Clinical Potential of YY1-Hypoxia Axis for Vascular Normalization and to Improve Immunotherapy
Abstract
:Simple Summary
Abstract
1. Introduction
2. Vascular and Immune Crosstalk Normalization
3. Vascular Signaling by YY1
3.1. HIF-YY1 Vascular Signaling Axis
3.2. YY1 Promotes Tumor Angiogenesis in HIF-1α-Independent Manners
3.3. YY1-Related Non-Coding RNAs (ncRNAs) in Angiogenic Mechanisms
4. Immune Cells Regulate Tumor Angiogenesis
5. YY1 in B and T Cells
6. Preclinical Model and Clinical Findings of Vascular Normalization and Immune Modulation
7. Novel Strategies: Targeted Therapy
8. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Folkman, J. Tumor angiogenesis: Therapeutic implications. N. Engl. J. Med. 1971, 285, 1182–1186. [Google Scholar]
- Welti, J.; Loges, S.; Dimmeler, S.; Carmeliet, P. Recent molecular discoveries in angiogenesis and antiangiogenic therapies in cancer. J. Clin. Invest. 2013, 123, 3190–3200. [Google Scholar] [CrossRef]
- Liu, Z.L.; Chen, H.H.; Zheng, L.L.; Sun, L.P.; Shi, L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct. Target. Ther. 2023, 8, 198. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
- Gabrilovich, D.I.; Chen, H.L.; Girgis, K.R.; Cunningham, H.T.; Meny, G.M.; Nadaf, S.; Kavanaugh, D.; Carbone, D.P. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat. Med. 1996, 2, 1096–1103. [Google Scholar] [CrossRef]
- Albini, A.; Bruno, A.; Noonan, D.M.; Mortara, L. Contribution to Tumor Angiogenesis From Innate Immune Cells within the Tumor Microenvironment: Implications for Immunotherapy. Front. Immunol. 2018, 9, 527. [Google Scholar] [CrossRef] [PubMed]
- Zhao, C.C.; Han, Q.J.; Ying, H.Y.; Gu, X.X.; Yang, N.; Li, L.Y.; Zhang, Q.Z. TNFSF15 facilitates differentiation and polarization of macrophages toward M1 phenotype to inhibit tumor growth. Oncoimmunology 2022, 11, 2032918. [Google Scholar] [CrossRef] [PubMed]
- Ge, Z.; Zhang, Q.; Lin, W.; Jiang, X.; Zhang, Y. The role of angiogenic growth factors in the immune microenvironment of glioma. Front. Oncol. 2023, 13, 1254694. [Google Scholar] [CrossRef]
- Facciabene, A.; Motz, G.T.; Coukos, G. T-regulatory cells: Key players in tumor immune escape and angiogenesis. Cancer Res. 2012, 72, 2162–2171. [Google Scholar] [CrossRef]
- Bruno, A.; Pagani, A.; Pulze, L.; Albini, A.; Dallaglio, K.; Noonan, D.M.; Mortara, L. Orchestration of angiogenesis by immune cells. Front. Oncol. 2014, 4, 131. [Google Scholar] [CrossRef]
- Cao, Y.; Langer, R.; Ferrara, N. Targeting angiogenesis in oncology, ophthalmology and beyond. Nat. Rev. Drug Discov. 2023, 22, 476–495. [Google Scholar] [CrossRef]
- Jayson, G.C.; Kerbel, R.; Ellis, L.M.; Harris, A.L. Antiangiogenic therapy in oncology: Current status and future directions. Lancet 2016, 388, 518–529. [Google Scholar] [CrossRef]
- Jain, R.K. Normalizing tumor vasculature with anti-angiogenic therapy: A new paradigm for combination therapy. Nat. Med. 2001, 7, 987–989. [Google Scholar] [CrossRef]
- Fang, J.; Lu, Y.; Zheng, J.; Jiang, X.; Shen, H.; Shang, X.; Lu, Y.; Fu, P. Exploring the crosstalk between endothelial cells, immune cells, and immune checkpoints in the tumor microenvironment: New insights and therapeutic implications. Cell Death Dis. 2023, 14, 586. [Google Scholar] [CrossRef]
- Fan, P.; Zhang, N.; Candi, E.; Agostini, M.; Piacentini, M.; Centre, T.O.R.; Shi, Y.; Huang, Y.; Melino, G. Alleviating hypoxia to improve cancer immunotherapy. Oncogene 2023, 42, 3591–3604. [Google Scholar] [CrossRef]
- Choi, Y.; Jung, K. Normalization of the tumor microenvironment by harnessing vascular and immune modulation to achieve enhanced cancer therapy. Exp. Mol. Med. 2023, 55, 2308–2319. [Google Scholar] [CrossRef]
- Binnewies, M.; Roberts, E.W.; Kersten, K.; Chan, V.; Fearon, D.F.; Merad, M.; Coussens, L.M.; Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Hedrick, C.C.; et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 2018, 24, 541–550. [Google Scholar] [CrossRef]
- Fukumura, D.; Kloepper, J.; Amoozgar, Z.; Duda, D.G.; Jain, R.K. Enhancing cancer immunotherapy using antiangiogenics: Opportunities and challenges. Nat. Rev. Clin. Oncol. 2018, 15, 325–340. [Google Scholar] [CrossRef] [PubMed]
- Mpekris, F.; Voutouri, C.; Baish, J.W.; Duda, D.G.; Munn, L.L.; Stylianopoulos, T.; Jain, R.K. Combining microenvironment normalization strategies to improve cancer immunotherapy. Proc. Natl. Acad. Sci. USA 2020, 117, 3728–3737. [Google Scholar] [CrossRef]
- Lee, W.S.; Yang, H.; Chon, H.J.; Kim, C. Combination of anti-angiogenic therapy and immune checkpoint blockade normalizes vascular-immune crosstalk to potentiate cancer immunity. Exp. Mol. Med. 2020, 52, 1475–1485. [Google Scholar] [CrossRef]
- Patel, S.A.; Nilsson, M.B.; Le, X.; Cascone, T.; Jain, R.K.; Heymach, J.V. Molecular Mechanisms and Future Implications of VEGF/VEGFR in Cancer Therapy. Clin. Cancer Res. 2023, 29, 30–39. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.W.; Bae, S.H.; Jeong, J.W.; Kim, S.H.; Kim, K.W. Hypoxia-inducible factor (HIF-1)alpha: Its protein stability and biological functions. Exp. Mol. Med. 2004, 36, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Hashimoto, T.; Shibasaki, F. Hypoxia-inducible factor as an angiogenic master switch. Front. Pediatr. 2015, 3, 33. [Google Scholar] [CrossRef]
- Infantino, V.; Santarsiero, A.; Convertini, P.; Todisco, S.; Iacobazzi, V. Cancer Cell Metabolism in Hypoxia: Role of HIF-1 as Key Regulator and Therapeutic Target. Int. J. Mol. Sci. 2021, 22, 5703. [Google Scholar] [CrossRef] [PubMed]
- Thomas, M.J.; Seto, E. Unlocking the mechanisms of transcription factor YY1: Are chromatin modifying enzymes the key? Gene 1999, 236, 197–208. [Google Scholar] [CrossRef] [PubMed]
- Gordon, S.; Akopyan, G.; Garban, H.; Bonavida, B. Transcription factor YY1: Structure, function, and therapeutic implications in cancer biology. Oncogene 2006, 25, 1125–1142. [Google Scholar] [CrossRef]
- Nicholson, S.; Whitehouse, H.; Naidoo, K.; Byers, R.J. Yin Yang 1 in human cancer. Crit. Rev. Oncog. 2011, 16, 245–260. [Google Scholar] [CrossRef] [PubMed]
- Weintraub, A.S.; Li, C.H.; Zamudio, A.V.; Sigova, A.A.; Hannett, N.M.; Day, D.S.; Abraham, B.J.; Cohen, M.A.; Nabet, B.; Buckley, D.L.; et al. YY1 Is a Structural Regulator of Enhancer-Promoter Loops. Cell 2017, 171, 1573–1588.e28. [Google Scholar] [CrossRef]
- Seligson, D.; Horvath, S.; Huerta-Yepez, S.; Hanna, S.; Garban, H.; Roberts, A.; Shi, T.; Liu, X.; Chia, D.; Goodglick, L.; et al. Expression of transcription factor Yin Yang 1 in prostate cancer. Int. J. Oncol. 2005, 27, 131–141. [Google Scholar] [CrossRef] [PubMed]
- De Nigris, F.; Botti, C.; de Chiara, A.; Rossiello, R.; Apice, G.; Fazioli, F.; Fiorito, C.; Sica, V.; Napoli, C. Expression of transcription factor Yin Yang 1 in human osteosarcomas. Eur. J. Cancer 2006, 42, 2420–2424. [Google Scholar] [CrossRef]
- Allouche, A.; Nolens, G.; Tancredi, A.; Delacroix, L.; Mardaga, J.; Fridman, V.; Winkler, R.; Boniver, J.; Delvenne, P.; Begon, D.Y. The combined immunodetection of AP-2alpha and YY1 transcription factors is associated with ERBB2 gene overexpression in primary breast tumors. Breast Cancer Res. 2008, 10, R9. [Google Scholar] [CrossRef] [PubMed]
- Baritaki, S.; Chatzinikola, A.M.; Vakis, A.F.; Soulitzis, N.; Karabetsos, D.A.; Neonakis, I.; Bonavida, B.; Spandidos, D.A. YY1 Over-expression in human brain gliomas and meningiomas correlates with TGF-beta1, IGF-1 and FGF-2 mRNA levels. Cancer Invest. 2009, 27, 184–192. [Google Scholar] [CrossRef] [PubMed]
- Chinnappan, D.; Xiao, D.; Ratnasari, A.; Andry, C.; King, T.C.; Weber, H.C. Transcription factor YY1 expression in human gastrointestinal cancer cells. Int. J. Oncol. 2009, 34, 1417–1423. [Google Scholar] [PubMed]
- Zhang, J.J.; Zhu, Y.; Xie, K.L.; Peng, Y.P.; Tao, J.Q.; Tang, J.; Li, Z.; Xu, Z.K.; Dai, C.C.; Qian, Z.Y.; et al. Yin Yang-1 suppresses invasion and metastasis of pancreatic ductal adenocarcinoma by downregulating MMP10 in a MUC4/ErbB2/p38/MEF2C-dependent mechanism. Mol. Cancer 2014, 13, 130. [Google Scholar] [CrossRef] [PubMed]
- Zhao, G.; Li, Q.; Wang, A.; Jiao, J. YY1 regulates melanoma tumorigenesis through a miR-9~RYBP axis. J. Exp. Clin. Cancer Res. 2015, 34, 66. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Li, D.; Sui, G. YY1 Is an Inducer of Cancer Metastasis. Crit. Rev. Oncog. 2017, 22, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Riggs, K.J.; Saleque, S.; Wong, K.K.; Merrell, K.T.; Lee, J.S.; Shi, Y.; Calame, K. Yin-yang 1 activates the c-myc promoter. Mol. Cell. Biol. 1993, 13, 7487–7495. [Google Scholar] [PubMed]
- Zhang, Q.; Wan, M.; Shi, J.; Horita, D.A.; Miller, L.D.; Kute, T.E.; Kridel, S.J.; Kulik, G.; Sui, G. Yin Yang 1 promotes mTORC2-mediated AKT phosphorylation. J. Mol. Cell Biol. 2016, 8, 232–243. [Google Scholar] [CrossRef] [PubMed]
- Sui, G.; Affar, B.; Shi, Y.; Brignone, C.; Wall, N.R.; Yin, P.; Donohoe, M.; Luke, M.P.; Calvo, D.; Grossman, S.R.; et al. Yin Yang 1 is a negative regulator of p53. Cell 2004, 117, 859–872. [Google Scholar] [CrossRef]
- Gronroos, E.; Terentiev, A.A.; Punga, T.; Ericsson, J. YY1 inhibits the activation of the p53 tumor suppressor in response to genotoxic stress. Proc. Natl. Acad. Sci. USA 2004, 101, 12165–12170. [Google Scholar] [CrossRef]
- De Nigris, F.; Rossiello, R.; Schiano, C.; Arra, C.; Williams-Ignarro, S.; Barbieri, A.; Lanza, A.; Balestrieri, A.; Giuliano, M.T.; Ignarro, L.J.; et al. Deletion of Yin Yang 1 protein in osteosarcoma cells on cell invasion and CXCR4/angiogenesis and metastasis. Cancer Res. 2008, 68, 1797–1808. [Google Scholar] [CrossRef] [PubMed]
- De Nigris, F.; Crudele, V.; Giovane, A.; Casamassimi, A.; Giordano, A.; Garban, H.J.; Cacciatore, F.; Pentimalli, F.; Marquez-Garban, D.C.; Petrillo, A.; et al. CXCR4/YY1 inhibition impairs VEGF network and angiogenesis during malignancy. Proc. Natl. Acad. Sci. USA 2010, 107, 14484–14489. [Google Scholar] [CrossRef]
- Wu, S.; Kasim, V.; Kano, M.R.; Tanaka, S.; Ohba, S.; Miura, Y.; Miyata, K.; Liu, X.; Matsuhashi, A.; Chung, U.I.; et al. Transcription factor YY1 contributes to tumor growth by stabilizing hypoxia factor HIF-1alpha in a p53-independent manner. Cancer Res. 2013, 73, 1787–1799. [Google Scholar] [CrossRef]
- Yang, W.; Li, Z.; Qin, R.; Wang, X.; An, H.; Wang, Y.; Zhu, Y.; Liu, Y.; Cai, S.; Chen, S.; et al. YY1 Promotes Endothelial Cell-Dependent Tumor Angiogenesis in Hepatocellular Carcinoma by Transcriptionally Activating VEGFA. Front. Oncol. 2019, 9, 1187. [Google Scholar] [CrossRef]
- Infante, T.; Mancini, F.P.; Lanza, A.; Soricelli, A.; de Nigris, F.; Napoli, C. Polycomb YY1 is a critical interface between epigenetic code and miRNA machinery after exposure to hypoxia in malignancy. Biochim. Biophys. Acta 2015, 1853, 975–986. [Google Scholar] [CrossRef]
- Korbecki, J.; Kojder, K.; Kapczuk, P.; Kupnicka, P.; Gawronska-Szklarz, B.; Gutowska, I.; Chlubek, D.; Baranowska-Bosiacka, I. The Effect of Hypoxia on the Expression of CXC Chemokines and CXC Chemokine Receptors—A Review of Literature. Int. J. Mol. Sci. 2021, 22, 843. [Google Scholar] [CrossRef]
- Zhang, Y.; Liu, J.; Wang, S.; Luo, X.; Li, Y.; Lv, Z.; Zhu, J.; Lin, J.; Ding, L.; Ye, Q. The DEK oncogene activates VEGF expression and promotes tumor angiogenesis and growth in HIF-1alpha-dependent and -independent manners. Oncotarget 2016, 7, 23740–23756. [Google Scholar] [CrossRef]
- Han, S.; Xuan, Y.; Liu, S.; Zhang, M.; Jin, D.; Jin, R.; Lin, Z. Clinicopathological significance of DEK overexpression in serous ovarian tumors. Pathol. Int. 2009, 59, 443–447. [Google Scholar] [CrossRef]
- Shibata, T.; Kokubu, A.; Miyamoto, M.; Hosoda, F.; Gotoh, M.; Tsuta, K.; Asamura, H.; Matsuno, Y.; Kondo, T.; Imoto, I.; et al. DEK oncoprotein regulates transcriptional modifiers and sustains tumor initiation activity in high-grade neuroendocrine carcinoma of the lung. Oncogene 2010, 29, 4671–4681. [Google Scholar] [CrossRef]
- Wang, X.; Lin, L.; Ren, X.; Lin, Z.; Li, Z.; Li, C.; Jin, T. High expression of oncoprotein DEK predicts poor prognosis of small cell lung cancer. Int. J. Clin. Exp. Pathol. 2014, 7, 5016–5023. [Google Scholar]
- Deutzmann, A.; Ganz, M.; Schonenberger, F.; Vervoorts, J.; Kappes, F.; Ferrando-May, E. The human oncoprotein and chromatin architectural factor DEK counteracts DNA replication stress. Oncogene 2015, 34, 4270–4277. [Google Scholar] [CrossRef]
- McGarvey, T.; Rosonina, E.; McCracken, S.; Li, Q.; Arnaout, R.; Mientjes, E.; Nickerson, J.A.; Awrey, D.; Greenblatt, J.; Grosveld, G.; et al. The acute myeloid leukemia-associated protein, DEK, forms a splicing-dependent interaction with exon-product complexes. J. Cell Biol. 2000, 150, 309–320. [Google Scholar] [CrossRef]
- Ageberg, M.; Gullberg, U.; Lindmark, A. The involvement of cellular proliferation status in the expression of the human proto-oncogene DEK. Haematologica 2006, 91, 268–269. [Google Scholar]
- Wise-Draper, T.M.; Allen, H.V.; Jones, E.E.; Habash, K.B.; Matsuo, H.; Wells, S.I. Apoptosis inhibition by the human DEK oncoprotein involves interference with p53 functions. Mol. Cell. Biol. 2006, 26, 7506–7519. [Google Scholar] [CrossRef]
- Von Lindern, M.; Fornerod, M.; van Baal, S.; Jaegle, M.; de Wit, T.; Buijs, A.; Grosveld, G. The translocation (6;9), associated with a specific subtype of acute myeloid leukemia, results in the fusion of two genes, dek and can, and the expression of a chimeric, leukemia-specific dek-can mRNA. Mol. Cell. Biol. 1992, 12, 1687–1697. [Google Scholar]
- Sitwala, K.V.; Adams, K.; Markovitz, D.M. YY1 and NF-Y binding sites regulate the transcriptional activity of the dek and dek-can promoter. Oncogene 2002, 21, 8862–8870. [Google Scholar] [CrossRef]
- Sparmann, A.; Bar-Sagi, D. Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. Cancer Cell 2004, 6, 447–458. [Google Scholar] [CrossRef]
- Ancrile, B.B.; O’Hayer, K.M.; Counter, C.M. Oncogenic ras-induced expression of cytokines: A new target of anti-cancer therapeutics. Mol. Interv. 2008, 8, 22–27. [Google Scholar] [CrossRef]
- Watnick, R.S.; Cheng, Y.N.; Rangarajan, A.; Ince, T.A.; Weinberg, R.A. Ras modulates Myc activity to repress thrombospondin-1 expression and increase tumor angiogenesis. Cancer Cell 2003, 3, 219–231. [Google Scholar] [CrossRef]
- Lin, C.C.; Kuo, I.Y.; Wu, L.T.; Kuan, W.H.; Liao, S.Y.; Jen, J.; Yang, Y.E.; Tang, C.W.; Chen, Y.R.; Wang, Y.C. Dysregulated Kras/YY1/ZNF322A/Shh transcriptional axis enhances neo-angiogenesis to promote lung cancer progression. Theranostics 2020, 10, 10001–10015. [Google Scholar] [CrossRef]
- Boareto, M.; Jolly, M.K.; Ben-Jacob, E.; Onuchic, J.N. Jagged mediates differences in normal and tumor angiogenesis by affecting tip-stalk fate decision. Proc. Natl. Acad. Sci. USA 2015, 112, E3836–E3844. [Google Scholar] [CrossRef]
- Benedito, R.; Roca, C.; Sorensen, I.; Adams, S.; Gossler, A.; Fruttiger, M.; Adams, R.H. The notch ligands Dll4 and Jagged1 have opposing effects on angiogenesis. Cell 2009, 137, 1124–1135. [Google Scholar] [CrossRef]
- Zhang, S.; Kim, J.Y.; Xu, S.; Liu, H.; Yin, M.; Koroleva, M.; Guo, J.; Pei, X.; Jin, Z.G. Endothelial-specific YY1 governs sprouting angiogenesis through directly interacting with RBPJ. Proc. Natl. Acad. Sci. USA 2020, 117, 4792–4801. [Google Scholar] [CrossRef]
- Li, Z.J.; Cheng, J.; Song, Y.; Li, H.H.; Zheng, J.F. LncRNA SNHG5 upregulation induced by YY1 contributes to angiogenesis via miR-26b/CTGF/VEGFA axis in acute myelogenous leukemia. Lab. Invest. 2021, 101, 341–352. [Google Scholar] [CrossRef]
- Chen, Y.; Jacamo, R.; Konopleva, M.; Garzon, R.; Croce, C.; Andreeff, M. CXCR4 downregulation of let-7a drives chemoresistance in acute myeloid leukemia. J. Clin. Invest. 2013, 123, 2395–2407. [Google Scholar] [CrossRef]
- Morales-Martinez, M.; Vega, G.G.; Neri, N.; Nambo, M.J.; Alvarado, I.; Cuadra, I.; Duran-Padilla, M.A.; Huerta-Yepez, S.; Vega, M.I. MicroRNA-7 Regulates Migration and Chemoresistance in Non-Hodgkin Lymphoma Cells Through Regulation of KLF4 and YY1. Front. Oncol. 2020, 10, 588893. [Google Scholar] [CrossRef]
- Li, X.; Yu, M.; Yang, C. YY1-mediated overexpression of long noncoding RNA MCM3AP-AS1 accelerates angiogenesis and progression in lung cancer by targeting miR-340-5p/KPNA4 axis. J. Cell. Biochem. 2020, 121, 2258–2267. [Google Scholar] [CrossRef]
- Xu, Y.; Leng, K.; Yao, Y.; Kang, P.; Liao, G.; Han, Y.; Shi, G.; Ji, D.; Huang, P.; Zheng, W.; et al. A Circular RNA, Cholangiocarcinoma-Associated Circular RNA 1, Contributes to Cholangiocarcinoma Progression, Induces Angiogenesis, and Disrupts Vascular Endothelial Barriers. Hepatology 2021, 73, 1419–1435. [Google Scholar] [CrossRef]
- Ji, Z.Z.; Chan, M.K.; Chan, A.S.; Leung, K.T.; Jiang, X.; To, K.F.; Wu, Y.; Tang, P.M. Tumour-associated macrophages: Versatile players in the tumour microenvironment. Front. Cell Dev. Biol. 2023, 11, 1261749. [Google Scholar] [CrossRef]
- Qian, B.Z.; Pollard, J.W. Macrophage diversity enhances tumor progression and metastasis. Cell 2010, 141, 39–51. [Google Scholar] [CrossRef]
- De Palma, M.; Murdoch, C.; Venneri, M.A.; Naldini, L.; Lewis, C.E. Tie2-expressing monocytes: Regulation of tumor angiogenesis and therapeutic implications. Trends Immunol. 2007, 28, 519–524. [Google Scholar] [CrossRef]
- Yang, L.; DeBusk, L.M.; Fukuda, K.; Fingleton, B.; Green-Jarvis, B.; Shyr, Y.; Matrisian, L.M.; Carbone, D.P.; Lin, P.C. Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell 2004, 6, 409–421. [Google Scholar] [CrossRef]
- Kammertoens, T.; Friese, C.; Arina, A.; Idel, C.; Briesemeister, D.; Rothe, M.; Ivanov, A.; Szymborska, A.; Patone, G.; Kunz, S.; et al. Tumour ischaemia by interferon-gamma resembles physiological blood vessel regression. Nature 2017, 545, 98–102. [Google Scholar] [CrossRef]
- Bromley, S.K.; Mempel, T.R.; Luster, A.D. Orchestrating the orchestrators: Chemokines in control of T cell traffic. Nat. Immunol. 2008, 9, 970–980. [Google Scholar] [CrossRef]
- Tian, L.; Goldstein, A.; Wang, H.; Ching Lo, H.; Sun Kim, I.; Welte, T.; Sheng, K.; Dobrolecki, L.E.; Zhang, X.; Putluri, N.; et al. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature 2017, 544, 250–254. [Google Scholar] [CrossRef]
- Mosca, L.; de Angelis, A.; Ronchi, A.; De Chiara, A.; Fazioli, F.; Ruosi, C.; Altucci, L.; Conte, M.; de Nigris, F. Sarcoma Common MHC-I Haplotype Restricts Tumor-Specific CD8+ T Cell Response. Cancers 2022, 14, 3414. [Google Scholar] [CrossRef]
- Voron, T.; Colussi, O.; Marcheteau, E.; Pernot, S.; Nizard, M.; Pointet, A.L.; Latreche, S.; Bergaya, S.; Benhamouda, N.; Tanchot, C.; et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J. Exp. Med. 2015, 212, 139–148. [Google Scholar] [CrossRef]
- Sanmamed, M.F.; Chen, L. A Paradigm Shift in Cancer Immunotherapy: From Enhancement to Normalization. Cell 2018, 175, 313–326. [Google Scholar] [CrossRef]
- Kleiman, E.; Jia, H.; Loguercio, S.; Su, A.I.; Feeney, A.J. YY1 plays an essential role at all stages of B-cell differentiation. Proc. Natl. Acad. Sci. USA 2016, 113, E3911–E3920. [Google Scholar] [CrossRef]
- Banerjee, A.; Sindhava, V.; Vuyyuru, R.; Jha, V.; Hodewadekar, S.; Manser, T.; Atchison, M.L. YY1 Is Required for Germinal Center B Cell Development. PLoS ONE 2016, 11, e0155311. [Google Scholar] [CrossRef]
- Liu, H.; Schmidt-Supprian, M.; Shi, Y.; Hobeika, E.; Barteneva, N.; Jumaa, H.; Pelanda, R.; Reth, M.; Skok, J.; Rajewsky, K.; et al. Yin Yang 1 is a critical regulator of B-cell development. Genes Dev. 2007, 21, 1179–1189. [Google Scholar] [CrossRef] [PubMed]
- Green, M.R.; Monti, S.; Dalla-Favera, R.; Pasqualucci, L.; Walsh, N.C.; Schmidt-Supprian, M.; Kutok, J.L.; Rodig, S.J.; Neuberg, D.S.; Rajewsky, K.; et al. Signatures of murine B-cell development implicate YY1 as a regulator of the germinal center-specific program. Proc. Natl. Acad. Sci. USA 2011, 108, 2873–2878. [Google Scholar] [CrossRef] [PubMed]
- Arribas Arranz, J.; Winter, D.N.; Drexler, H.G.; Eberth, S. Suitability of Yin Yang 1 transcript and protein levels for biomarker studies in B cell non-Hodgkin lymphoma. Biomark Res. 2018, 6, 11. [Google Scholar] [CrossRef]
- Sakhinia, E.; Glennie, C.; Hoyland, J.A.; Menasce, L.P.; Brady, G.; Miller, C.; Radford, J.A.; Byers, R.J. Clinical quantitation of diagnostic and predictive gene expression levels in follicular and diffuse large B-cell lymphoma by RT-PCR gene expression profiling. Blood 2007, 109, 3922–3928. [Google Scholar] [CrossRef]
- Castellano, G.; Torrisi, E.; Ligresti, G.; Nicoletti, F.; Malaponte, G.; Traval, S.; McCubrey, J.A.; Canevari, S.; Libra, M. Yin Yang 1 overexpression in diffuse large B-cell lymphoma is associated with B-cell transformation and tumor progression. Cell Cycle 2010, 9, 557–563. [Google Scholar] [CrossRef]
- Ramkumar, C.; Cui, H.; Kong, Y.; Jones, S.N.; Gerstein, R.M.; Zhang, H. Smurf2 suppresses B-cell proliferation and lymphomagenesis by mediating ubiquitination and degradation of YY1. Nat. Commun. 2013, 4, 2598. [Google Scholar] [CrossRef]
- Morales-Martinez, M.; Valencia-Hipolito, A.; Vega, G.G.; Neri, N.; Nambo, M.J.; Alvarado, I.; Cuadra, I.; Duran-Padilla, M.A.; Martinez-Maza, O.; Huerta-Yepez, S.; et al. Regulation of Kruppel-Like Factor 4 (KLF4) expression through the transcription factor Yin-Yang 1 (YY1) in non-Hodgkin B-cell lymphoma. Oncotarget 2019, 10, 2173–2188. [Google Scholar] [CrossRef] [PubMed]
- Assumpcao, A.; Fu, G.; Singh, D.K.; Lu, Z.; Kuehnl, A.M.; Welch, R.; Ong, I.M.; Wen, R.; Pan, X. A lineage-specific requirement for YY1 Polycomb Group protein function in early T cell development. Development 2021, 148, dev197319. [Google Scholar] [CrossRef]
- Hwang, S.S.; Jang, S.W.; Kim, M.K.; Kim, L.K.; Kim, B.S.; Kim, H.S.; Kim, K.; Lee, W.; Flavell, R.A.; Lee, G.R. YY1 inhibits differentiation and function of regulatory T cells by blocking Foxp3 expression and activity. Nat. Commun. 2016, 7, 10789. [Google Scholar] [CrossRef]
- Balkhi, M.Y.; Wittmann, G.; Xiong, F.; Junghans, R.P. YY1 Upregulates Checkpoint Receptors and Downregulates Type I Cytokines in Exhausted, Chronically Stimulated Human T Cells. iScience 2018, 2, 105–122. [Google Scholar] [CrossRef]
- Li, C.; Jiang, P.; Wei, S.; Xu, X.; Wang, J. Regulatory T cells in tumor microenvironment: New mechanisms, potential therapeutic strategies and future prospects. Mol. Cancer 2020, 19, 116. [Google Scholar] [CrossRef]
- Butterfield, L.H.; Najjar, Y.G. Immunotherapy combination approaches: Mechanisms, biomarkers and clinical observations. Nat. Rev. Immunol. 2023. [Google Scholar] [CrossRef]
- Schiano, C.; Soricelli, A.; De Nigris, F.; Napoli, C. New challenges in integrated diagnosis by imaging and osteo-immunology in bone lesions. Expert Rev. Clin. Immunol. 2019, 15, 289–301. [Google Scholar] [CrossRef]
- Kwiatkowska, D.; Mazur, E.; Reich, A. YY1 Is a Key Player in Melanoma Immunotherapy/Targeted Treatment Resistance. Front. Oncol. 2022, 12, 856963. [Google Scholar] [CrossRef]
- Tang, X.Y.; Xiong, Y.L.; Zhao, Y.B.; Yang, J.; Shi, A.P.; Zheng, K.F.; Liu, Y.J.; Shu, C.; Jiang, T.; Ma, N.; et al. Dual immunological and proliferative regulation of immune checkpoint FGL1 in lung adenocarcinoma: The pivotal role of the YY1-FGL1-MYH9 axis. Front. Immunol. 2022, 13, 1014053. [Google Scholar] [CrossRef]
- Hays, E.; Bonavida, B. YY1 regulates cancer cell immune resistance by modulating PD-L1 expression. Drug Resist. Updates 2019, 43, 10–28. [Google Scholar] [CrossRef]
- Shigeta, K.; Datta, M.; Hato, T.; Kitahara, S.; Chen, I.X.; Matsui, A.; Kikuchi, H.; Mamessier, E.; Aoki, S.; Ramjiawan, R.R.; et al. Dual Programmed Death Receptor-1 and Vascular Endothelial Growth Factor Receptor-2 Blockade Promotes Vascular Normalization and Enhances Antitumor Immune Responses in Hepatocellular Carcinoma. Hepatology 2020, 71, 1247–1261. [Google Scholar] [CrossRef]
- Zheng, X.; Fang, Z.; Liu, X.; Deng, S.; Zhou, P.; Wang, X.; Zhang, C.; Yin, R.; Hu, H.; Chen, X.; et al. Increased vessel perfusion predicts the efficacy of immune checkpoint blockade. J. Clin. Invest. 2018, 128, 2104–2115. [Google Scholar] [CrossRef]
- Motzer, R.J.; Penkov, K.; Haanen, J.; Rini, B.; Albiges, L.; Campbell, M.T.; Venugopal, B.; Kollmannsberger, C.; Negrier, S.; Uemura, M.; et al. Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2019, 380, 1103–1115. [Google Scholar] [CrossRef]
- Cheng, A.L.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.Y.; Lim, H.Y.; Kudo, M.; Breder, V.; Merle, P.; et al. Updated efficacy and safety data from IMbrave150: Atezolizumab plus bevacizumab vs. sorafenib for unresectable hepatocellular carcinoma. J. Hepatol. 2022, 76, 862–873. [Google Scholar] [CrossRef] [PubMed]
- Schmittnaegel, M.; Rigamonti, N.; Kadioglu, E.; Cassara, A.; Wyser Rmili, C.; Kiialainen, A.; Kienast, Y.; Mueller, H.J.; Ooi, C.H.; Laoui, D.; et al. Dual angiopoietin-2 and VEGFA inhibition elicits antitumor immunity that is enhanced by PD-1 checkpoint blockade. Sci. Transl. Med. 2017, 9, eaak9670. [Google Scholar] [CrossRef]
- Makker, V.; Rasco, D.; Vogelzang, N.J.; Brose, M.S.; Cohn, A.L.; Mier, J.; Di Simone, C.; Hyman, D.M.; Stepan, D.E.; Dutcus, C.E.; et al. Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer: An interim analysis of a multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol. 2019, 20, 711–718. [Google Scholar] [CrossRef]
- Cleveland, A.H.; Fan, Y. Reprogramming endothelial cells to empower cancer immunotherapy. Trends Mol. Med. 2023. [Google Scholar] [CrossRef]
- Huang, Y.; Yuan, J.; Righi, E.; Kamoun, W.S.; Ancukiewicz, M.; Nezivar, J.; Santosuosso, M.; Martin, J.D.; Martin, M.R.; Vianello, F.; et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc. Natl. Acad. Sci. USA 2012, 109, 17561–17566. [Google Scholar] [CrossRef]
- Sung, Y.C.; Jin, P.R.; Chu, L.A.; Hsu, F.F.; Wang, M.R.; Chang, C.C.; Chiou, S.J.; Qiu, J.T.; Gao, D.Y.; Lin, C.C.; et al. Delivery of nitric oxide with a nanocarrier promotes tumour vessel normalization and potentiates anti-cancer therapies. Nat. Nanotechnol. 2019, 14, 1160–1169. [Google Scholar] [CrossRef]
- Huerta-Yepez, S.; Vega, M.; Jazirehi, A.; Garban, H.; Hongo, F.; Cheng, G.; Bonavida, B. Nitric oxide sensitizes prostate carcinoma cell lines to TRAIL-mediated apoptosis via inactivation of NF-kappa B and inhibition of Bcl-xl expression. Oncogene 2004, 23, 4993–5003. [Google Scholar] [CrossRef]
- Hongo, F.; Garban, H.; Huerta-Yepez, S.; Vega, M.; Jazirehi, A.R.; Mizutani, Y.; Miki, T.; Bonavida, B. Inhibition of the transcription factor Yin Yang 1 activity by S-nitrosation. Biochem. Biophys. Res. Commun. 2005, 336, 692–701. [Google Scholar] [CrossRef]
- Bonavida, B. Therapeutic YY1 Inhibitors in Cancer: ALL in ONE. Crit. Rev. Oncog. 2017, 22, 37–47. [Google Scholar] [CrossRef]
- Kao, T.W.; Bai, G.H.; Wang, T.L.; Shih, I.M.; Chuang, C.M.; Lo, C.L.; Tsai, M.C.; Chiu, L.Y.; Lin, C.C.; Shen, Y.A. Novel cancer treatment paradigm targeting hypoxia-induced factor in conjunction with current therapies to overcome resistance. J. Exp. Clin. Cancer Res. 2023, 42, 171. [Google Scholar] [CrossRef]
- Erin, N.; Grahovac, J.; Brozovic, A.; Efferth, T. Tumor microenvironment and epithelial mesenchymal transition as targets to overcome tumor multidrug resistance. Drug Resist. Updates 2020, 53, 100715. [Google Scholar] [CrossRef]
- Liu, X.; Jutooru, I.; Lei, P.; Kim, K.; Lee, S.O.; Brents, L.K.; Prather, P.L.; Safe, S. Betulinic acid targets YY1 and ErbB2 through cannabinoid receptor-dependent disruption of microRNA-27a:ZBTB10 in breast cancer. Mol. Cancer Ther. 2012, 11, 1421–1431. [Google Scholar] [CrossRef] [PubMed]
- Huerta-Yepez, S.; Baritaki, S.; Baay-Guzman, G.; Hernandez-Luna, M.A.; Hernandez-Cueto, A.; Vega, M.I.; Bonavida, B. Contribution of either YY1 or BclXL-induced inhibition by the NO-donor DETANONOate in the reversal of drug resistance, both in vitro and in vivo. YY1 and BclXL are overexpressed in prostate cancer. Nitric Oxide 2013, 29, 17–24. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; He, S.; Mei, R.; Kang, Y.; Duan, J.; Wei, R.; Xiang, C.; Wu, Y.; Lu, X.; Cai, Z.; et al. miR-29a suppresses IL-13-induced cell invasion by inhibiting YY1 in the AKT pathway in lung adenocarcinoma A549 cells. Oncol. Rep. 2018, 39, 2613–2623. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Garzon, R.; Sun, H.; Ladner, K.J.; Singh, R.; Dahlman, J.; Cheng, A.; Hall, B.M.; Qualman, S.J.; Chandler, D.S.; et al. NF-kappaB-YY1-miR-29 regulatory circuitry in skeletal myogenesis and rhabdomyosarcoma. Cancer Cell 2008, 14, 369–381. [Google Scholar] [CrossRef]
- Huang, T.; Wang, G.; Yang, L.; Peng, B.; Wen, Y.; Ding, G.; Wang, Z. MiR-186 inhibits proliferation, migration, and invasion of non-small cell lung cancer cells by downregulating Yin Yang 1. Cancer Biomark. 2017, 21, 221–228. [Google Scholar] [CrossRef] [PubMed]
- Su, H.; Liu, L.; Zhang, Y.; Wang, J.; Zhao, Y. Long noncoding RNA NPCCAT1 promotes nasopharyngeal carcinoma progression via upregulating YY1. Biochimie 2019, 157, 184–194. [Google Scholar] [CrossRef] [PubMed]
- Xu, C.; Tsai, Y.H.; Galbo, P.M.; Gong, W.; Storey, A.J.; Xu, Y.; Byrum, S.D.; Xu, L.; Whang, Y.E.; Parker, J.S.; et al. Cistrome analysis of YY1 uncovers a regulatory axis of YY1:BRD2/4-PFKP during tumorigenesis of advanced prostate cancer. Nucleic Acids Res. 2021, 49, 4971–4988. [Google Scholar] [CrossRef]
- Rosenberg, S.A.; Restifo, N.P.; Yang, J.C.; Morgan, R.A.; Dudley, M.E. Adoptive cell transfer: A clinical path to effective cancer immunotherapy. Nat. Rev. Cancer 2008, 8, 299–308. [Google Scholar] [CrossRef]
- Sterner, R.C.; Sterner, R.M. CAR-T cell therapy: Current limitations and potential strategies. Blood Cancer J. 2021, 11, 69. [Google Scholar] [CrossRef]
- Benmebarek, M.R.; Karches, C.H.; Cadilha, B.L.; Lesch, S.; Endres, S.; Kobold, S. Killing Mechanisms of Chimeric Antigen Receptor (CAR) T Cells. Int. J. Mol. Sci. 2019, 20, 1283. [Google Scholar] [CrossRef]
- Safarzadeh Kozani, P.; Safarzadeh Kozani, P.; Ahmadi Najafabadi, M.; Yousefi, F.; Mirarefin, S.M.J.; Rahbarizadeh, F. Recent Advances in Solid Tumor CAR-T Cell Therapy: Driving Tumor Cells From Hero to Zero? Front. Immunol. 2022, 13, 795164. [Google Scholar] [CrossRef] [PubMed]
- Kochenderfer, J.N.; Wilson, W.H.; Janik, J.E.; Dudley, M.E.; Stetler-Stevenson, M.; Feldman, S.A.; Maric, I.; Raffeld, M.; Nathan, D.A.; Lanier, B.J.; et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood 2010, 116, 4099–4102. [Google Scholar] [CrossRef] [PubMed]
- T Cell Receptor Immunotherapy Targeting VEGFR2 for Patients with Metastatic Cancer. 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT01218867CAR (accessed on 6 December 2023).
- Chinnasamy, D.; Yu, Z.; Theoret, M.R.; Zhao, Y.; Shrimali, R.K.; Morgan, R.A.; Feldman, S.A.; Restifo, N.P.; Rosenberg, S.A. Gene therapy using genetically modified lymphocytes targeting VEGFR-2 inhibits the growth of vascularized syngenic tumors in mice. J. Clin. Investig. 2010, 120, 3953–3968. [Google Scholar] [CrossRef]
- Wang, W.; Ma, Y.; Li, J.; Shi, H.S.; Wang, L.Q.; Guo, F.C.; Zhang, J.; Li, D.; Mo, B.H.; Wen, F.; et al. Specificity redirection by CAR with human VEGFR-1 affinity endows T lymphocytes with tumor-killing ability and anti-angiogenic potency. Gene Ther. 2013, 20, 970–978. [Google Scholar] [CrossRef] [PubMed]
- Xing, H.; Yang, X.; Xu, Y.; Tang, K.; Tian, Z.; Chen, Z.; Zhang, Y.; Xue, Z.; Rao, Q.; Wang, M.; et al. Anti-tumor effects of vascular endothelial growth factor/vascular endothelial growth factor receptor binding domain-modified chimeric antigen receptor T cells. Cytotherapy 2021, 23, 810–819. [Google Scholar] [CrossRef] [PubMed]
- Byrd, T.T.; Fousek, K.; Pignata, A.; Szot, C.; Samaha, H.; Seaman, S.; Dobrolecki, L.; Salsman, V.S.; Oo, H.Z.; Bielamowicz, K.; et al. TEM8/ANTXR1-Specific CAR T Cells as a Targeted Therapy for Triple-Negative Breast Cancer. Cancer Res. 2018, 78, 489–500. [Google Scholar] [CrossRef] [PubMed]
- Fierle, J.K.; Brioschi, M.; de Tiani, M.; Wetterwald, L.; Atsaves, V.; Abram-Saliba, J.; Petrova, T.V.; Coukos, G.; Dunn, S.M. Soluble trivalent engagers redirect cytolytic T cell activity toward tumor endothelial marker 1. Cell Rep. Med. 2021, 2, 100362. [Google Scholar] [CrossRef]
- Zhuang, X.; Maione, F.; Robinson, J.; Bentley, M.; Kaul, B.; Whitworth, K.; Jumbu, N.; Jinks, E.; Bystrom, J.; Gabriele, P.; et al. CAR T cells targeting tumor endothelial marker CLEC14A inhibit tumor growth. JCI Insight 2020, 5, e138808. [Google Scholar] [CrossRef]
- Wagner, J.; Wickman, E.; Shaw, T.I.; Anido, A.A.; Langfitt, D.; Zhang, J.; Porter, S.N.; Pruett-Miller, S.M.; Tillman, H.; Krenciute, G.; et al. Antitumor Effects of CAR T Cells Redirected to the EDB Splice Variant of Fibronectin. Cancer Immunol. Res. 2021, 9, 279–290. [Google Scholar] [CrossRef]
- Xie, Y.J.; Dougan, M.; Jailkhani, N.; Ingram, J.; Fang, T.; Kummer, L.; Momin, N.; Pishesha, N.; Rickelt, S.; Hynes, R.O.; et al. Nanobody-based CAR T cells that target the tumor microenvironment inhibit the growth of solid tumors in immunocompetent mice. Proc. Natl. Acad. Sci. USA 2019, 116, 7624–7631. [Google Scholar] [CrossRef]
- Wallstabe, L.; Mades, A.; Frenz, S.; Einsele, H.; Rader, C.; Hudecek, M. CAR T cells targeting alpha(v)beta(3) integrin are effective against advanced cancer in preclinical models. Adv. Cell Gene Ther. 2018, 1, e11. [Google Scholar] [CrossRef] [PubMed]
- Fu, X.; Rivera, A.; Tao, L.; Zhang, X. Genetically modified T cells targeting neovasculature efficiently destroy tumor blood vessels, shrink established solid tumors and increase nanoparticle delivery. Int. J. Cancer 2013, 133, 2483–2492. [Google Scholar] [CrossRef] [PubMed]
- Chen, Z.; Li, Y.; Tan, B.; Zhao, Q.; Fan, L.; Li, F.; Zhao, X. Progress and current status of molecule-targeted therapy and drug resistance in gastric cancer. Drugs Today 2020, 56, 469–482. [Google Scholar] [CrossRef] [PubMed]
- Rini, B.I.; Atkins, M.B. Resistance to targeted therapy in renal-cell carcinoma. Lancet Oncol. 2009, 10, 992–1000. [Google Scholar] [CrossRef] [PubMed]
- Oladipupo, S.S.; Hu, S.; Santeford, A.C.; Yao, J.; Kovalski, J.R.; Shohet, R.V.; Maslov, K.; Wang, L.V.; Arbeit, J.M. Conditional HIF-1 induction produces multistage neovascularization with stage-specific sensitivity to VEGFR inhibitors and myeloid cell independence. Blood 2011, 117, 4142–4153. [Google Scholar] [CrossRef] [PubMed]
- Bottsford-Miller, J.N.; Coleman, R.L.; Sood, A.K. Resistance and escape from antiangiogenesis therapy: Clinical implications and future strategies. J. Clin. Oncol. 2012, 30, 4026–4034. [Google Scholar] [CrossRef] [PubMed]
- Lai, X.M.; Liu, S.Y.; Tsai, Y.T.; Sun, G.H.; Chang, S.Y.; Huang, S.M.; Cha, T.L. HAF mediates the evasive resistance of anti-angiogenesis TKI through disrupting HIF-1alpha and HIF-2alpha balance in renal cell carcinoma. Oncotarget 2017, 8, 49713–49724. [Google Scholar] [CrossRef] [PubMed]
- Zhao, D.; Zhai, B.; He, C.; Tan, G.; Jiang, X.; Pan, S.; Dong, X.; Wei, Z.; Ma, L.; Qiao, H.; et al. Upregulation of HIF-2alpha induced by sorafenib contributes to the resistance by activating the TGF-alpha/EGFR pathway in hepatocellular carcinoma cells. Cell. Signal. 2014, 26, 1030–1039. [Google Scholar] [CrossRef]
- Shi, X.; Wang, M.; Zhang, Y.; Guo, X.; Liu, M.; Zhou, Z.; Zhao, Y.; He, R.; Gao, Y.; Liu, Y.; et al. Hypoxia activated HGF expression in pancreatic stellate cells confers resistance of pancreatic cancer cells to EGFR inhibition. EBioMedicine 2022, 86, 104352. [Google Scholar] [CrossRef]
- Lu, Y.; Liu, Y.; Oeck, S.; Zhang, G.J.; Schramm, A.; Glazer, P.M. Hypoxia Induces Resistance to EGFR Inhibitors in Lung Cancer Cells via Upregulation of FGFR1 and the MAPK Pathway. Cancer Res. 2020, 80, 4655–4667. [Google Scholar] [CrossRef]
- Sarvagalla, S.; Kolapalli, S.P.; Vallabhapurapu, S. The Two Sides of YY1 in Cancer: A Friend and a Foe. Front. Oncol. 2019, 9, 1230. [Google Scholar] [CrossRef] [PubMed]
Study | CAR T Cell Design | Study Data | Ref. | |||
---|---|---|---|---|---|---|
Vessel Target | Tumor Type | Study Type/Model | Ag Recognition; Clone | Construct | End Points | |
VEGFR2 | Metastatic cancer | Phase I/II | scFv anti-human VEGFR-2; KDR1121 | CD8/CD28/4–1-BB/CD3ζ | No response/Progression Disease | [123] |
VEGFR2 | Melanoma (B16F10) | Syngeneic | scFv anti-mouse VEGFR-2; DC101 | CD8/CD28/4–1-BB/CD3ζ | Inhibition of tumor growth; increased survival | [124] |
Fibrosarcoma (MCA205) | ||||||
Colon (MC38) | ||||||
Colon (CT26) | ||||||
kidney (RENCA) | ||||||
VEGFR1 | Lung carcinoma (A549) | Xenograft | scFv anti-human VEGFR1; IMC-18F1 | IgG-Fc/CD4/CD3ζ | Inhibition of tumor growth; increased survival; metastasis inhibition | [125] |
VEGFR2/3 | Breast (MDA-MB-231) | Xenograft | N-terminus VEGF-C | CD8/CD28/CD3ζ | Inhibition of tumor growth; metastasis inhibition | [126] |
Breast (HCC1806) | ||||||
TEM8 | Breast (MDA-MB-468) | Xenograft | scFv anti-human TEM8; L2 | IgG-Fc/CD28/4–1-BB/CD3ζ | Inhibition of tumor growth; increased survival; vascular disruption | [127] |
Breast (LM231) | ||||||
Breast (BCM-2665) | Patient-derived xenograft | |||||
Breast (WHIM12) | ||||||
TEM1 | Ewing sarcoma (A673) | Xenograft | scFv anti-human and anti-mouse TEM1; L1C1m | Trilobite engager (CD3/TEM1) | Inhibition of tumor growth | [128] |
CLEC14a | Healthy mice | N/A Transgenic | scFV anti-mouse and anti-human CLEC14a; CRT3, CRT5 | CD28/CD3ζ | Inhibition of tumor growth; increased survival | [129] |
RipTag2 | ||||||
Pancreas (mPDAC) | Syngeneic | |||||
Lung (LLC) | Syngeneic | |||||
ED-B | Glioma (U87) | Xenograft | scFv anti-human ED-B; L19 | CD28/CD3ζ | Inhibition of tumor growth; increased survival | [130] |
Lung (A549) | ||||||
Ewing sarcoma (A673) | ||||||
ED-B | Melanoma (B16F10) | Syngeneic | VHH; NJB2; camelid | CD8/CD28/CD3ζ | Inhibition of tumor growth; increased survival | [131] |
Colon (MC38) | ||||||
Integrin αvβ3 | Melanoma (A375) | Xenograft | scFv anti-human αvβ3; LM609 | CD28/CD3ζ | Tumor regression; increased survival | [132] |
Integrin αvβ3 | Melanoma (B16F10) | Syngeneic | Echistatin (Disintegrin in snake venom) | CD28/CD3ζ | Inhibition of tumor growth; vascular disruption | [133] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 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
Meo, C.; de Nigris, F. Clinical Potential of YY1-Hypoxia Axis for Vascular Normalization and to Improve Immunotherapy. Cancers 2024, 16, 491. https://doi.org/10.3390/cancers16030491
Meo C, de Nigris F. Clinical Potential of YY1-Hypoxia Axis for Vascular Normalization and to Improve Immunotherapy. Cancers. 2024; 16(3):491. https://doi.org/10.3390/cancers16030491
Chicago/Turabian StyleMeo, Concetta, and Filomena de Nigris. 2024. "Clinical Potential of YY1-Hypoxia Axis for Vascular Normalization and to Improve Immunotherapy" Cancers 16, no. 3: 491. https://doi.org/10.3390/cancers16030491
APA StyleMeo, C., & de Nigris, F. (2024). Clinical Potential of YY1-Hypoxia Axis for Vascular Normalization and to Improve Immunotherapy. Cancers, 16(3), 491. https://doi.org/10.3390/cancers16030491