Cripto-1 as a Potential Target of Cancer Stem Cells for Immunotherapy
Abstract
:Simple Summary
Abstract
1. Introduction
2. Cripto-1 as a Novel Target for Immunotherapy
3. CSCs as a Therapeutic Target
4. Prospect of Antibody Drugs in Immunotherapy
5. Functions of Cripto-1 in CSCs
5.1. Nodal/ALK4,7/Smad2 Signaling Pathway
5.2. Glypican-1/c-Src/MAPK/AKT Signaling Pathway
6. Role of Cripto-1 in Metastasis and Recurrence
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
References
- van der Bruggen, P.; Traversari, C.; Chomez, P.; Lurquin, C.; De Plaen, E.; Van den Eynde, B.; Knuth, A.; Boon, T. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science 1991, 254, 1643. [Google Scholar] [CrossRef] [PubMed]
- Blankenstein, T.; Coulie, P.G.; Gilboa, E.; Jaffee, E.M. The determinants of tumour immunogenicity. Nat. Rev. Cancer 2012, 12, 307–313. [Google Scholar] [CrossRef]
- Ishida, Y.; Agata, Y.; Shibahara, K.; Honjo, T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992, 11, 3887–3895. [Google Scholar] [CrossRef]
- Kumagai, S.; Togashi, Y.; Kamada, T.; Sugiyama, E.; Nishinakamura, H.; Takeuchi, Y.; Vitaly, K.; Itahashi, K.; Maeda, Y.; Matsui, S.; et al. The PD-1 expression balance between effector and regulatory T cells predicts the clinical efficacy of PD-1 blockade therapies. Nat. Immunol. 2020, 21, 1346–1358. [Google Scholar] [CrossRef] [PubMed]
- Freeman, G.J.; Long, A.J.; Iwai, Y.; Bourque, K.; Chernova, T.; Nishimura, H.; Fitz, L.J.; Malenkovich, N.; Okazaki, T.; Byrne, M.C.; et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 2000, 192, 1027–1034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef] [Green Version]
- Egen, J.G.; Kuhns, M.S.; Allison, J.P. CTLA-4: New insights into its biological function and use in tumor immunotherapy. Nat. Immunol. 2002, 3, 611–618. [Google Scholar] [CrossRef]
- Leach, D.R.; Krummel, M.F.; Allison, J.P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 1996, 271, 1734–1736. [Google Scholar] [CrossRef] [Green Version]
- Catakovic, K.; Klieser, E.; Neureiter, D.; Geisberger, R. T cell exhaustion: From pathophysiological basics to tumor immunotherapy. Cell Commun. Signal. 2017, 15, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Okazaki, T.; Honjo, T. PD-1 and PD-1 ligands: From discovery to clinical application. Int. Immunol. 2007, 19, 813–824. [Google Scholar] [CrossRef] [Green Version]
- Keenan, T.E.; Burke, K.P.; Van Allen, E.M. Genomic correlates of response to immune checkpoint blockade. Nat. Med. 2019, 25, 389–402. [Google Scholar] [CrossRef]
- Sanjabi, S.; Oh, S.A.; Li, M.O. Regulation of the Immune Response by TGF-β: From Conception to Autoimmunity and Infection. Cold Spring Harb. Perspect. Biol. 2017, 9, a022236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Batlle, E.; Massagué, J. Transforming Growth Factor-β Signaling in Immunity and Cancer. Immunity 2019, 50, 924–940. [Google Scholar] [CrossRef]
- Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Cañellas, A.; Hernando-Momblona, X.; et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel Iii, E.E.; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef]
- Sandomenico, A.; Ruvo, M. Targeting Nodal and Cripto-1: Perspectives Inside Dual Potential Theranostic Cancer Biomarkers. Curr. Med. Chem. 2019, 26, 1994–2050. [Google Scholar] [CrossRef] [PubMed]
- Bianco, C.; Rangel, M.C.; Castro, N.P.; Nagaoka, T.; Rollman, K.; Gonzales, M.; Salomon, D.S. Role of Cripto-1 in stem cell maintenance and malignant progression. Am. J. Pathol. 2010, 177, 532–540. [Google Scholar] [CrossRef]
- Watanabe, K.; Hamada, S.; Bianco, C.; Mancino, M.; Nagaoka, T.; Gonzales, M.; Bailly, V.; Strizzi, L.; Salomon, D.S. Requirement of glycosylphosphatidylinositol anchor of Cripto-1 for trans activity as a Nodal co-receptor. J. Biol. Chem. 2007, 282, 35772–35786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, G.H.; Fujita, M.; Takaoka, K.; Murakami, Y.; Fujihara, Y.; Kanzawa, N.; Murakami, K.I.; Kajikawa, E.; Takada, Y.; Saito, K.; et al. A GPI processing phospholipase A2, PGAP6, modulates Nodal signaling in embryos by shedding CRIPTO. J. Cell Biol. 2016, 215, 705–718. [Google Scholar] [CrossRef]
- Lonardo, E.; Hermann, P.C.; Mueller, M.-T.; Huber, S.; Balic, A.; Miranda-Lorenzo, I.; Zagorac, S.; Alcala, S.; Rodriguez-Arabaolaza, I.; Ramirez, J.C.; et al. Nodal/Activin Signaling Drives Self-Renewal and Tumorigenicity of Pancreatic Cancer Stem Cells and Provides a Target for Combined Drug Therapy. Cell Stem Cell 2011, 9, 433–446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gong, W.; Sun, B.; Sun, H.; Zhao, X.; Zhang, D.; Liu, T.; Zhao, N.; Gu, Q.; Dong, X.; Liu, F. Nodal signaling activates the Smad2/3 pathway to regulate stem cell-like properties in breast cancer cells. Am. J. Cancer Res. 2017, 7, 503–517. [Google Scholar]
- Fiorenzano, A.; Pascale, E.; D’Aniello, C.; Acampora, D.; Bassalert, C.; Russo, F.; Andolfi, G.; Biffoni, M.; Francescangeli, F.; Zeuner, A.; et al. Cripto is essential to capture mouse epiblast stem cell and human embryonic stem cell pluripotency. Nat. Commun. 2016, 7, 12589. [Google Scholar] [CrossRef] [PubMed]
- Lo, R.C.-L.; Leung, C.O.-N.; Chan, K.K.-S.; Ho, D.W.-H.; Wong, C.-M.; Lee, T.K.-W.; Ng, I.O.-L. Cripto-1 contributes to stemness in hepatocellular carcinoma by stabilizing Dishevelled-3 and activating Wnt/β-catenin pathway. Cell Death Differ. 2018, 25, 1426–1441. [Google Scholar] [CrossRef] [Green Version]
- Francescangeli, F.; Contavalli, P.; De Angelis, M.L.; Baiocchi, M.; Gambara, G.; Pagliuca, A.; Fiorenzano, A.; Prezioso, C.; Boe, A.; Todaro, M.; et al. Dynamic regulation of the cancer stem cell compartment by Cripto-1 in colorectal cancer. Cell Death Differ. 2015, 22, 1700–1713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Q.; Cui, X.; Yu, X.; Bian, B.-S.-J.; Qian, F.; Hu, X.-G.; Ji, C.-d.; Yang, L.; Ren, Y.; Cui, W.; et al. Cripto-1 acts as a functional marker of cancer stem-like cells and predicts prognosis of the patients in esophageal squamous cell carcinoma. Mol. Cancer 2017, 16, 81. [Google Scholar] [CrossRef] [PubMed]
- Singh, A.; Settleman, J. EMT, cancer stem cells and drug resistance: An emerging axis of evil in the war on cancer. Oncogene 2010, 29, 4741–4751. [Google Scholar] [CrossRef] [Green Version]
- Hermann, P.C.; Huber, S.L.; Herrler, T.; Aicher, A.; Ellwart, J.W.; Guba, M.; Bruns, C.J.; Heeschen, C. Distinct Populations of Cancer Stem Cells Determine Tumor Growth and Metastatic Activity in Human Pancreatic Cancer. Cell Stem Cell 2007, 1, 313–323. [Google Scholar] [CrossRef] [Green Version]
- Oskarsson, T.; Batlle, E.; Massagué, J. Metastatic Stem Cells: Sources, Niches, and Vital Pathways. Cell Stem Cell 2014, 14, 306–321. [Google Scholar] [CrossRef] [Green Version]
- Peitzsch, C.; Tyutyunnykova, A.; Pantel, K.; Dubrovska, A. Cancer stem cells: The root of tumor recurrence and metastases. Semin. Cancer Biol. 2017, 44, 10–24. [Google Scholar] [CrossRef]
- Budhu, S.; Schaer, D.A.; Li, Y.; Toledo-Crow, R.; Panageas, K.; Yang, X.; Zhong, H.; Houghton, A.N.; Silverstein, S.C.; Merghoub, T.; et al. Blockade of surface-bound TGF-β on regulatory T cells abrogates suppression of effector T cell function in the tumor microenvironment. Sci. Signal. 2017, 10, eaak9702. [Google Scholar] [CrossRef] [Green Version]
- Bellomo, C.; Caja, L.; Moustakas, A. Transforming growth factor β as regulator of cancer stemness and metastasis. Br. J. Cancer 2016, 115, 761–769. [Google Scholar] [CrossRef]
- Oshimori, N.; Oristian, D.; Fuchs, E. TGF-β promotes heterogeneity and drug resistance in squamous cell carcinoma. Cell 2015, 160, 963–976. [Google Scholar] [CrossRef] [Green Version]
- Shimizu, T.; Ishikawa, T.; Sugihara, E.; Kuninaka, S.; Miyamoto, T.; Mabuchi, Y.; Matsuzaki, Y.; Tsunoda, T.; Miya, F.; Morioka, H.; et al. c-MYC overexpression with loss of Ink4a/Arf transforms bone marrow stromal cells into osteosarcoma accompanied by loss of adipogenesis. Oncogene 2010, 29, 5687–5699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Strizzi, L.; Hardy, K.M.; Kirschmann, D.A.; Ahrlund-Richter, L.; Hendrix, M.J.C. Nodal expression and detection in cancer: Experience and challenges. Cancer Res. 2012, 72, 1915–1920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalyan, A.; Carneiro, B.A.; Chandra, S.; Kaplan, J.; Chae, Y.K.; Matsangou, M.; Hendrix, M.J.C.; Giles, F. Nodal Signaling as a Developmental Therapeutics Target in Oncology. Mol. Cancer Ther. 2017, 16, 787–792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Watanabe, K.; Bianco, C.; Strizzi, L.; Hamada, S.; Mancino, M.; Bailly, V.; Mo, W.; Wen, D.; Miatkowski, K.; Gonzales, M.; et al. Growth factor induction of Cripto-1 shedding by glycosylphosphatidylinositol-phospholipase D and enhancement of endothelial cell migration. J. Biol. Chem. 2007, 282, 31643–31655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ligtenberg, M.A.; Witt, K.; Galvez-Cancino, F.; Sette, A.; Lundqvist, A.; Lladser, A.; Kiessling, R. Cripto-1 vaccination elicits protective immunity against metastatic melanoma. Oncoimmunology 2016, 5, e1128613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Witt, K.; Ligtenberg, M.A.; Conti, L.; Lanzardo, S.; Ruiu, R.; Wallmann, T.; Tufvesson-Stiller, H.; Chambers, B.J.; Rolny, C.; Lladser, A.; et al. Cripto-1 Plasmid DNA Vaccination Targets Metastasis and Cancer Stem Cells in Murine Mammary Carcinoma. Cancer Immunol. Res. 2018, 6, 1417–1425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, D.M.; Bao, Y.L.; Yu, C.L.; Wang, Y.M.; Song, Z.B. Cripto-1 modulates macrophage cytokine secretion and phagocytic activity via NF-κB signaling. Immunol. Res. 2016, 64, 104–114. [Google Scholar] [CrossRef]
- Stifter, K.; Krieger, J.; Ruths, L.; Gout, J.; Mulaw, M.; Lechel, A.; Kleger, A.; Seufferlein, T.; Wagner, M.; Schirmbeck, R. IFN-γ treatment protocol for MHC-I(lo)/PD-L1(+) pancreatic tumor cells selectively restores their TAP-mediated presentation competence and CD8 T-cell priming potential. J. Immunother. Cancer 2020, 8. [Google Scholar] [CrossRef]
- Strizzi, L.; Postovit, L.-M.; Margaryan, N.V.; Seftor, E.A.; Abbott, D.E.; Seftor, R.E.B.; Salomon, D.S.; Hendrix, M.J.C. Emerging roles of nodal and Cripto-1: From embryogenesis to breast cancer progression. Breast Dis. 2008, 29, 91–103. [Google Scholar] [CrossRef] [Green Version]
- Daraghma, H.; Untiveros, G.; Raskind, A.; Iaccarino, E.; Sandomenico, A.; Ruvo, M.; Arnouk, H.; Ciancio, M.J.; Cuevas-Nunez, M.; Strizzi, L. The role of Nodal and Cripto-1 in human oral squamous cell carcinoma. Oral Dis. 2020. [Google Scholar] [CrossRef]
- Alowaidi, F.; Hashimi, S.M.; Alqurashi, N.; Wood, S.A.; Wei, M.Q. Cripto-1 overexpression in U87 glioblastoma cells activates MAPK, focal adhesion and ErbB pathways. Oncol. Lett. 2019, 18, 3399–3406. [Google Scholar] [CrossRef] [Green Version]
- Bianco, C.; Strizzi, L.; Ebert, A.; Chang, C.; Rehman, A.; Normanno, N.; Guedez, L.; Salloum, R.; Ginsburg, E.; Sun, Y.; et al. Role of human cripto-1 in tumor angiogenesis. J. Natl. Cancer Inst. 2005, 97, 132–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bianco, C.; Castro, N.P.; Baraty, C.; Rollman, K.; Held, N.; Rangel, M.C.; Karasawa, H.; Gonzales, M.; Strizzi, L.; Salomon, D.S. Regulation of human Cripto-1 expression by nuclear receptors and DNA promoter methylation in human embryonal and breast cancer cells. J. Cell. Physiol. 2013, 228, 1174–1188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Castro, N.P.; Fedorova-Abrams, N.D.; Merchant, A.S.; Rangel, M.C.; Nagaoka, T.; Karasawa, H.; Klauzinska, M.; Hewitt, S.M.; Biswas, K.; Sharan, S.K.; et al. Cripto-1 as a novel therapeutic target for triple negative breast cancer. Oncotarget 2015, 6, 11910–11929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alowaidi, F.; Hashimi, S.M.; Nguyen, M.; Meshram, M.; Alqurashi, N.; Cavanagh, B.L.; Bellette, B.; Ivanovski, S.; Meedenyia, A.; Wood, S.A. Investigating the role of CRIPTO-1 (TDGF-1) in glioblastoma multiforme U87 cell line. J. Cell. Biochem. 2018. [Google Scholar] [CrossRef] [PubMed]
- Gudbergsson, J.M.; Duroux, M. Cripto-1 localizes to dynamic and shed filopodia associated with cellular migration in glioblastoma cells. Eur. J. Cell Biol. 2019, 98, 151044. [Google Scholar] [CrossRef]
- Liu, Y.; Wang, J.; Yang, T.; Liu, R.; Xu, Y. Overexpression levels of cripto-1 predict poor prognosis in patients with prostate cancer following radical prostatectomy. Oncol. Lett. 2019, 18, 2584–2591. [Google Scholar] [CrossRef] [Green Version]
- De Luca, A.; Lamura, L.; Strizzi, L.; Roma, C.; D’Antonio, A.; Margaryan, N.; Pirozzi, G.; Hsu, M.Y.; Botti, G.; Mari, E.; et al. Expression and functional role of CRIPTO-1 in cutaneous melanoma. Br. J. Cancer 2011, 105, 1030–1038. [Google Scholar] [CrossRef] [Green Version]
- Alam, M.J.; Takahashi, R.; Afify, S.M.; Oo, A.K.K.; Kumon, K.; Nawara, H.M.; Khayrani, A.C.; Du, J.; Zahra, M.H.; Seno, A.; et al. Exogenous Cripto-1 Suppresses Self-Renewal of Cancer Stem Cell Model. Int. J. Mol. Sci. 2018, 19, 3345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karkampouna, S.; van der Helm, D.; Gray, P.C.; Chen, L.; Klima, I.; Grosjean, J.; Burgmans, M.C.; Farina-Sarasqueta, A.; Snaar-Jagalska, E.B.; Stroka, D.M.; et al. CRIPTO promotes an aggressive tumour phenotype and resistance to treatment in hepatocellular carcinoma. J. Pathol. 2018, 245, 297–310. [Google Scholar] [CrossRef] [PubMed]
- Diehn, M.; Cho, R.W.; Lobo, N.A.; Kalisky, T.; Dorie, M.J.; Kulp, A.N.; Qian, D.; Lam, J.S.; Ailles, L.E.; Wong, M.; et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 2009, 458, 780–783. [Google Scholar] [CrossRef] [PubMed]
- Chakravarti, A.; Chakladar, A.; Delaney, M.A.; Latham, D.E.; Loeffler, J.S. The epidermal growth factor receptor pathway mediates resistance to sequential administration of radiation and chemotherapy in primary human glioblastoma cells in a RAS-dependent manner. Cancer Res. 2002, 62, 4307–4315. [Google Scholar]
- Donnenberg, V.S.; Donnenberg, A.D. Multiple drug resistance in cancer revisited: The cancer stem cell hypothesis. J. Clin. Pharmacol. 2005, 45, 872–877. [Google Scholar] [CrossRef]
- Ishiwata, T. Cancer stem cells and epithelial-mesenchymal transition: Novel therapeutic targets for cancer. Pathol. Int. 2016, 66, 601–608. [Google Scholar] [CrossRef]
- Lambert, A.W.; Weinberg, R.A. Linking EMT programmes to normal and neoplastic epithelial stem cells. Nat. Rev. Cancer 2021. [Google Scholar] [CrossRef]
- Pützer, B.M.; Solanki, M.; Herchenröder, O. Advances in cancer stem cell targeting: How to strike the evil at its root. Adv. Drug Deliv. Rev. 2017, 120, 89–107. [Google Scholar] [CrossRef]
- Wang, A.; Qu, L.; Wang, L. At the crossroads of cancer stem cells and targeted therapy resistance. Cancer Lett. 2017, 385, 87–96. [Google Scholar] [CrossRef]
- Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124–1134. [Google Scholar] [CrossRef]
- Desai, A.; Yan, Y.; Gerson, S.L. Concise Reviews: Cancer Stem Cell Targeted Therapies: Toward Clinical Success. Stem Cells Transl. Med. 2019, 8, 75–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, T.; Shigdar, S.; Gantier, M.P.; Hou, Y.; Wang, L.; Li, Y.; Shamaileh, H.A.; Yin, W.; Zhou, S.F.; Zhao, X.; et al. Cancer stem cell targeted therapy: Progress amid controversies. Oncotarget 2015, 6, 44191–44206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ajani, J.A.; Song, S.; Hochster, H.S.; Steinberg, I.B. Cancer stem cells: The promise and the potential. Semin. Oncol. 2015, 42 (Suppl. S1), S3–S17. [Google Scholar] [CrossRef] [PubMed]
- Ramos, E.K.; Hoffmann, A.D.; Gerson, S.L.; Liu, H. New Opportunities and Challenges to Defeat Cancer Stem Cells. Trends Cancer 2017, 3, 780–796. [Google Scholar] [CrossRef]
- Afify, S.M.; Seno, M. Conversion of Stem Cells to Cancer Stem Cells: Undercurrent of Cancer Initiation. Cancers 2019, 11, 345. [Google Scholar] [CrossRef] [Green Version]
- Schofield, R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells 1978, 4, 7–25. [Google Scholar]
- Scadden, D.T. Nice neighborhood: Emerging concepts of the stem cell niche. Cell 2014, 157, 41–50. [Google Scholar] [CrossRef] [Green Version]
- Plaks, V.; Kong, N.; Werb, Z. The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 2015, 16, 225–238. [Google Scholar] [CrossRef] [Green Version]
- Gascard, P.; Tlsty, T.D. Carcinoma-associated fibroblasts: Orchestrating the composition of malignancy. Genes Dev. 2016, 30, 1002–1019. [Google Scholar] [CrossRef]
- Relation, T.; Dominici, M.; Horwitz, E.M. Concise Review: An (Im)Penetrable Shield: How the Tumor Microenvironment Protects Cancer Stem Cells. Stem Cells 2017, 35, 1123–1130. [Google Scholar] [CrossRef] [Green Version]
- Ravindran, S.; Rasool, S.; Maccalli, C. The Cross Talk between Cancer Stem Cells/Cancer Initiating Cells and Tumor Microenvironment: The Missing Piece of the Puzzle for the Efficient Targeting of these Cells with Immunotherapy. Cancer Microenviron. 2019, 12, 133–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, Z.; Fu, Y.X.; Peng, H. Targeting tumor cells with antibodies enhances anti-tumor immunity. Biophys. Rep. 2018, 4, 243–253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gajria, D.; Chandarlapaty, S. HER2-amplified breast cancer: Mechanisms of trastuzumab resistance and novel targeted therapies. Expert Rev. Anticancer 2011, 11, 263–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ferrara, N.; Hillan, K.J.; Novotny, W. Bevacizumab (Avastin), a humanized anti-VEGF monoclonal antibody for cancer therapy. Biochem. Biophys. Res. Commun. 2005, 333, 328–335. [Google Scholar] [CrossRef] [PubMed]
- Linke, R.; Klein, A.; Seimetz, D. Catumaxomab: Clinical development and future directions. MAbs 2010, 2, 129–136. [Google Scholar] [CrossRef] [Green Version]
- Weiner, L.M.; Dhodapkar, M.V.; Ferrone, S. Monoclonal antibodies for cancer immunotherapy. Lancet 2009, 373, 1033–1040. [Google Scholar] [CrossRef] [Green Version]
- Andrews, L.P.; Yano, H.; Vignali, D.A.A. Inhibitory receptors and ligands beyond PD-1, PD-L1 and CTLA-4: Breakthroughs or backups. Nat. Immunol. 2019, 20, 1425–1434. [Google Scholar] [CrossRef]
- Adkins, H.B.; Bianco, C.; Schiffer, S.G.; Rayhorn, P.; Zafari, M.; Cheung, A.E.; Orozco, O.; Olson, D.; De Luca, A.; Chen, L.L.; et al. Antibody blockade of the Cripto CFC domain suppresses tumor cell growth in vivo. J. Clin. Investig. 2003, 112, 575–587. [Google Scholar] [CrossRef] [Green Version]
- Kelly, R.K.; Olson, D.L.; Sun, Y.; Wen, D.; Wortham, K.A.; Antognetti, G.; Cheung, A.E.; Orozco, O.E.; Yang, L.; Bailly, V.; et al. An antibody-cytotoxic conjugate, BIIB015, is a new targeted therapy for Cripto positive tumours. Eur. J. Cancer 2011, 47, 1736–1746. [Google Scholar] [CrossRef]
- Xing, P.X.; Hu, X.F.; Pietersz, G.A.; Hosick, H.L.; McKenzie, I.F. Cripto: A novel target for antibody-based cancer immunotherapy. Cancer Res. 2004, 64, 4018–4023. [Google Scholar] [CrossRef] [Green Version]
- Hu, X.F.; Li, J.; Yang, E.; Vandervalk, S.; Xing, P.X. Anti-Cripto Mab inhibit tumour growth and overcome MDR in a human leukaemia MDR cell line by inhibition of Akt and activation of JNK/SAPK and bad death pathways. Br. J. Cancer 2007, 96, 918–927. [Google Scholar] [CrossRef] [Green Version]
- Ishii, H.; Zahra, M.H.; Takayanagi, A.; Seno, M. A Novel Artificially Humanized Anti-Cripto-1 Antibody Suppressing Cancer Cell Growth. Int. J. Mol. Sci. 2021, 22, 1709. [Google Scholar] [CrossRef]
- Deng, Z.; Wu, Y.; Ma, W.; Zhang, S.; Zhang, Y.Q. Adoptive T-cell therapy of prostate cancer targeting the cancer stem cell antigen EpCAM. BMC Immunol. 2015, 16, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Emlet, D.R.; Gupta, P.; Holgado-Madruga, M.; Del Vecchio, C.A.; Mitra, S.S.; Han, S.Y.; Li, G.; Jensen, K.C.; Vogel, H.; Xu, L.W.; et al. Targeting a glioblastoma cancer stem-cell population defined by EGF receptor variant III. Cancer Res. 2014, 74, 1238–1249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ciccodicola, A.; Dono, R.; Obici, S.; Simeone, A.; Zollo, M.; Persico, M.G. Molecular characterization of a gene of the ‘EGF family’ expressed in undifferentiated human NTERA2 teratocarcinoma cells. EMBO J. 1989, 8, 1987–1991. [Google Scholar] [CrossRef] [PubMed]
- Normanno, N.; Qi, C.F.; Gullick, W.J.; Persico, G.; Yarden, Y.; Wen, D.Z.; Plowman, G.; Kenney, N.; Johnson, G.; Kim, N.; et al. Expression of amphiregulin, Cripto-1, and heregulin-alpha in human breast-cancer cells. Int. J. Oncol. 1993, 2, 903–911. [Google Scholar] [CrossRef]
- Qi, C.F.; Liscia, D.S.; Normanno, N.; Merlo, G.; Johnson, G.R.; Gullick, W.J.; Ciardiello, F.; Saeki, T.; Brandt, R.; Kim, N.; et al. Expression of transforming growth factor alpha, amphiregulin and cripto-1 in human breast carcinomas. Br. J. Cancer 1994, 69, 903–910. [Google Scholar] [CrossRef] [Green Version]
- Friess, H.; Yamanaka, Y.; Büchler, M.; Kobrin, M.S.; Tahara, E.; Korc, M. Cripto, a member of the epidermal growth factor family, is over-expressed in human pancreatic cancer and chronic pancreatitis. Int. J. Cancer 1994, 56, 668–674. [Google Scholar] [CrossRef]
- Baldassarre, G.; Romano, A.; Armenante, F.; Rambaldi, M.; Paoletti, I.; Sandomenico, C.; Pepe, S.; Staibano, S.; Salvatore, G.; De Rosa, G.; et al. Expression of teratocarcinoma-derived growth factor-1 (TDGF-1) in testis germ cell tumors and its effects on growth and differentiation of embryonal carcinoma cell line NTERA2/D1. Oncogene 1997, 15, 927–936. [Google Scholar] [CrossRef] [Green Version]
- Byrne, R.L.; Autzen, P.; Birch, P.; Robinson, M.C.; Gullick, W.J.; Neal, D.E.; Hamdy, F.C. The immunohistochemical detection of cripto-1 in benign and malignant human bladder. J. Pathol. 1998, 185, 108–111. [Google Scholar] [CrossRef]
- Ding, J.; Yang, L.; Yan, Y.T.; Chen, A.; Desai, N.; Wynshaw-Boris, A.; Shen, M.M. Cripto is required for correct orientation of the anterior-posterior axis in the mouse embryo. Nature 1998, 395, 702–707. [Google Scholar] [CrossRef]
- Strizzi, L.; Bianco, C.; Normanno, N.; Salomon, D. Cripto-1: A multifunctional modulator during embryogenesis and oncogenesis. Oncogene 2005, 24, 5731–5741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Klauzinska, M.; Castro, N.P.; Rangel, M.C.; Spike, B.T.; Gray, P.C.; Bertolette, D.; Cuttitta, F.; Salomon, D. The multifaceted role of the embryonic gene Cripto-1 in cancer, stem cells and epithelial-mesenchymal transition. Semin. Cancer Biol. 2014, 29, 51–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hu, X.F.; Xing, P.X. Cripto as a target for cancer immunotherapy. Expert Opin. Targets 2005, 9, 383–394. [Google Scholar] [CrossRef] [PubMed]
- Gong, W.; Sun, B.; Zhao, X.; Zhang, D.; Sun, J.; Liu, T.; Gu, Q.; Dong, X.; Liu, F.; Wang, Y.; et al. Nodal signaling promotes vasculogenic mimicry formation in breast cancer via the Smad2/3 pathway. Oncotarget 2016, 7, 70152–70167. [Google Scholar] [CrossRef] [Green Version]
- Kirsammer, G.; Strizzi, L.; Margaryan, N.V.; Gilgur, A.; Hyser, M.; Atkinson, J.; Kirschmann, D.A.; Seftor, E.A.; Hendrix, M.J. Nodal signaling promotes a tumorigenic phenotype in human breast cancer. Semin. Cancer Biol. 2014, 29, 40–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vo, B.T.; Cody, B.; Cao, Y.; Khan, S.A. Differential role of Sloan-Kettering Institute (Ski) protein in Nodal and transforming growth factor-beta (TGF-β)-induced Smad signaling in prostate cancer cells. Carcinogenesis 2012, 33, 2054–2064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reissmann, E.; Jörnvall, H.; Blokzijl, A.; Andersson, O.; Chang, C.; Minchiotti, G.; Persico, M.G.; Ibáñez, C.F.; Brivanlou, A.H. The orphan receptor ALK7 and the Activin receptor ALK4 mediate signaling by Nodal proteins during vertebrate development. Genes Dev. 2001, 15, 2010–2022. [Google Scholar] [CrossRef] [Green Version]
- Yeo, C.; Whitman, M. Nodal signals to Smads through Cripto-dependent and Cripto-independent mechanisms. Mol. Cell 2001, 7, 949–957. [Google Scholar] [CrossRef]
- Bianco, C.; Adkins, H.B.; Wechselberger, C.; Seno, M.; Normanno, N.; De Luca, A.; Sun, Y.; Khan, N.; Kenney, N.; Ebert, A.; et al. Cripto-1 activates nodal- and ALK4-dependent and -independent signaling pathways in mammary epithelial Cells. Mol. Cell. Biol. 2002, 22, 2586–2597. [Google Scholar] [CrossRef] [Green Version]
- Schier, A.F. Nodal signaling in vertebrate development. Annu. Rev. Cell Dev. Biol. 2003, 19, 589–621. [Google Scholar] [CrossRef]
- Gray, P.C.; Harrison, C.A.; Vale, W. Cripto forms a complex with activin and type II activin receptors and can block activin signaling. Proc. Natl. Acad. Sci. USA 2003, 100, 5193–5198. [Google Scholar] [CrossRef] [Green Version]
- Bianco, C.; Normanno, N.; De Luca, A.; Maiello, M.R.; Wechselberger, C.; Sun, Y.; Khan, N.; Adkins, H.; Sanicola, M.; Vonderhaar, B.; et al. Detection and localization of Cripto-1 binding in mouse mammary epithelial cells and in the mouse mammary gland using an immunoglobulin-cripto-1 fusion protein. J. Cell. Physiol. 2002, 190, 74–82. [Google Scholar] [CrossRef] [PubMed]
- Attisano, L.; Silvestri, C.; Izzi, L.; Labbé, E. The transcriptional role of Smads and FAST (FoxH1) in TGFbeta and activin signalling. Mol. Cell. Endocrinol. 2001, 180, 3–11. [Google Scholar] [CrossRef]
- Yamamoto, M.; Meno, C.; Sakai, Y.; Shiratori, H.; Mochida, K.; Ikawa, Y.; Saijoh, Y.; Hamada, H. The transcription factor FoxH1 (FAST) mediates Nodal signaling during anterior-posterior patterning and node formation in the mouse. Genes Dev. 2001, 15, 1242–1256. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Zhang, L.; Meng, Y.; Huang, L. Benzyl isothiocyanate inhibits breast cancer cell tumorigenesis via repression of the FoxH1-Mediated Wnt/β-catenin pathway. Int. J. Clin. Exp. Med. 2015, 8, 17601–17611. [Google Scholar]
- Bianco, C.; Strizzi, L.; Rehman, A.; Normanno, N.; Wechselberger, C.; Sun, Y.; Khan, N.; Hirota, M.; Adkins, H.; Williams, K.; et al. A Nodal- and ALK4-independent signaling pathway activated by Cripto-1 through Glypican-1 and c-Src. Cancer Res. 2003, 63, 1192–1197. [Google Scholar]
- Kannan, S.; De Santis, M.; Lohmeyer, M.; Riese, D.J., 2nd; Smith, G.H.; Hynes, N.; Seno, M.; Brandt, R.; Bianco, C.; Persico, G.; et al. Cripto enhances the tyrosine phosphorylation of Shc and activates mitogen-activated protein kinase (MAPK) in mammary epithelial cells. J. Biol. Chem. 1997, 272, 3330–3335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bianco, C.; Kannan, S.; De Santis, M.; Seno, M.; Tang, C.K.; Martinez-Lacaci, I.; Kim, N.; Wallace-Jones, B.; Lippman, M.E.; Ebert, A.D.; et al. Cripto-1 indirectly stimulates the tyrosine phosphorylation of erb B-4 through a novel receptor. J. Biol. Chem. 1999, 274, 8624–8629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Watanabe, K.; Yamada, H.; Yamaguchi, Y. K-glypican: A novel GPI-anchored heparan sulfate proteoglycan that is highly expressed in developing brain and kidney. J. Cell Biol. 1995, 130, 1207–1218. [Google Scholar] [CrossRef] [Green Version]
- Bianco, C.; Mysliwiec, M.; Watanabe, K.; Mancino, M.; Nagaoka, T.; Gonzales, M.; Salomon, D.S. Activation of a Nodal-independent signaling pathway by Cripto-1 mutants with impaired activation of a Nodal-dependent signaling pathway. FEBS Lett. 2008, 582, 3997–4002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Normanno, N.; De Luca, A.; Bianco, C.; Maiello, M.R.; Carriero, M.V.; Rehman, A.; Wechselberger, C.; Arra, C.; Strizzi, L.; Sanicola, M.; et al. Cripto-1 overexpression leads to enhanced invasiveness and resistance to anoikis in human MCF-7 breast cancer cells. J. Cell. Physiol. 2004, 198, 31–39. [Google Scholar] [CrossRef]
- Zhang, W.; Liu, H.T. MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Res. 2002, 12, 9–18. [Google Scholar] [CrossRef] [PubMed]
- Dillekås, H.; Rogers, M.S.; Straume, O. Are 90% of deaths from cancer caused by metastases? Cancer Med. 2019, 8, 5574–5576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dillekås, H.; Demicheli, R.; Ardoino, I.; Jensen, S.A.H.; Biganzoli, E.; Straume, O. The recurrence pattern following delayed breast reconstruction after mastectomy for breast cancer suggests a systemic effect of surgery on occult dormant micrometastases. Breast Cancer Res. Treat. 2016, 158, 169–178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hu, Z.; Li, Z.; Ma, Z.; Curtis, C. Multi-cancer analysis of clonality and the timing of systemic spread in paired primary tumors and metastases. Nat. Genet. 2020, 52, 701–708. [Google Scholar] [CrossRef]
- Mahvi, D.A.; Liu, R.; Grinstaff, M.W.; Colson, Y.L.; Raut, C.P. Local Cancer Recurrence: The Realities, Challenges, and Opportunities for New Therapies. CA Cancer J. Clin. 2018, 68, 488–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zoni, E.; Chen, L.; Karkampouna, S.; Granchi, Z.; Verhoef, E.I.; La Manna, F.; Kelber, J.; Pelger, R.C.M.; Henry, M.D.; Snaar-Jagalska, E.; et al. CRIPTO and its signaling partner GRP78 drive the metastatic phenotype in human osteotropic prostate cancer. Oncogene 2017, 36, 4739–4749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, K.-S.; Raffeld, M.; Moon, Y.W.; Xi, L.; Bianco, C.; Pham, T.; Lee, L.C.; Mitsudomi, T.; Yatabe, Y.; Okamoto, I.; et al. CRIPTO1 expression in EGFR-mutant NSCLC elicits intrinsic EGFR-inhibitor resistance. J. Clin. Investig. 2014, 124, 3003–3015. [Google Scholar] [CrossRef] [Green Version]
- Focà, G.; Iaccarino, E.; Focà, A.; Sanguigno, L.; Untiveros, G.; Cuevas-Nunez, M.; Strizzi, L.; Leonardi, A.; Ruvo, M.; Sandomenico, A. Development of conformational antibodies targeting Cripto-1 with neutralizing effects in vitro. Biochimie 2019, 158, 246–256. [Google Scholar] [CrossRef]
- Watanabe, K.; Meyer, M.J.; Strizzi, L.; Lee, J.M.; Gonzales, M.; Bianco, C.; Nagaoka, T.; Farid, S.S.; Margaryan, N.; Hendrix, M.J.C.; et al. Cripto-1 is a cell surface marker for a tumorigenic, undifferentiated subpopulation in human embryonal carcinoma cells. Stem Cells 2010, 28, 1303–1314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Organ | Cancer Cells | CSCs |
---|---|---|
Breast | Regulation of human Cripto-1 expression by nuclear receptors and DNA promoter methylation in human embryonal and breast cancer cells [45] | Cripto-1 Plasmid DNA Vaccination Targets Metastasis and Cancer Stem Cells in Murine Mammary Carcinoma [38] |
Cripto-1 as a novel therapeutic target for triple-negative breast cancer [46] | ||
Brain | Cripto-1 overexpression in U87 glioblastoma cells activates MAPK, focal adhesion, and ErbB pathways [43] | |
Investigating the role of CRIPTO-1 (TDGF-1) in glioblastoma multiforme U87 cell line [47] | ||
Cripto-1 localizes to dynamic and shed filopodia associated with cellular migration in glioblastoma cells [48] | ||
The others | Overexpression levels of cripto-1 predict poor prognosis in patients with prostate cancer following radical prostatectomy [49] | Cripto-1 acts as a functional marker of cancer stem-like cells and predicts prognosis of the patients in esophageal squamous cell carcinoma [25] |
Expression and functional role of CRIPTO-1 in cutaneous melanoma [50] | Cripto-1 contributes to stemness in hepatocellular carcinoma by stabilizing Dishevelled-3 and activating Wnt/β-catenin pathway [23] | |
The role of Nodal and Cripto-1 in human oral squamous cell carcinoma [42] | Exogenous Cripto-1 Suppresses Self-Renewal of Cancer Stem Cell Model [51] | |
CRIPTO promotes an aggressive tumor phenotype and resistance to treatment in hepatocellular carcinoma [52] | Dynamic regulation of the cancer stem cell compartment by Cripto-1 in colorectal cancer [24] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Ishii, H.; Afify, S.M.; Hassan, G.; Salomon, D.S.; Seno, M. Cripto-1 as a Potential Target of Cancer Stem Cells for Immunotherapy. Cancers 2021, 13, 2491. https://doi.org/10.3390/cancers13102491
Ishii H, Afify SM, Hassan G, Salomon DS, Seno M. Cripto-1 as a Potential Target of Cancer Stem Cells for Immunotherapy. Cancers. 2021; 13(10):2491. https://doi.org/10.3390/cancers13102491
Chicago/Turabian StyleIshii, Hiroko, Said M. Afify, Ghmkin Hassan, David S. Salomon, and Masaharu Seno. 2021. "Cripto-1 as a Potential Target of Cancer Stem Cells for Immunotherapy" Cancers 13, no. 10: 2491. https://doi.org/10.3390/cancers13102491
APA StyleIshii, H., Afify, S. M., Hassan, G., Salomon, D. S., & Seno, M. (2021). Cripto-1 as a Potential Target of Cancer Stem Cells for Immunotherapy. Cancers, 13(10), 2491. https://doi.org/10.3390/cancers13102491