Immunotherapy and the Combination with Targeted Therapies for Advanced Hepatocellular Carcinoma
Abstract
:Simple Summary
Abstract
1. Introduction
2. PD-1/PD-L1 Inhibitors
2.1. Single Agents
2.1.1. Nivolumab
2.1.2. Pembrolizumab
2.1.3. Other PD-1/PD-L1 Inhibitors
2.2. PD-1/PD-L1 Inhibitors plus Antiangiogenetic Therapies
2.2.1. Atezolizumab and Bevacizumab
2.2.2. Sintilimab and Bevacizumab
2.2.3. Pembrolizumab and Lenvatinib
2.2.4. Nivolumab and Lenvatinib
2.2.5. Avelumab and Axitinib
2.2.6. Camrelizumab and Apatinib
2.3. PD-1/PD-L1 Inhibitors Plus Other Immunotherapies
2.3.1. Nivolumab and Ipilimumab
2.3.2. Tremelimumab and Durvalumab
3. Adoptive Cell Transfer
4. Vaccines
5. Notch Signaling Pathway: The Role in HCC Development and Response to Cancer Treatments
6. Conclusions and Future Perspectives
Author Contributions
Funding
Conflicts of Interest
Abbreviations
Adoptive Cell Transfer | ACT |
Adverse Event | AE |
Angiopoietin 2 | ANG2 |
Antigen-Presenting Cell | APC |
Barcelona Clinic HCC | BCLC |
Chimeric Antigen Receptor T Cell | CAR-T Cell |
Cytokine-Induced Killer Cell | CIK |
Cytotoxic T-Lymphocyte | CTL |
Cytotoxic T Lymphocyte-Associated Antigen 4 | CTLA-4 |
Dendritic Cell | DC |
Disease Control Rate | DCR |
ECOG Performance Status | ECOG |
Hepatocellular Carcinoma | HCC |
Immune Checkpoints | ICPs |
Intercellular adhesion molecule 1 | ICAM1 |
Lymphocyte Activation Gene-3 | LAG-3 |
Major Histocompatibility Complex Class 1 | MHC-1 |
Median Time to Progression | mTTP |
Monoclonal Antibody | mAb |
Natural Killer Cell | NK |
Notch pathway-related genes | NPRGs |
Objective Response Rate | ORR |
Overall Survival | OS |
Programmed Cell Death 1 | PD-1 |
Progression-Free Survival | PFS |
Regulatory T Cell | Treg |
Stem/progenitor cells | FLSPCs |
T Cell Receptor | TCR |
T-Cell Immunoglobulin Mucin-3 | TIM-3 |
Transforming Growth Factor-Β | TGF-Β |
Tumor-Associated Antigen | TAA |
Tumor-Infiltrating Lymphocyte | TIL |
Tumor-Associated Macrophage | TAM |
Tumor Microenvironment | TME |
References
- Wei, Q.; Taskén, K. Immunoregulatory signal networks and tumor immune evasion mechanisms: Insights into therapeutic targets and agents in clinical development. Biochem. J. 2022, 479, 2219–2260. [Google Scholar] [CrossRef] [PubMed]
- Jiang, X.; Wang, J.; Deng, X.; Xiong, F.; Ge, J.; Xiang, B.; Wu, X.; Ma, J.; Zhou, M.; Li, X.; et al. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol. Cancer 2019, 18, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.; Cao, X. Immunosuppressive cells in tumor immune escape and metastasis. J. Mol. Med. 2016, 94, 509–522. [Google Scholar] [CrossRef] [PubMed]
- Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y. Cancer immunotherapy: Harnessing the immune system to battle cancer. J. Clin. Investig. 2015, 125, 3335–3337. [Google Scholar] [CrossRef] [Green Version]
- Beatty, G.L.; Gladney, W.L. Immune escape mechanisms as a guide for cancer immunotherapy. Clin. Cancer Res. 2015, 21, 687–692. [Google Scholar] [CrossRef] [Green Version]
- Kong, X. Discovery of New Immune Checkpoints: Family Grows Up. Adv. Exp. Med. Biol. 2020, 1248, 61–82. [Google Scholar] [CrossRef]
- Li, B.; Chan, H.L.; Chen, P. Immune Checkpoint Inhibitors: Basics and Challenges. Curr. Med. Chem. 2019, 26, 3009–3025. [Google Scholar] [CrossRef]
- Zhang, Y.; Zheng, J. Functions of Immune Checkpoint Molecules beyond Immune Evasion. Adv. Exp. Med. Biol. 2020, 1248, 201–226. [Google Scholar] [CrossRef]
- Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am. J. Clin. Oncol. 2016, 39, 98–106. [Google Scholar] [CrossRef]
- Rowshanravan, B.; Halliday, N.; Sansom, D.M. CTLA-4: A moving target in immunotherapy. Blood 2018, 131, 58–67. [Google Scholar] [CrossRef]
- Van Coillie, S.; Wiernicki, B.; Xu, J. Molecular and Cellular Functions of CTLA-4. Adv. Exp. Med. Biol. 2020, 1248, 7–32. [Google Scholar] [CrossRef]
- Han, Y.; Liu, D.; Li, L. PD-1/PD-L1 pathway: Current researches in cancer. Am. J. Cancer Res. 2020, 10, 727–742. [Google Scholar]
- Ai, L.; Xu, A.; Xu, J. Roles of PD-1/PD-L1 Pathway: Signaling, Cancer, and Beyond. Adv. Exp. Med. Biol. 2020, 1248, 33–59. [Google Scholar] [CrossRef]
- Jiang, Y.; Chen, M.; Nie, H.; Yuan, Y. PD-1 and PD-L1 in cancer immunotherapy: Clinical implications and future considerations. Hum. Vaccines Immunother. 2019, 15, 1111–1122. [Google Scholar] [CrossRef]
- Doherty, D.G. Immunity, tolerance and autoimmunity in the liver: A comprehensive review. J. Autoimmun. 2016, 66, 60–75. [Google Scholar] [CrossRef]
- Gnyawali, B.; Pusateri, A.; Nickerson, A.; Jalil, S.; Mumtaz, K. Epidemiologic and socioeconomic factors impacting hepatitis B virus and related hepatocellular carcinoma. World J. Gastroenterol. 2022, 28, 3793–3802. [Google Scholar] [CrossRef]
- Huang, D.Q.; Mathurin, P.; Cortez-Pinto, H.; Loomba, R. Global epidemiology of alcohol-associated cirrhosis and HCC: Trends, projections and risk factors. Nat. Rev. Gastroenterol. Hepatol. 2022, 20, 37–49. [Google Scholar] [CrossRef]
- Stella, L.; Santopaolo, F.; Gasbarrini, A.; Pompili, M.; Ponziani, F.R. Viral hepatitis and hepatocellular carcinoma: From molecular pathways to the role of clinical surveillance and antiviral treatment. World J. Gastroenterol. 2022, 28, 2251–2281. [Google Scholar] [CrossRef]
- Ueno, M.; Takeda, H.; Takai, A.; Seno, H. Risk factors and diagnostic biomarkers for nonalcoholic fatty liver disease-associated hepatocellular carcinoma: Current evidence and future perspectives. World J. Gastroenterol. 2022, 28, 3410–3421. [Google Scholar] [CrossRef]
- Najafi, M.; Farhood, B.; Mortezaee, K. Contribution of regulatory T cells to cancer: A review. J. Cell Physiol. 2019, 234, 7983–7993. [Google Scholar] [CrossRef] [PubMed]
- Valzasina, B.; Piconese, S.; Guiducci, C.; Colombo, M.P. Tumor-induced expansion of regulatory T cells by conversion of CD4+CD25- lymphocytes is thymus and proliferation independent. Cancer Res. 2006, 66, 4488–4495. [Google Scholar] [CrossRef] [Green Version]
- Zong, Z.; Zou, J.; Mao, R.; Ma, C.; Li, N.; Wang, J.; Wang, X.; Zhou, H.; Zhang, L.; Shi, Y. M1 Macrophages Induce PD-L1 Expression in Hepatocellular Carcinoma Cells Through IL-1β Signaling. Front. Immunol. 2019, 10, 1643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cha, J.H.; Chan, L.C.; Li, C.W.; Hsu, J.L.; Hung, M.C. Mechanisms Controlling PD-L1 Expression in Cancer. Mol. Cell 2019, 76, 359–370. [Google Scholar] [CrossRef] [PubMed]
- Xu, M.; Zhao, Z.; Song, J.; Lan, X.; Lu, S.; Chen, M.; Wang, Z.; Chen, W.; Fan, X.; Wu, F.; et al. Interactions between interleukin-6 and myeloid-derived suppressor cells drive the chemoresistant phenotype of hepatocellular cancer. Exp. Cell Res. 2017, 351, 142–149. [Google Scholar] [CrossRef] [PubMed]
- Lu, L.C.; Chang, C.J.; Hsu, C.H. Targeting myeloid-derived suppressor cells in the treatment of hepatocellular carcinoma: Current state and future perspectives. J. Hepatocell. Carcinoma 2019, 6, 71–84. [Google Scholar] [CrossRef] [Green Version]
- Zhou, G.; Sprengers, D.; Boor, P.P.C.; Doukas, M.; Schutz, H.; Mancham, S.; Pedroza-Gonzalez, A.; Polak, W.G.; de Jonge, J.; Gaspersz, M.; et al. Antibodies Against Immune Checkpoint Molecules Restore Functions of Tumor-Infiltrating T Cells in Hepatocellular Carcinomas. Gastroenterology 2017, 153, 1107–1119. [Google Scholar] [CrossRef] [PubMed]
- Huang, M.; He, M.; Guo, Y.; Li, H.; Shen, S.; Xie, Y.; Li, X.; Xiao, H.; Fang, L.; Li, D.; et al. The Influence of Immune Heterogeneity on the Effectiveness of Immune Checkpoint Inhibitors in Multifocal Hepatocellular Carcinomas. Clin. Cancer Res. 2020, 26, 4947–4957. [Google Scholar] [CrossRef]
- Bian, J.; Lin, J.; Long, J.; Yang, X.; Yang, X.; Lu, X.; Sang, X.; Zhao, H. T lymphocytes in hepatocellular carcinoma immune microenvironment: Insights into human immunology and immunotherapy. Am. J. Cancer Res. 2020, 10, 4585–4606. [Google Scholar]
- Tellapuri, S.; Sutphin, P.D.; Beg, M.S.; Singal, A.G.; Kalva, S.P. Staging systems of hepatocellular carcinoma: A review. Indian J. Gastroenterol. 2018, 37, 481–491. [Google Scholar] [CrossRef]
- Reig, M.; Forner, A.; Rimola, J.; Ferrer-Fàbrega, J.; Burrel, M.; Garcia-Criado, Á.; Kelley, R.K.; Galle, P.R.; Mazzaferro, V.; Salem, R.; et al. BCLC strategy for prognosis prediction and treatment recommendation: The 2022 update. J. Hepatol. 2022, 76, 681–693. [Google Scholar] [CrossRef]
- Gadaleta, C.D.; Ranieri, G. Trans-arterial chemoembolization as a therapy for liver tumours: New clinical developments and suggestions for combination with angiogenesis inhibitors. Crit. Rev. Oncol./Hematol. 2011, 80, 40–53. [Google Scholar] [CrossRef]
- Goffredo, V.; Paradiso, A.; Ranieri, G.; Gadaleta, C.D. Yttrium-90 (90Y) in the principal radionuclide therapies: An efficacy correlation between peptide receptor radionuclide therapy, radioimmunotherapy and transarterial radioembolization therapy. Ten years of experience (1999–2009). Crit. Rev. Oncol./Hematol. 2011, 80, 393–410. [Google Scholar] [CrossRef]
- Ranieri, G.; Ammendola, M.; Marech, I.; Laterza, A.; Abbate, I.; Oakley, C.; Vacca, A.; Sacco, R.; Gadaleta, C.D. Vascular endothelial growth factor and tryptase changes after chemoembolization in hepatocarcinoma patients. World J. Gastroenterol. 2015, 21, 6018–6025. [Google Scholar] [CrossRef]
- Ranieri, G.; Laface, C. Loco-Regional and Systemic Chemotherapies for Hepato-Pancreatic Tumors: Integrated Treatments. Cancers 2020, 12, 2737. [Google Scholar] [CrossRef]
- Ranieri, G.; Laface, C.; Fazio, V.; De Ceglia, D.; Macina, F.; Gisone, V.; Porcelli, M.; Vinciarelli, G.; Carella, C.; Molinari, P.; et al. Local treatment with deep percutaneous electrochemotherapy of different tumor lesions: Pain relief and objective response results from an observational study. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 7764–7775. [Google Scholar] [CrossRef]
- Ranieri, G.; Marech, I.; Lorusso, V.; Goffredo, V.; Paradiso, A.; Ribatti, D.; Gadaleta, C.D. Molecular targeting agents associated with transarterial chemoembolization or radiofrequency ablation in hepatocarcinoma treatment. World J. Gastroenterol. 2014, 20, 486–497. [Google Scholar] [CrossRef]
- Kulik, L.; El-Serag, H.B. Epidemiology and Management of Hepatocellular Carcinoma. Gastroenterology 2019, 156, 477–491. [Google Scholar] [CrossRef]
- Yin, X.; Wu, T.; Lan, Y.; Yang, W. Current progress of immune checkpoint inhibitors in the treatment of advanced hepatocellular carcinoma. Biosci. Rep. 2022, 42, BSR20212304. [Google Scholar] [CrossRef]
- El-Khoueiry, A.B.; Sangro, B.; Yau, T.; Crocenzi, T.S.; Kudo, M.; Hsu, C.; Kim, T.-Y.; Choo, S.-P.; Trojan, J.; Welling, T.H., 3rd; et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): An open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017, 389, 2492–2502. [Google Scholar] [CrossRef]
- Yau, T.; Park, J.-W.; Finn, R.S.; Cheng, A.-L.; Mathurin, P.; Edeline, J.; Kudo, M.; Harding, J.J.; Merle, P.; Rosmorduc, O.; et al. Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): A randomised, multicentre, open-label, phase 3 trial. Lancet Oncol. 2022, 23, 77–90. [Google Scholar] [CrossRef] [PubMed]
- Zhu, A.X.; Finn, R.S.; Edeline, J.; Cattan, S.; Ogasawara, S.; Palmer, D.; Verslype, C.; Zagonel, V.; Fartoux, L.; Vogel, A.; et al. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): A non-randomised, open-label phase 2 trial. Lancet Oncol. 2018, 19, 940–952. [Google Scholar] [CrossRef] [PubMed]
- Finn, R.S.; Ryoo, B.Y.; Merle, P.; Kudo, M.; Bouattour, M.; Lim, H.Y.; Breder, V.; Edeline, J.; Chao, Y.; Ogasawara, S.; et al. Pembrolizumab As Second-Line Therapy in Patients with Advanced Hepatocellular Carcinoma in KEYNOTE-240: A Randomized, Double-Blind, Phase III Trial. J. Clin. Oncol. 2020, 38, 193–202. [Google Scholar] [CrossRef] [PubMed]
- Qin, S.; Chen, Z.; Fang, W.; Ren, Z.; Xu, R.; Ryoo, B.-Y.; Meng, Z.; Bai, Y.; Chen, X.; Liu, X.; et al. Pembrolizumab plus best supportive care versus placebo plus best supportive care as second-line therapy in patients in Asia with advanced hepatocellular carcinoma (HCC): Phase 3 KEYNOTE-394 study. J. Clin. Oncol. 2022, 40, 383. [Google Scholar] [CrossRef]
- Qin, S.; Ren, Z.; Meng, Z.; Chen, Z.; Chai, X.; Xiong, J.; Bai, Y.; Yang, L.; Zhu, H.; Fang, W.; et al. Camrelizumab in patients with previously treated advanced hepatocellular carcinoma: A multicentre, open-label, parallel-group, randomised, phase 2 trial. Lancet Oncol. 2020, 21, 571–580. [Google Scholar] [CrossRef]
- Qin, S.; Finn, R.S.; Kudo, M.; Meyer, T.; Vogel, A.; Ducreux, M.; Macarulla, T.M.; Tomasello, G.; Boisserie, F.; Hou, J.; et al. RATIONALE 301 study: Tislelizumab versus sorafenib as first-line treatment for unresectable hepatocellular carcinoma. Future Oncol. 2019, 15, 1811–1822. [Google Scholar] [CrossRef] [Green Version]
- Wainberg, Z.A.; Segal, N.H.; Jaeger, D.; Lee, K.-H.; Marshall, J.; Antonia, S.J.; Butler, M.; Sanborn, R.E.; Nemunaitis, J.J.; Carlson, C.A.; et al. Safety and clinical activity of durvalumab monotherapy in patients with hepatocellular carcinoma (HCC). J. Clin. Oncol. 2017, 35, 4071. [Google Scholar] [CrossRef]
- Alsaab, H.O.; Sau, S.; Alzhrani, R.; Tatiparti, K.; Bhise, K.; Kashaw, S.K.; Iyer, A.K. PD-1 and PD-L1 Checkpoint Signaling Inhibition for Cancer Immunotherapy: Mechanism, Combinations, and Clinical Outcome. Front. Pharmacol. 2017, 8, 561. [Google Scholar] [CrossRef] [Green Version]
- Kudo, M.; Matilla, A.; Santoro, A.; Melero, I.; Gracián, A.C.; Acosta-Rivera, M.; Choo, S.-P.; El-Khoueiry, A.B.; Kuromatsu, R.; El-Rayes, B.; et al. CheckMate 040 cohort 5: A phase I/II study of nivolumab in patients with advanced hepatocellular carcinoma and Child-Pugh B cirrhosis. J. Hepatol. 2021, 75, 600–609. [Google Scholar] [CrossRef]
- El-Khoueiry, A.B.; Melero, I.; Yau, T.C.; Crocenzi, T.S.; Kudo, M.; Hsu, C.; Choo, S.; Trojan, J.; Welling, T.; Meyer, T.; et al. Impact of antitumor activity on survival outcomes, and nonconventional benefit, with nivolumab (NIVO) in patients with advanced hepatocellular carcinoma (aHCC): Subanalyses of CheckMate-040. J. Clin. Oncol. 2018, 36, 475. [Google Scholar] [CrossRef]
- Lee, C.H.; Lee, Y.B.; Kim, M.A.; Jang, H.; Oh, H.; Kim, S.W.; Cho, E.J.; Lee, K.H.; Lee, J.H.; Yu, S.J.; et al. Effectiveness of nivolumab versus regorafenib in hepatocellular carcinoma patients who failed sorafenib treatment. Clin. Mol. Hepatol. 2020, 26, 328–339. [Google Scholar] [CrossRef]
- Kuo, Y.H.; Yen, Y.H.; Chen, Y.Y.; Kee, K.M.; Hung, C.H.; Lu, S.N.; Hu, T.H.; Chen, C.H.; Wang, J.H. Nivolumab Versus Regorafenib in Patients with Hepatocellular Carcinoma After Sorafenib Failure. Front. Oncol. 2021, 11, 1998. [Google Scholar] [CrossRef]
- Ammerata, G.; Filippo, R.; Laface, C.; Memeo, R.; Solaini, L.; Cavaliere, D.; Navarra, G.; Ranieri, G.; Currò, G.; Ammendola, M. Hyperthermic intraperitoneal chemotherapy and colorectal cancer: From physiology to surgery. World J. Clin. Cases 2022, 10, 10852–10861. [Google Scholar] [CrossRef]
- Ranieri, G.; Laface, C.; Laforgia, M.; De Summa, S.; Porcelli, M.; Macina, F.; Ammendola, M.; Molinari, P.; Lauletta, G.; Di Palo, A.; et al. Bevacizumab Plus FOLFOX-4 Combined with Deep Electro-Hyperthermia as First-line Therapy in Metastatic Colon Cancer: A Pilot Study. Front. Oncol. 2020, 10, 2387. [Google Scholar] [CrossRef]
- Gadaleta-Caldarola, G.; Infusino, S.; Galise, I.; Ranieri, G.; Vinciarelli, G.; Fazio, V.; Divella, R.; Daniele, A.; Filippelli, G.; Gadaleta, C.D. Sorafenib and locoregional deep electro-hyperthermia in advanced hepatocellular carcinoma: A phase II study. Oncol. Lett. 2014, 8, 1783–1787. [Google Scholar] [CrossRef] [Green Version]
- Goffredo, V.; Gadaleta, C.D.; Laterza, A.; Vacca, A.; Ranieri, G. Tryptase serum levels in patients suffering from hepatocellular carcinoma undergoing intra-arterial chemoembolization: Possible predictive role of response to treatment. Mol. Clin. Oncol. 2013, 1, 385–389. [Google Scholar] [CrossRef] [Green Version]
- Ranieri, G.; Gadaleta-Caldarola, G.; Goffredo, V.; Patruno, R.; Mangia, A.; Rizzo, A.; Sciorsci, R.L.; Gadaleta, C.D. Sorafenib (BAY 43-9006) in hepatocellular carcinoma patients: From discovery to clinical development. Curr. Med. Chem. 2012, 19, 938–944. [Google Scholar] [CrossRef]
- Ammendola, M.; Sacco, R.; Sammarco, G.; Piardi, T.; Zuccalà, V.; Patruno, R.; Zullo, A.; Zizzo, N.; Nardo, B.; Marech, I.; et al. Mast cells positive to tryptase, endothelial cells positive to protease-activated receptor-2, and microvascular density correlate among themselves in hepatocellular carcinoma patients who have undergone surgery. OncoTargets Ther. 2016, 9, 4465–4471. [Google Scholar] [CrossRef] [Green Version]
- Gadaleta, C.; Catino, A.; Ranieri, G.; Fazio, V.; Gadaleta-Caldarola, G.; Cramarossa, A.; Armenise, F.; Canniello, E.; Vinciarelli, G.; Laricchia, G.; et al. Single-step therapy–Feasibility and safety of simultaneous transarterial chemoembolization and radiofrequency ablation for hepatic malignancies. In Vivo 2009, 23, 813–820. [Google Scholar]
- Gadaleta, C.; Coviello, M.; Catino, A.; Venneri, M.T.; Stea, B.; Quaranta, M.; Mattioli, V.; Ranieri, G. Serum vascular endothelial growth factor concentrations in hepatocellular cancer patients undergoing percutaneously radiofrequency thermal ablation. J. Chemother. 2004, 16, 7–10. [Google Scholar] [CrossRef]
- Khan, K.A.; Kerbel, R.S. Improving immunotherapy outcomes with anti-angiogenic treatments and vice versa. Nat. Rev. Clin. Oncol. 2018, 15, 310–324. [Google Scholar] [CrossRef] [PubMed]
- Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O.; et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905. [Google Scholar] [CrossRef] [PubMed]
- Ren, Z.; Xu, J.; Bai, Y.; Xu, A.; Cang, S.; Du, C.; Li, Q.; Lu, Y.; Chen, Y.; Guo, Y.; et al. Sintilimab plus a bevacizumab biosimilar (IBI305) versus sorafenib in unresectable hepatocellular carcinoma (ORIENT-32): A randomised, open-label, phase 2–3 study. Lancet Oncol. 2021, 22, 977–990. [Google Scholar] [CrossRef] [PubMed]
- Llovet, J.M.; Kudo, M.; Cheng, A.-L.; Finn, R.S.; Galle, P.R.; Kaneko, S.; Meyer, T.; Qin, S.; Dutcus, C.E.; Chen, E.; et al. Lenvatinib (len) plus pembrolizumab (pembro) for the first-line treatment of patients (pts) with advanced hepatocellular carcinoma (HCC): Phase 3 LEAP-002 study. J. Clin. Oncol. 2019, 37, TPS4152. [Google Scholar] [CrossRef]
- Kudo, M.; Ikeda, M.; Motomura, K.; Okusaka, T.; Kato, N.; Dutcus, C.E.; Hisai, T.; Suzuki, M.; Ikezawa, H.; Iwata, T.; et al. A phase Ib study of lenvatinib (LEN) plus nivolumab (NIV) in patients (pts) with unresectable hepatocellular carcinoma (uHCC): Study 117. J. Clin. Oncol. 2020, 38, 513. [Google Scholar] [CrossRef]
- Kudo, M.; Motomura, K.; Wada, Y.; Inaba, Y.; Sakamoto, Y.; Kurosaki, M.; Umeyama, Y.; Kamei, Y.; Yoshimitsu, J.; Fujii, Y.; et al. Avelumab in Combination with Axitinib as First-Line Treatment in Patients with Advanced Hepatocellular Carcinoma: Results from the Phase 1b VEGF Liver 100 Trial. Liver Cancer 2021, 10, 249–259. [Google Scholar] [CrossRef]
- Xu, J.; Shen, J.; Gu, S.; Zhang, Y.; Wu, L.; Wu, J.; Shao, G.; Zhang, Y.; Xu, L.; Yin, T.; et al. Camrelizumab in Combination with Apatinib in Patients with Advanced Hepatocellular Carcinoma (RESCUE): A Nonrandomized, Open-label, Phase II Trial. Clin. Cancer Res. 2021, 27, 1003–1011. [Google Scholar] [CrossRef] [PubMed]
- Finn, R.S.; Ikeda, M.; Zhu, A.X.; Sung, M.W.; Baron, A.D.; Kudo, M.; Okusaka, T.; Kobayashi, M.; Kumada, H.; Kaneko, S.; et al. Phase Ib Study of Lenvatinib Plus Pembrolizumab in Patients with Unresectable Hepatocellular Carcinoma. J. Clin. Oncol. 2020, 38, 2960–2970. [Google Scholar] [CrossRef]
- Kudo, M. Combination Cancer Immunotherapy in Hepatocellular Carcinoma. Liver Cancer 2018, 7, 20–27. [Google Scholar] [CrossRef]
- Ott, P.A.; Hodi, F.S.; Kaufman, H.L.; Wigginton, J.M.; Wolchok, J.D. Combination immunotherapy: A road map. J. ImmunoTher. Cancer 2017, 5, 16. [Google Scholar] [CrossRef] [Green Version]
- Yau, T.; Kang, Y.-K.; Kim, T.-Y.; El-Khoueiry, A.B.; Santoro, A.; Sangro, B.; Melero, I.; Kudo, M.; Hou, M.-M.; Matilla, A.; et al. Efficacy and Safety of Nivolumab Plus Ipilimumab in Patients with Advanced Hepatocellular Carcinoma Previously Treated with Sorafenib: The CheckMate 040 Randomized Clinical Trial. JAMA Oncol. 2020, 6, e204564. [Google Scholar] [CrossRef]
- Abou-Alfa, G.K.; Lau, G.; Kudo, M.; Chan, S.L.; Kelley, R.K.; Furuse, J.; Sukeepaisarnjaroen, W.; Kang, Y.-K.; Dao, T.V.; Toni, E.N.D.; et al. Tremelimumab plus Durvalumab in Unresectable Hepatocellular Carcinoma. NEJM Evid. 2022, 1, EVIDoa2100070. [Google Scholar] [CrossRef]
- Zhang, R.; Zhang, Z.; Liu, Z.; Wei, D.; Wu, X.; Bian, H.; Chen, Z. Adoptive cell transfer therapy for hepatocellular carcinoma. Front. Med. 2019, 13, 3–11. [Google Scholar] [CrossRef]
- Kershaw, M.H.; Westwood, J.A.; Darcy, P.K. Gene-engineered T cells for cancer therapy. Nat. Rev. Cancer 2013, 13, 525–541. [Google Scholar] [CrossRef]
- Mizukoshi, E.; Kaneko, S. Immune cell therapy for hepatocellular carcinoma. J. Hematol. Oncol. 2019, 12, 52. [Google Scholar] [CrossRef]
- Rochigneux, P.; Chanez, B.; De Rauglaudre, B.; Mitry, E.; Chabannon, C.; Gilabert, M. Adoptive Cell Therapy in Hepatocellular Carcinoma: Biological Rationale and First Results in Early Phase Clinical Trials. Cancers 2021, 13, 271. [Google Scholar] [CrossRef]
- Ping, Y.; Liu, C.; Zhang, Y. T-cell receptor-engineered T cells for cancer treatment: Current status and future directions. Protein Cell 2018, 9, 254–266. [Google Scholar] [CrossRef] [Green Version]
- Sun, L.; Guo, H.; Jiang, R.; Lu, L.; Liu, T.; He, X. Engineered cytotoxic T lymphocytes with AFP-specific TCR gene for adoptive immunotherapy in hepatocellular carcinoma. Tumor Biol. 2016, 37, 799–806. [Google Scholar] [CrossRef]
- Dargel, C.; Bassani-Sternberg, M.; Hasreiter, J.; Zani, F.; Bockmann, J.-H.; Thiele, F.; Bohne, F.; Wisskirchen, K.; Wilde, S.; Sprinzl, M.F.; et al. T Cells Engineered to Express a T-Cell Receptor Specific for Glypican-3 to Recognize and Kill Hepatoma Cells In Vitro and in Mice. Gastroenterology 2015, 149, 1042–1052. [Google Scholar] [CrossRef] [Green Version]
- June, C.H.; O’Connor, R.S.; Kawalekar, O.U.; Ghassemi, S.; Milone, M.C. CAR T cell immunotherapy for human cancer. Science 2018, 359, 1361–1365. [Google Scholar] [CrossRef] [Green Version]
- Shi, D.; Shi, Y.; Kaseb, A.O.; Qi, X.; Zhang, Y.; Chi, J.; Lu, Q.; Gao, H.; Jiang, H.; Wang, H.; et al. Chimeric Antigen Receptor-Glypican-3 T-Cell Therapy for Advanced Hepatocellular Carcinoma: Results of Phase I Trials. Clin. Cancer Res. 2020, 26, 3979–3989. [Google Scholar] [CrossRef] [PubMed]
- Dai, H.; Tong, C.; Shi, D.; Chen, M.; Guo, Y.; Chen, D.; Han, X.; Wang, H.; Wang, Y.; Shen, P. Efficacy and biomarker analysis of CD133-directed CAR T cells in advanced hepatocellular carcinoma: A single-arm, open-label, phase II trial. OncoImmunology 2020, 9, 1846926. [Google Scholar] [CrossRef] [PubMed]
- Jiang, J.; Wu, C.; Lu, B. Cytokine-induced killer cells promote antitumor immunity. J. Transl. Med. 2013, 11, 83. [Google Scholar] [CrossRef] [PubMed]
- Gustafsson, K.; Ingelsten, M.; Bergqvist, L.; Nyström, J.; Andersson, B.; Karlsson-Parra, A. Recruitment and Activation of Natural Killer Cells In vitro by a Human Dendritic Cell Vaccine. Cancer Res. 2008, 68, 5965–5971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Palmer, D.H.; Midgley, R.S.; Mirza, N.; Torr, E.E.; Ahmed, F.; Steele, J.C.; Steven, N.M.; Kerr, D.J.; Young, L.S.; Adams, D.H. A phase II study of adoptive immunotherapy using dendritic cells pulsed with tumor lysate in patients with hepatocellular carcinoma. Hepatology 2009, 49, 124–132. [Google Scholar] [CrossRef] [PubMed]
- Tada, F.; Abe, M.; Hirooka, M.; Ikeda, Y.; Hiasa, Y.; Lee, Y.; Jung, N.-C.; Lee, W.-B.; Lee, H.-S.; Bae, Y.-S.; et al. Phase I/II study of immunotherapy using tumor antigen-pulsed dendritic cells in patients with hepatocellular carcinoma. Int. J. Oncol. 2012, 41, 1601–1609. [Google Scholar] [CrossRef] [Green Version]
- Nakagawa, H.; Mizukoshi, E.; Kobayashi, E.; Tamai, T.; Hamana, H.; Ozawa, T.; Kishi, H.; Kitahara, M.; Yamashita, T.; Arai, K.; et al. Association between High-Avidity T-Cell Receptors, Induced by α-Fetoprotein−Derived Peptides, and Anti-Tumor Effects in Patients with Hepatocellular Carcinoma. Gastroenterology 2017, 152, 1395–1406. [Google Scholar] [CrossRef]
- Sawada, Y.; Yoshikawa, T.; Ofuji, K.; Yoshimura, M.; Tsuchiya, N.; Takahashi, M.; Nobuoka, D.; Gotohda, N.; Takahashi, S.; Kato, Y.; et al. Phase II study of the GPC3-derived peptide vaccine as an adjuvant therapy for hepatocellular carcinoma patients. OncoImmunology 2016, 5, e1129483. [Google Scholar] [CrossRef]
- Mizukoshi, E.; Nakagawa, H.; Kitahara, M.; Yamashita, T.; Arai, K.; Sunagozaka, H.; Iida, N.; Fushimi, K.; Kaneko, S. Phase I trial of multidrug resistance-associated protein 3-derived peptide in patients with hepatocellular carcinoma. Cancer Lett. 2015, 369, 242–249. [Google Scholar] [CrossRef] [Green Version]
- Raja, J.; Ludwig, J.M.; Gettinger, S.N.; Schalper, K.A.; Kim, H.S. Oncolytic virus immunotherapy: Future prospects for oncology. J. ImmunoTher. Cancer 2018, 6, 140. [Google Scholar] [CrossRef]
- Russell, S.J.; Peng, K.-W.; Bell, J.C. Oncolytic virotherapy. Nat. Biotechnol. 2012, 30, 658–670. [Google Scholar] [CrossRef] [Green Version]
- Moehler, M.; Heo, J.; Lee, H.C.; Tak, W.Y.; Chao, Y.; Paik, S.W.; Yim, H.J.; Byun, K.S.; Baron, A.; Ungerechts, G.; et al. Vaccinia-based oncolytic immunotherapy Pexastimogene Devacirepvec in patients with advanced hepatocellular carcinoma after sorafenib failure: A randomized multicenter Phase IIb trial (TRAVERSE). OncoImmunology 2019, 8, 1615817. [Google Scholar] [CrossRef] [Green Version]
- Abou-Alfa, G.K.; Galle, P.R.; Chao, Y.; Brown, K.T.; Heo, J.; Borad, M.J.; Luca, A.; Pelusio, A.; Agathon, D.; Lusky, M.; et al. PHOCUS: A phase 3 randomized, open-label study comparing the oncolytic immunotherapy Pexa-Vec followed by sorafenib (SOR) vs. SOR in patients with advanced hepatocellular carcinoma (HCC) without prior systemic therapy. J. Clin. Oncol. 2016, 34, TPS4146. [Google Scholar] [CrossRef]
- LaRocca, C.J.; Warner, S.G. Oncolytic viruses and checkpoint inhibitors: Combination therapy in clinical trials. Clin. Transl. Med. 2018, 7, 35. [Google Scholar] [CrossRef] [Green Version]
- Hunter, G.L.; Hadjivasiliou, Z.; Bonin, H.; He, L.; Perrimon, N.; Charras, G.; Baum, B. Coordinated control of Notch/Delta signalling and cell cycle progression drives lateral inhibition-mediated tissue patterning. Development 2016, 143, 2305–2310. [Google Scholar] [CrossRef] [Green Version]
- Lubman, O.Y.; Korolev, S.V.; Kopan, R. Anchoring notch genetics and biochemistry; structural analysis of the ankyrin domain sheds light on existing data. Mol. Cell 2004, 13, 619–626. [Google Scholar] [CrossRef]
- Kopan, R.; Ilagan, M.X. The canonical Notch signaling pathway: Unfolding the activation mechanism. Cell 2009, 137, 216–233. [Google Scholar] [CrossRef] [Green Version]
- Lim, K.J.; Brandt, W.D.; Heth, J.A.; Muraszko, K.M.; Fan, X.; Bar, E.E.; Eberhart, C.G. Lateral inhibition of Notch signaling in neoplastic cells. Oncotarget 2015, 6, 1666–1677. [Google Scholar] [CrossRef] [Green Version]
- Yamamoto, S.; Schulze, K.L.; Bellen, H.J. Introduction to Notch signaling. Methods Mol. Biol. 2014, 1187, 1–14. [Google Scholar] [CrossRef]
- Radtke, F.; Raj, K. The role of Notch in tumorigenesis: Oncogene or tumour suppressor? Nat. Rev. Cancer 2003, 3, 756–767. [Google Scholar] [CrossRef]
- Lim, S.O.; Kim, H.S.; Quan, X.; Ahn, S.M.; Kim, H.; Hsieh, D.; Seong, J.K.; Jung, G. Notch1 binds and induces degradation of Snail in hepatocellular carcinoma. BMC Biol. 2011, 9, 83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Villanueva, A.; Alsinet, C.; Yanger, K.; Hoshida, Y.; Zong, Y.; Toffanin, S.; Rodriguez-Carunchio, L.; Solé, M.; Thung, S.; Stanger, B.Z.; et al. Notch signaling is activated in human hepatocellular carcinoma and induces tumor formation in mice. Gastroenterology 2012, 143, 1660–1669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Giovannini, C.; Minguzzi, M.; Genovese, F.; Baglioni, M.; Gualandi, A.; Ravaioli, M.; Milazzo, M.; Tavolari, S.; Bolondi, L.; Gramantieri, L. Molecular and proteomic insight into Notch1 characterization in hepatocellular carcinoma. Oncotarget 2016, 7, 39609–39626. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.S.; Jung, G. Notch1 increases Snail expression under high reactive oxygen species conditions in hepatocellular carcinoma cells. Free. Radic. Res. 2014, 48, 806–813. [Google Scholar] [CrossRef] [PubMed]
- Hu, Y.J.; Li, H.Y.; Qiu, K.J.; Li, D.C.; Zhou, J.H.; Hu, Y.H.; Zhang, F.M. Downregulation of Notch1 inhibits the invasion of human hepatocellular carcinoma HepG2 and MHCC97H cells through the regulation of PTEN and FAK. Int. J. Mol. Med. 2014, 34, 1081–1086. [Google Scholar] [CrossRef] [Green Version]
- Ahn, S.; Hyeon, J.; Park, C.K. Notch1 and Notch4 are markers for poor prognosis of hepatocellular carcinoma. Hepatobiliary Pancreat. Dis. Int. 2013, 12, 286–294. [Google Scholar] [CrossRef]
- Gramantieri, L.; Giovannini, C.; Lanzi, A.; Chieco, P.; Ravaioli, M.; Venturi, A.; Grazi, G.L.; Bolondi, L. Aberrant Notch3 and Notch4 expression in human hepatocellular carcinoma. Liver Int. 2007, 27, 997–1007. [Google Scholar] [CrossRef]
- Hayashi, Y.; Osanai, M.; Lee, G.H. NOTCH2 signaling confers immature morphology and aggressiveness in human hepatocellular carcinoma cells. Oncol. Rep. 2015, 34, 1650–1658. [Google Scholar] [CrossRef] [Green Version]
- Wu, W.R.; Zhang, R.; Shi, X.D.; Yi, C.; Xu, L.B.; Liu, C. Notch2 is a crucial regulator of self-renewal and tumorigenicity in human hepatocellular carcinoma cells. Oncol. Rep. 2016, 36, 181–188. [Google Scholar] [CrossRef] [Green Version]
- Ortica, S.; Tarantino, N.; Aulner, N.; Israël, A.; Gupta-Rossi, N. The 4 Notch receptors play distinct and antagonistic roles in the proliferation and hepatocytic differentiation of liver progenitors. FASEB J. 2014, 28, 603–614. [Google Scholar] [CrossRef]
- Giovannini, C.; Bolondi, L.; Gramantieri, L. Targeting Notch3 in Hepatocellular Carcinoma: Molecular Mechanisms and Therapeutic Perspectives. Int. J. Mol. Sci. 2017, 18, 56. [Google Scholar] [CrossRef]
- Hu, L.; Xue, F.; Shao, M.; Deng, A.; Wei, G. Aberrant expression of Notch3 predicts poor survival for hepatocellular carcinomas. Biosci. Trends 2013, 7, 152–156. [Google Scholar]
- Gao, J.; Song, Z.; Chen, Y.; Xia, L.; Wang, J.; Fan, R.; Du, R.; Zhang, F.; Hong, L.; Song, J.; et al. Deregulated expression of Notch receptors in human hepatocellular carcinoma. Dig. Liver Dis. 2008, 40, 114–121. [Google Scholar] [CrossRef]
- McAuliffe, S.M.; Morgan, S.L.; Wyant, G.A.; Tran, L.T.; Muto, K.W.; Chen, Y.S.; Chin, K.T.; Partridge, J.C.; Poole, B.B.; Cheng, K.H.; et al. Targeting Notch, a key pathway for ovarian cancer stem cells, sensitizes tumors to platinum therapy. Proc. Natl. Acad. Sci. USA 2012, 109, E2939–E2948. [Google Scholar] [CrossRef]
- Ye, Q.F.; Zhang, Y.C.; Peng, X.Q.; Long, Z.; Ming, Y.Z.; He, L.Y. siRNA-mediated silencing of Notch-1 enhances docetaxel induced mitotic arrest and apoptosis in prostate cancer cells. Asian Pac. J. Cancer Prev. APJCP 2012, 13, 2485–2489. [Google Scholar] [CrossRef] [Green Version]
- Giovannini, C.; Gramantieri, L.; Chieco, P.; Minguzzi, M.; Lago, F.; Pianetti, S.; Ramazzotti, E.; Marcu, K.B.; Bolondi, L. Selective ablation of Notch3 in HCC enhances doxorubicin’s death promoting effect by a p53 dependent mechanism. J. Hepatol. 2009, 50, 969–979. [Google Scholar] [CrossRef]
- Giovannini, C.; Baglioni, M.; Toaldo, M.B.; Ventrucci, C.; D’Adamo, S.; Cipone, M.; Chieco, P.; Gramantieri, L.; Bolondi, L. Notch3 inhibition enhances sorafenib cytotoxic efficacy by promoting GSK3b phosphorylation and p21 down-regulation in hepatocellular carcinoma. Oncotarget 2013, 4, 1618–1631. [Google Scholar] [CrossRef] [Green Version]
- Ma, P.; Zou, C.; Xia, S. Oncogenic signaling pathway mediated by Notch pathway-related genes induces immunosuppression and immunotherapy resistance in hepatocellular carcinoma. Immunogenetics 2022, 74, 539–557. [Google Scholar] [CrossRef]
- Laface, C.; Fedele, P.; Maselli, F.M.; Ambrogio, F.; Foti, C.; Molinari, P.; Ammendola, M.; Lioce, M.; Ranieri, G. Targeted Therapy for Hepatocellular Carcinoma: Old and New Opportunities. Cancers 2022, 14, 4028. [Google Scholar] [CrossRef]
- Laface, C.; Laforgia, M.; Molinari, P.; Ugenti, I.; Gadaleta, C.D.; Porta, C.; Ranieri, G. Hepatic Arterial Infusion of Chemotherapy for Advanced Hepatobiliary Cancers: State of the Art. Cancers 2021, 13, 3091. [Google Scholar] [CrossRef]
- Laforgia, M.; Laface, C.; Calabrò, C.; Ferraiuolo, S.; Ungaro, V.; Tricarico, D.; Gadaleta, C.D.; Nardulli, P.; Ranieri, G. Peripheral Neuropathy under Oncologic Therapies: A Literature Review on Pathogenetic Mechanisms. Int. J. Mol. Sci. 2021, 22, 1980. [Google Scholar] [CrossRef] [PubMed]
- Sia, D.; Jiao, Y.; Martinez-Quetglas, I.; Kuchuk, O.; Villacorta-Martin, C.; Castro de Moura, M.; Putra, J.; Camprecios, G.; Bassaganyas, L.; Akers, N.; et al. Identification of an Immune-specific Class of Hepatocellular Carcinoma, Based on Molecular Features. Gastroenterology 2017, 153, 812–826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Llovet, J.M.; Montal, R.; Sia, D.; Finn, R.S. Molecular therapies and precision medicine for hepatocellular carcinoma. Nat. Rev. Clin. Oncol. 2018, 15, 599–616. [Google Scholar] [CrossRef]
- Harding, J.J.; Nandakumar, S.; Armenia, J.; Khalil, D.N.; Albano, M.; Ly, M.; Shia, J.; Hechtman, J.F.; Kundra, R.; El Dika, I.; et al. Prospective Genotyping of Hepatocellular Carcinoma: Clinical Implications of Next-Generation Sequencing for Matching Patients to Targeted and Immune Therapies. Clin. Cancer Res. 2019, 25, 2116–2126. [Google Scholar] [CrossRef] [PubMed]
- Pinyol, R.; Sia, D.; Llovet, J.M. Immune Exclusion-Wnt/CTNNB1 Class Predicts Resistance to Immunotherapies in HCC. Clin. Cancer Res. 2019, 25, 2021–2023. [Google Scholar] [CrossRef] [Green Version]
- Jung, K.H.; Yoo, W.; Stevenson, H.L.; Deshpande, D.; Shen, H.; Gagea, M.; Yoo, S.-Y.; Wang, J.; Eckols, T.K.; Bharadwaj, U.; et al. Multifunctional Effects of a Small-Molecule STAT3 Inhibitor on NASH and Hepatocellular Carcinoma in Mice. Clin. Cancer Res. 2017, 23, 5537–5546. [Google Scholar] [CrossRef] [Green Version]
- Tsimberidou, A.M.; de Achaval, S.; Alibhai, I.; Kaseb, A.O. First-in-man phase I clinical trial evaluating TTI-101, an orally bioavailable, small molecule inhibitor of STAT3, in patients with advanced solid tumors. J. Clin. Oncol. 2021, 39, TPS3158. [Google Scholar] [CrossRef]
- Yang, Y.; Li, G.; Lu, Z.; Liu, Y.; Kong, J.; Liu, J. Progression of Prothrombin Induced by Vitamin K Absence-II in Hepatocellular Carcinoma. Front. Oncol. 2021, 11, 4997. [Google Scholar] [CrossRef]
- Rizzo, A.; Brandi, G. Biochemical predictors of response to immune checkpoint inhibitors in unresectable hepatocellular carcinoma. Cancer Treat. Res. Commun. 2021, 27, 100328. [Google Scholar] [CrossRef]
PD-1/PD-L1 Inhibitors | Trial | Comparison | Setting | OS | PFS/TTP * | ORR/DCR ** | Grade 3–4 AEs |
---|---|---|---|---|---|---|---|
Nivolumab (Opdivo) | CheckMate-040 (Phase I–II) [40] | / | ≥Second-line | / | 3.4 months * | 15–20% 58–64% ** | 25% |
Nivolumab (Opdivo) | CheckMate-459 (Phase III) [41] | Sorafenib | First-line | 16.4 vs. 14.7 months; HR = 0.85; p = 0.0752 | / | 15% vs. 7% | 22% vs. 49% |
Pembrolizumab (Keytruda) | KEYNOTE-224 (Phase II) [42] | / | Second-line | 12.9 months | 4.9 months | 17% | 25% |
Pembrolizumab (Keytruda) | KEYNOTE-240 (Phase III) [43] | Placebo | Second/Third-line | 13.9 vs. 10.6 months; HR, 0.781; p = 0.0238 | 3.0 vs. 2.8 months HR = 0.718; p = 0.0022 | 18.3% vs. 4.4% p = 0.00007 | 52.7% vs. 46.3% |
Pembrolizumab (Keytruda) | KEYNOTE-394 (Phase III) [44] | Placebo | Second-line | 13.6 vs. 13.0 months; HR 0.79, p = 0.0180 | 2.6 vs. 2.3 months; HR 0.74, p = 0.0032 | 13.7% vs. 1.3% | 14.4% vs. 5.9% |
Camrelizumab (SHR-1210) | Phase II [45] | / | Second-line | 74.4% alive at six months | / | 14.7% | 22% |
Tislelizumab (BGB-A317) | RATIONALE-301 (Phase III) [46] | Sorafenib | First-line | 15.9 vs. 14.1 months; HR: 0.85 | / | 25% vs. 10.3% | 48.2% vs. 65.4% |
Durvalumab (Imfizi) | Phase I–II [47] | / | Second-line | 19.3 vs. 13.2 months | / | 25% vs. 10.3% | 24% |
Combination | Trial | Comparison | Setting | OS | PFS/TTRP * | ORR/DCR ** | Grade 3–4 AEs |
---|---|---|---|---|---|---|---|
Bevacizumab (Avastin) + Atezolizumab (Tecentriq) | IMBrave150 (Phase III) [62] | Sorafenib | First-line | 19.2 vs. 13.4 months; HR = 0.66; p = 0.0009 | 6.8 vs. 4.3 months; HR 0.59 | 43% vs. 32%; p = 0.002 ** | 56.5% vs. 55.1% |
Sintilimab (Tyvyt) + Bevacizumab (IBI305) | ORIENT-32 (Phase II–III) [63] | Sorafenib | First-line | Median not reached vs. 10.4 months; HR 0.57; p < 0.0001 | 4.6 vs. 2.8 months, HR 0.56, p < 0.0001 | / | 14% vs. 6% |
Pembrolizumab (Keyruda) + Lenvatinib (Lenviva) | LEAP002 (Phase III) [64] | Sorafenib | First-line | 21.2 vs. 19 months HR 0.840, p = 0.0227 | / | 26.1% vs. 17.5% | 62.5% vs. 57.5% |
Nivolumab (Opdivo) + Lenvatinib (Lenviva) | Study 117 (Phase Ib) [65] | / | First-line | / | / | 76.7% | 55% |
Avelumab (Bavencio) + Axitinib (Inlyta) | VEGF Liver 100 (Phase Ib) [66] | / | First-line | / | / | 13.6% | 50% |
Camrelizumab (AiRuiKa) + Apatinib (Rivoceranib) | RESCUE (Phase II) [67] | / | First/second-line | The 12-month survival rate, was 74.7% vs. 68.2% in first- vs. second-line groups, respectively. | 5.7 vs. 5.5 months in first- vs. second-line groups, respectively | 34.3% vs. 22.5% in first- vs. second-line groups, respectively | 77.4% |
Combinations | Trial | Comparison | Setting | OS | PFS/TTRP * | ORR/DCR ** | Grade 3–4 AEs |
---|---|---|---|---|---|---|---|
Nivolumab (Opdivo) + Ipilimumab (Yervoy) | CheckMate 040 (Phase I–II) [71] | Three different dosing arms | Second-line | 22.8 months | / | 32% | 25% |
Nivolumab (Opdivo) + Ipilimumab (Yervoy) | CheckMate 9DW (Phase III) | Sorafenib or Lenvatinib | First-line | Ongoing | Ongoing | Ongoing | Ongoing |
Tremelimumab (a single dose of 300 mg) + Durvalumab (1500 mg every 4 weeks) | HIMALAYA (Phase III) [72] | Sorafenib | First-line | 16.4 vs. 13.8 months p = 0.0035 | 3.8 vs. 4.1 months | 20% vs. 5.1% | 26% vs. 37% |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Laface, C.; Ranieri, G.; Maselli, F.M.; Ambrogio, F.; Foti, C.; Ammendola, M.; Laterza, M.; Cazzato, G.; Memeo, R.; Mastrandrea, G.; et al. Immunotherapy and the Combination with Targeted Therapies for Advanced Hepatocellular Carcinoma. Cancers 2023, 15, 654. https://doi.org/10.3390/cancers15030654
Laface C, Ranieri G, Maselli FM, Ambrogio F, Foti C, Ammendola M, Laterza M, Cazzato G, Memeo R, Mastrandrea G, et al. Immunotherapy and the Combination with Targeted Therapies for Advanced Hepatocellular Carcinoma. Cancers. 2023; 15(3):654. https://doi.org/10.3390/cancers15030654
Chicago/Turabian StyleLaface, Carmelo, Girolamo Ranieri, Felicia Maria Maselli, Francesca Ambrogio, Caterina Foti, Michele Ammendola, Marigia Laterza, Gerardo Cazzato, Riccardo Memeo, Giovanni Mastrandrea, and et al. 2023. "Immunotherapy and the Combination with Targeted Therapies for Advanced Hepatocellular Carcinoma" Cancers 15, no. 3: 654. https://doi.org/10.3390/cancers15030654
APA StyleLaface, C., Ranieri, G., Maselli, F. M., Ambrogio, F., Foti, C., Ammendola, M., Laterza, M., Cazzato, G., Memeo, R., Mastrandrea, G., Lioce, M., & Fedele, P. (2023). Immunotherapy and the Combination with Targeted Therapies for Advanced Hepatocellular Carcinoma. Cancers, 15(3), 654. https://doi.org/10.3390/cancers15030654