Drug co-administration in the tumor immune microenvironment of Hepatocellular carcinoma
Yingying Shao, Ranran Su, Yu Wang, Shuangshuang Yin, Weiling Pu, Sangho Koo, Haiyang Yu
Drug co-administration in the tumor immune microenvironment of Hepatocellular carcinoma
The etiology and exact molecular mechanisms of primary hepatocellular carcinoma (HCC) remain unclear, and its incidence has continued to increase in recent years. Despite tremendous advances in systemic therapies such as molecularly targeted drugs, HCC has some of the worst prognoses owing to drug resistance, frequent recurrence, and metastasis. Hepatocellular carcinoma is a widespread disease and its progression is regulated by the immune system. Traditional Chinese medicine (TCM) has been gradually theorized and systematized to have a holistic regulatory role for use in the prevention and treatment of tumors. Although half of the patients with HCC receive systemic therapy, traditionally sorafenib or lenvatinib are used as first-line treatment modalities. TCM is also widely used in the treatment of HCC, and the same immune checkpoint inhibitors (ICIs) such as PD-L1 have also received much focus in the field of continuously changing cancer treatment. Owing to the high probability of resistance to specific drugs and unsatisfactory efficacy due to administration of chemotherapy in single doses, the combination of drugs is the newest therapeutic option for patients with tumors and has become increasingly prominent for treatment. In this article, the research progress on combination therapy in the immunology of HCC is reviewed and the unique advantages of synergistic anti-tumor therapy with combination drugs are highlighted to provide new solutions for the clinical treatment of tumors.
Combination drug / Hepatocellular carcinoma / Immune checkpoint inhibitors / Traditional Chinese medicine / Tumor microenvironment
[[1]] |
Llovet JM, Castet F, Heikenwalder M, et al. Immunotherapies for hepatocellular carcinoma. Nat Rev Clin Oncol 2022;19(3):151-172.
|
[[2]] |
Sangro B, Sarobe P, Hervas-Stubbs S, et al. Advances in immunotherapy for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2021;18(8):525-543.
|
[[3]] |
Yang JD, Hainaut P, Gores GJ, et al. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol 2019;16(10):589-604.
|
[[4]] |
Donne R, Lujambio A. The liver cancer immune microenvironment: therapeutic implications for hepatocellular carcinoma. Hepatology (Baltimore, Md.) 2022;77(5):1773-1796.
|
[[5]] |
Nedeljković M, Damjanović A. Mechanisms of chemotherapy resistance in triple-negative breast cancer—how we can rise to the challenge. Cells 2019;8(9):957.
|
[[6]] |
Villanueva A, Longo DL. Hepatocellular carcinoma. N Engl J Med 2019;380(15):1450-1462.
|
[[7]] |
Llovet JM, Ricci S, Mazzaferro V, et al. SHARP Investigators Study Group. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 2008;359(4):378-390.
|
[[8]] |
Galle PR, Forner A, Llovet JM, et al. EASL clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol 2018;69(1):182-236.
|
[[9]] |
Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity 2013;39(1):1-10.
|
[[10]] |
Predina J, Eruslanov E, Judy B, et al. Changes in the local tumor microenvironment in recurrent cancers may explain the failure of vaccines after surgery. Proc Natl Acad Sci USA 2013;110(5):415-424.
|
[[11]] |
Wang L, Qian J, Lu Y, et al. Immune evasion of mantle cell lymphoma: expression of B7-H1 leads to inhibited T-cell response to and killing of tumor cells. Haematologica 2013;98(9):1458-1466.
|
[[12]] |
Harkus U, Wankell M, Palamuthusingam P, et al. Immune checkpoint inhibitors in HCC: cellular, molecular and systemic data. Semin Cancer Biol 2022;86(Pt 3):799-815.
|
[[13]] |
Turner MD, Nedjai B, Hurst T, et al. Cytokines and chemokines: at the crossroads of cell signalling and inflammatory disease. Biochim Biophys Acta 2014;1843(11):2563-2582.
|
[[14]] |
Feng GS, Hanley KL, Liang Y, et al. Improving the efficacy of liver cancer immunotherapy: the power of combined preclinical and clinical studies. Hepatology 2021;73(S1):104-114.
|
[[15]] |
Xu J, Niu T. Natural killer cell-based immunotherapy for acute myeloid leukemia. J Hematol Oncol 2020;13(1):167.
|
[[16]] |
Yilmaz A, Cui H, Caligiuri MA, et al. Chimeric antigen receptor-engineered natural killer cells for cancer immunotherapy. J Hematol Oncol 2020;13(1):168.
|
[[17]] |
Wang Y, Xiang Y, Xin VW, et al. Dendritic cell biology and its role in tumor immunotherapy. J Hematol Oncol 2020;13(1):107.
|
[[18]] |
Lin Y, Xu J, Lan H. Tumor-associated macrophages in tumor metastasis: biological roles and clinical therapeutic applications. J Hematol Oncol 2019;12(1):76.
|
[[19]] |
Lee J, Liao R, Wang G, et al. Preventive inhibition of liver tumorigenesis by systemic activation of innate immune functions. Cell Rep 2017;21(7):1870-1882.
|
[[20]] |
Hato T, Goyal L, Greten TF, et al. Immune checkpoint blockade in hepatocellular carcinoma: current progress and future directions. Hepatology 2014;60(5):1776-1782.
|
[[21]] |
Sangro B, Gomez-Martin C, de la Mata M, et al. A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C. J Hepatol 2013;59(1):81-88.
|
[[22]] |
El-Khoueiry AB, Sangro B, Yau T, 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(10088):2492-2502.
|
[[23]] |
Tian M, Shi Y, Liu W, et al. Immunotherapy of hepatocellular carcinoma: strategies for combinatorial intervention. Sci China Life Sci 2019;62(9):1138-1143.
|
[[24]] |
Abdu S, Juaid N, Amin A, et al. Effects of sorafenib and quercetin alone or in combination in treating hepatocellular carcinoma: in vitro and in vivo approaches. Molecules 2022;27(22):8082.
|
[[25]] |
Abdu S, Juaid N, Amin A, et al. Therapeutic effects of crocin alone or in combination with sorafenib against hepatocellular carcinoma: in vivo & in vitro insights. Antioxidants (Basel) 2022;11(9):1645.
|
[[26]] |
Septembre-Malaterre A, Lalarizo Rakoto M, Marodon C, et al. Artemisia annua, a traditional plant brought to light. Int J Mol Sci 2020;21(14):4986.
|
[[27]] |
Hou J, Wang D, Zhang R, et al. Experimental therapy of hepatoma with artemisinin and its derivatives: in vitro and in vivo activity, chemosensitization, and mechanisms of action. Clin Cancer Res 2008;14(17):5519-5530.
|
[[28]] |
Crespo-Ortiz MP, Wei MQ. Antitumor activity of artemisinin and its derivatives: from a well-known antimalarial agent to a potential anticancer drug. J Biomed Biotechnol 2012;2012:1-18.
|
[[29]] |
Li H, Xu K, Pian G, et al. Artesunate and sorafenib: combinatorial inhibition of liver cancer cell growth. Oncol Lett 2019, 18(5): 4735-4743.
|
[[30]] |
Xie X, Yang Y, Wang Q, et al. Targeting ATAD3A-PINK1-mitophagy axis overcomes chemoimmunotherapy resistance by redirecting PD-L1 to mitochondria. Cell Res 2023;33(3):215-228.
|
[[31]] |
Man S, Yao J, Lv P, et al. Curcumin-enhanced antitumor effects of sorafenib via regulating the metabolism and tumor microenvironment. Food Funct 2020;11(7):6422-6432.
|
[[32]] |
Kudo M, Finn RS, Qin S, et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. Lancet 2018;391(10126):1163-1173.
|
[[33]] |
Bruix J, Qin S, Merle P, et al. RESORCE Investigators. Regorafenib for patients with hepatocellular carcinoma who progressed onsorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;389(10064):56-66.
|
[[34]] |
Abou-Alfa GK, Meyer T, Cheng AL, et al. Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. N Engl J Med 2018;379(1):54-63.
|
[[35]] |
Wang J, Hu F, Yu P, et al. Sorafenib inhibits doxorubicin-induced PD-L1 upregulation to improve immunosuppressive microenvironment in Osteosarcoma. J Cancer Res Clin Oncol 2022;149(8):5127-5138.
|
[[36]] |
Cheng CC, Ho AS, Peng CL, et al. Sorafenib suppresses radioresistance and synergizes radiotherapy-mediated CD8(+) T cell activation to eradicate hepatocellular carcinoma. Int Immunopharmacol 2022;112:109110.
|
[[37]] |
Kikuchi H, Matsui A, Morita S, et al. Increased CD8+ T-cell infiltration and efficacy for multikinase inhibitors after PD-1 blockade in hepatocellular carcinoma. J Natl Cancer Inst 2022;114(9):1301-1305.
|
[[38]] |
Wang X, Hu R, Song Z, et al. Sorafenib combined with STAT3 knockdown triggers ER stress-induced HCC apoptosis and cGAS-STING-mediated anti-tumor immunity. Cancer Lett 2022;547:215880.
|
[[39]] |
Liu B, Fang X, Kwong DL, et al. Targeting TROY-mediated P85a/AKT/TBX3 signaling attenuates tumor stemness and elevates treatment response in hepatocellular carcinoma. J Exp Clin Cancer Res 2022;41(1):182.
|
[[40]] |
Lu LC, Lee YH, Chang CJ, et al. Increased expression of programmed death-ligand 1 in infiltrating immune cells in hepatocellular carcinoma tissues after sorafenib treatment. Liver Cancer 2019;8(2):110-120.
|
[[41]] |
Mei Z, Gao X, Pan C, et al. Lenvatinib enhances antitumor immunity by promoting the infiltration of TCF1(+) CD8(+) T cells in HCC via blocking VEGFR2. Cancer Sci 2023;114(4):1284-1296.
|
[[42]] |
Wei CY, Zhu MX, Zhang PF, et al. PKCalpha/ZFP64/CSF1 axis resets the tumor microenvironment and fuels anti-PD1 resistance in hepatocellular carcinoma. J Hepatol 2022;77(1):163-176.
|
[[43]] |
Yi C, Chen L, Lin Z, et al. Lenvatinib targets FGF receptor 4 to enhance antitumor immune response of anti-programmed cell death-1 in HCC. Hepatology 2021;74(5):2544-2560.
|
[[44]] |
Ou DL, Chen CW, Hsu CL, et al. Regorafenib enhances antitumor immunity via inhibition of p38 kinase/Creb1/Klf4 axis in tumor-associated macrophages. J ImmunoTher Cancer 2021;9(3):e001657.
|
[[45]] |
Shigeta K, Matsui A, Kikuchi H, et al. Regorafenib combined with PD1 blockade increases CD8 T-cell infiltration by inducing CXCL10 expression in hepatocellular carcinoma. J ImmunoTher Cancer 2020;8(2):e001435.
|
[[46]] |
Shang R, Song X, Wang P, et al. Cabozantinib-based combination therapy for the treatment of hepatocellular carcinoma. Gut 2021;70(9):1746-1757.
|
[[47]] |
Esteban-Fabro R, Willoughby CE, Pique-Gili M, et al. Cabozantinib enhances anti-PD1 activity and elicits a neutrophil-based immune response in hepatocellular carcinoma. Clin Cancer Res 2022;28(11):2449-2460.
|
[[48]] |
Chan CY, Chiu DK, Yuen VW, et al. CFI-402257, a TTK inhibitor, effectively suppresses hepatocellular carcinoma. Proc Natl Acad Sci U S A 2022;119(32):e2119514119.
|
[[49]] |
Sheng H, Huang Y, Xiao Y, et al. ATR inhibitor AZD6738 enhances the antitumor activity of radiotherapy and immune checkpoint inhibitors by potentiating the tumor immune microenvironment in hepatocellular carcinoma. J ImmunoTher Cancer 2020;8(1):e000340.
|
[[50]] |
Guo Y, Wang J, Benedict B, et al. Targeting CDC7 potentiates ATR-CHK1 signaling inhibition through induction of DNA replication stress in liver cancer. Genome Med 2021;13(1):166.
|
[[51]] |
Yang Y, Sun M, Yao W, et al. Compound kushen injection relieves tumor-associated macrophage-mediated immunosuppression through TNFR1 and sensitizes hepatocellular carcinoma to sorafenib. J ImmunoTher Cancer 2020;8(1):e000317.
|
[[52]] |
Galluzzi L, Vitale I, Aaronson SA, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ 2018;25(3):486-541.
|
[[53]] |
Patel SA, Minn AJ. Combination cancer therapy with immune checkpoint blockade: mechanisms and strategies. Immunity 2018;48(3):417-433.
|
[[54]] |
Galluzzi L, Vitale I, Warren S, et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J ImmunoTher Cancer 2020;8(1):e000337.
|
[[55]] |
Gravett AM, Trautwein N, Stevanovic S, et al. Gemcitabine alters the proteasome composition and immunopeptidome of tumour cells. Oncoimmunology 2018;7(6):e1438107.
|
[[56]] |
Wan S, Pestka S, Jubin RG, et al. Chemotherapeutics and radiation stimulate MHC class I expression through elevated interferon-beta signaling in breast cancer cells. PLoS One 2012;7(3):e32542.
|
[[57]] |
Suzuki E, Kapoor V, Jassar AS, et al. Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res 2005;11(18):6713-6721.
|
[[58]] |
Vincent J, Mignot G, Chalmin F, et al. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res 2010;70(8):3052-3061.
|
[[59]] |
Dimeloe S, Frick C, Fischer M, et al. Human regulatory T cells lack the cyclophosphamide-extruding transporter ABCB1 and are more susceptible to cyclophosphamide-induced apoptosis. Eur J Immunol 2014;44(12):3614-3620.
|
[[60]] |
Welters MJ, van der Sluis TC, van Meir H, et al. Vaccination during myeloid cell depletion by cancer chemotherapy fosters robust T cell responses. Sci Transl Med 2016;8(334):334ra52.
|
[[61]] |
Germano G, Frapolli R, Belgiovine C, et al. Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell 2013;23(2):249-262.
|
[[62]] |
Park SE, Lee SH, Ahn JS, et al. Increased response rates to salvage chemotherapy administered after PD-1/PD-L1 inhibitors in patients with non-small cell lung cancer. J Thorac Oncol 2018;13(1):106-111.
|
[[63]] |
Moschella F, Valentini M, Arico E, et al. Unraveling cancer chemoimmunotherapy mechanisms by gene and protein expression profiling of responses to cyclophosphamide. Cancer Res 2011;71(10):3528-3539.
|
[[64]] |
Sautes-Fridman C, Lawand M, Giraldo NA, et al. Tertiary lymphoid structures in cancers: prognostic value, regulation, and manipulation for therapeutic intervention. Front Immunol 2016;7:407.
|
[[65]] |
Sautes-Fridman C, Petitprez F, Calderaro J, et al. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer 2019;19(6):307-325.
|
[[66]] |
Lin L, Hu X, Zhang H, et al. Tertiary lymphoid organs in cancer immunology: mechanisms and the new strategy for immunotherapy. Front Immunol 2019;10:1398.
|
[[67]] |
Silina K, Soltermann A, Attar FM, et al. Germinal centers determine the prognostic relevance of tertiary lymphoid structures and are impaired by corticosteroids in lung squamous cell carcinoma. Cancer Res 2018;78(5):1308-1320.
|
[[68]] |
West H, McCleod M, Hussein M, et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2019;20(7):924-937.
|
[[69]] |
Paz-Ares L, Dvorkin M, Chen Y, et al. CASPIAN Investigators. Durvalumab plus platinum-etoposide versus platinum-etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): a randomised, controlled, open-label, phase 3 trial. Lancet 2019;394(10212):1929-1939.
|
[[70]] |
Schmid P, Adams S, Rugo HS, et al. IMpassion130 Trial Investigators. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med 2018;379(22):2108-2121.
|
[[71]] |
Powles T, Park SH, Voog E, et al. Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma. N Engl J Med 2020;383(13):1218-1230.
|
[[72]] |
Jotte R, Cappuzzo F, Vynnychenko I, et al. Atezolizumab in combination with carboplatin and Nab-paclitaxel in advanced squamous NSCLC (IMpower131): results from a randomized phase III trial. J Thorac Oncol 2020;15(8):1351-1360.
|
[[73]] |
Qin S, Cheng Y, Liang J, et al. Efficacy and safety of the FOLFOX4 regimen versus doxorubicin in Chinese patients with advanced hepatocellular carcinoma: a subgroup analysis of the EACH study. Oncologist 2014;19(11):1169-1178.
|
[[74]] |
Casares N, Pequignot MO, Tesniere A, et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J Exp Med 2005;202(12):1691-1701.
|
[[75]] |
Dhanasekaran R, Kooby DA, Staley CA, et al. Comparison of conventional transarterial chemoembolization (TACE) and chemoembolization with doxorubicin drug eluting beads (DEB) for unresectable hepatocelluar carcinoma (HCC). J Surg Oncol 2010;101(6):476-480.
|
[[76]] |
Kwong TT, Wong CH, Zhou JY, et al. Chemotherapy-induced recruitment of myeloid-derived suppressor cells abrogates efficacy of immune checkpoint blockade. JHEP Rep 2021;3(2):100224.
|
[[77]] |
Brandi G, Di Federico A, Rizzo A, et al. The power of kindness: curative treatment with metronomic combination in advanced hepatocellular carcinoma. Anticancer Drugs 2022;33(1):e781-e783.
|
[[78]] |
Koyama S, Akbay EA, Li YY, et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun 2016;7:10501.
|
[[79]] |
Zhu S, Zhang T, Zheng L, et al. Combination strategies to maximize the benefits of cancer immunotherapy. J Hematol Oncol 2021;14(1):156.
|
[[80]] |
Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363(8):711-723.
|
[[81]] |
Nicolo E, Giugliano F, Ascione L, et al. Combining antibody-drug conjugates with immunotherapy in solid tumors: current landscape and future perspectives. Cancer Treat Rev 2022;106:102395.
|
[[82]] |
Lv T, Xiong X, Yan W, et al. Targeting of GSDMD sensitizes HCC to anti-PD-1 by activating cGAS pathway and downregulating PD-L1 expression. J ImmunoTher Cancer 2022;10(6):e004763.
|
[[83]] |
Yu M, Chen Z, Zhou Q, et al. PARG inhibition limits HCC progression and potentiates the efficacy of immune checkpoint therapy. J Hepatol 2022;77(1):140-151.
|
[[84]] |
Xiao Y, Chen J, Zhou H, et al. Combining p53 mRNA nanotherapy with immune checkpoint blockade reprograms the immune microenvironment for effective cancer therapy. Nat Commun 2022;13(1):758.
|
[[85]] |
Finn RS, Qin S, Ikeda M, et al. IMbrave150 Investigators. Atezolizumab plus Bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med 2020;382(20):1894-1905.
|
[[86]] |
Galle PR, Finn RS, Qin S, et al. Patient-reported outcomes with atezolizumab plus bevacizumab versus sorafenib in patients with unresectable hepatocellular carcinoma (IMbrave150): an open-label, randomised, phase 3 trial. Lancet Oncol 2021;22(7):991-1001.
|
[[87]] |
Yau T, Kang YK, Kim TY, 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(11):e204564.
|
[[88]] |
Finn RS, Ryoo BY, Merle P, et al. KEYNOTE-240 Investigators. 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(3):193-202.
|
[[89]] |
Zhu AX, Finn RS, Edeline J, et al. KEYNOTE-224 Investigators. 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(7):940-952.
|
[[90]] |
Finn RS, Ikeda M, Zhu AX, et al. Phase Ib study of lenvatinib plus pembrolizumab in patients with unresectable hepatocellular carcinoma. J Clin Oncol 2020;38(26):2960-2970.
|
[[91]] |
Li H, Qin S, Liu Y, et al. Camrelizumab combined with FOLFOX4 regimen as first-line therapy for advanced hepatocellular carcinomas: a sub-cohort of a multicenter phase Ib/II study. Drug Des Devel Ther 2021;15:1873-1882.
|
[[92]] |
Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature 2011;480(7378):480-489.
|
/
〈 | 〉 |