yuko Takami
- Department of Hepato-Biliary-Pancreatic Surgery, National Hospital Organization Kyushu Medical Center, Clinical Research Institute, Fukuoka, Japan
| | - Masatoshi Kudo
- Department of Medicine, Kindai University Faculty of Medicine, Osakasayama, Japan
| |
Collapse
|
35
|
Rizzo A, Dadduzio V, Ricci AD, Massari F, Di Federico A, Gadaleta-Caldarola G, Brandi G. Lenvatinib plus pembrolizumab: the next frontier for the treatment of hepatocellular carcinoma? Expert Opin Investig Drugs 2021; 31:371-378. [PMID: 34167433 DOI: 10.1080/13543784.2021.1948532] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Introduction: While sorafenib monotherapy represented the mainstay of medical treatment for advanced hepatocellular carcinoma (HCC) patients for more than a decade, novel agents and combination therapies have recently produced unprecedented paradigm shifts. The combination of lenvatinib plus pembrolizumab is now being evaluated as a front-line treatment in advanced HCC patients; early phase clinical trials have already reported promising results.Areas covered: This paper reviews the combination of lenvatinib plus pembrolizumab for the treatment of advanced HCC. The preclinical rationale and completed and ongoing trials are examined and later, the authors reflect on biomarkers of predictive of response to immune-based combinations and future treatment decision-making on the basis of tolerability and clinical benefits provided by these novel therapeutics. A literature search was conducted in April 2021 of Pubmed/Medline, Cochrane library and Scopus databases; moreover, abstracts of international cancer meetings were reviewed.Expert opinion: The landscape of new agents and combinations continues to expand. Recently, immune-based combinations have reported important results in advanced HCC, as witnessed by the landmark IMbrave150 trial. Based on the promising results of early phase clinical trials, lenvatinib plus pembrolizumab has the potential to represent a novel treatment option in this setting.
Collapse
Affiliation(s)
- Alessandro Rizzo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italia.,Medical Oncology Unit, "Mons. R. Dimiccoli" Hospital, Barletta (BT), Italy
| | - Vincenzo Dadduzio
- Medical Oncology Unit, "Mons. R. Dimiccoli" Hospital, Barletta (BT), Italy
| | - Angela Dalia Ricci
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italia.,Medical Oncology Unit, "Mons. R. Dimiccoli" Hospital, Barletta (BT), Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italia
| | | | | | - Giovanni Brandi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italia
| |
Collapse
|
36
|
Damiris K, Abbad H, Pyrsopoulos N. Cellular based treatment modalities for unresectable hepatocellular carcinoma. World J Clin Oncol 2021; 12:290-308. [PMID: 34131562 PMCID: PMC8173328 DOI: 10.5306/wjco.v12.i5.290] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary malignancy of the liver and is unfortunately associated with an overall poor prognosis and high mortality. Early and intermediate stages of HCC allow for treatment with surgical resection, ablation and even liver transplantation, however disease progression warrants conventional systemic therapy. For years treatment options were limited to molecular-targeting medications, of which sorafenib remains the standard of care. The recent development and success of immune checkpoint inhibitors has proven to be a breakthrough in the treatment of HCC, but there is an urgent need for the development of further novel therapeutic treatments that prolong overall survival and minimize recurrence. Current investigation is focused on adoptive cell therapy including chimeric antigen receptor-T cells (CAR-T cells), T cell receptor (TCR) engineered T cells, dendritic cells, natural killer cells, and tumor infiltrating lymphocyte cells, which have shown remarkable success in the treatment of hematological and solid tumor malignancies. In this review we briefly introduce readers to the currently approved systemic treatment options and present clinical and experimental evidence of HCC immunotherapeutic treatments that will hopefully one day allow for revolutionary change in the treatment modalities used for unresectable HCC. We also provide an up-to-date compilation of ongoing clinical trials investigating CAR-T cells, TCR engineered T cells, cancer vaccines and oncolytic viruses, while discussing strategies that can help overcome commonly faced challenges when utilizing cellular based treatments.
Collapse
Affiliation(s)
- Konstantinos Damiris
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, United States
| | - Hamza Abbad
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, United States
| | - Nikolaos Pyrsopoulos
- Department of Medicine, Division of Gastroenterology and Hepatology, Rutgers New Jersey Medical School, Newark, NJ 07103, United States
| |
Collapse
|
37
|
Kim S, Kim KH, Kim BK, Park JY, Ahn SH, Kim DY, Kim SU. Lenvatinib is independently associated with the reduced risk of progressive disease when compared with sorafenib in patients with advanced hepatocellular carcinoma. J Gastroenterol Hepatol 2021; 36:1317-1325. [PMID: 33217054 DOI: 10.1111/jgh.15355] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/08/2020] [Accepted: 11/14/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Recently, lenvatinib demonstrated non-inferiority to sorafenib in terms of overall survival (OS) in a randomized phase III study that was conducted at 154 sites in 20 countries. Here, we investigated treatment outcomes and safety of lenvatinib compared with sorafenib and identified independent predictors of poor outcomes, including shorter progression-free survival (PFS) and OS in Korean patients with unresectable hepatocellular carcinoma (HCC). METHODS Patients with advanced HCC treated with lenvatinib or sorafenib at Yonsei Liver Center, Severance Hospital, Yonsei University College of Medicine between October 2018 to October 2019 were considered eligible. Response evaluation was performed according to the modified Response Evaluation Criteria in Solid Tumors. RESULTS The lenvatinib arm had a significantly lower proportion of patients who received prior anti-HCC treatments (47.7% vs 78.7%; P < 0.001) than those in the sorafenib arm. Univariate analysis showed that ECOG 1 (vs 0), serum albumin, alpha-fetoprotein (AFP), previous anti-HCC treatments, and lenvatinib (vs sorafenib) were significant predictors of progressive disease (all P < 0.05). In the subsequent multivariate analysis, ECOG 1 (vs 0) (hazard ratio [HR] = 4.721, 95% confidence interval [CI] 1.371-16.259; P = 0.014), higher AFP level (HR = 1.000, 95% CI 1.000-1.000; P = 0.015), and lenvatinib treatment (vs sorafenib) (HR = 0.461, 95% CI 0.264-0.804; P = 0.006) independently predicted a higher probability of progressive disease. CONCLUSIONS Patients treated with lenvatinib demonstrated significantly longer PFS than those treated with sorafenib. Furthermore, no significant differences were observed in mortality rates between the two groups, which indicated that lenvatinib is non-inferior to sorafenib in terms of OS.
Collapse
Affiliation(s)
- Soojin Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung Hyun Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Beom Kyung Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Jun Yong Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Sang Hoon Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Do Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Seung Up Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| |
Collapse
|
38
|
Radiomics, Radiogenomics, and Next-Generation Molecular Imaging to Augment Diagnosis of Hepatocellular Carcinoma. ACTA ACUST UNITED AC 2021; 26:108-115. [PMID: 32205534 DOI: 10.1097/ppo.0000000000000435] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ultrasound, computed tomography, magnetic resonance imaging, and [F]F-fluorodeoxyglucose positron emission tomography are invaluable in the clinical evaluation of human cancers. Radiomics and radiogenomics tools may allow clinicians to standardize interpretation of these conventional imaging modalities, while better linking radiographic hallmarks to disease biology and prognosis. These advances, coupled with next-generation positron emission tomography imaging tracers capable of providing biologically relevant tumor information, may further expand the tools available in our armamentarium against human cancers. We present current imaging methods and explore emerging research that may improve diagnosis and monitoring of local, oligometastatic, and disseminated cancers exhibiting heterogeneous uptake of [F]F-fluorodeoxyglucose, using hepatocellular carcinoma as an example.
Collapse
|
39
|
Liver Cancer: Therapeutic Challenges and the Importance of Experimental Models. Can J Gastroenterol Hepatol 2021; 2021:8837811. [PMID: 33728291 PMCID: PMC7937489 DOI: 10.1155/2021/8837811] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 01/16/2021] [Accepted: 02/16/2021] [Indexed: 02/06/2023] Open
Abstract
Liver cancer is one of the main causes of death related to cancer worldwide; its etiology is related with infections by C or B hepatitis virus, alcohol consumption, smoking, obesity, nonalcoholic fatty liver disease, diabetes, and iron overload, among other causes. Several kinds of primary liver cancer occur, but we will focus on hepatocellular carcinoma (HCC). Numerous cellular signaling pathways are implicated in hepatocarcinogenesis, including YAP-HIPPO, Wnt-β-catenin, and nuclear factor-κB (NF-κB); these in turn are considered novel therapeutic targets. In this review, the role of lipid metabolism regulated by peroxisome proliferator-activated receptor gamma (PPARγ) in the development of HCC will also be discussed. Moreover, recent evidence has been obtained regarding the participation of epigenetic changes such as acetylation and methylation of histones and DNA methylation in the development of HCC. In this review, we provide detailed and current information about these topics. Experimental models represent useful tools for studying the different stages of liver cancer and help to develop new pharmacologic treatments. Each model in vivo and in vitro has several characteristics and advantages to offer for the study of this disease. Finally, the main therapies approved for the treatment of HCC patients, first- and second-line therapies, are described in this review. We also describe a novel option, pirfenidone, which due to its pharmacological properties could be considered in the future as a therapeutic option for HCC treatment.
Collapse
|
40
|
Malik A, Thanekar U, Amarachintha S, Mourya R, Nalluri S, Bondoc A, Shivakumar P. "Complimenting the Complement": Mechanistic Insights and Opportunities for Therapeutics in Hepatocellular Carcinoma. Front Oncol 2021; 10:627701. [PMID: 33718121 PMCID: PMC7943925 DOI: 10.3389/fonc.2020.627701] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/22/2020] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary malignancy of the liver and a leading cause of death in the US and worldwide. HCC remains a global health problem and is highly aggressive with unfavorable prognosis. Even with surgical interventions and newer medical treatment regimens, patients with HCC have poor survival rates. These limited therapeutic strategies and mechanistic understandings of HCC immunopathogenesis urgently warrant non-palliative treatment measures. Irrespective of the multitude etiologies, the liver microenvironment in HCC is intricately associated with chronic necroinflammation, progressive fibrosis, and cirrhosis as precedent events along with dysregulated innate and adaptive immune responses. Central to these immunological networks is the complement cascade (CC), a fundamental defense system inherent to the liver which tightly regulates humoral and cellular responses to noxious stimuli. Importantly, the liver is the primary source for biosynthesis of >80% of complement components and expresses a variety of complement receptors. Recent studies implicate the complement system in liver inflammation, abnormal regenerative responses, fibrosis, carcinogenesis, and development of HCC. Although complement activation differentially promotes immunosuppressive, stimulant, and angiogenic microenvironments conducive to HCC development, it remains under-investigated. Here, we review derangement of specific complement proteins in HCC in the context of altered complement regulatory factors, immune-activating components, and their implications in disease pathogenesis. We also summarize how complement molecules regulate cancer stem cells (CSCs), interact with complement-coagulation cascades, and provide therapeutic opportunities for targeted intervention in HCC.
Collapse
Affiliation(s)
- Astha Malik
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Unmesha Thanekar
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Surya Amarachintha
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Reena Mourya
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Shreya Nalluri
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Alexander Bondoc
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Pranavkumar Shivakumar
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
41
|
Zhang J, Song Q, Wu M, Zheng W. The Emerging Roles of Exosomes in the Chemoresistance of Hepatocellular Carcinoma. Curr Med Chem 2021; 28:93-109. [PMID: 32000636 DOI: 10.2174/0929867327666200130103206] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/08/2019] [Accepted: 10/23/2019] [Indexed: 11/22/2022]
Abstract
Hepatocellular carcinoma (HCC) is a common gastrointestinal malignancy with a leading incidence of cancer-related mortality worldwide. Despite the progress of treatment options, there remains low efficacy for patients with intermediate-advanced HCC, due to tumor metastasis, recurrence and chemoresistance. Increasing evidence suggests that exosomes in the tumor microenvironment (TME), along with other extracellular vesicles (EVs) and cytokines, contribute to the drug chemosensitivity of cancer cells. Exosomes, the intercellular communicators in various biological activities, have shown to play important roles in HCC progression. This review summarizes the underlying associations between exosomes and chemoresistance of HCC cells. The exosomes derived from distinct cell types mediate the drug resistance by regulating drug efflux, epithelial-mesenchymal transition (EMT), cancer stem cell (CSC) properties, autophagic phenotypes, as well as the immune response. In summary, TME-related exosomes can be a potential target to reverse chemoresistance and a candidate biomarker of drug efficacy in HCC patients.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Chemotherapy, Affiliated Hospital of Nantong University, 20 Xisi Road, 226001 Nantong, Jiangsu, China
| | - Qianqian Song
- Department of Radiology, Wake Forest School of Medicine, One Medical Center Boulevard, Winston-Salem, 27157 NC, United States
| | - Mengna Wu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, 226001 Nantong, Jiangsu, China
| | - Wenjie Zheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, 226001 Nantong, Jiangsu, China
| |
Collapse
|
42
|
Hatanaka T, Naganuma A, Kakizaki S. Lenvatinib for Hepatocellular Carcinoma: A Literature Review. Pharmaceuticals (Basel) 2021; 14:36. [PMID: 33418941 PMCID: PMC7825021 DOI: 10.3390/ph14010036] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/01/2021] [Accepted: 01/03/2021] [Indexed: 12/11/2022] Open
Abstract
Lenvatinib, which is an oral multikinase inhibitor, showed non-inferiority to the sorafenib in terms of overall survival (OS) and a higher objective response rate (ORR) and better progression-free survival (PFS) in patients with hepatocellular carcinoma (HCC). A good liver function and Barcelona Clinic Liver Cancer (BCLC) intermediate stage were the key factors in achieving therapeutic efficacy. The management of adverse events plays an important role in continuing lenvatinib treatment. While sequential therapies contributed to prolonging overall survival, effective molecular targeted agents for the administration after lenvatinib have not been established. Repeated transcatheter arterial chemoembolization (TACE) was associated with a decline in the liver function and poor therapeutic response in BCLC intermediate patients. Recently, the Asia-Pacific Primary Liver Cancer Expert (APPLE) Consensus Statement proposed the criteria for TACE unsuitability. Upfront systemic therapy may be better for the BCLC intermediate stage HCC patients with a high tumor burden, while selective TACE will be recommended for obtaining a curative response in patients with a low tumor burden. This article reviews the therapeutic response, management of adverse events, post-progression treatment after Lenvatinib, and treatment strategy for BCLC intermediate stage HCC.
Collapse
Affiliation(s)
- Takeshi Hatanaka
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, 564-1 Kamishindenmachi, Maebashi, Gunma 371-0821, Japan
| | - Atsushi Naganuma
- Department of Gastroenterology, National Hospital Organization Takasaki General Medical Center, 36 Takamatsucho, Takasaki, Gunma 370-0829, Japan;
| | - Satoru Kakizaki
- Department of Clinical Research, National Hospital Organization Takasaki General Medical Center, 36 Takamatsucho, Takasaki, Gunma 370-0829, Japan;
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, 3-39-15 Showa, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
43
|
Kulik L, da Fonseca LG, He AR, Rimola J, Wilson Woods A, Zöllner YF, Galle PR. Potential Impact of IMbrave150 Results in the Evolving Treatment Landscape of Advanced Hepatocellular Carcinoma: A Multidisciplinary Expert Opinion. J Hepatocell Carcinoma 2020; 7:423-433. [PMID: 33376711 PMCID: PMC7762763 DOI: 10.2147/jhc.s274930] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/10/2020] [Indexed: 12/22/2022] Open
Abstract
A virtual expert roundtable was convened on April 16, 2020, to discuss the evolving landscape of care for treating patients with advanced hepatocellular carcinoma (HCC) and discuss questions related to patient care and treatment selection. This commentary presents highlights from this discussion and provides an expert opinion about approaches to treatment for HCC in the Americas and the European Union. We anticipate that atezolizumab plus bevacizumab will become the standard of care for advanced HCC patients. However, this approach will make decisions regarding the sequencing of treatments for second-line therapies and beyond more challenging. Therapy will require individualization based on patient characteristics and preferences, while insurance coverage decisions and requirements may also impact the options that patients can access. Additional research regarding prognostic and predictive biomarkers is needed to help better identify optimal treatment approaches for specific patient populations. Multidisciplinary tumor boards will continue to play a critical role in guiding treatment selection for individual patients. Atezolizumab plus bevacizumab offers a promising new first-line therapeutic option for patients with advanced HCC, but more research is needed to optimize and individualize patient therapy.
Collapse
Affiliation(s)
| | | | | | - Jordi Rimola
- Radiology Department, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain
| | - Andrea Wilson Woods
- Blue Faery: The Adrienne Wilson Liver Cancer Association, Birmingham, AL, USA
| | - York F Zöllner
- Hamburg University of Applied Sciences, Competence Center Health, Hamburg, Germany
| | | |
Collapse
|
44
|
Nia A, Dhanasekaran R. Genomic Landscape of HCC. CURRENT HEPATOLOGY REPORTS 2020; 19:448-461. [PMID: 33816052 PMCID: PMC8015384 DOI: 10.1007/s11901-020-00553-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/23/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is a leading cause of cancer related mortality in the world and it has limited treatment options. Understanding the molecular drivers of HCC is important to develop novel biomarkers and therapeutics. PURPOSE OF REVIEW HCC arises in a complex background of chronic hepatitis, fibrosis and liver regeneration which lead to genomic changes. Here, we summarize studies that have expanded our understanding of the molecular landscape of HCC. RECENT FINDINGS Recent technological advances in next generation sequencing (NGS) have elucidated specific genetic and molecular programs involved in hepatocarcinogenesis. We summarize the major somatic mutations and epigenetic changes have been identified in NGS-based studies. We also describe promising molecular therapies and immunotherapies which target specific genetic and epigenetic molecular events. SUMMARY The genomic landscape of HCC is incredibly complex and heterogeneous. Promising new developments are helping us decipher the molecular drivers of HCC and leading to new therapies.
Collapse
|
45
|
Lin C, Yang T, Yen C, Cheng R, Liu J, Hsu C. Safety and Preliminary Efficacy of Ramucirumab in Combination with FOLFOX4 in Patients with Advanced Hepatocellular Carcinoma: A Nonrandomized, Open-Label, Phase Ib Study. Oncologist 2020; 25:e1921-e1929. [PMID: 33017497 PMCID: PMC8108062 DOI: 10.1002/onco.13550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022] Open
Abstract
LESSONS LEARNED The combination of ramucirumab (8 mg/kg intravenous, day 1 every 2 weeks) and FOLFOX4 as first-line treatment in patients with advanced hepatocellular carcinoma (HCC) was not sufficiently tolerated. Preliminary efficacy data suggest that the combination may provide clinical benefit to patients with HCC. Dose modification and patient selection should be considered for the future development of ramucirumab plus FOLFOX chemotherapy for advanced HCC. BACKGROUND The objective of this study was to investigate the safety, preliminary efficacy, pharmacokinetics, and immunogenicity of ramucirumab plus FOLFOX4 as first-line treatment in patients with advanced hepatocellular carcinoma (HCC). METHODS Patients received ramucirumab (8 mg/kg) intravenously (IV) on day 1, followed by FOLFOX4 (oxaliplatin 85 mg/m2 IV on day 1, folinic acid 200 mg/m2 IV, bolus fluorouracil [5-FU] 400 mg/m2 , and a continuous infusion of 5-FU 600 mg/m2 over 22 hours, on days 1 and 2) every 2 weeks. The primary endpoint was to assess the safety and tolerability of the combination therapy. RESULTS Eight patients (6 men, 2 women) were treated; all eight patients experienced at least one treatment-emergent adverse event (TEAE) of grade ≥3. Dose-limiting toxicities occurred in three patients (37.5%): hepatic hemorrhage (grade 4), blood bilirubin increased (grade 3), and febrile neutropenia (grade 3). Two patients discontinued study because of hepatic hemorrhage (grade 4) and blood bilirubin increase (grade 3). Six deaths occurred due to progressive disease, and no deaths due to TEAEs. CONCLUSION There were no unexpected safety findings with ramucirumab plus FOLFOX4 based on the known safety and toxicity of this regimen. The combination was not sufficiently tolerated in patients with advanced HCC at the specified dose and schedule.
Collapse
Affiliation(s)
- Chia‐Chi Lin
- Department of Oncology, National Taiwan University HospitalTaipeiTaiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of MedicineTaipeiTaiwan
| | | | - Chia‐Jui Yen
- Institute of Clinical Medicine, National Cheng Kung UniversityTainanTaiwan
| | | | | | - Chiun Hsu
- Department of Oncology, National Taiwan University HospitalTaipeiTaiwan
| |
Collapse
|
46
|
Polidoro MA, Mikulak J, Cazzetta V, Lleo A, Mavilio D, Torzilli G, Donadon M. Tumor microenvironment in primary liver tumors: A challenging role of natural killer cells. World J Gastroenterol 2020; 26:4900-4918. [PMID: 32952338 PMCID: PMC7476172 DOI: 10.3748/wjg.v26.i33.4900] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/24/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
In the last years, several studies have been focused on elucidate the role of tumor microenvironment (TME) in cancer development and progression. Within TME, cells from adaptive and innate immune system are one of the main abundant components. The dynamic interactions between immune and cancer cells lead to the activation of complex molecular mechanisms that sustain tumor growth. This important cross-talk has been elucidate for several kind of tumors and occurs also in patients with liver cancer, such as hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA). Liver is well-known to be an important immunological organ with unique microenvironment. Here, in normal conditions, the rich immune-infiltrating cells cooperate with non-parenchymal cells, such as liver sinusoidal endothelial cells and Kupffer cells, favoring self-tolerance against gut antigens. The presence of underling liver immunosuppressive microenvironment highlights the importance to dissect the interaction between HCC and iCCA cells with immune infiltrating cells, in order to understand how this cross-talk promotes tumor growth. Deeper attention is, in fact, focused on immune-based therapy for these tumors, as promising approach to counteract the intrinsic anti-tumor activity of this microenvironment. In this review, we will examine the key pathways underlying TME cell-cell communications, with deeper focus on the role of natural killer cells in primary liver tumors, such as HCC and iCCA, as new opportunities for immune-based therapeutic strategies.
Collapse
Affiliation(s)
- Michela Anna Polidoro
- Hepatobiliary Immunopathology Laboratory, Humanitas Clinical and Research Center – IRCCS, Rozzano 20089, Milan, Italy
| | - Joanna Mikulak
- Laboratory of Clinical and Experimental Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano 20089, Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Rozzano 20089, Milan, Italy
| | - Valentina Cazzetta
- Laboratory of Clinical and Experimental Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano 20089, Milan, Italy
| | - Ana Lleo
- Hepatobiliary Immunopathology Laboratory, Humanitas Clinical and Research Center – IRCCS, Rozzano 20089, Milan, Italy
- Department of Biomedical Science, Humanitas University, Pieve Emanuele 20090, Milan, Italy
- Department of Internal Medicine, Humanitas Clinical and Research Center – IRCCS, Rozzano 20089, Milan, Italy
| | - Domenico Mavilio
- Laboratory of Clinical and Experimental Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano 20089, Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Rozzano 20089, Milan, Italy
| | - Guido Torzilli
- Department of Biomedical Science, Humanitas University, Pieve Emanuele 20090, Milan, Italy
- Department of Hepatobiliary and General Surgery, Humanitas Clinical and Research Center - IRCCS, Rozzano 20089, Milan, Italy
| | - Matteo Donadon
- Department of Biomedical Science, Humanitas University, Pieve Emanuele 20090, Milan, Italy
- Department of Hepatobiliary and General Surgery, Humanitas Clinical and Research Center - IRCCS, Rozzano 20089, Milan, Italy
| |
Collapse
|
47
|
Liu S, Chu B, Cai C, Wu X, Yao W, Wu Z, Yang Z, Li F, Liu Y, Dong P, Gong W. DGCR5 Promotes Gallbladder Cancer by Sponging MiR-3619-5p via MEK/ERK1/2 and JNK/p38 MAPK Pathways. J Cancer 2020; 11:5466-5477. [PMID: 32742494 PMCID: PMC7391188 DOI: 10.7150/jca.46351] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/15/2020] [Indexed: 02/05/2023] Open
Abstract
Gallbladder cancer (GBC) is a highly aggressive malignant cancer with poor prognosis. Long noncoding RNA (lncRNA) DiGeorge syndrome critical region gene (DGCR5) has been reported to participate in various types of cancers, but its role in GBC remains largely unknown. This study aimed to explore the functions and mechanisms of DGCR5 in GBC. Here, we found that DGCR5 was upregulated in GBC tissues and cell lines. Through functional experiments, it was demonstrated that silence of DGCR5 significantly suppressed the cell proliferation, migration, invasion, and induced apoptosis and cell cycle arrest in GBC cells. In addition, miR-3619-5p was predicted and further verified as the target of DGCR5. Moreover, miR-3619-5p was observed downregulated in GBC tissues and cell lines, and miR-3619-5p mimics repressed the GBC cell proliferation, migration, invasion and could be rescued by DGCR5 overexpression. Mechanistically, it was found that DGCR5 knockdown and miR-3619-5p mimics inactivated the MEK/ERK1/2 and JNK/p38 MAPK pathways. In addition, rescue experiments indicated that inhibition of MEK/ERK1/2 and JNK/p38 MAPK pathways could reverse the effects of DGCR5 overexpression on cell proliferation, migration and invasion. Finally, xenograft model assay was used to validate that knockdown of DGCR5 suppressed GBC via regulating MEK/ERK1/2 and JNK/p38 MAPK pathways in vivo. Taken together, it was uncovered in our study that DGCR5 exerts an oncogenic role by sponging miR-3619-5p and activating MEK/ERK1/2 and JNK/p38 MAPK pathways in GBC progression.
Collapse
Affiliation(s)
- Shilei Liu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Bingfeng Chu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Chen Cai
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Xiangsong Wu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Wenyan Yao
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Ziyou Wu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Ziyi Yang
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Fengnan Li
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Yingbin Liu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Ping Dong
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai 200092, China
| | - Wei Gong
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, No. 1665 Kongjiang Road, Shanghai 200092, China
| |
Collapse
|
48
|
Thylur RP, Roy SK, Shrivastava A, LaVeist TA, Shankar S, Srivastava RK. Assessment of risk factors, and racial and ethnic differences in hepatocellular carcinoma. JGH OPEN 2020; 4:351-359. [PMID: 32514436 PMCID: PMC7273694 DOI: 10.1002/jgh3.12336] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/05/2020] [Accepted: 03/24/2020] [Indexed: 12/24/2022]
Abstract
Despite improved screening and surveillance guidelines, significant race/ethnicity‐specific disparities in hepatocellular carcinoma (HCC) continue to exist and disproportionately affect minority and disadvantaged populations. This trend indicates that social determinants, genetic, and environmental factors are driving the epidemic at the population level. Race and geography had independent associations with risk of mortality among patients with HCC. The present review discusses the risk factors and issues related to disparities in HCC. The underlying etiologies for these disparities are complex and multifactorial. Some of the risk factors for developing HCC include hepatitis B (HBV) and hepatitis C (HCV) viral infection, nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, smoking and alcohol consumption. In addition, population genetics; socioeconomic and health care access; treatment and prevention differences; and genetic, behavioral, and biological influences can contribute to HCC. Acculturation of ethnic minorities, insurance status, and access to health care may further contribute to the observed disparities in HCC. By increasing awareness, better modalities for screening and surveillance, improving access to health care, and adapting targeted preventive and therapeutic interventions, disparities in HCC outcomes can be reduced or eliminated.
Collapse
Affiliation(s)
- Ramesh P Thylur
- Stanley S. Scott Cancer Center Louisiana State University Health-New Orleans School of Medicine New Orleans Louisiana USA
| | - Sanjit K Roy
- Stanley S. Scott Cancer Center Louisiana State University Health-New Orleans School of Medicine New Orleans Louisiana USA
| | | | - Thomas A LaVeist
- Department of Health Policy and Management Tulane University School of Public Health and Tropical Medicine New Orleans Louisiana USA
| | - Sharmila Shankar
- Stanley S. Scott Cancer Center Louisiana State University Health-New Orleans School of Medicine New Orleans Louisiana USA.,Department of Genetics Louisiana State University Health Sciences Center-New Orleans New Orleans Louisiana USA
| | - Rakesh K Srivastava
- Stanley S. Scott Cancer Center Louisiana State University Health-New Orleans School of Medicine New Orleans Louisiana USA.,Department of Genetics Louisiana State University Health Sciences Center-New Orleans New Orleans Louisiana USA
| |
Collapse
|
49
|
Stoiber P, Ekladious I, Zhao Q, Colson YL, Schaus SE, Hansen U, Grinstaff MW. Expansile Nanoparticles Encapsulate Factor Quinolinone Inhibitor 1 and Accumulate in Murine Liver upon Intravenous Administration. Biomacromolecules 2020; 21:1499-1506. [PMID: 32101401 DOI: 10.1021/acs.biomac.0c00064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Expansile nanoparticles (eNPs) are a promising pH-responsive polymeric drug delivery vehicle, as demonstrated in multiple intraperitoneal cancer models. However, previous delivery routes were limited to intraperitoneal injection and to a single agent, paclitaxel. In this study, we preliminarily evaluate the biodistribution and in vivo toxicity of eNPs in mice after intravenous injection. The eNPs localize predominantly to the liver, without detectable acute toxicity in the liver or other key organs. On the basis of these results, we encapsulated FQI1, a promising lead compound for treatment of hepatocellular carcinoma, in eNPs. eNPs are taken up by cancerous and noncancerous human liver cells in vitro, although at different rates. FQI1-loaded eNPs release FQI1 in a pH-dependent manner and limit proliferation equivalently to unencapsulated FQI1 in immortalized hepatocytes in vitro. eNPs are a versatile platform delivery system for therapeutic compounds and have potential utility in the treatment of liver disease.
Collapse
Affiliation(s)
- Patrick Stoiber
- MCBB Graduate Program and Department of Biology, Boston University, Boston, Massachusetts 02215, United States
| | - Iriny Ekladious
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
| | - Qing Zhao
- Department of Pathology and Laboratory Medicine, Boston University Medical Center, Boston, Massachusetts 02118, United States
| | - Yolonda L Colson
- Division of Thoracic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Scott E Schaus
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States.,Center for Molecular Discovery, Boston University, Boston, Massachusetts 02215, United States
| | - Ulla Hansen
- MCBB Graduate Program and Department of Biology, Boston University, Boston, Massachusetts 02215, United States
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States.,Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| |
Collapse
|
50
|
Kiruthiga C, Devi KP, Nabavi SM, Bishayee A. Autophagy: A Potential Therapeutic Target of Polyphenols in Hepatocellular Carcinoma. Cancers (Basel) 2020; 12:cancers12030562. [PMID: 32121322 PMCID: PMC7139730 DOI: 10.3390/cancers12030562] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a conserved biological phenomenon that maintains cellular homeostasis through the clearing of damaged cellular components under cellular stress and offers the cell building blocks for cellular survival. Aberrations in autophagy subsidize to various human pathologies, such as dementia, cardiovascular diseases, leishmaniosis, influenza, hepatic diseases, and cancer, including hepatocellular carcinoma (HCC). HCC is the fifth common mortal type of liver cancer globally, with an inhomogeneous topographical distribution and highest incidence tripled in men than women. Existing treatment procedures with liver cancer patients result in variable success rates and poor prognosis due to their drug resistance and toxicity. One of the pathophysiological mechanisms that are targeted during the development of anti-liver cancer drugs is autophagy. Generally, overactivated autophagy may lead to a non-apoptotic form of programmed cell death (PCD) or autophagic cell death or type II PCD. Emerging evidence suggests that manipulation of autophagy could induce type II PCD in cancer cells, acting as a potential tumor suppressor. Hence, altering autophagic signaling offers new hope for the development of novel drugs for the therapy of resistant cancer cells. Natural polyphenolic compounds, including flavonoids and non-flavonoids, execute their anticarcinogenic mechanism through upregulating tumor suppressors and autophagy by modulating canonical (Beclin-1-dependent) and non-canonical (Beclin-1-independent) signaling pathways. Additionally, there is evidence signifying that plant polyphenols target angiogenesis and metastasis in HCC via interference with multiple intracellular signals and decrease the risk against HCC. The current review offers a comprehensive understanding of how natural polyphenolic compounds exhibit their anti-HCC effects through regulation of autophagy, the non-apoptotic mode of cell death.
Collapse
Affiliation(s)
- Chandramohan Kiruthiga
- Department of Biotechnology, Alagappa University (Science Campus), Karaikudi 630 003, Tamil Nadu, India;
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University (Science Campus), Karaikudi 630 003, Tamil Nadu, India;
- Correspondence: (K.P.D.); or (A.B.); Tel.: +91-4565223325 (K.P.D.); +1-941-782-5950 (A.B.)
| | - Seyed M. Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran;
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
- Correspondence: (K.P.D.); or (A.B.); Tel.: +91-4565223325 (K.P.D.); +1-941-782-5950 (A.B.)
| |
Collapse
|