Systemic Therapies for Hepatocellular Carcinoma: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor:

Clinical treatment of Hepatocellular carcinoma (HCC) in initial stages includes surgical therapies, resection or tumor ablation, transplantation and transarterial chemoembolization (TACE). In advanced HCC, useful systemic therapies were not available for patients until 2007, when sorafenib was approved. After years without major therapeutic improvements and with increasing incidence, finally advances are arriving for hepatocellular carcinoma (HCC) treatment. Novel drugs and combinations are already working in clinical trials, reaching  hospitals to help in HCC management.

  • Hepatocellular carcinoma (HCC)
  • Atezolizumab
  • Sorafenib
  • Therapies
  • multikinase inhiitors

1. First-Line Therapies

1.1. Atezolizumab-Bevacizumab (Atezo-Bev)

Atezolizumab and bevacizumab combination is the first treatment superior to sorafenib demonstrating prolonged overall survival (67.2% vs. 54.6%; hazard ratio [HR] 0.58) and progression-free survival (6.8 months vs. 4.3 months; HR 0.59) [1]. The success of IMbrave 150 clinical trial has changed the paradigm of hepatocellular carcinoma (HCC) treatment, and atezo-bev has become the recommended systemic therapy if no contraindications are present [2].
Atezolizumab (Tecentriq) is a humanized IgG1 monoclonal antibody that targets PD-L1 to prevent its binding with PD-1 and B7-1 receptors, thus reversing T-cell suppression [3]. Bevacizumab (Avastin) is a monoclonal antibody that targets vascular endothelial growth factor (VEGF), inhibiting angiogenesis and tumor growth [4]. Anti-VEGF therapy also enhances anti-PD-1/PD-L1 activity by reducing VEGF-mediated immunosuppression and promoting T-cell infiltration in tumors [5]. Of note, other immune checkpoint inhibitors [6],[7], as well as atezolizumab [8], in monotherapy, did not reach better outcome in HCC patients, highlighting the synergistic efficacy of immunotherapy and anti-angiogenic combination.
Regarding adverse effects, hypertension and increased AST or ALT are grade 3 or 4 adverse events frequently observed after atezo–bev treatment. Moreover, bleeding, a life-threatening risk for cirrhotic patients, is a common reaction to bevacizumab. In this sense, risk of bleeding, comorbidities such as arterial hypertension and cardiovascular disease, and prior autoimmune conditions may become limiting parameters for the indication of atezo-bev. If the patient has contraindications to atezo-bev, alternative therapies should be considered, such as sorafenib or lenvatinib.
Another immune-based therapy that will likely be included in the first line soon is the tremelimumab/durvalumab combination. Although the peer review data have not yet been published, a significant survival benefit over sorafenib has been announced in the HIMALAYA phase III trial. Once the study data are fully available, changes in clinical decision-making are expected in such a setting, although these are now difficult to foresee.

1.2. Sorafenib

Sorafenib (Nexavar) is a small molecule that inhibits the phosphorylation of up to 40 tyrosine kinases, including VEGFR1, 2 and 3, PDGFRβ, KIT, and RET. This tyrosine kinase inhibitor (TKI) also suppresses Raf kinase isoforms, such as wild-type Raf1, B-Raf, and mutant b-raf V600E. Sorafenib displayed anti-proliferative, anti-angiogenic, and pro-apoptotic properties in HCC cell lines [9], anti-tumor activity in tumor xenograft nude mice [10], and anti-metastatic effect in preventing postsurgical recurrence in an orthotopic mouse model [11]. The efficacy of sorafenib possibly lays in its capacity to target both tumor cells and their microenvironment. As an example, it has been described that sorafenib also had an impact on HSCs proliferation by the suppression of α-SMA and PDGF-related pathways, which decreased HCC cell viability [12]. However, a high dose of sorafenib has been described as promoting immunosuppression through the induction of PD-1 expression in infiltrating immune cells in a murine HCC model [13]; whether this could affect patients, particularly those under immunotherapy, is an aspect that deserves to be studied.
Sorafenib was the first compound that demonstrated survival benefit in HCC in a phase 3, double-blind trial versus placebo (SHARP trial). The median overall survival for patients in the sorafenib arm was 10.7 months compared to 7.9 months in the control group (HR 0.69, 95% confidence interval 0.55–0.87, p < 0.001) [14]. In a parallel trial conducted in the Asian-Pacific population, sorafenib showed a similar survival benefit [15]. The most common adverse effects are diarrhea (8–9% patients) and a hand–foot skin reaction (8–16% patients). Sorafenib has been recommended as the standard systemic therapy for HCC in the first line setting in patients with well-preserved liver function (Child–Pugh A or early B class), with advanced tumors, BCLC-C, or tumors that progressed after loco-regional therapies [16]. The appearance of dermatologic reactions has been linked to better survival following sorafenib administration [17].
Among the molecular mechanisms responsible for sorafenib effectivity in HCC cells is the activation of programmed cell death, apoptosis, provoked by the downregulation of myeloid cell leukemia sequence 1 (MCL-1) expression, an anti-apoptotic member of the BCL-2 family [18]. Recent data have shown that the mitochondrial link with sorafenib activity is more profound. Sorafenib induces mitochondrial reactive oxygen species (ROS), depletes mitochondrial membrane potential, and induces changes in the BCL-2/MCL-1 ratio [19],[20]. In fact, continuous sorafenib exposure altered the levels of anti-apoptotic BCL-2 proteins allowing HCC cell death escape. In contrast, surviving cells are sensitized against BH3-mimetics, inhibitors of specific BCL-2 proteins such as navitoclax [20]. Sorafenib has also been described as involved in the autophagy pathway. The administration of autophagy inhibitors, such as chloroquine or pemetrexed, improved sorafenib efficacy in tumor cells and nude mice hepatoma tumors [21]. Additionally, MCL-1 downregulation was found to disrupt the MCL-1:Beclin 1 complex and induce autophagic cell death in HCC cell lines [22]. In fact, as a consequence of the mitochondrial damage induced by sorafenib, mitophagy is also activated by a triggering mechanism that involves mitochondrial ROS production [23], allowing sorafenib activity to be modulated by antioxidant administration [24]. Acquired drug resistance, which reduces sorafenib effectiveness in patients, may depend on these or other mechanisms. HCC is highly heterogeneous, within the tumor and among individuals, and this influences disease progression, classification, prognosis, and, naturally, cellular susceptibility to drug resistance. In this sense, long-term exposure to sorafenib of hepatoma cells provoked the acquisition of chemoresistance, as well as EMT features [25],[26]. Hypoxia has been described to be involved in sorafenib resistance due to HIF-1α and NF-κB activation [27]. Moreover, M2 macrophages have been found to participate in sorafenib resistance by the release of HGF [28].

1.3. Lenvatinib

Lenvatinib (Lenvima) is an inhibitor of VEGFRs, RET, KIT, PDGFRα, and FGFR1-FGFR4 [29]. It also displayed anti-angiogenic properties and anti-FGFRs activity in hepatoma cells and xenografts [30],[31]. Lenvatinib has been described to exert an immunomodulatory effect through the increase of CD8+ T cell population while diminishing macrophages and monocytes populations in HCC cells [32].
In a phase 3 clinical trial, lenvatinib showed to be non-inferior to sorafenib in terms of overall survival. Hypertension, diarrhea, or a decrease in appetite or weight were among the most common adverse events [33]. In a small group of patients, the levels of AFP were found to decrease in the next two weeks following treatment, suggesting that AFP levels could be predictive of patients’ response [34]. Furthermore, circulating FGF-19 and Ang-2 have been proposed as predictors of clinical response to lenvatinib in HCC patients [35],[36], as well as an early tumor shrinkage [37]. However, like sorafenib, HCC has been described as displaying resistance against lenvatinib. The HGF/c-MET signaling activation was identified as one mechanism of lenvatinib tolerance [38].

2. Second-Line Therapies

2.1. Regorafenib

Regorafenib (Stivarga) is a multikinase inhibitor (MKI) against VEGFR-2, VEGFR-3, KIT, RET, wild-type, and mutant (V600E) B-Raf, PDGFR, FGFR1, angiopoietin 1 receptor (TIE2), RET, and p-38-alpha. Its inhibitory profile is slightly different from sorafenib, since regorafenib has stronger potency targeting VEGFR and TIE2, KIT, and RET [39]. Like sorafenib, regorafenib inhibits angiogenesis, oncogenesis, and tumor microenvironment. Regorafenib was shown to block cell growth and invasion in hepatoma cell lines [40]. This MKI also targeted MAPK pathway, induced caspase cleavage and activated the autophagic pathway [41],[42], and mitophagy as a consequence of its mitochondrial activity [24]. In fact, regorafenib alteration of mitochondrial proteins such as BCL-xL is related to regorafenib resistance, pointing to BH3 mimetics for combined therapies [43]. Moreover, both intrinsic and extrinsic apoptotic pathways were activated by regorafenib [44]. The treatment with regorafenib provoked a decrease in the expression of metastasis-related proteins in HCC cells[45]. Regorafenib was demonstrated to block EMT activation and overcome the acquired resistance to sorafenib [46].
The RESORCE trial was the first phase 3 clinical trial that showed that patients who progressed on sorafenib benefited from oral regorafenib administration versus placebo in a second line setting [47]. Median survival was 10.6 months for the regorafenib arm, while 7.8 months for the control group (HR 0.63; 95% 0.50–0.79; p < 0.0001). Manageable adverse events consisted of a hand–foot skin reaction, hypertension, and fatigue. Additional analyses of the RESORCE trial have suggested that the administration of regorafenib following sorafenib may extend survival [48].

2.2. Cabozantinib

Cabozantinib (Cometriq, Cabometyx) is a small molecule with tyrosine kinase inhibitory prolife against VEGFR-2, RET, KIT, FLT-3, TIE2, and AXL. Cabozantinib differs from sorafenib and regorafenib in that it is capable to also block c-Met [49]. Cabozantinib has demonstrated anti-tumor activity in HCC cells by inhibiting tumor growth, angiogenesis, invasion, and migration. It also reduced the number of HCC metastatic nodules in the lungs and liver in mice [50]. In a phase two clinical trial, cabozantinib demonstrated effectivity in HCC patients[51]. Those promising results led to the conduction of a phase 3 clinical trial in patients who progressed after sorafenib treatment. Cabozantinib increased overall survival (10.2 months) compared to placebo (8.0 months, HR 0.76; 95% CI, 0.63–0.92; p = 0.005). The most frequent side effects were palmar-plantar erythrodysesthesia, hypertension, increase AST, fatigue, and diarrhea [52].

2.3. Nivolumab

Nivolumab (Opdivo) is a human monoclonal antibody that targets programmed cell death protein 1 (PD-1). It is an immune checkpoint inhibitor, since nivolumab impedes the signaling that blocks T cell anti-tumor activity [53]. A phase 1/2 dose escalation study performed with advanced HCC with or without previous sorafenib treatment showed the potential of nivolumab for the treatment of HCC (CheckMate 040 trial) [54]. A further analysis of the CheckMate 040 trial highlighted that some inflammatory biomarkers trended with improved survival and an anti-tumor immune response [55]. Nevertheless, a subset of patients with hyperprogressive disease (HPD) was identified after nivolumab treatment in HCC patients [56]. Furthermore, administration of nivolumab plus ipilimumab, which targets CTLA-4, a inhibitory T-cell receptor, also showed to be a promising therapeutic strategy in HCC patients who progressed on sorafenib [57].

2.4. Pembrolizumab

The humanized monoclonal antibody pembrolizumab (Keytruda) blocks PD-1 as well. In a non-randomized phase 2 clinical trial, pembrolizumab was effective in patients who were treated previously with sorafenib (KEYNOTE-224) [58]. These results led to testing pembrolizumab compared to placebo in a phase 3 randomized clinical trial. Although median overall survival was longer for the pembrolizumab arm, 13.9 months (95% CI, 11.6 to 16.0 months) and 10.6 months (95% CI, 8.3 to 13.5 months) for placebo, the results were not statistically significant [6].

2.5. Ramucirumab

Regarding antiangiogenic therapies, ramucirumab (Cyramza), a monoclonal antibody against VEGFR2 [59],[60],[61], failed to improve survival in the REACH trial in patients treated previously with sorafenib. However, the authors identified AFP serum levels as a prognostic marker showing that patients with high levels of AFP (≥400 ng/mL) benefit from ramucirumab treatment. These observations were validated in REACH-2, a double-blind phase III trial, wherein only patients treated with sorafenib with high AFP levels were included [60]. Ramucirumab improved overall survival (8.5 versus 7.3 months HR 0.710, 95% CI 0.531–0.949; p = 0.0199) and has become the first HCC therapy with biomarker-guided patient selection. Hypertension, liver failure, and hyponatremia were the most common grade 3–4 adverse events.

2.6. Combination Therapies

Regarding ongoing clinical studies, several combinations of treatment regimens are being tested in patients with HCC in both the first line and second line: the RENOBATE study (combination of regorafenib and nivolumab administered as first-line therapy in unresectable HCC), the REGOMUNE trial (avelumab, which targets PD-L1, will be studied together with regorafenib), the GOING trial (second-line treatment with regorafenib, followed by nivolumab treatment in patients who have progressed on sorafenib administration), the ACTION trial (will evaluate the effectivity of cabozantinib in patients who are sorafenib-intolerant or who do not meet the RESORCE criteria), and the COSMIC-312 clinical trial (administration of cabozantinib in combination with the immune checkpoint inhibitor atezolizumab), among some others. So, new therapies are coming and updates on  novel strategies and treatment recommendations [62] will be needed soon.
This entry is adapted from 10.3390/cancers14030621

References

  1. Richard S. Finn; Shukui Qin; Masafumi Ikeda; Peter R. Galle; Michel Ducreux; Tae-You Kim; Masatoshi Kudo; Valeriy Breder; Philippe Merle; Ahmed O. Kaseb; et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. New England Journal of Medicine 2020, 382, 1894-1905, 10.1056/nejmoa1915745.
  2. Jordi Bruix; Stephen L. Chan; Peter R. Galle; Lorenza Rimassa; Bruno Sangro; Systemic treatment of hepatocellular carcinoma: An EASL position paper. Journal of Hepatology 2021, 75, 960-974, 10.1056/NEJMoa1915745.
  3. Roy S. Herbst; Jean-Charles Soria; Marcin Kowanetz; Gregg D. Fine; Omid Hamid; Michael S. Gordon; Jeffery A. Sosman; David F. McDermott; John D. Powderly; Scott N. Gettinger; et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014, 515, 563-567, 10.1038/nature14011.
  4. Michael A. Morse; Weijing Sun; Richard Kim; Aiwu Ruth He; Paolo B. Abada; Michelle Mynderse; Richard S. Finn; The Role of Angiogenesis in Hepatocellular Carcinoma. Clinical Cancer Research 2018, 25, 912-920, 10.1158/1078-0432.ccr-18-1254.
  5. Jeffrey J. Wallin; Johanna C. Bendell; Roel Funke; Mario Sznol; Konstanty Korski; Suzanne Jones; Genevive Hernandez; James Mier; Xian He; F. Stephen Hodi; et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nature Communications 2016, 7, 12624, 10.1038/ncomms12624.
  6. Richard S. Finn; Baek-Yeol Ryoo; Philippe Merle; Masatoshi Kudo; Mohamed Bouattour; Ho Yeong Lim; Valeriy Breder; Julien Edeline; Yee Chao; Sadahisa Ogasawara; et al. Pembrolizumab As Second-Line Therapy in Patients With Advanced Hepatocellular Carcinoma in KEYNOTE-240: A Randomized, Double-Blind, Phase III Trial. Journal of Clinical Oncology 2020, 38, 193-202, 10.1200/jco.19.01307.
  7. T. Yau; J.W. Park; R.S. Finn; A.-L. Cheng; P. Mathurin; J. Edeline; M. Kudo; K.-H. Han; J.J. Harding; P. Merle; et al. CheckMate 459: A randomized, multi-center phase III study of nivolumab (NIVO) vs sorafenib (SOR) as first-line (1L) treatment in patients (pts) with advanced hepatocellular carcinoma (aHCC). Annals of Oncology 2019, 30, v874-v875, 10.1093/annonc/mdz394.029.
  8. Michael S Lee; Baek-Yeol Ryoo; Chih-Hung Hsu; Kazushi Numata; Stacey Stein; Wendy Verret; Stephen P Hack; Jessica Spahn; Bo Liu; Heba Abdullah; et al. Atezolizumab with or without bevacizumab in unresectable hepatocellular carcinoma (GO30140): an open-label, multicentre, phase 1b study. The Lancet Oncology 2020, 21, 808-820, 10.1016/s1470-2045(20)30156-x.
  9. Li Liu; Yichen Cao; Charles Chen; Xiaomei Zhang; Angela McNabola; Dean Wilkie; Scott Wilhelm; Mark Lynch; Christopher Carter; Sorafenib Blocks the RAF/MEK/ERK Pathway, Inhibits Tumor Angiogenesis, and Induces Tumor Cell Apoptosis in Hepatocellular Carcinoma Model PLC/PRF/5. Cancer Research 2006, 66, 11851-11858, 10.1158/0008-5472.can-06-1377.
  10. Scott M. Wilhelm; Christopher Carter; Liya Tang; Dean Wilkie; Angela McNabola; Hong Rong; Charles Chen; Xiaomei Zhang; Patrick Vincent; Mark McHugh; et al. BAY 43-9006 Exhibits Broad Spectrum Oral Antitumor Activity and Targets the RAF/MEK/ERK Pathway and Receptor Tyrosine Kinases Involved in Tumor Progression and Angiogenesis. Cancer Research 2004, 64, 7099-7109, 10.1158/0008-5472.can-04-1443.
  11. Yu-Xiong Feng; Tao Wang; Yue-Zhen Deng; Pengyuan Yang; Jing-Jing Li; Dong-Xian Guan; Fan Yao; Yin-Qiu Zhu; Ying Qin; Hui Wang; et al. Sorafenib suppresses postsurgical recurrence and metastasis of hepatocellular carcinoma in an orthotopic mouse model. Hepatology 2010, 53, 483-492, 10.1002/hep.24075.
  12. Zhi-Min Geng; Rajiv Kumar Jha; Bo Li; Chen Chen; Wen-Zhi Li; Jian-Bao Zheng; Lin Wang; Sha Huanchen; Sorafenib Inhibition of Hepatic Stellate Cell Proliferation in Tumor Microenvironment of Hepatocellular Carcinoma: A Study of the Sorafenib Mechanisms. Cell Biochemistry and Biophysics 2014, 69, 717-724, 10.1007/s12013-014-9858-y.
  13. Renuka V. Iyer; Orla Maguire; Minhyung Kim; Leslie I. Curtin; Sandra Sexton; Daniel T. Fisher; Sarah A. Schihl; Gerald Fetterly; Stephan Menne; Hans Minderman; et al. Dose-Dependent Sorafenib-Induced Immunosuppression Is Associated with Aberrant NFAT Activation and Expression of PD-1 in T Cells.. Cancers 2019, 11, 681, 10.3390/cancers11050681.
  14. Josep M. Llovet; Sergio Ricci; Vincenzo Maria Mazzaferro; Philip Hilgard; Edward Gane; Jean-Frédéric Blanc; Andre Cosme De Oliveira; Armando Santoro; Jean-Luc Raoul; Alejandro Forner; et al. Sorafenib in Advanced Hepatocellular Carcinoma. New England Journal of Medicine 2008, 359, 378-390, 10.1056/nejmoa0708857.
  15. Ann-Lii Cheng; Yoon-Koo Kang; Zhendong Chen; Chao-Jung Tsao; Shukui Qin; Jun Suk Kim; Rongcheng Luo; Jifeng Feng; Shenglong Ye; Tsai-Sheng Yang; et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. The Lancet Oncology 2008, 10, 25-34, 10.1016/s1470-2045(08)70285-7.
  16. Peter R. Galle; Alejandro Forner; Josep M Llovet; Vincenzo Maria Mazzaferro; Fabio Piscaglia; Jean-Luc Raoul; Peter Schirmacher; Valérie Vilgrain; EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. Journal of Hepatology 2018, 69, 182-236, 10.1016/j.jhep.2018.03.019.
  17. Maria Reig; Ferran Torres; Carlos Rodriguez-Lope; Alejandro Forner; Neus Llarch; Jordi Rimola; Anna Darnell; José Ríos; Carmen Ayuso; Jordi Bruix; et al. Early dermatologic adverse events predict better outcome in HCC patients treated with sorafenib. Journal of Hepatology 2014, 61, 318-324, 10.1016/j.jhep.2014.03.030.
  18. Mohamed Rahmani; Eric Maynard Davis; Cheryl Bauer; Paul Dent; Steven Grant; Apoptosis Induced by the Kinase Inhibitor BAY 43-9006 in Human Leukemia Cells Involves Down-regulation of Mcl-1 through Inhibition of Translation. Journal of Biological Chemistry 2005, 280, 35217-35227, 10.1074/jbc.m506551200.
  19. Milica Stefanovic; Anna Tutusaus; Guillermo A. Martinez-Nieto; Cristina Bárcena; Estefania De Gregorio; Catia Moutinho; Elisabet Barbero-Camps; Alberto Villanueva; Anna Colell; Montserrat Mari; et al. Targeting glucosylceramide synthase upregulation reverts sorafenib resistance in experimental hepatocellular carcinoma. Oncotarget 2016, 7, 8253-8267, 10.18632/oncotarget.6982.
  20. Anna Tutusaus; Milica Stefanovic; Loreto Boix; Blanca Cucarull; Aynara Zamora; Laura Blasco; Pablo García de Frutos; Maria Reig; Jose C. Fernandez-Checa; Montserrat Mari; et al. Antiapoptotic BCL-2 proteins determine sorafenib/regorafenib resistance and BH3-mimetic efficacy in hepatocellular carcinoma. Oncotarget 2018, 9, 16701-16717, 10.18632/oncotarget.24673.
  21. Satoshi Shimizu; Tetsuo Takehara; Hayato Hikita; Takahiro Kodama; Hinako Tsunematsu; Takuya Miyagi; Atsushi Hosui; Hisashi Ishida; Tomohide Tatsumi; Tatsuya Kanto; et al. Inhibition of autophagy potentiates the antitumor effect of the multikinase inhibitor sorafenib in hepatocellular carcinoma. International Journal of Cancer 2011, 131, 548-557, 10.1002/ijc.26374.
  22. W-T Tai; C-W Shiau; H-L Chen; C-Y Liu; Chen-Si Lin; Ann-Lii Cheng; Pei-Jer Chen; K-F Chen; Mcl-1-dependent activation of Beclin 1 mediates autophagic cell death induced by sorafenib and SC-59 in hepatocellular carcinoma cells. Cell Death & Disease 2013, 4, e485-e485, 10.1038/cddis.2013.18.
  23. Néstor Prieto-Domínguez; Raquel Ordóñez; Anna Fernández; Carolina Méndez Blanco; Anna Baulies; Carmen Garcia-Ruiz; José Carlos Fernández-Checa; Jose L Mauriz; Javier González-Gallego; Melatonin-induced increase in sensitivity of human hepatocellular carcinoma cells to sorafenib is associated with reactive oxygen species production and mitophagy. Journal of Pineal Research 2016, 61, 396-407, 10.1111/jpi.12358.
  24. Blanca Cucarull; Anna Tutusaus; Tania Hernáez-Alsina; Pablo García de Frutos; María Reig; Anna Colell; Montserrat Marí; Albert Morales; Antioxidants Threaten Multikinase Inhibitor Efficacy against Liver Cancer by Blocking Mitochondrial Reactive Oxygen Species. Antioxidants 2021, 10, 1336, 10.3390/antiox10091336.
  25. Hannah van Malenstein; Jeroen Dekervel; Chris Verslype; Eric Van Cutsem; Petra Windmolders; Frederik Nevens; Jos van Pelt; Long-term exposure to sorafenib of liver cancer cells induces resistance with epithelial-to-mesenchymal transition, increased invasion and risk of rebound growth. Cancer Letters 2013, 329, 74-83, 10.1016/j.canlet.2012.10.021.
  26. Ariel Ka-Man Chow; Lui Ng; Colin Siu-Chi Lam; Sunny Kit-Man Wong; Timothy Ming-Hun Wan; Nathan Shiu-Man Cheng; Thomas Chung Cheung Yau; Ronnie Tung-Ping Poon; Roberta Wen-Chi Pang; The Enhanced Metastatic Potential of Hepatocellular Carcinoma (HCC) Cells with Sorafenib Resistance. PLOS ONE 2013, 8, e78675, 10.1371/journal.pone.0078675.
  27. Yingjian Liang; Tongsen Zheng; Ruipeng Song; Jiabei Wang; Dalong Yin; Luoluo Wang; Haitao Liu; Lantian Tian; Xiang Fang; Xianzhi Meng; et al. Hypoxia-mediated sorafenib resistance can be overcome by EF24 through Von Hippel-Lindau tumor suppressor-dependent HIF-1α inhibition in hepatocellular carcinoma. Hepatology 2013, 57, 1847-1857, 10.1002/hep.26224.
  28. Ningning Dong; Xiangyi Shi; Suihai Wang; Yanjun Gao; Zhenzhan Kuang; Qian Xie; Yonglong Li; Huan Deng; Yingsong Wu; Ming Li; et al. M2 macrophages mediate sorafenib resistance by secreting HGF in a feed-forward manner in hepatocellular carcinoma. British Journal of Cancer 2019, 121, 22-33, 10.1038/s41416-019-0482-x.
  29. Junji Matsui; Yasuhiro Funahashi; Toshimitsu Uenaka; Tatsuo Watanabe; Akihiko Tsuruoka; Makoto Asada; Multi-Kinase Inhibitor E7080 Suppresses Lymph Node and Lung Metastases of Human Mammary Breast Tumor MDA-MB-231 via Inhibition of Vascular Endothelial Growth Factor-Receptor (VEGF-R) 2 and VEGF-R3 Kinase. Clinical Cancer Research 2008, 14, 5459-5465, 10.1158/1078-0432.ccr-07-5270.
  30. Yusuke Adachi; Masahiro Matsuki; Hideki Watanabe; Kazuma Takase; Kotaro Kodama; Junji Matsui; Yasuhiro Funahashi; Kenichi Nomoto; Antitumor and Antiangiogenic Activities of Lenvatinib in Mouse Xenograft Models of Vascular Endothelial Growth Factor-Induced Hypervascular Human Hepatocellular Carcinoma.. Cancer Investigation 2019, 37, 185-198, 10.1080/07357907.2019.1601209.
  31. Taisuke Hoshi; Saori Watanabe Miyano; Hideki Watanabe; Regina Mikie Kanada Sonobe; Yuki Seki; Etsuko Ohta; Kenichi Nomoto; Junji Matsui; Yasuhiro Funahashi; Lenvatinib induces death of human hepatocellular carcinoma cells harboring an activated FGF signaling pathway through inhibition of FGFR–MAPK cascades. Biochemical and Biophysical Research Communications 2019, 513, 1-7, 10.1016/j.bbrc.2019.02.015.
  32. Takayuki Kimura; Yu Kato; Yoichi Ozawa; Kotaro Kodama; Junichi Ito; Kenji Ichikawa; Kazuhiko Yamada; Yusaku Hori; Kimiyo Tabata; Kazuma Takase; et al. Immunomodulatory activity of lenvatinib contributes to antitumor activity in the Hepa1‐6 hepatocellular carcinoma model. Cancer Science 2018, 109, 3993-4002, 10.1111/cas.13806.
  33. Masatoshi Kudo; Richard S Finn; Shukui Qin; Kwang-Hyub Han; Kenji Ikeda; Fabio Piscaglia; Ari Baron; Joong-Won Park; Guohong Han; Jacek Jassem; et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. The Lancet 2018, 391, 1163-1173, 10.1016/s0140-6736(18)30207-1.
  34. Kenichiro Kodama; Tomokazu Kawaoka; Maiko Namba; Shinsuke Uchikawa; Kazuki Ohya; Kei Morio; Takashi Nakahara; Eisuke Murakami; Masami Yamauchi; Akira Hiramatsu; et al. Correlation between Early Tumor Marker Response and Imaging Response in Patients with Advanced Hepatocellular Carcinoma Treated with Lenvatinib. Oncology 2019, 97, 75-81, 10.1159/000499715.
  35. Makoto Chuma; Haruki Uojima; Kazushi Numata; Hisashi Hidaka; Hidenori Toyoda; Atsushi Hiraoka; Toshifumi Tada; Shunji Hirose; Masanori Atsukawa; Norio Itokawa; et al. Early Changes in Circulating FGF19 and Ang-2 Levels as Possible Predictive Biomarkers of Clinical Response to Lenvatinib Therapy in Hepatocellular Carcinoma. Cancers 2020, 12, 293, 10.3390/cancers12020293.
  36. Issei Saeki; Takahiro Yamasaki; Satoyoshi Yamashita; Tadasuke Hanazono; Yohei Urata; Takakazu Furutani; Yuichiro Yokoyama; Toshiyuki Oishi; Masaki Maeda; Teruaki Kimura; et al. Early Predictors of Objective Response in Patients with Hepatocellular Carcinoma Undergoing Lenvatinib Treatment. Cancers 2020, 12, 779, 10.3390/cancers12040779.
  37. Aya Takahashi; Michihisa Moriguchi; Yuya Seko; Toshihide Shima; Yasuhide Mitsumoto; Hidetaka Takashima; Hiroyuki Kimura; Hideki Fujii; Hiroki Ishikawa; Takaharu Yo; et al. Early Tumor Shrinkage as a Predictive Factor for Outcomes in Hepatocellular Carcinoma Patients Treated with Lenvatinib: A Multicenter Analysis. Cancers 2020, 12, 754, 10.3390/cancers12030754.
  38. RongDang Fu; Shaotao Jiang; Jieyuan Li; Huanwei Chen; Xiaohong Zhang; Activation of the HGF/c-MET axis promotes lenvatinib resistance in hepatocellular carcinoma cells with high c-MET expression. Medical Oncology 2020, 37, 24-7, 10.1007/s12032-020-01350-4.
  39. Dirk Strumberg; Beate Schultheis; Regorafenib for cancer. Expert Opinion on Investigational Drugs 2012, 21, 879-889, 10.1517/13543784.2012.684752.
  40. Brian I. Carr; Rosalba D'Alessandro; Maria G. Refolo; Palma A. Iacovazzi; Catia Lippolis; Caterina Messa; Aldo Cavallini; Mario Correale; Antonio Di Carlo; Effects of low concentrations of Regorafenib and Sorafenib on human HCC cell AFP, migration, invasion, and growth in vitro. Journal of Cellular Physiology 2012, 228, 1344-1350, 10.1002/jcp.24291.
  41. Brian I. Carr; Aldo Cavallini; Catia Lippolis; Rosalba D'Alessandro; Caterina Messa; Maria G. Refolo; Angela Tafaro; Fluoro-Sorafenib (Regorafenib) effects on hepatoma cells: Growth inhibition, quiescence, and recovery. Journal of Cellular Physiology 2012, 228, 292-297, 10.1002/jcp.24148.
  42. Rui Han Rui Han; Shixin Li Shixin Li; Regorafenib delays the proliferation of hepatocellular carcinoma by inducing autophagy. Pharmazie 2018, 73, 218-222, 10.1691/PH.2018.7988.
  43. Blanca Cucarull; Anna Tutusaus; Miguel Subías; Milica Stefanovic; Tania Hernáez-Alsina; Loreto Boix; María Reig; Pablo García De Frutos; Montserrat Mari; Anna Colell; et al. Regorafenib Alteration of the BCL-xL/MCL-1 Ratio Provides a Therapeutic Opportunity for BH3-Mimetics in Hepatocellular Carcinoma Models. Cancers 2020, 12, 332, 10.3390/cancers12020332.
  44. Jai-Jen Tsai; Po-Jung Pan; Fei-Ting Hsu; Regorafenib induces extrinsic and intrinsic apoptosis through inhibition of ERK/NF-?B activation in hepatocellular carcinoma cells. Oncology Reports 2016, 37, 1036-1044, 10.3892/or.2016.5328.
  45. Yu-Chang Liu; Reng-Hong Wu; Wei-Shu Wang; Regorafenib diminishes the expression and secretion of angiogenesis and metastasis associated proteins and inhibits cell invasion via NF-κB inactivation in SK-Hep1 cells. Oncology Letters 2017, 14, 461-467, 10.3892/ol.2017.6142.
  46. Weibo Chen; Junsheng Yang; Yue Zhang; Huihua Cai; Xuemin Chen; Donglin Sun; Regorafenib reverses HGF ‐induced sorafenib resistance by inhibiting epithelial‐mesenchymal transition in hepatocellular carcinoma. FEBS Open Bio 2018, 9, 335-347, 10.1002/2211-5463.12578.
  47. Jordi Bruix; Shukui Qin; Philippe Merle; Alessandro Granito; Yi-Hsiang Huang; György Bodoky; Marc Pracht; Osamu Yokosuka; Olivier Rosmorduc; Valeriy Breder; et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet 2016, 389, 56-66, 10.1016/s0140-6736(16)32453-9.
  48. Richard S. Finn; Philippe Merle; Alessandro Granito; Yi-Hsiang Huang; György Bodoky; Marc Pracht; Osamu Yokosuka; Olivier Rosmorduc; René Gerolami; Chiara Caparello; et al. Outcomes of sequential treatment with sorafenib followed by regorafenib for HCC: Additional analyses from the phase III RESORCE trial. Journal of Hepatology 2018, 69, 353-358, 10.1016/j.jhep.2018.04.010.
  49. F. Michael Yakes; Jason Chen; Jenny Tan; Kyoko Yamaguchi; Yongchang Shi; Peiwen Yu; Fawn Qian; Felix Chu; Frauke Bentzien; Belinda Cancilla; et al. Cabozantinib (XL184), a Novel MET and VEGFR2 Inhibitor, Simultaneously Suppresses Metastasis, Angiogenesis, and Tumor Growth. Molecular Cancer Therapeutics 2011, 10, 2298-2308, 10.1158/1535-7163.mct-11-0264.
  50. Qingfeng Xiang; Weiqiang Chen; Meng Ren; Jingnan Wang; Hongwu Zhang; David Y.B. Deng; Lei Zhang; Changzhen Shang; Yajin Chen; Cabozantinib Suppresses Tumor Growth and Metastasis in Hepatocellular Carcinoma by a Dual Blockade of VEGFR2 and MET. Clinical Cancer Research 2014, 20, 2959-2970, 10.1158/1078-0432.ccr-13-2620.
  51. R. K. Kelley; C. Verslype; A. L. Cohn; T.-S. Yang; Wu-Chou Su; H. Burris; F. Braiteh; N. Vogelzang; A. Spira; P. Foster; et al. Cabozantinib in hepatocellular carcinoma: results of a phase 2 placebo-controlled randomized discontinuation study. Annals of Oncology 2017, 28, 528-534, 10.1093/annonc/mdw651.
  52. Ghassan K. Abou-Alfa; Tim Meyer; Ann-Lii Cheng; Anthony B. El-Khoueiry; Lorenza Rimassa; Baek-Yeol Ryoo; Irfan Cicin; Philippe Merle; YenHsun Chen; Joong-Won Park; et al. Cabozantinib in Patients with Advanced and Progressing Hepatocellular Carcinoma. New England Journal of Medicine 2018, 379, 54-63, 10.1056/nejmoa1717002.
  53. Julie R. Brahmer; Charles G. Drake; Ira Wollner; John D. Powderly; Joel Picus; William H. Sharfman; Elizabeth Stankevich; Alice Pons; Theresa M. Salay; Tracee L. McMiller; et al. Phase I Study of Single-Agent Anti–Programmed Death-1 (MDX-1106) in Refractory Solid Tumors: Safety, Clinical Activity, Pharmacodynamics, and Immunologic Correlates. Journal of Clinical Oncology 2010, 28, 3167-3175, 10.1200/jco.2009.26.7609.
  54. Anthony B El-Khoueiry; Bruno Sangro; Thomas Yau; Todd S Crocenzi; Masatoshi Kudo; Chiun Hsu; Tae-You Kim; Su-Pin Choo; Jörg Trojan; Theodore H Welling; et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. The Lancet 2017, 389, 2492-2502, 10.1016/s0140-6736(17)31046-2.
  55. Bruno Sangro; Ignacio Melero; Samir Wadhawan; Richard S. Finn; Ghassan K. Abou-Alfa; Ann-Lii Cheng; Thomas Yau; Junji Furuse; Joong-Won Park; Zachary Boyd; et al. Association of inflammatory biomarkers with clinical outcomes in nivolumab-treated patients with advanced hepatocellular carcinoma. Journal of Hepatology 2020, 73, 1460-1469, 10.1016/j.jhep.2020.07.026.
  56. Chang Gon Kim; Chan Kim; Sang Eun Yoon; Kyung Hwan Kim; Seong Jin Choi; Beodeul Kang; Hye Ryun Kim; Su-Hyung Park; Eui-Cheol Shin; Yeun-Yoon Kim; et al. Hyperprogressive disease during PD-1 blockade in patients with advanced hepatocellular carcinoma. Journal of Hepatology 2020, 74, 350-359, 10.1016/j.jhep.2020.08.010.
  57. Thomas Yau; Yoon-Koo Kang; Tae-You Kim; Anthony B. El-Khoueiry; Armando Santoro; Bruno Sangro; Ignacio Melero; Masatoshi Kudo; Ming-Mo Hou; Ana Matilla; et al. Efficacy and Safety of Nivolumab Plus Ipilimumab in Patients With Advanced Hepatocellular Carcinoma Previously Treated With Sorafenib. JAMA Oncology 2020, 6, e204564, 10.1001/jamaoncol.2020.4564.
  58. Andrew X Zhu; Richard S Finn; Julien Edeline; Stephane Cattan; Sadahisa Ogasawara; Daniel Palmer; Chris Verslype; Vittorina Zagonel; Laetitia Fartoux; Arndt Vogel; et al. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): a non-randomised, open-label phase 2 trial. The Lancet Oncology 2018, 19, 940-952, 10.1016/s1470-2045(18)30351-6.
  59. Masatoshi Kudo; Etsuro Hatano; Shinichi Ohkawa; Hirofumi Fujii; Akihide Masumoto; Junji Furuse; Yoshiyuki Wada; Hiroshi Ishii; Shuntaro Obi; Shuichi Kaneko; et al. Ramucirumab as second-line treatment in patients with advanced hepatocellular carcinoma: Japanese subgroup analysis of the REACH trial. Journal of Gastroenterology 2016, 52, 494-503, 10.1007/s00535-016-1247-4.
  60. Andrew X Zhu; Yoon-Koo Kang; Chia-Jui Yen; Richard S Finn; Peter R Galle; Josep M Llovet; Eric Assenat; Giovanni Brandi; Marc Pracht; Ho Yeong Lim; et al. Ramucirumab after sorafenib in patients with advanced hepatocellular carcinoma and increased α-fetoprotein concentrations (REACH-2): a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet Oncology 2019, 20, 282-296, 10.1016/s1470-2045(18)30937-9.
  61. Masatoshi Kudo; Takuji Okusaka; Kenta Motomura; Izumi Ohno; Manabu Morimoto; Satoru Seo; Yoshiyuki Wada; Shinpei Sato; Tatsuya Yamashita; Masayuki Furukawa; et al. Ramucirumab after prior sorafenib in patients with advanced hepatocellular carcinoma and elevated alpha-fetoprotein: Japanese subgroup analysis of the REACH-2 trial. Journal of Gastroenterology 2020, 55, 627-639, 10.1007/s00535-020-01668-w.
  62. Maria Reig; Alejandro Forner; Jordi Rimola; Joana Ferrer-Fàbrega; Marta Burrel; Ángeles Garcia-Criado; Robin K. Kelley; Peter R. Galle; Vincenzo Mazzaferro; Riad Salem; et al. BCLC strategy for prognosis prediction and treatment recommendation: The 2022 update. Journal of Hepatology 2021, 76, 681-693, 10.1016/j.jhep.2021.11.018.
More
This entry is offline, you can click here to edit this entry!
Video Production Service