882
Views
24
CrossRef citations to date
0
Altmetric
Review

Novel immunotherapeutic approaches for hepatocellular carcinoma treatment

, , , , &
Pages 453-470 | Received 06 Feb 2019, Accepted 20 Mar 2019, Published online: 14 Apr 2019

References

  • Llovet JM, Zucman-Rossi J, Pikarsky E, et al. Hepatocellular carcinoma. Nat Rev Dis Primer. 2016;2:16018.
  • European Association For The Study Of The Liver, European Organisation For Research And Treatment Of Cancer. EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2012;56:908–943.
  • Lumsden AB, Henderson JM, Kutner MH. Endotoxin levels measured by a chromogenic assay in portal, hepatic and peripheral venous blood in patients with cirrhosis. Hepatology. 1988;8:232–236.
  • Jenne CN, Kubes P. Immune surveillance by the liver. Nat Immunol. 2013;14:996–1006.
  • Protzer U, Maini MK, Knolle PA. Living in the liver: hepatic infections. Nat Rev Immunol. 2012;12:201–213.
  • Robinson MW, Harmon C, O’Farrelly C. Liver immunology and its role in inflammation and homeostasis. Cell Mol Immunol. 2016;13:267–276.
  • Heymann F, Peusquens J, Ludwig-Portugall I, et al. Liver inflammation abrogates immunological tolerance induced by Kupffer cells. Hepatology. 2015;62:279–291.
  • Daher S, Massarwa M, Benson AA, et al. Current and future treatment of hepatocellular carcinoma: an updated comprehensive review. J Clin Transl Hepatol. 2018;6:69–78.
  • 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 Lond Engl. 2017;389:2492–2502.
  • Cancer Genome Atlas Research Network. Electronic address: [email protected], Cancer Genome Atlas Research Network. Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell. 2017;169:1327–1341.
  • Ringelhan M, Pfister D, O’Connor T, et al. The immunology of hepatocellular carcinoma. Nat Immunol. 2018;19:222–232.
  • Ishiguro T, Sugimoto M, Kinoshita Y, et al. Anti-glypican 3 antibody as a potential antitumor agent for human liver cancer. Cancer Res. 2008;68:9832–9838.
  • Yong CSM, Dardalhon V, Devaud C, et al. CAR T-cell therapy of solid tumors. Immunol Cell Biol. 2017;95:356–363.
  • Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378:439–448.
  • Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large b-cell lymphoma. N Engl J Med. 2017;377:2531–2544.
  • Hu Z, Ott PA, Wu CJ. Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat Rev Immunol. 2017;18:168–182.
  • Consortium HEPAVAC, Buonaguro L. Developments in cancer vaccines for hepatocellular carcinoma. Cancer Immunol Immunother. 2016;65:93–99.
  • Buonaguro L, Mayer-Mokler A, Accolla R, et al. HepaVac-101 first-in-man therapeutic cancer vaccine phase I/II clinical trial for hepatocellular carcinoma patients. Cancer Immunol Immunother. 2018;36:TPS3135–TPS3135.
  • Weiner GJ. Building better monoclonal antibody-based therapeutics. Nat Rev Cancer. 2015;15:361–370.
  • Xu F, Jin T, Zhu Y, et al. Immune checkpoint therapy in liver cancer. J Exp Clin Cancer Res. 2018;37:110.
  • Bhandaru M, Rotte A. Blockade of programmed cell death protein-1 pathway for the treatment of melanoma. J Dermatol Res Ther. 2017;1:1–11.
  • Iñarrairaegui M, Melero I, Sangro B. Immunotherapy of hepatocellular carcinoma: facts and hopes. Clin Cancer Res. 2018;24:1518–1524.
  • Stone JD, Chervin AS, Kranz DM. T-cell receptor binding affinities and kinetics: impact on T-cell activity and specificity. Immunology. 2009;126:165–176.
  • Krummel MF, Allison JP. CTLA-4 engagement inhibits IL-2 accumulation and cell cycle progression upon activation of resting T cells. J Exp Med. 1996;183:2533–2540.
  • Chambers CA, Kuhns MS, Egen JG, et al. CTLA-4-mediated inhibition in regulation of T cell responses: mechanisms and manipulation in tumor immunotherapy. Annu Rev Immunol. 2001;19:565–594.
  • Collins AV, Brodie DW, Gilbert RJC, et al. The interaction properties of costimulatory molecules revisited. Immunity. 2002;17:201–210.
  • Fallarino F, Fields PE, Gajewski TF. B7–1 engagement of cytotoxic T lymphocyte antigen 4 inhibits T cell activation in the absence of CD28. J Exp Med. 1998;188:205–210.
  • Masteller EL, Chuang E, Mullen AC, et al. Structural analysis of CTLA-4 function in vivo. J Immunol. 2000;164:5319–5327.
  • Krummel MF, Allison JP. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med. 1995;182:459–465.
  • Balzano C, Buonavista N, Rouvier E, et al. CTLA-4 and CD28: similar proteins, neighbouring genes. Int J Cancer. 1992;7:28–32.
  • Read S, Malmström V, Powrie F. Cytotoxic T lymphocyte–associated antigen 4 plays an essential role in the function of cd25+cd4+ regulatory cells that control intestinal inflammation. J Exp Med. 2000;192:295–302.
  • Linsley PS, Bradshaw J, Greene J, et al. Intracellular trafficking of CTLA-4 and focal localization towards sites of TCR engagement. Immunity. 1996;4:535–543.
  • Fife BT, Bluestone JA. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunol Rev. 2008;224:166–182.
  • Egen JG, Kuhns MS, Allison JP. CTLA-4: new insights into its biological function and use in tumor immunotherapy. Nat Immunol. 2002;3:611–618.
  • Schneider H, Downey J, Smith A, et al. Reversal of the TCR stop signal by CTLA-4. Science. 2006;313:1972–1975.
  • Han Y, Chen Z, Yang Y, et al. Human CD14+CTLA‐4+ regulatory dendritic cells suppress T‐cell response by cytotoxic T‐lymphocyte antigen‐4‐dependent IL‐10 and indoleamine‐2,3‐dioxygenase production in hepatocellular carcinoma. Hepatology. 2014;59:567–579.
  • Ishida Y, Agata Y, Shibahara K, et al. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. Embo J. 1992;11:3887–3895.
  • Keir ME, Butte MJ, Freeman GJ, et al. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704.
  • Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol Rev. 2010;236:219–242.
  • Cheng X, Veverka V, Radhakrishnan A, et al. Structure and interactions of the human programmed cell death 1 receptor. J Biol Chem. 2013;288:11771–11785.
  • Hui E, Cheung J, Zhu J, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science. 2017;355:1428–1433.
  • Rotte A, Jin JY, Lemaire V. Mechanistic overview of immune checkpoints to support the rational design of their combinations in cancer immunotherapy. Ann Oncol. 2018;29:71–83.
  • Latchman Y, Wood CR, Chernova T, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2:261–268.
  • Francisco LM, Salinas VH, Brown KE, et al. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med. 2009;206:3015–3029.
  • Butte MJ, Keir ME, Phamduy TB, et al. Programmed death-1 ligand 1 interacts specifically with the B7–1 costimulatory molecule to inhibit T cell responses. Immunity. 2007;27:111–122.
  • Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12:492–499.
  • Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–264.
  • Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14:847–856.
  • Wu K, Kryczek I, Chen L, Zou W, Welling TH. Kupffer cell suppression of CD8+ T cells in human hepatocellular carcinoma is mediated by B7-H1/PD-1 interactions. Cancer Res. 2009;69:8067–8075.
  • Semaan A, Dietrich D, Bergheim D, et al. CXCL12 expression and PD-L1 expression serve as prognostic biomarkers in HCC and are induced by hypoxia. Virchows Arch. 2017;470:185–196.
  • Dai X, Xue J, Hu J, et al. Positive expression of programmed death ligand 1 in peritumoral liver tissue is associated with poor survival after curative resection of hepatocellular carcinoma. Transl Oncol. 2017;10:511–517.
  • Parry RV, Chemnitz JM, Frauwirth KA, et al. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol Cell Biol. 2005;25:9543–9553.
  • Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004;21:137–148.
  • Morse MA. Technology evaluation: ipilimumab, Medarex/Bristol-Myers Squibb. Curr Opin Mol Ther. 2005;7:588–597.
  • Jain S, Clark JI. Ipilimumab for the treatment of melanoma. Melanoma Manag. 2015;2:33–39.
  • Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–723.
  • Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364:2517–2526.
  • Eggermont AMM, Chiarion-Sileni V, Grob -J-J, et al. Adjuvant ipilimumab versus placebo after complete resection of high-risk stage III melanoma (EORTC 18071): a randomised, double-blind, phase 3 trial. Lancet Oncol. 2015;16:522–530.
  • Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373:23–34.
  • Motzer RJ, Tannir NM, McDermott DF, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378:1277–1290.
  • Kaseb AO, Carmagnani Pestana R, Vence LM, et al. Randomized, open-label, perioperative phase II study evaluating nivolumab alone versus nivolumab plus ipilimumab in patients with resectable HCC. J Clin Oncol. 2019;37:185.
  • Hanson DC, Canniff PC, Primiano MJ, et al. Preclinical in vitro characterization of anti-CTLA4 therapeutic antibody CP-675,206. Cancer Res. 2004;64:877.
  • ClinicalTrials.gov [Internet]. [ cited 2019 Mar 18]. Available from: https://clinicaltrials.gov/ct2/results?cond=&term=tremelimumab&cntry=&state=&city=&dist
  • 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;59:81–88.
  • Duffy AG, Ulahannan SV, Makorova-Rusher O, et al. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J Hepatol. 2017;66:545–551.
  • Kelley RK, Abou-Alfa GK, Bendell JC, et al. Phase I/II study of durvalumab and tremelimumab in patients with unresectable hepatocellular carcinoma (HCC): phase I safety and efficacy analyses. J Clin Oncol. 2017;35:4073.
  • Floudas CS, Xie C, Brar G, et al. Combined immune checkpoint inhibition (ICI) with tremelimumab and durvalumab in patients with advanced hepatocellular carcinoma (HCC) or biliary tract carcinomas (BTC). J Clin Oncol. 2019;37:336.
  • Brahmer JR, Drake CG, Wollner I, 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. J Clin Oncol. 2010;28:3167–3175.
  • fda.gov. Approved drugs - FDA grants accelerated approval to nivolumab for HCC previously treated with sorafenib [Internet]. [ cited 2019 Mar 18]. Available from: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm577166.htm
  • Esteve M, Saro C, González-Huix F, et al. Chronic hepatitis B reactivation following infliximab therapy in Crohn’s disease patients: need for primary prophylaxis. Gut. 2004;53:1363–1365.
  • Patnaik A, Kang SP, Rasco D, et al. Phase I Study of pembrolizumab (MK-3475; anti–PD-1 monoclonal antibody) in patients with advanced solid tumors. Clin Cancer Res. 2015;21:4286–4293.
  • fda.gov. Drugs@FDA: FDA approved drug products [Internet]. [ cited 2019 Mar 18]. Available from: https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview.process&applno=125514
  • Robert C, Ribas A, Wolchok JD, et al. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet. 2014;384:1109–1117.
  • fda.gov. Approved drugs - FDA approves pembrolizumab for advanced cervical cancer with disease progression during or after chemotherapy [Internet]. [ cited 2019 Mar 18]. Available from: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm610572.htm
  • fda.gov. Approved drugs - FDA grants accelerated approval to pembrolizumab for hepatocellular carcinoma [Internet]. [ cited 2019 Mar 18]. Available from: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm625705.htm
  • Zhu AX, Finn RS, Edeline J, 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.
  • Ikeda M, Sung MW, Kudo M, et al. A phase 1b trial of lenvatinib (LEN) plus pembrolizumab (PEM) in patients (pts) with unresectable hepatocellular carcinoma (uHCC). J Clin Oncol. 2018;36:4076.
  • Chung VM, Kos F, Hardwick N, et al. A phase 1 study of p53MVA vaccine in combination with pembrolizumab. J Clin Oncol. 2018;36:206.
  • Gangadhar TC, Hamid O, Smith DC, et al. Preliminary results from a Phase I/II study of epacadostat (incb024360) in combination with pembrolizumab in patients with selected advanced cancers. J Immunother Cancer. 2015;3:O7.
  • Friedlander M, Meniawy T, Markman B, et al. A phase 1b study of the anti-PD-1 monoclonal antibody BGB-A317 (A317) in combination with the PARP inhibitor BGB-290 (290) in advanced solid tumors. J Clin Oncol. 2018;36:48.
  • Zhang T, Song X, Xu L, et al. The binding of an anti-PD-1 antibody to FcγRΙ has a profound impact on its biological functions. Cancer Immunol Immunother. 2018;67:1079–1090.
  • Li Y, Li F, Jiang F, et al. A mini-review for cancer immunotherapy: molecular understanding of pd-1/pd-l1 pathway & translational blockade of immune checkpoints. Int J Mol Sci. 2016;17:1151.
  • Xu J, Zhang Y, Jia R, et al. Anti-PD-1 antibody shr-1210 combined with apatinib for advanced hepatocellular carcinoma, gastric, or esophagogastric junction cancer: an open-label, dose escalation and expansion study. Clin Cancer Res. 2019;25:515–523.
  • Stewart R, Morrow M, Hammond SA, et al. Identification and characterization of MEDI4736, an antagonistic anti–PD-L1 monoclonal antibody. Cancer Immunol Res. 2015;3:1052–1062.
  • Karzai F, VanderWeele D, Madan RA, et al. Activity of durvalumab plus olaparib in metastatic castration-resistant prostate cancer in men with and without DNA damage repair mutations. J Immunother Cancer. 2018;6:141.
  • Powles T, O’Donnell PH, Massard C, et al. Efficacy and safety of durvalumab in locally advanced or metastatic urothelial carcinoma: updated results from a phase 1/2 open-label study. JAMA Oncol. 2017;3:e172411.
  • fda.gov. Approved drugs - durvalumab (imfinzi) [Internet]. [ cited 2019 Mar 18]. Available from: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm555930.htm
  • Antonia SJ, Villegas A, Daniel D, et al. Durvalumab after chemoradiotherapy in stage iii non-small-cell lung cancer. N Engl J Med. 2017;377:1919–1929.
  • fda.gov. Approved Drugs - FDA approves durvalumab after chemoradiation for unresectable stage III NSCLC [Internet]. [ cited 2019 Mar 18]. Available from: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm597248.htm
  • Boyerinas B, Jochems C, Fantini M, et al. Antibody-dependent cellular cytotoxicity activity of a novel anti–PD-L1 antibody avelumab (MSB0010718C) on human tumor cells. Cancer Immunol Res. 2015;3:1148–1157.
  • Kaufman HL, Russell J, Hamid O, et al. Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial. Lancet Oncol. 2016;17:1374–1385.
  • fda.gov. Approved drugs - avelumab (BAVENCIO) [Internet]. [ cited 2019 Mar 18]. Available from: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm547965.htm
  • Apolo AB, Infante JR, Balmanoukian A, et al. Avelumab, an anti–programmed death-ligand 1 antibody, in patients with refractory metastatic urothelial carcinoma: results from a multicenter, phase ib study. J Clin Oncol. 2017;35:2117–2124.
  • fda.gov. Approved drugs - FDA grants accelerated approval to avelumab for urothelial carcinoma [Internet]. [ cited 2019 Mar 18]. Available from: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm557162.htm
  • Shah NJ, Kelly WJ, Liu SV, et al. Product review on the Anti-PD-L1 antibody atezolizumab. Hum Vaccines Immunother. 2018;14:269–276.
  • Herbst RS, Soria J-C, Kowanetz M, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–567.
  • fda.gov. Press announcements - FDA approves new, targeted treatment for bladder cancer [Internet]. [ cited 2019 Mar 18]. Available from: https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm501762.htm
  • Rosenberg JE, Hoffman-Censits J, Powles T, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet Lond Engl. 2016;387:1909–1920.
  • fda.gov. Approved drugs - FDA approves atezolizumab with chemotherapy and bevacizumab for first-line treatment of metastatic non-squamous NSCLC [Internet]. [ cited 2019 Mar 18]. Available from: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm627874.htm
  • Yamashita T, Forgues M, Wang W, et al. EpCAM and alpha-fetoprotein expression defines novel prognostic subtypes of hepatocellular carcinoma. Cancer Res. 2008;68:1451–1461.
  • Abelev GI. Alpha-fetoprotein in ontogenesis and its association with malignant tumors. Adv Cancer Res. 1971;14:295–358.
  • Thimme R, Neagu M, Boettler T, et al. Comprehensive analysis of the alpha-fetoprotein-specific CD8+ T cell responses in patients with hepatocellular carcinoma. Hepatology. 2008;48:1821–1833.
  • Korangy F, Ormandy LA, Bleck JS, et al. Spontaneous tumor-specific humoral and cellular immune responses to NY-ESO-1 in hepatocellular carcinoma. Clin Cancer Res. 2004;10:4332–4341.
  • Bricard G, Bouzourene H, Martinet O, et al. Naturally acquired MAGE-A10- and SSX-2-specific CD8+ T cell responses in patients with hepatocellular carcinoma. J Immunol. 2005;174:1709–1716.
  • Kerzerho J, Adotevi O, Castelli FA, et al. The angiogenic growth factor and biomarker midkine is a tumor-shared antigen. J Immunol. 2010;185:418–423.
  • Jia H-L, Ye Q-H, Qin L-X, et al. Gene expression profiling reveals potential biomarkers of human hepatocellular carcinoma. Clin Cancer Res. 2007;13:1133–1139.
  • Lin Y-S, Jung S-M, Yeh C-N, et al. MUC1, MUC2 and MUC5AC expression in hepatocellular carcinoma with cardiac metastasis. Mol Med Rep. 2009;2:291–294.
  • Yuan S-F, Li K-Z, Wang L, et al. Expression of MUC1 and its significance in hepatocellular and cholangiocarcinoma tissue. World J Gastroenterol. 2005;11:4661–4666.
  • Sawabu N, Wakabayashi T, Ozaki K, et al. Serum tumor markers in patients with hepatocellular carcinoma: diagnosis of alpha-fetoprotein -low or -negative patients. Gastroenterol Jpn. 1985;20:209–215.
  • Yoshikawa M, Morine Y, Ikemoto T, et al. Elevated preoperative serum CEA level is associated with poor prognosis in patients with hepatocellular carcinoma through the epithelial-mesenchymal transition. Anticancer Res. 2017;37:1169–1175.
  • Ogawa K, Tanaka S, Matsumura S, et al. EpCAM-targeted therapy for human hepatocellular carcinoma. Ann Surg Oncol. 2014;21:1314–1322.
  • Yamashita T, Ji J, Budhu A, et al. EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features. Gastroenterology. 2009;136:1012–1024.
  • Ross JS, Sheehan CE, Fisher HAG, et al. Correlation of primary tumor prostate-specific membrane antigen expression with disease recurrence in prostate cancer. Clin Cancer Res. 2003;9:6357–6362.
  • Capurro M, Wanless IR, Sherman M, et al. Glypican-3: a novel serum and histochemical marker for hepatocellular carcinoma. Gastroenterology. 2003;125:89–97.
  • Zhu Z-W, Friess H, Wang L, et al. Enhanced glypican-3 expression differentiates the majority of hepatocellular carcinomas from benign hepatic disorders. Gut. 2001;48:558–564.
  • Hsu HC, Cheng W, Lai PL. Cloning and expression of a developmentally regulated transcript MXR7 in hepatocellular carcinoma: biological significance and temporospatial distribution. Cancer Res. 1997;57:5179–5184.
  • Anatelli F, Chuang S-T, Yang XJ, et al. Value of glypican 3 immunostaining in the diagnosis of hepatocellular carcinoma on needle biopsy. Am J Clin Pathol. 2008;130:219–223.
  • Zhang L, Liu H, Sun L, et al. Glypican-3 as a potential differential diagnosis marker for hepatocellular carcinoma: a tissue microarray-based study. Acta Histochem. 2012;114:547–552.
  • Yamauchi N, Watanabe A, Hishinuma M, et al. The glypican 3 oncofetal protein is a promising diagnostic marker for hepatocellular carcinoma. Mod Pathol. 2005;18:1591–1598.
  • Valdez Y, Maia M, Conway EM. CD248: reviewing its role in health and disease. Curr Drug Targets. 2012;13:432–439.
  • Tomkowicz B, Rybinski K, Foley B, et al. Interaction of endosialin/TEM1 with extracellular matrix proteins mediates cell adhesion and migration. Proc Natl Acad Sci U S A. 2007;104:17965–17970.
  • Becker R, Lenter MC, Vollkommer T, et al. Tumor stroma marker endosialin (Tem1) is a binding partner of metastasis-related protein Mac-2 BP/90K. FASEB J. 2008;22:3059–3067.
  • Nanda A, Karim B, Peng Z, et al. Tumor endothelial marker 1 (Tem1) functions in the growth and progression of abdominal tumors. Proc Natl Acad Sci U S A. 2006;103:3351–3356.
  • Lee JH, Lee S-W. The roles of carcinoembryonic antigen in liver metastasis and therapeutic approaches. Gastroenterol Res Pract. 2017;2017:7521987.
  • Sasikumar A, Joy A, Nanabala R, et al. (68)Ga-PSMA PET/CT imaging in primary hepatocellular carcinoma. Eur J Nucl Med Mol Imaging. 2016;43:795–796.
  • Huang HL, Zhen Loh TJ, Hoe Chow PK. A case of well-differentiated hepatocellular carcinoma identified on gallium-68 prostate-specific membrane antigen positron emission tomography/computed tomography. World J Nucl Med. 2018;17:102–105.
  • Frigerio B, Fracasso G, Luison E, et al. A single-chain fragment against prostate specific membrane antigen as a tool to build theranostic reagents for prostate cancer. Eur J Cancer Oxf Engl 1990. 2013;49:2223–2232.
  • Zuccolotto G, Fracasso G, Merlo A, et al. PSMA-specific CAR-engineered T cells eradicate disseminated prostate cancer in preclinical models. PloS One. 2014;9:e109427.
  • Filmus J, Capurro M, Rast J. Glypicans. Genome Biol. 2008;9:224.
  • De Cat B, Muyldermans S-Y, Coomans C, et al. Processing by proprotein convertases is required for glypican-3 modulation of cell survival, Wnt signaling, and gastrulation movements. J Cell Biol. 2003;163:625–635.
  • Filmus J. Glypicans in growth control and cancer. Glycobiology. 2001;11:19R–23R.
  • Filmus J, Selleck SB. Glypicans: proteoglycans with a surprise. J Clin Invest. 2001;108:497–501.
  • Gallagher JT. Heparan sulfate: growth control with a restricted sequence menu. J Clin Invest. 2001;108:357–361.
  • Capurro MI, Xu P, Shi W, et al. Glypican-3 inhibits Hedgehog signaling during development by competing with patched for Hedgehog binding. Dev Cell. 2008;14:700–711.
  • Capurro MI, Xiang -Y-Y, Lobe C, et al. Glypican-3 promotes the growth of hepatocellular carcinoma by stimulating canonical Wnt signaling. Cancer Res. 2005;65:6245–6254.
  • Song HH, Shi W, Xiang -Y-Y, et al. the loss of glypican-3 induces alterations in Wnt signaling. J Biol Chem. 2005;280:2116–2125.
  • Zittermann SI, Capurro MI, Shi W, et al. Soluble glypican 3 inhibits the growth of hepatocellular carcinoma in vitro and in vivo. Int J Cancer. 2010;126:1291–1301.
  • Capurro M, Martin T, Shi W, et al. Glypican-3 binds to Frizzled and plays a direct role in the stimulation of canonical Wnt signaling. J Cell Sci. 2014;127:1565–1575.
  • Liu H. Diagnostic value of glypican-3 in serum and liver for primary hepatocellular carcinoma. World J Gastroenterol. 2010;16:4410.
  • Christian S, Ahorn H, Koehler A, et al. Molecular cloning and characterization of endosialin, a C-type lectin-like cell surface receptor of tumor endothelium. J Biol Chem. 2001;276:7408–7414.
  • Christian S, Winkler R, Helfrich I, et al. Endosialin (Tem1) is a marker of tumor-associated myofibroblasts and tumor vessel-associated mural cells. Am J Pathol. 2008;172:486–494.
  • Bagley RG, Weber W, Rouleau C, et al. Human mesenchymal stem cells from bone marrow express tumor endothelial and stromal markers. Int J Oncol. 2009;34:619–627.
  • Bagley RG, Rouleau C, St Martin T, et al. Human endothelial precursor cells express tumor endothelial marker 1/endosialin/CD248. Mol Cancer Ther. 2008;7:2536–2546.
  • Rettig WJ, Garin-Chesa P, Healey JH, et al. Identification of endosialin, a cell surface glycoprotein of vascular endothelial cells in human cancer. Proc Natl Acad Sci U S A. 1992;89:10832–10836.
  • Zhu AX, Gold PJ, El-Khoueiry AB, et al. First-in-man phase I study of GC33, a novel recombinant humanized antibody against glypican-3, in patients with advanced hepatocellular carcinoma. Clin Cancer Res. 2013;19:920–928.
  • Abou-Alfa GK, Puig O, Daniele B, et al. Randomized phase II placebo controlled study of codrituzumab in previously treated patients with advanced hepatocellular carcinoma. J Hepatol. 2016;65:289–295.
  • Abou-Alfa GK, Yen C-J, Hsu C-H, et al. Phase Ib study of codrituzumab in combination with sorafenib in patients with non-curable advanced hepatocellular carcinoma (HCC). Cancer Chemother Pharmacol. 2017;79:421–429.
  • Phung Y, Gao W, Man Y-G, et al. High-affinity monoclonal antibodies to cell surface tumor antigen glypican-3 generated through a combination of peptide immunization and flow cytometry screening. mAbs. 2012;4:592–599.
  • Zhang Y-F, Ho M. Humanization of high-affinity antibodies targeting glypican-3 in hepatocellular carcinoma. Sci Rep. 2016;6:33878.
  • Feng M, Gao W, Wang R, et al. Therapeutically targeting glypican-3 via a conformation-specific single-domain antibody in hepatocellular carcinoma. Proc Natl Acad Sci U S A. 2013;110:E1083–91.
  • Diaz LA, Coughlin CM, Weil SC, et al. A first-in-human phase I study of MORAb-004, a monoclonal antibody to endosialin in patients with advanced solid tumors. Clin Cancer Res. 2015;21:1281–1288.
  • Doi T, Aramaki T, Yasui H, et al. A phase I study of ontuxizumab, a humanized monoclonal antibody targeting endosialin, in Japanese patients with solid tumors. Invest New Drugs. 2019.
  • Hoseini SS, Cheung N-KV. Immunotherapy of hepatocellular carcinoma using chimeric antigen receptors and bispecific antibodies. Cancer Lett. 2017;399:44–52.
  • Mackall CL, Merchant MS, Fry TJ. Immune-based therapies for childhood cancer. Nat Rev Clin Oncol. 2014;11:693–703.
  • Yu S, Li A, Liu Q, et al. Recent advances of bispecific antibodies in solid tumors. J Hematol Oncol. 2017;10:155.
  • May C, Sapra P, Gerber H-P. Advances in bispecific biotherapeutics for the treatment of cancer. Biochem Pharmacol. 2012;84:1105–1112.
  • Suryadevara CM, Gedeon PC, Sanchez-Perez L, et al. Are BiTEs the “missing link” in cancer therapy? OncoImmunology. 2015;4:e1008339.
  • Löffler A, Kufer P, Lutterbüse R, et al. A recombinant bispecific single-chain antibody, CD19 × CD3, induces rapid and high lymphoma-directed cytotoxicity by unstimulated T lymphocytes. Blood. 2000;95:2098–2103.
  • Wu J, Fu J, Zhang M, et al. Blinatumomab: a bispecific T cell engager (BiTE) antibody against CD19/CD3 for refractory acute lymphoid leukemia. J Hematol Oncol. 2015;8:104.
  • fda.gov. Approved Drugs - FDA grants regular approval to blinatumomab and expands indication to include Philadelphia chromosome-positive B cell [Internet]. [ cited 2019 Mar 18]. Available from: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm566708.htm
  • fda.gov. Approved drugs - FDA granted accelerated approval to blinatumomab (Blincyto, Amgen Inc.) for the treatment of adult and pediatric patients with B-cell precursor acute lymphoblastic leukemia [Internet]. [ cited 2019 Mar 18]. Available from: https://www.fda.gov/drugs/informationondrugs/approveddrugs/ucm603171.htm
  • Goéré D, Flament C, Rusakiewicz S, et al. Potent immunomodulatory effects of the trifunctional antibody catumaxomab. Cancer Res. 2013;73:4663–4673.
  • ema.europa.eu. Removab [Internet]. 2018 [cited 2019 Mar 18]. Available from: https://www.ema.europa.eu/en/medicines/human/EPAR/removab
  • Ishiguro T, Sano Y, Komatsu S, et al. An anti–glypican 3/CD3 bispecific T cell–redirecting antibody for treatment of solid tumors. Sci Transl Med. 2017;9:eaal4291.
  • Hedvat M, Bonzon C, Bernett MJ, et al. Abstract 2784: simultaneous checkpoint-checkpoint or checkpoint-costimulatory receptor targeting with bispecific antibodies promotes enhanced human T cell activation. Cancer Res. 2018;78:2784.
  • Yamashita T, Budhu A, Forgues M, et al. Activation of hepatic stem cell marker EpCAM by Wnt–β-catenin signaling in hepatocellular carcinoma. Cancer Res. 2007;67:10831–10839.
  • Zhang P, Shi B, Gao H, et al. An EpCAM/CD3 bispecific antibody efficiently eliminates hepatocellular carcinoma cells with limited galectin-1 expression. Cancer Immunol Immunother. 2014;63:121–132.
  • Beck A, Goetsch L, Dumontet C, et al. Strategies and challenges for the next generation of antibody–drug conjugates. Nat Rev Drug Discov. 2017;16:315–337.
  • Lambert JM, Morris CQ. Antibody–drug conjugates (ADCs) for personalized treatment of solid tumors: a review. Adv Ther. 2017;34:1015–1035.
  • Fu Y, Urban DJ, Nani RR, et al. Glypican-3-specific antibody drug conjugates targeting hepatocellular carcinoma. Hepatology. 2018. Epub ahead of print.
  • fda.gov. Approved Drugs - FDA approves inotuzumab ozogamicin for relapsed or refractory B-cell precursor ALL [Internet]. [ cited 2019 Mar 18]. Available from: https://www.fda.gov/drugs/informationondrugs/approveddrugs/ucm572133.htm
  • Cardillo TM, Govindan SV, Sharkey RM, et al. Humanized anti-trop-2 IgG-SN-38 conjugate for effective treatment of diverse epithelial cancers: preclinical studies in human cancer xenograft models and monkeys. Clin Cancer Res. 2011;17:3157–3169.
  • Pastan I, Hassan R, FitzGerald DJ, et al. Immunotoxin therapy of cancer. Nat Rev Cancer. 2006;6:559–565.
  • Batra JK, Kasprzyk PG, Bird RE, et al. Recombinant anti-erbB2 immunotoxins containing Pseudomonas exotoxin. Proc Natl Acad Sci U S A. 1992;89:5867–5871.
  • Hassan R, Alewine C, Pastan I. New life for immunotoxin cancer therapy. Clin Cancer Res. 2016;22:1055–1058.
  • Weldon JE, Xiang L, Zhang J, et al. A recombinant immunotoxin against the tumor-associated antigen mesothelin reengineered for high activity, low off-target toxicity, and reduced antigenicity. Mol Cancer Ther. 2013;12:48–57.
  • Liu W, Onda M, Lee B, et al. Recombinant immunotoxin engineered for low immunogenicity and antigenicity by identifying and silencing human B-cell epitopes. Proc Natl Acad Sci U S A. 2012;109:11782–11787.
  • Onda M, Nagata S, FitzGerald DJ, et al. Characterization of the B cell epitopes associated with a truncated form of pseudomonas exotoxin (PE38) used to make immunotoxins for the treatment of cancer patients. J Immunol. 2006;177:8822–8834.
  • Schmohl JU, Todhunter D, Oh S, et al. Mutagenic deimmunization of diphtheria toxin for use in biologic drug development. Toxins (Basel). 2015;7:4067–4082.
  • Schmohl JU, Todhunter D, Taras E, et al. Development of a deimmunized bispecific immunotoxin dDT2219 against B-cell malignancies. Toxins (Basel). 2018;10:32.
  • Mazor R, Zhang J, Xiang L, et al. Recombinant immunotoxin with T cell epitope mutations that greatly reduce immunogenicity for treatment of mesothelin expressing tumors. Mol Cancer Ther. 2015;14:2789–2796.
  • Kreitman RJ, Pastan I. Accumulation of a recombinant immunotoxin in a tumor in vivo: fewer than 1000 molecules per cell are sufficient for complete responses. Cancer Res. 1998;58:968–975.
  • Alewine C, Xiang L, Yamori T, et al. Efficacy of RG7787, a next generation mesothelin-targeted immunotoxin, against triple-negative breast and gastric cancers. Mol Cancer Ther. 2014;13:2653–2661.
  • Hollevoet K, Mason-Osann E, Liu X, et al. In vitro and in vivo activity of the low-immunogenic anti-mesothelin immunotoxin RG7787 in pancreatic cancer. Mol Cancer Ther. 2014;13:2040–2049.
  • Moskowitz CH, Nademanee A, Masszi T, et al. Brentuximab vedotin as consolidation therapy after autologous stem-cell transplantation in patients with Hodgkin’s lymphoma at risk of relapse or progression (AETHERA): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2015;385:1853–1862.
  • Pai-Scherf LH, Villa J, Pearson D, et al. Hepatotoxicity in cancer patients receiving erb-38, a recombinant immunotoxin that targets the erbb2 receptor. Clin Cancer Res. 1999;5:2311–2315.
  • Gao W, Tang Z, Zhang Y, et al. Immunotoxin targeting glypican-3 regresses liver cancer via dual inhibition of Wnt signaling and protein synthesis. Nat Commun. 2015;6:6536.
  • Wang C, Gao W, Feng M, et al. Construction of an immunotoxin, HN3-mPE24, targeting glypican-3 for liver cancer therapy. Oncotarget. 2016;8:32450–32460.
  • Weldon JE, Xiang L, Chertov O, et al. A protease-resistant immunotoxin against CD22 with greatly increased activity against CLL and diminished animal toxicity. Blood. 2009;113:3792–3800.
  • June CH, O’Connor RS, Kawalekar OU, et al. CAR T cell immunotherapy for human cancer. Science. 2018;359:1361–1365.
  • Pang Y, Hou X, Yang C, et al. Advances on chimeric antigen receptor-modified T-cell therapy for oncotherapy. Mol Cancer. 2018;17:91.
  • Kalos M, June CH. Adoptive T cell transfer for cancer immunotherapy in the era of synthetic biology. Immunity. 2013;39:49–60.
  • fda.gov. Approved Drugs - FDA approves tisagenlecleucel for B-cell ALL and tocilizumab for cytokine release syndrome [Internet]. [ cited 2019 Mar 18]. Available from: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm574154.htm
  • Milone MC, Fish JD, Carpenito C, et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of t cells and increased antileukemic efficacy in vivo. Mol Ther. 2009;17:1453–1464.
  • Kochenderfer JN, Feldman SA, Zhao Y, et al. Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J Immunother. 1997;2009(32):689–702.
  • fda.gov. Approved drugs - FDA approves axicabtagene ciloleucel for large B-cell lymphoma [Internet]. [ cited 2019 Mar 18]. Available from: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm581296.htm
  • Gao H, Li K, Tu H, et al. Development of T cells redirected to glypican-3 for the treatment of hepatocellular carcinoma. Clin Cancer Res. 2014;20:6418–6428.
  • Burga RA, Thorn M, Point GR, et al. Liver myeloid-derived suppressor cells expand in response to liver metastases in mice and inhibit the anti-tumor efficacy of anti-CEA CAR-T. Cancer Immunol Immunother. 2015;64:817–829.
  • Katz SC, Burga RA, McCormack E, et al. Phase I Hepatic Immunotherapy for Metastases study of intra-arterial chimeric antigen receptor modified T cell therapy for CEA+ liver metastases. Clin Cancer Res. 2015;21:3149–3159.
  • Jiang Z, Jiang X, Chen S, et al. Anti-GPC3-CAR T cells suppress the growth of tumor cells in patient-derived xenografts of hepatocellular carcinoma. Front Immunol. 2016;7:690.
  • Zhang Q, Zhang Z, Peng M, et al. CAR-T cell therapy in gastrointestinal tumors and hepatic carcinoma: from bench to bedside. Oncoimmunology. 2016;5:e1251539.
  • Li K, Lan Y, Wang J, et al. Chimeric antigen receptor–engineered T cells for liver cancers, progress and obstacles. Tumor Biol. 2017;39:1010428317692229.
  • Zhai B, Shi D, Gao H, et al. A phase I study of anti-GPC3 chimeric antigen receptor modified T cells (GPC3 CAR-T) in Chinese patients with refractory or relapsed GPC3+ hepatocellular carcinoma (r/r GPC3+ HCC). J Clin Oncol. 2017;35:3049.
  • Guo C, Manjili MH, Subjeck JR, et al. Therapeutic cancer vaccines: past, present and future. Adv Cancer Res. 2013;119:421–475.
  • Boisguérin V, Castle JC, Loewer M, et al. Translation of genomics-guided RNA-based personalised cancer vaccines: towards the bedside. Br J Cancer. 2014;111:1469–1475.
  • Prieto J, Melero I, Sangro B. Immunological landscape and immunotherapy of hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2015;12:681–700.
  • cancer.gov. Definition of mRNA-based personalized cancer vaccine NCI-4650 - NCI drug dictionary - national cancer institute [Internet]. [ cited 2019 Mar 18]. Available from: https://www.cancer.gov/publications/dictionaries/cancer-drug/def/795447
  • Schlom J. Therapeutic cancer vaccines: current status and moving forward. J Natl Cancer Inst. 2012;104:599–613.
  • Niethammer AG, Xiang R, Becker JC, et al. A DNA vaccine against VEGF receptor 2 prevents effective angiogenesis and inhibits tumor growth. Nat Med. 2002;8:1369–1375.
  • Ishizaki H, Tsunoda T, Wada S, et al. Inhibition of tumor growth with antiangiogenic cancer vaccine using epitope peptides derived from human vascular endothelial growth factor receptor 1. Clin Cancer Res. 2006;12:5841–5849.
  • Wada S, Tsunoda T, Baba T, et al. Rationale for antiangiogenic cancer therapy with vaccination using epitope peptides derived from human vascular endothelial growth factor receptor 2. Cancer Res. 2005;65:4939–4946.
  • Tang J, Yu JX, Hubbard-Lucey VM, et al. Trial watch: the clinical trial landscape for PD1/PDL1 immune checkpoint inhibitors. Nat Rev Drug Discov. 2018;17:854–855.
  • Gajewski TF, Schreiber H, Fu Y-X. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 2013;14:1014–1022.
  • Onyshchenko M. The puzzle of predicting response to immune checkpoint blockade. EBioMedicine. 2018;33:18–19.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.