Glypican-3 Targeting Immunotoxins for the Treatment of Liver Cancer
Abstract
:1. Introduction
2. Anti-GPC3 Recombinant Immunotoxins
3. Perspective and Future Directions
Acknowledgments
Conflicts of Interest
Abbreviations
ABC | ATP-Binding Cassette |
ADC | Antibody Drug Conjugate |
ADCC | Antibody Dependent Cellular Cytotoxicity |
ADP | Adenosine Diphosphate |
ALK | Activin Receptor-Like Kinase |
ARC | Antibody Radioisotope Conjugate |
CAR-T | Chimeric Antigen Receptor T Cell |
CD | Cluster of Differentiation |
CDC | Complement Dependent Cytotoxicity |
CTLA | Cytotoxic T-Lymphocyte Associated Protein |
EGFR | Epidermal Growth Factor Receptor |
EpCAM | Epithelial Cell Adhesion Molecule |
ERAD | Endoplasmic Reticulum-Associated Protein Degradation |
FDA | Food and Drug Administration |
Fv | Variable Fragment |
GPC3 | Glypican-3 |
HCC | Hepatocellular Carcinoma |
IGF(R) | Insulin-Like Growth Factor (Receptor) |
MHC | Major Histocompatibility Complex |
MMAE | Monomethyl Auristatin E |
Mono | Monoclonal |
mPE24 | Mutated PE24 |
mRNA | Messenger Ribonucleic Acid |
NCI | National Cancer Institute |
PDGFRA | Platelet-Derived Growth Factor Receptor Alpha Chain |
PD-1 | Programmed Cell Death Protein 1 |
PD-L1 | Programmed Cell Death Protein Ligand 1 |
PEG | Polyethylene Glycol |
PE38 | Pseudomonas Exotoxin A Domain II and Domain III |
PGF | Placental Growth Factor |
RIT | Recombinant Immunotoxin |
TCR | T-Cell Receptor |
VEGF(R) | Vascular Endothelial Growth Factor (Receptor) |
VH | Heavy Chain Variable Region |
References
- Sanchez, L.; Wang, Y.; Siegel, D.S.; Wang, M.L. Daratumumab: A first-in-class CD38 monoclonal antibody for the treatment of multiple myeloma. J. Hematol. Oncol. 2016, 9, 51. [Google Scholar] [CrossRef] [PubMed]
- Vacchelli, E.; Pol, J.; Bloy, N.; Eggermont, A.; Cremer, I.; Fridman, W.H.; Galon, J.; Marabelle, A.; Kohrt, H.; Zitvogel, L.; et al. Trial watch: Tumor-targeting monoclonal antibodies for oncological indications. Oncoimmunology 2015, 4, E985940. [Google Scholar] [CrossRef] [PubMed]
- Suzuki, S.; Ishida, T.; Yoshikawa, K.; Ueda, R. Current status of immunotherapy. Jpn. J. Clin. Oncol. 2016, 46, 191–203. [Google Scholar] [CrossRef] [PubMed]
- Bedognetti, D.; Maccalli, C.; Bader, S.B.; Marincola, F.M.; Seliger, B. Checkpoint inhibitors and their application in breast cancer. Breast Care 2016, 11, 108–115. [Google Scholar] [CrossRef] [PubMed]
- Todd, P.A.; Brogden, R.N. Muromonab CD3. A review of its pharmacology and therapeutic potential. Drugs 1989, 37, 871–899. [Google Scholar] [CrossRef] [PubMed]
- Gramatzki, M.; Burger, R.; Strobel, G.; Trautmann, U.; Bartram, C.R.; Helm, G.; Horneff, G.; Alsalameh, S.; Jonker, M.; Gebhart, E. Therapy with OKT3 monoclonal antibody in refractory T cell acute lymphoblastic leukemia induces interleukin-2 responsiveness. Leukemia 1995, 9, 382–390. [Google Scholar] [PubMed]
- Li, J.; Zhu, Z. Research and development of next generation of antibody-based therapeutics. Acta Pharmacol. Sin. 2010, 31, 1198–1207. [Google Scholar] [CrossRef] [PubMed]
- Maloney, D.G.; Grillo-López, A.J.; White, C.A.; Bodkin, D.; Schilder, R.J.; Neidhart, J.A.; Janakiraman, N.; Foon, K.A.; Liles, T.M.; Dallaire, B.K.; et al. IDEC-C2B8 (Rituximab) anti-CD20 monoclonal antibody therapy in patients with relapsed low-grade non-Hodgkin’s lymphoma. Blood 1997, 90, 2188–2195. [Google Scholar] [PubMed]
- Maloney, D.G.; Grillo-López, A.J.; Bodkin, D.J.; White, C.A.; Liles, T.M.; Royston, I.; Varns, C.; Rosenberg, J.; Levy, R. IDEC-C2B8: Results of a phase I multiple-dose trial in patients with relapsed non-Hodgkin’s lymphoma. J. Clin. Oncol. 1997, 15, 3266–3274. [Google Scholar] [PubMed]
- Maloney, D.G.; Liles, T.M.; Czerwinski, D.K.; Waldichuk, C.; Rosenberg, J.; Grillo-Lopez, A.; Levy, R. Phase I clinical trial using escalating single-dose infusion of chimeric anti-CD20 monoclonal antibody (IDEC-C2B8) in patients with recurrent B-cell lymphoma. Blood 1994, 84, 2457–2466. [Google Scholar] [PubMed]
- Reichert, J.M. Antibodies to watch in 2016. Mabs 2016, 8, 197–204. [Google Scholar] [CrossRef] [PubMed]
- Hoy, S.M. Dinutuximab: A review in high-risk neuroblastoma. Target. Oncol. 2016, 11, 247–253. [Google Scholar] [CrossRef] [PubMed]
- Lokhorst, H.M.; Plesner, T.; Laubach, J.P.; Nahi, H.; Gimsing, P.; Hansson, M.; Minnema, M.C.; Lassen, U.; Krejcik, J.; Palumbo, A.; et al. Targeting CD38 with daratumumab monotherapy in multiple myeloma. N. Engl. J. Med. 2015, 373, 1207–1219. [Google Scholar] [CrossRef] [PubMed]
- Zaza, G.; Tomei, P.; Granata, S.; Boschiero, L.; Lupo, A. Monoclonal antibody therapy and renal transplantation: Focus on adverse effects. Toxins 2014, 6, 869–891. [Google Scholar] [CrossRef] [PubMed]
- Winkler, U.; Jensen, M.; Manzke, O.; Schulz, H.; Diehl, V.; Engert, A. Cytokine-release syndrome in patients with B-cell chronic lymphocytic leukemia and high lymphocyte counts after treatment with an anti-CD20 monoclonal antibody (rituximab, IDEC-C2B8). Blood 1999, 94, 2217–2224. [Google Scholar] [PubMed]
- Baldo, B.A. Adverse events to monoclonal antibodies used for cancer therapy: Focus on hypersensitivity responses. Oncoimmunology 2013, 2, E26333. [Google Scholar] [CrossRef] [PubMed]
- Thomas, A.; Teicher, B.A.; Hassan, R. Antibody-drug conjugates for cancer therapy. Lancet Oncol. 2016, 17, E254–E262. [Google Scholar] [CrossRef]
- Zinzani, P.L.; Sasse, S.; Radford, J.; Gautam, A.; Bonthapally, V. Brentuximab vedotin in relapsed/refractory Hodgkin lymphoma: An updated review of published data from the named patient program. Crit. Rev. Oncol. Hematol. 2016, 104, 65–70. [Google Scholar] [CrossRef] [PubMed]
- Recondo, G.; de la Vega, M.; Galanternik, F.; Díaz-Cantón, E.; Leone, B.A.; Leone, J.P. Novel approaches to target HER2-positive breast cancer: Trastuzumab emtansine. Cancer Manag. Res. 2016, 8, 57–65. [Google Scholar] [PubMed]
- Bakhtiar, R. Antibody drug conjugates. Biotechnol. Lett. 2016, in press. [Google Scholar] [CrossRef] [PubMed]
- Prabhu, S.; Boswell, C.A.; Leipold, D.; Khawli, L.A.; Li, D.; Lu, D.; Theil, F.P.; Joshi, A.; Lum, B.L. Antibody delivery of drugs and radionuclides: Factors influencing clinical pharmacology. Ther. Deliv. 2011, 2, 769–791. [Google Scholar] [CrossRef] [PubMed]
- Almåsbak, H.; Aarvak, T.; Vemuri, M.C. CAR T cell therapy: A game changer in cancer treatment. J. Immunol. Res. 2016, 2016, 5474602. [Google Scholar] [CrossRef] [PubMed]
- Douglas, C.M.; Collier, R.J. Exotoxin A of Pseudomonas aeruginosa: Substitution of glutamic acid 553 with aspartic acid drastically reduces toxicity and enzymatic activity. J. Bacteriol. 1987, 169, 4967–4971. [Google Scholar] [PubMed]
- Weldon, J.E.; Pastan, I. A guide to taming a toxin-recombinant immunotoxins constructed from Pseudomonas exotoxin A for the treatment of cancer. FEBS J. 2011, 278, 4683–4700. [Google Scholar] [CrossRef] [PubMed]
- Wedekind, J.E.; Trame, C.B.; Dorywalska, M.; Koehl, P.; Raschke, T.M.; Mckee, M.; Fitzgerald, D.; Collier, R.J.; Mckay, D.B. Refined crystallographic structure of Pseudomonas aeruginosa exotoxin A and its implications for the molecular mechanism of toxicity. J. Mol. Biol. 2001, 314, 823–837. [Google Scholar] [CrossRef] [PubMed]
- Michalska, M.; Wolf, P. Pseudomonas exotoxin A: Optimized by evolution for effective killing. Front. Microbiol. 2015, 6, 963. [Google Scholar] [CrossRef] [PubMed]
- Kreitman, R.J.; Pastan, I. Antibody fusion proteins: Anti-CD22 recombinant immunotoxin moxetumomab pasudotox. Clin. Cancer Res. 2011, 17, 6398–6405. [Google Scholar] [CrossRef] [PubMed]
- Jackson, M.E.; Simpson, J.C.; Girod, A.; Pepperkok, R.; Roberts, L.M.; Lord, J.M. The KDEL retrieval system is exploited by Pseudomonas exotoxin A, but not by shiga-like toxin-1, during retrograde transport from the Golgi complex to the endoplasmic reticulum. J. Cell Sci. 1999, 112 Pt 4, 467–475. [Google Scholar] [PubMed]
- Kreitman, R.J.; Pastan, I. Importance of the glutamate residue of KDEL in increasing the cytotoxicity of Pseudomonas exotoxin derivatives and for increased binding to the KDEL receptor. Biochem. J. 1995, 307 Pt 1, 29–37. [Google Scholar] [CrossRef] [PubMed]
- Chaudhary, V.K.; Jinno, Y.; Fitzgerald, D.; Pastan, I. Pseudomonas exotoxin contains a specific sequence at the carboxyl terminus that is required for cytotoxicity. Proc. Natl. Acad. Sci. USA 1990, 87, 308–312. [Google Scholar] [CrossRef] [PubMed]
- Zdanovsky, A.G.; Chiron, M.; Pastan, I.; Fitzgerald, D.J. Mechanism of action of Pseudomonas exotoxin. Identification of a rate-limiting step. J. Biol. Chem. 1993, 268, 21791–21799. [Google Scholar] [PubMed]
- Koopmann, J.O.; Albring, J.; Hüter, E.; Bulbuc, N.; Spee, P.; Neefjes, J.; Hämmerling, G.J.; Momburg, F. Export of antigenic peptides from the endoplasmic reticulum intersects with retrograde protein translocation through the Sec61p channel. Immunity 2000, 13, 117–127. [Google Scholar] [CrossRef]
- Hazes, B.; Read, R.J. Accumulating evidence suggests that several AB-toxins subvert the endoplasmic reticulum-associated protein degradation pathway to enter target cells. Biochemistry 1997, 36, 11051–11054. [Google Scholar] [CrossRef] [PubMed]
- Lord, J.M.; Roberts, L.M.; Lencer, W.I. Entry of protein toxins into mammalian cells by crossing the endoplasmic reticulum membrane: Co-opting basic mechanisms of endoplasmic reticulum-associated degradation. Curr. Top. Microbiol. Immunol. 2005, 300, 149–168. [Google Scholar] [PubMed]
- Ogata, M.; Fryling, C.M.; Pastan, I.; Fitzgerald, D.J. Cell-mediated cleavage of Pseudomonas exotoxin between Arg279 and Gly280 generates the enzymatically active fragment which translocates to the cytosol. J. Biol. Chem. 1992, 267, 25396–25401. [Google Scholar] [PubMed]
- Fryling, C.; Ogata, M.; Fitzgerald, D. Characterization of a cellular protease that cleaves Pseudomonas exotoxin. Infect. Immun. 1992, 60, 497–502. [Google Scholar] [PubMed]
- Kreitman, R.J.; Wilson, W.H.; Bergeron, K.; Raggio, M.; Stetler-Stevenson, M.; Fitzgerald, D.J.; Pastan, I. Efficacy of the anti-CD22 recombinant immunotoxin BL22 in chemotherapy-resistant hairy-cell leukemia. N. Engl. J. Med. 2001, 345, 241–247. [Google Scholar] [CrossRef] [PubMed]
- Kreitman, R.J.; Tallman, M.S.; Robak, T.; Coutre, S.; Wilson, W.H.; Stetler-Stevenson, M.; Fitzgerald, D.J.; Lechleider, R.; Pastan, I. Phase I trial of anti-CD22 recombinant immunotoxin moxetumomab pasudotox (CAT-8015 or HA22) In patients with hairy cell leukemia. J. Clin. Oncol. 2012, 30, 1822–1828. [Google Scholar] [CrossRef] [PubMed]
- Kreitman, R.J.; Hassan, R.; Fitzgerald, D.J.; Pastan, I. Phase I trial of continuous infusion anti-mesothelin recombinant immunotoxin SS1P. Clin. Cancer Res. 2009, 15, 5274–5279. [Google Scholar] [CrossRef] [PubMed]
- Hassan, R.; Bullock, S.; Premkumar, A.; Kreitman, R.J.; Kindler, H.; Willingham, M.C.; Pastan, I. Phase I study of SS1P, a recombinant anti-mesothelin immunotoxin given as a bolus I.V. infusion to patients with mesothelin-expressing mesothelioma, ovarian, and pancreatic cancers. Clin. Cancer Res. 2007, 13, 5144–5149. [Google Scholar] [CrossRef] [PubMed]
- Hassan, R.; Sharon, E.; Thomas, A.; Zhang, J.; Ling, A.; Miettinen, M.; Kreitman, R.J.; Steinberg, S.M.; Hollevoet, K.; Pastan, I. Phase 1 study of the antimesothelin immunotoxin SS1P in combination with pemetrexed and cisplatin for front-line therapy of pleural mesothelioma and correlation of tumor response with serum mesothelin, megakaryocyte potentiating factor, and cancer antigen 125. Cancer 2014, 120, 3311–3319. [Google Scholar] [PubMed]
- Chandramohan, V.; Bao, X.; Keir, S.T.; Pegram, C.N.; Szafranski, S.E.; Piao, H.; Wikstrand, C.J.; Mclendon, R.E.; Kuan, C.T.; Pastan, I.H.; et al. Construction of an immunotoxin, D2C7-(scdsFv)-PE38KDEL, targeting EGFRwt and EGFRvIII for brain tumor therapy. Clin. Cancer Res. 2013, 19, 4717–4727. [Google Scholar] [CrossRef] [PubMed]
- Wei, X.; Juan, Z.X.; Min, F.X.; Nan, C.; Hua, Z.X.; Qing, F.Z.; Zheng, L. Recombinant immunotoxin anti-c-MET/PE38KDEL inhibits proliferation and promotes apoptosis of gastric cancer cells. J. Exp. Clin. Cancer Res. 2011, 30, 67. [Google Scholar] [CrossRef] [PubMed]
- Lv, M.; Qiu, F.; Li, T.; Sun, Y.; Zhang, C.; Zhu, P.; Qi, X.; Wan, J.; Yang, K.; Zhang, K. Construction, expression, and characterization of a recombinant immunotoxin targeting EpCAM. Mediat. Inflamm 2015, 2015, 460264. [Google Scholar] [CrossRef] [PubMed]
- Ogawa, K.; Tanaka, S.; Matsumura, S.; Murakata, A.; Ban, D.; Ochiai, T.; Irie, T.; Kudo, A.; Nakamura, N.; Tanabe, M.; et al. EpCAM-targeted therapy for human hepatocellular carcinoma. Ann. Surg. Oncol. 2014, 21, 1314–1322. [Google Scholar] [CrossRef] [PubMed]
- Frankel, A.E.; Woo, J.H.; Ahn, C.; Foss, F.M.; Duvic, M.; Neville, P.H.; Neville, D.M. Resimmune, an anti-CD3ε recombinant immunotoxin, induces durable remissions in patients with cutaneous T-cell lymphoma. Haematologica 2015, 100, 794–800. [Google Scholar] [CrossRef] [PubMed]
- Girbés, T.; Citores, L.; Iglesias, R.; Ferreras, J.M.; Muñoz, R.; Rojo, M.A.; Arias, F.J.; García, J.R.; Méndez, E.; Calonge, M. Ebulin 1, a nontoxic novel type 2 ribosome-inactivating protein from Sambucus ebulus L. leaves. J. Biol. Chem. 1993, 268, 18195–18199. [Google Scholar] [PubMed]
- Pizzo, E.; di Maro, A. A new age for biomedical applications of ribosome inactivating proteins (RIPs): From bioconjugate to nanoconstructs. J. Biomed. Sci 2016, 23, 54. [Google Scholar] [CrossRef] [PubMed]
- Aruna, G. Immunotoxins: A review of their use in cancer treatment. J. Stem Cells Regen. Med. 2006, 1, 31–36. [Google Scholar] [PubMed]
- Polito, L.; Bortolotti, M.; Pedrazzi, M.; Bolognesi, A. Immunotoxins and other conjugates containing saporin-S6 for cancer therapy. Toxins 2011, 3, 697–720. [Google Scholar] [CrossRef] [PubMed]
- Bortolotti, M.; Bolognesi, A.; Battelli, M.G.; Polito, L. High in vitro anti-tumor efficacy of dimeric rituximab/saporin-S6 immunotoxin. Toxins 2016, 8, E192. [Google Scholar] [CrossRef] [PubMed]
- Guo, J.L.; Cheng, Y.L.; Qiu, Y.; Shen, C.H.; Yi, B.; Peng, C. Purification and characterization of a novel type I ribosome inactivating protein, pachyerosin, from Pachyrhizus erosus seeds, and preparation of its immunotoxin against human hepatoma cells. Planta Med. 2014, 80, 896–901. [Google Scholar] [CrossRef] [PubMed]
- Forner, A.; Llovet, J.M.; Bruix, J. Hepatocellular carcinoma. Lancet 2012, 379, 1245–1255. [Google Scholar] [CrossRef]
- Bosch, F.X.; Ribes, J.; Díaz, M.; Cléries, R. Primary liver cancer: Worldwide incidence and trends. Gastroenterology 2004, 127, S5–S16. [Google Scholar] [CrossRef] [PubMed]
- Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer incidence and mortality worldwide: Sources, methods and major patterns in globocan 2012. Int. J. Cancer 2015, 136, E359–E386. [Google Scholar] [CrossRef] [PubMed]
- Chen, Z.; Shi, T.; Zhang, L.; Zhu, P.; Deng, M.; Huang, C.; Hu, T.; Jiang, L.; Li, J. Mammalian drug efflux transporters of the ATP binding cassette (ABC) family in multidrug resistance: A review of the past decade. Cancer Lett. 2016, 370, 153–164. [Google Scholar] [CrossRef] [PubMed]
- Wong, R.; Frenette, C. Updates in the management of hepatocellular carcinoma. Gastroenterol. Hepatol. 2011, 7, 16–24. [Google Scholar]
- Cheng, A.L.; Kang, Y.K.; Chen, Z.; Tsao, C.J.; Qin, S.; Kim, J.S.; Luo, R.; Feng, J.; Ye, S.; Yang, T.S.; et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: A phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009, 10, 25–34. [Google Scholar] [CrossRef]
- Llovet, J.M.; Ricci, S.; Mazzaferro, V.; Hilgard, P.; Gane, E.; Blanc, J.F.; de Oliveira, A.C.; Santoro, A.; Raoul, J.L.; Forner, A.; et al. Sorafenib in advanced hepatocellular carcinoma. N. Engl. J. Med. 2008, 359, 378–390. [Google Scholar] [CrossRef] [PubMed]
- Gauthier, A.; Ho, M. Role of sorafenib in the treatment of advanced hepatocellular carcinoma: An update. Hepatol. Res. 2013, 43, 147–154. [Google Scholar] [CrossRef] [PubMed]
- Keating, G.M.; Santoro, A. Sorafenib: A review of its use in advanced hepatocellular carcinoma. Drugs 2009, 69, 223–240. [Google Scholar] [CrossRef] [PubMed]
- Leonetti, A.; Bersanelli, M.; Castagneto, B.; Masini, C.; di Meglio, G.; Pellegrino, B.; Buti, S. Outcome and safety of sorafenib in metastatic renal cell carcinoma dialysis patients: A systematic review. Clin. Genitourin. Cancer 2016, 14, 277–283. [Google Scholar] [CrossRef] [PubMed]
- Chen, K.F.; Chen, H.L.; Tai, W.T.; Feng, W.C.; Hsu, C.H.; Chen, P.J.; Cheng, A.L. Activation of phosphatidylinositol 3-kinase/Akt signaling pathway mediates acquired resistance to sorafenib in hepatocellular carcinoma cells. J. Pharmacol. Exp. Ther. 2011, 337, 155–161. [Google Scholar] [CrossRef] [PubMed]
- Franklin, M.C.; Navarro, E.C.; Wang, Y.; Patel, S.; Singh, P.; Zhang, Y.; Persaud, K.; Bari, A.; Griffith, H.; Shen, L.; et al. The structural basis for the function of two anti-VEGF receptor 2 antibodies. Structure 2011, 19, 1097–1107. [Google Scholar] [CrossRef] [PubMed]
- Zhu, A.X.; Park, J.O.; Ryoo, B.Y.; Yen, C.J.; Poon, R.; Pastorelli, D.; Blanc, J.F.; Chung, H.C.; Baron, A.D.; Pfiffer, T.E.; et al. Ramucirumab versus placebo as second-line treatment in patients with advanced hepatocellular carcinoma following first-line therapy with sorafenib (Reach): A randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2015, 16, 859–870. [Google Scholar] [CrossRef]
- Pinter, M.; Ulbrich, G.; Sieghart, W.; Kölblinger, C.; Reiberger, T.; Li, S.; Ferlitsch, A.; Müller, C.; Lammer, J.; Peck-Radosavljevic, M. Hepatocellular carcinoma: A phase II randomized controlled double-blind trial of transarterial chemoembolization in combination with biweekly intravenous administration of bevacizumab or a placebo. Radiology 2015, 277, 903–912. [Google Scholar] [CrossRef] [PubMed]
- Roskoski, R. Vascular endothelial growth factor (VEGF) signaling in tumor progression. Crit. Rev. Oncol. Hematol. 2007, 62, 179–213. [Google Scholar] [CrossRef] [PubMed]
- Kudo, M. Signaling pathway/molecular targets and new targeted agents under development in hepatocellular carcinoma. World J. Gastroenterol. 2012, 18, 6005–6017. [Google Scholar] [CrossRef] [PubMed]
- Haruyama, Y.; Kataoka, H. Glypican-3 is a prognostic factor and an immunotherapeutic target in hepatocellular carcinoma. World J. Gastroenterol. 2016, 22, 275–283. [Google Scholar] [CrossRef] [PubMed]
- Qi, X.S.; Guo, X.Z.; Han, G.H.; Li, H.Y.; Chen, J. Met inhibitors for treatment of advanced hepatocellular carcinoma: A review. World J. Gastroenterol. 2015, 21, 5445–5453. [Google Scholar] [CrossRef] [PubMed]
- Choi, K.J.; Baik, I.H.; Ye, S.K.; Lee, Y.H. Molecular targeted therapy for hepatocellular carcinoma: Present status and future directions. Biol. Pharm. Bull. 2015, 38, 986–991. [Google Scholar] [CrossRef] [PubMed]
- Enguita-Germán, M.; Fortes, P. Targeting the insulin-like growth factor pathway in hepatocellular carcinoma. World J. Hepatol. 2014, 6, 716–737. [Google Scholar] [CrossRef] [PubMed]
- Rosen, L.S.; Gordon, M.S.; Robert, F.; Matei, D.E. Endoglin for targeted cancer treatment. Curr. Oncol. Rep. 2014, 16, 365. [Google Scholar] [CrossRef] [PubMed]
- Doi, T.; Lee, K.H.; Kim, T.M.; Ohtsu, A.; Kim, T.Y.; Ikeda, M.; Yoh, K.; Gallo Stampino, C.; Hirohashi, T.; Suzuki, A.; et al. A phase I study of the human anti-activin receptor-like kinase 1 antibody PF-03446962 in Asian patients with advanced solid tumors. Cancer Med. 2016, 22, 2146–2154. [Google Scholar] [CrossRef] [PubMed]
- Shvartsur, A.; Bonavida, B. Trop2 and its overexpression in cancers: Regulation and clinical/therapeutic implications. Genes Cancer 2015, 6, 84–105. [Google Scholar] [PubMed]
- Yamashita, T.; Forgues, M.; Wang, W.; Kim, J.W.; Ye, Q.; Jia, H.; Budhu, A.; Zanetti, K.A.; Chen, Y.; Qin, L.X.; et al. EpCAM and alpha-fetoprotein expression defines novel prognostic subtypes of hepatocellular carcinoma. Cancer Res. 2008, 68, 1451–1461. [Google Scholar] [CrossRef] [PubMed]
- Yamashita, T.; Ji, J.; Budhu, A.; Forgues, M.; Yang, W.; Wang, H.Y.; Jia, H.; Ye, Q.; Qin, L.X.; Wauthier, E.; et al. EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features. Gastroenterology 2009, 136, 1012–1024. [Google Scholar] [CrossRef] [PubMed]
- Yang, X.R.; Xu, Y.; Yu, B.; Zhou, J.; Qiu, S.J.; Shi, G.M.; Zhang, B.H.; Wu, W.Z.; Shi, Y.H.; Wu, B.; et al. High expression levels of putative hepatic stem/progenitor cell biomarkers related to tumour angiogenesis and poor prognosis of hepatocellular carcinoma. Gut 2010, 59, 953–962. [Google Scholar] [CrossRef] [PubMed]
- Hsu, H.C.; Cheng, W.; Lai, P.L. Cloning and expression of a developmentally regulated transcript MXR7 in hepatocellular carcinoma: Biological significance and temporospatial distribution. Cancer Res. 1997, 57, 5179–5184. [Google Scholar] [PubMed]
- Capurro, M.; Wanless, I.R.; Sherman, M.; Deboer, G.; Shi, W.; Miyoshi, E.; Filmus, J. Glypican-3: A novel serum and histochemical marker for hepatocellular carcinoma. Gastroenterology 2003, 125, 89–97. [Google Scholar] [CrossRef]
- Jia, H.L.; Ye, Q.H.; Qin, L.X.; Budhu, A.; Forgues, M.; Chen, Y.; Liu, Y.K.; Sun, H.C.; Wang, L.; Lu, H.Z.; et al. Gene expression profiling reveals potential biomarkers of human hepatocellular carcinoma. Clin. Cancer Res. 2007, 13, 1133–1139. [Google Scholar] [CrossRef] [PubMed]
- Capurro, M.I.; Xiang, Y.Y.; Lobe, C.; Filmus, J. Glypican-3 promotes the growth of hepatocellular carcinoma by stimulating canonical Wnt signaling. Cancer Res. 2005, 65, 6245–6254. [Google Scholar] [CrossRef] [PubMed]
- Gao, W.; Kim, H.; Feng, M.; Phung, Y.; Xavier, C.P.; Rubin, J.S.; Ho, M. Inactivation of Wnt signaling by a human antibody that recognizes the heparan sulfate chains of glypican-3 for liver cancer therapy. Hepatology 2014, 60, 576–587. [Google Scholar] [CrossRef] [PubMed]
- Feng, M.; Gao, W.; Wang, R.; Chen, W.; Man, Y.G.; Figg, W.D.; Wang, X.W.; Dimitrov, D.S.; Ho, M. Therapeutically targeting glypican-3 via a conformation-specific single-domain antibody in hepatocellular carcinoma. Proc. Natl. Acad. Sci. USA 2013, 110, E1083–E1091. [Google Scholar] [CrossRef] [PubMed]
- Gao, W.; Tang, Z.; Zhang, Y.F.; Feng, M.; Qian, M.; Dimitrov, D.S.; Ho, M. Immunotoxin targeting glypican-3 regresses liver cancer via dual inhibition of Wnt signaling and protein synthesis. Nat. Commun. 2015, 6, 6536. [Google Scholar] [CrossRef] [PubMed]
- Phung, Y.; Gao, W.; Man, Y.G.; Nagata, S.; Ho, M. 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. [Google Scholar] [CrossRef] [PubMed]
- De Genst, E.; Silence, K.; Decanniere, K.; Conrath, K.; Loris, R.; Kinne, J.; Muyldermans, S.; Wyns, L. Molecular basis for the preferential cleft recognition by dromedary heavy-chain antibodies. Proc. Natl. Acad. Sci. USA 2006, 103, 4586–4591. [Google Scholar] [CrossRef] [PubMed]
- Gao, W.; Kim, H.; Ho, M. Human monoclonal antibody targeting the heparan sulfate chains of glypican-3 inhibits HGF-mediated migration and motility of hepatocellular carcinoma cells. PLoS ONE 2015, 10, E0137664. [Google Scholar] [CrossRef] [PubMed]
- Weldon, J.E.; Xiang, L.; Chertov, O.; Margulies, I.; Kreitman, R.J.; Fitzgerald, D.J.; Pastan, I. A protease-resistant immunotoxin against CD22 with greatly increased activity against CLL and diminished animal toxicity. Blood 2009, 113, 3792–3800. [Google Scholar] [CrossRef] [PubMed]
- Weldon, J.E.; Xiang, L.; Zhang, J.; Beers, R.; Walker, D.A.; Onda, M.; Hassan, R.; Pastan, I. 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. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.; Gao, W.; Feng, M.; Pastan, I.; Ho, M. Construction of an immunotoxin, HN3-mPE24, targeting glypican-3 for liver cancer therapy. Oncotarget 2016. [Google Scholar] [CrossRef] [PubMed]
- Mazor, R.; Onda, M.; Pastan, I. Immunogenicity of therapeutic recombinant immunotoxins. Immunol. Rev. 2016, 270, 152–164. [Google Scholar] [CrossRef] [PubMed]
- Hassan, R.; Miller, A.C.; Sharon, E.; Thomas, A.; Reynolds, J.C.; Ling, A.; Kreitman, R.J.; Miettinen, M.M.; Steinberg, S.M.; Fowler, D.H.; et al. Major cancer regressions in mesothelioma after treatment with an anti-mesothelin immunotoxin and immune suppression. Sci. Transl. Med. 2013, 5, 208ra147. [Google Scholar] [CrossRef] [PubMed]
- Mazor, R.; Zhang, J.; Xiang, L.; Addissie, S.; Awuah, P.; Beers, R.; Hassan, R.; Pastan, I. Recombinant immunotoxin with T-cell epitope mutations that greatly reduce immunogenicity for treatment of mesothelin-expressing tumors. Mol. Cancer Ther. 2015, 14, 2789–2796. [Google Scholar] [CrossRef] [PubMed]
- Liao, W.W.; Arthur, J.W. Predicting peptide binding to major histocompatibility complex molecules. Autoimmun Rev. 2011, 10, 469–473. [Google Scholar] [CrossRef] [PubMed]
- Kreitman, R.J.; 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. [Google Scholar] [PubMed]
- Böttger, R.; Knappe, D.; Hoffmann, R. Readily adaptable release kinetics of prodrugs using protease-dependent reversible PEGylation. J. Control. Release 2016, 230, 88–94. [Google Scholar] [CrossRef] [PubMed]
- Benhar, I.; Reiter, Y.; Pai, L.H.; Pastan, I. Administration of disulfide-stabilized Fv-immunotoxins B1(dsFv)-PE38 and B3(dsFv)-PE38 by continuous infusion increases their efficacy in curing large tumor xenografts in nude mice. Int. J. Cancer 1995, 62, 351–355. [Google Scholar] [CrossRef] [PubMed]
- Lorberboum-Galski, H.; Lafyatis, R.; Case, J.P.; Fitzgerald, D.; Wilder, R.L.; Pastan, I. Administration of IL-2-PE40 via osmotic pumps prevents adjuvant induced arthritis in rats. Improved therapeutic index of IL-2-PE40 administered by continuous infusion. Int. J. Immunopharmacol. 1991, 13, 305–315. [Google Scholar] [CrossRef]
Identifer | Drug(s) | Type | Target | Phase | Status |
---|---|---|---|---|---|
NCT01911273 | PF-03446962 | Mono | ALK-1 | II | Terminated |
NCT01897038 | Onartuzumab with Sorafenib | Mono | c-Met/Tyrosine Kinases | I | Completed |
NCT01008358 | Tremelimumab | Mono | CTLA4 | II | Completed |
NCT00483405 | Cetuximab with chemotherapy | Mono | EGFR | II | Completed |
NCT01375569 | TRC105 | Mono | Endoglin | II | Completed |
NCT02560779 | TRC105 with Sorafenib | Mono | Endoglin/Tyrosine Kinases | II | Recruiting |
NCT01507168 | GC33 | Mono | GPC3 | II | Completed |
NCT00639509 | Cixutumumab | Mono | IGF-1R | II | Completed |
NCT00906373 | Cixutumumab with Sorfenib | Mono | IGF-1R/Tyrosine Kinases | II | Completed |
NCT02315066 | PF-04518600 | Mono | OX40(CD134) | I | Recruiting |
NCT02595866 | Pembrolizumab | Mono | PD-1 | I | Recruiting |
NCT00966251 | Pidilizumab | Mono | PD-1 | II | Terminated |
NCT02423343 | Nivolumab with Galunisertib | Mono | PD-1/TGF-βR1 Kinases | II | Recruiting |
NCT01102400 | MEDI-575 | Mono | PDGFRA | I | Completed |
NCT02519348 | Durvalumab with Tremelimumab | Mono | PD-L1/CTLA4 | II | Recruiting |
NCT01308723 | RO5323441 with Sorafenib | Mono | PGF/Tyrosine Kinases | I | Completed |
NCT01258608 | Mapatumumab with Sorafenib | Mono | TRAIL-R1/Tyrosine Kinases | II | Ongoing |
NCT00055692 | Bevacizumab | Mono | VEGF-A | II | Completed |
NCT00467194 | Bevacizumab with Rapamycin | Mono | VEGF-A/mTor | I | Completed |
NCT01010126 | Bevacizumab with Temsirolimus | Mono | VEGF-A/mTor | II | Ongoing |
NCT00365391 | Bevacizumab with Erlotinib | Mono | VEGF-A/Tyrosine Kinases | II | Completed |
NCT00867321 | Bevacizumab with Sorafenib | Mono | VEGF-A/Tyrosine Kinases | II | Completed |
NCT01140347 | Ramucirumab | Mono | VEGF-R2 | III | Completed |
NCT02069041 | Ramucirumab with Oxaliplatin | Mono | VEGF-R2/DNA | I | Recruiting |
NCT02572687 | Ramucirumab with Durvalumab | Mono | VEGF-R2/PD-L1 | I | Recruiting |
NCT01498952 | MEDI-573 with Sorafenib | Bispecific | IGF-I and IGF-II/Tyrosine Kinases | I | Completed |
NCT01631552 | Sacituzumab Govitecan | ADC | TROP-2/Topoisomerase | II | Recruiting |
NCT00829465 | Metuximab labeled with Iodine131 | ARC | CD147 | IV | Unknown |
NCT02723942 | T cells expressing αGPC3 Antibody | CAR-T | GPC3 | II | Recruiting |
NCT02632006 | T cells expressing PD-1 Antibody | CAR-T | PD-1 | II | Recruiting |
© 2016 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Fleming, B.D.; Ho, M. Glypican-3 Targeting Immunotoxins for the Treatment of Liver Cancer. Toxins 2016, 8, 274. https://doi.org/10.3390/toxins8100274
Fleming BD, Ho M. Glypican-3 Targeting Immunotoxins for the Treatment of Liver Cancer. Toxins. 2016; 8(10):274. https://doi.org/10.3390/toxins8100274
Chicago/Turabian StyleFleming, Bryan D., and Mitchell Ho. 2016. "Glypican-3 Targeting Immunotoxins for the Treatment of Liver Cancer" Toxins 8, no. 10: 274. https://doi.org/10.3390/toxins8100274
APA StyleFleming, B. D., & Ho, M. (2016). Glypican-3 Targeting Immunotoxins for the Treatment of Liver Cancer. Toxins, 8(10), 274. https://doi.org/10.3390/toxins8100274