Oncogenesis, Microenvironment Modulation and Clinical Potentiality of FAP in Glioblastoma: Lessons Learned from Other Solid Tumors
Abstract
:1. Introduction
2. FAP Is Expressed in Various Cell Types within the GBM Microenvironment
3. FAP Plays a Protumorigenic Role in GBM and Other Solid Tumors
3.1. Potential Substrates and Enzymatic Activity of FAP
3.2. Signaling Pathways and Nonenzymatic Activity of FAP
3.2.1. PI3K/AKT Signaling Pathway
3.2.2. SHH/GLI Signaling Pathway
3.2.3. FAK Signaling Pathway
3.3. Functional Roles of FAP in GBM
3.3.1. Tumor Proliferation and Invasion
3.3.2. Angiogenesis
3.3.3. Immunosuppression in the Tumor Microenvironment
3.3.4. EMT and TMZ Resistance
4. Future Clinical Applications of FAP
4.1. FAP as a Potential Imaging Biomarker
4.2. Prognostic Value of FAP
4.3. Advances in FAP-Targeted Therapy
5. Discussion
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Acknowledgments
Conflicts of Interest
References
- Ostrom, Q.T.; Cioffi, G.; Gittleman, H.; Patil, N.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2012–2016. Neuro Oncol. 2019, 21, v1–v100. [Google Scholar] [CrossRef] [PubMed]
- Tan, A.C.; Ashley, D.M.; Lopez, G.Y.; Malinzak, M.; Friedman, H.S.; Khasraw, M. Management of glioblastoma: State of the art and future directions. CA Cancer J. Clin. 2020, 70, 299–312. [Google Scholar] [CrossRef] [PubMed]
- Weller, M.; Le Rhun, E. How did lomustine become standard of care in recurrent glioblastoma? Cancer Treat. Rev. 2020, 87. [Google Scholar] [CrossRef] [PubMed]
- Lim, M.; Xia, Y.; Bettegowda, C.; Weller, M. Current state of immunotherapy for glioblastoma. Nat. Rev. Clin. Oncol. 2018, 15, 422–442. [Google Scholar] [CrossRef]
- Hombach-Klonisch, S.; Mehrpour, M.; Shojaei, S.; Harlos, C.; Pitz, M.; Hamai, A.; Siemianowicz, K.; Likus, W.; Wiechec, E.; Toyota, B.D.; et al. Glioblastoma and chemoresistance to alkylating agents: Involvement of apoptosis, autophagy, and unfolded protein response. Pharmacol. Ther. 2018, 184, 13–41. [Google Scholar] [CrossRef] [PubMed]
- Park, J.E.; Lenter, M.C.; Zimmermann, R.N.; Garin-Chesa, P.; Old, L.J.; Rettig, J.W. Fibroblast activation protein, a dual Specificity serine protease expressed in reactive human tumor stromal fibroblasts. J. Biol. Chem. 1999, 274, 36505–36512. [Google Scholar] [CrossRef] [Green Version]
- Juillerat-Jeanneret, L.; Tafelmeyer, P.; Golshayan, D. Fibroblast activation protein-alpha in fibrogenic disorders and cancer: More than a prolyl-specific peptidase? Expert Opin. Ther. Target. 2017, 21, 977–991. [Google Scholar] [CrossRef] [Green Version]
- Pure, E.; Blomberg, R. Pro-tumorigenic roles of fibroblast activation protein in cancer: Back to the basics. Oncogene 2018, 37, 4343–4357. [Google Scholar] [CrossRef]
- Busek, P.; Balaziova, E.; Matrasova, I.; Hilser, M.; Tomas, R.; Syrucek, M.; Zemanova, Z.; Krepela, E.; Belacek, J.; Sedo, A. Fibroblast activation protein alpha is expressed by transformed and stromal cells and is associated with mesenchymal features in glioblastoma. Tumour Biol. 2016, 37, 13961–13971. [Google Scholar] [CrossRef]
- Li, M.; Li, G.; Kiyokawa, J.; Tirmizi, Z.; Richardson, L.G.; Ning, J.; Das, S.; Martuza, R.L.; Stemmer-Rachamimov, A.; Rabkin, S.D.; et al. Characterization and oncolytic virus targeting of FAP-expressing tumor-associated pericytes in glioblastoma. Acta Neuropathol. Commun. 2020, 8. [Google Scholar] [CrossRef] [PubMed]
- Pandya, D.N.; Sinha, A.; Yuan, H.; Mutkus, L.; Stumpf, K.; Marini, F.C.; Wadas, T.J. Imaging of fibroblast activation protein alpha expression in a preclinical mouse model of glioma using positron emission tomography. Molecules 2020, 25, 3672. [Google Scholar] [CrossRef]
- Ebert, L.M.; Yu, W.; Gargett, T.; Toubia, J.; Kollis, P.M.; Tea, M.N.; Ebert, B.W.; Bardy, C.; van den Hurk, M.; Bonder, C.S.; et al. Endothelial, pericyte and tumor cell expression in glioblastoma identifies fibroblast activation protein (FAP) as an excellent target for immunotherapy. Clin. Transl. Immunol. 2020, 9. [Google Scholar] [CrossRef] [PubMed]
- Nurmik, M.; Ullmann, P.; Rodriguez, F.; Haan, S.; Letellier, E. In search of definitions: Cancer-associated fibroblasts and their markers. Int. J. Cancer 2020, 146, 895–905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kahounová, Z.; Kurfürstová, D.; Bouchal, J.; Kharaishvili, G.; Navrátil, J.; Remšík, J.; Šimečková, S.; Študent, V.; Kozubík, A.; Souček, K. The fibroblast surface markers FAP, anti-fibroblast, and FSP are expressed by cells of epithelial origin and may be altered during epithelial-to-mesenchymal transition. Cytom. Part A 2018, 93, 941–951. [Google Scholar] [CrossRef] [Green Version]
- Kraman, M.; Bambrough, P.J.; Arnold, J.N.; Roberts, E.W.; Magiera, L.; Jones, J.O.; Gopinathan, A.; Tuveson, D.A.; Fearon, D.T. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-alpha. Science 2010, 330, 827–830. [Google Scholar] [CrossRef] [Green Version]
- Kelly, T.; Huang, Y.; Simms, A.E.; Mazur, A. Fibroblast activation protein-alpha: A key modulator of the microenvironment in multiple pathologies. Int. Rev. Cell Mol. Biol. 2012, 297, 83–116. [Google Scholar] [CrossRef] [PubMed]
- Arnold, J.N.; Magiera, L.; Kraman, M.; Fearon, D.T. Tumoral immune suppression by macrophages expressing fibroblast activation protein-alpha and heme oxygenase-1. Cancer Immunol. Res. 2014, 2, 121–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tchou, J.; Zhang, P.J.; Bi, Y.; Satija, C.; Marjumdar, R.; Stephen, T.L.; Lo, A.; Chen, H.; Mies, C.; June, C.H.; et al. Fibroblast activation protein expression by stromal cells and tumor-associated macrophages in human breast cancer. Hum. Pathol. 2013, 44, 2549–2557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Su, S.; Chen, J.; Yao, H.; Liu, J.; Yu, S.; Lao, L.; Wang, M.; Luo, M.; Xing, Y.; Chen, F.; et al. CD10(+)GPR77(+) cancer-associated fibroblasts promote cancer formation and chemoresistance by sustaining cancer stemness. Cell 2018, 172, 841–856. [Google Scholar] [CrossRef]
- Ligorio, M.; Sil, S.; Malagon-Lopez, J.; Nieman, L.T.; Misale, S.; Di Pilato, M.; Ebright, R.Y.; Karabacak, M.N.; Kulkarni, A.S.; Liu, A.; et al. Stromal microenvironment shapes the intratumoral architecture of pancreatic cancer. Cell 2019, 178, 160–175. [Google Scholar] [CrossRef]
- Clavreul, A.; Etcheverry, A.; Chassevent, A.; Quillien, V.; Avril, T.; Jourdan, M.L.; Michalak, S.; Francois, P.; Carre, J.L.; Mosser, J.; et al. Isolation of a new cell population in the glioblastoma microenvironment. J. Neurooncol. 2012, 106, 493–504. [Google Scholar] [CrossRef]
- Sena, I.F.G.; Paiva, A.E.; Prazeres, P.; Azevedo, P.O.; Lousado, L.; Bhutia, S.K.; Salmina, A.B.; Mintz, A.; Birbrair, A. Glioblastoma-activated pericytes support tumor growth via immunosuppression. Cancer Med. 2018, 7, 1232–1239. [Google Scholar] [CrossRef] [PubMed]
- Valdor, R.; Garcia-Bernal, D.; Riquelme, D.; Martinez, C.M.; Moraleda, J.M.; Cuervo, A.M.; Macian, F.; Martinez, S. Glioblastoma ablates pericytes antitumor immune function through aberrant up-regulation of chaperone-mediated autophagy. Proc. Natl. Acad. Sci. USA 2019, 116, 20655–20665. [Google Scholar] [CrossRef] [Green Version]
- Rohrich, M.; Loktev, A.; Wefers, A.K.; Altmann, A.; Paech, D.; Adeberg, S.; Windisch, P.; Hielscher, T.; Flechsig, P.; Floca, R.; et al. IDH-wildtype glioblastomas and grade III/IV IDH-mutant gliomas show elevated tracer uptake in fibroblast activation protein-specific PET/CT. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 2569–2580. [Google Scholar] [CrossRef]
- Krepela, E.; Vanickova, Z.; Hrabal, P.; Zubal, M.; Chmielova, B.; Balaziova, E.; Vymola, P.; Matrasova, I.; Busek, P.; Sedo, A. Regulation of fibroblast activation protein by transforming growth factor beta-1 in glioblastoma microenvironment. Int. J. Mol. Sci. 2021, 22, 1046. [Google Scholar] [CrossRef] [PubMed]
- Sasaki, A.; Naganuma, H.; Satoh, E.; Nagasaka, M.; Isoe, S.; Nakano, S.; Nukui, H. Secretion of transforming growth factor-beta 1 and -beta 2 by malignant glioma cells. Neurol. Med. Chir. 1995, 35, 423–430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leitlein, J.; Aulwurm, S.; Waltereit, R.; Naumann, U.; Wagenknecht, B.; Garten, W.; Weller, M.; Platten, M. Processing of immunosuppressive pro-TGF-beta 1,2 by human glioblastoma cells involves cytoplasmic and secreted furin-like proteases. J. Immunol. 2001, 166, 7238–7243. [Google Scholar] [CrossRef] [Green Version]
- Mikheeva, S.A.; Mikheev, A.M.; Petit, A.; Beyer, R.; Oxford, R.G.; Khorasani, L.; Maxwell, J.P.; Glackin, C.A.; Wakimoto, H.; Gonzalez-Herrero, I.; et al. TWIST1 promotes invasion through mesenchymal change in human glioblastoma. Mol. Cancer 2010, 9. [Google Scholar] [CrossRef] [Green Version]
- Simkova, A.; Busek, P.; Sedo, A.; Konvalinka, J. Molecular recognition of fibroblast activation protein for diagnostic and therapeutic applications. Biochim. Biophys. Acta Proteins Proteom 2020, 1868. [Google Scholar] [CrossRef]
- Christiansen, V.J.; Jackson, K.W.; Lee, K.N.; McKee, P.A. Effect of fibroblast activation protein and Α2-Antiplasmin cleaving enzyme on collagen types I, III, and IV. Arch. Biochem. Biophys. 2007, 457, 177–186. [Google Scholar] [CrossRef] [Green Version]
- Aggarwal, S.; Brennen, W.N.; Kole, T.P.; Schneider, E.; Topaloglu, O.; Yates, M.; Cotter, R.J.; Denmeade, S.R. Fibroblast activation protein peptide substrates identified from human collagen I derived gelatin cleavage sites. Biochemistry 2008, 47, 1076–1086. [Google Scholar] [CrossRef] [Green Version]
- Fan, M.H.; Zhu, Q.; Li, H.H.; Ra, H.J.; Majumdar, S.; Gulick, D.L.; Jerome, J.A.; Madsen, D.H.; Christofidou-Solomidou, M.; Speicher, D.W.; et al. Fibroblast activation protein (FAP) accelerates collagen degradation and clearance from lungs in mice. J. Biol. Chem. 2016, 291, 8070–8089. [Google Scholar] [CrossRef] [Green Version]
- Muliaditan, T.; Caron, J.; Okesola, M.; Opzoomer, J.W.; Kosti, P.; Georgouli, M.; Gordon, P.; Lall, S.; Kuzeva, D.M.; Pedro, L.; et al. Macrophages are exploited from an innate wound healing response to facilitate cancer metastasis. Nat. Commun. 2018, 9. [Google Scholar] [CrossRef] [Green Version]
- Strepkos, D.; Markouli, M.; Klonou, A.; Piperi, C.; Papavassiliou, A.G. Insights in the immunobiology of glioblastoma. J. Mol. Med. 2020, 98, 1–10. [Google Scholar] [CrossRef]
- Dzikowski, L.; Mirzaei, R.; Sarkar, S.; Kumar, M.; Bose, P.; Bellail, A.; Hao, C.; Yong, V.W. Fibrinogen in the glioblastoma microenvironment contributes to the invasiveness of brain tumor-initiating cells. Brain Pathol. 2021. [Google Scholar] [CrossRef] [PubMed]
- Luo, Y.; Ye, S.; Chen, X.; Gong, F.; Lu, W.; Li, X. Rush to the fire: FGF21 extinguishes metabolic stress, metaflammation and tissue damage. Cytokine Growth Factor Rev. 2017, 38, 59–65. [Google Scholar] [CrossRef]
- Dunshee, D.R.; Bainbridge, T.W.; Kljavin, N.M.; Zavala-Solorio, J.; Schroeder, A.C.; Chan, R.; Corpuz, R.; Wong, M.; Zhou, W.; Deshmukh, G.; et al. Fibroblast activation protein cleaves and inactivates fibroblast growth factor 21. J. Biol. Chem. 2016, 291, 5986–5996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhen, E.Y.; Jin, Z.; Ackermann, B.L.; Thomas, M.K.; Gutierrez, J.A. Circulating FGF21 proteolytic processing mediated by fibroblast activation protein. Biochem. J. 2016, 473, 605–614. [Google Scholar] [CrossRef] [Green Version]
- Wong, P.F.; Gall, M.G.; Bachovchin, W.W.; McCaughan, G.W.; Keane, F.M.; Gorrell, M.D. Neuropeptide Y is a physiological substrate of fibroblast activation protein: Enzyme kinetics in blood plasma and expression of Y2R and Y5R in human liver cirrhosis and hepatocellular carcinoma. Peptides 2016, 75, 80–95. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Bijker, M.S.; Herzog, H. The neuropeptide Y system: Pathophysiological and therapeutic implications in obesity and cancer. Pharmacol. Ther. 2011, 131, 91–113. [Google Scholar] [CrossRef]
- Kitlinska, J. Neuropeptide Y (NPY) in neuroblastoma: Effect on growth and vascularization. Peptides 2007, 28, 405–412. [Google Scholar] [CrossRef]
- Ruscica, M.; Dozio, E.; Boghossian, S.; Bovo, G.; Martos Riano, V.; Motta, M.; Magni, P. Activation of the Y1 receptor by neuropeptide Y regulates the growth of prostate cancer cells. Endocrinology 2006, 147, 1466–1473. [Google Scholar] [CrossRef]
- Zhang, H.E.; Hamson, E.J.; Koczorowska, M.M.; Tholen, S.; Chowdhury, S.; Bailey, C.G.; Lay, A.J.; Twigg, S.M.; Lee, Q.; Roediger, B.; et al. Identification of novel natural substrates of fibroblast activation protein-alpha by differential degradomics and proteomics. Mol. Cell Proteom. 2019, 18, 65–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koczorowska, M.M.; Tholen, S.; Bucher, F.; Lutz, L.; Kizhakkedathu, J.N.; De Wever, O.; Wellner, U.F.; Biniossek, M.L.; Stahl, A.; Lassmann, S.; et al. Fibroblast activation protein-alpha, a stromal cell surface protease, shapes key features of cancer associated fibroblasts through proteome and degradome alterations. Mol. Oncol. 2016, 10, 40–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baird, S.K.; Allan, L.; Renner, C.; Scott, F.E.; Scott, A.M. Fibroblast activation protein increases metastatic potential of fibrosarcoma line HT1080 through upregulation of integrin-mediated signaling pathways. Clin. Exp. Metastasis 2015, 32, 507–516. [Google Scholar] [CrossRef] [PubMed]
- Yang, W.; Han, W.; Ye, S.; Liu, D.; Wu, J.; Liu, H.; Li, C.; Chen, H. Fibroblast activation protein-alpha promotes ovarian cancer cell proliferation and invasion via extracellular and intracellular signaling mechanisms. Exp. Mol. Pathol. 2013, 95, 105–110. [Google Scholar] [CrossRef] [PubMed]
- Bullock, M.D.; Pickard, K.M.; Nielsen, B.S.; Sayan, A.E.; Jenei, V.; Mellone, M.; Mitter, R.; Primrose, J.N.; Thomas, G.J.; Packham, G.K.; et al. Pleiotropic actions of miR-21 highlight the critical role of deregulated stromal microRNAs during colorectal cancer progression. Cell Death Dis. 2013, 4. [Google Scholar] [CrossRef] [Green Version]
- Wen, W.; Ding, J.; Sun, W.; Fu, J.; Chen, Y.; Wu, K.; Ning, B.; Han, T.; Huang, L.; Chen, C.; et al. Cyclin G1-mediated epithelial-mesenchymal transition via phosphoinositide 3-kinase/Akt signaling facilitates liver cancer progression. Hepatology 2012, 55, 1787–1798. [Google Scholar] [CrossRef]
- Ye, Y.; Xiao, Y.; Wang, W.; Yearsley, K.; Gao, J.X.; Shetuni, B.; Barsky, S.H. ERalpha signaling through slug regulates E-cadherin and EMT. Oncogene 2010, 29, 1451–1462. [Google Scholar] [CrossRef] [Green Version]
- Han, L.; Peng, B.; Ma, Q.; Ma, J.; Li, J.; Li, W.; Duan, W.; Chen, C.; Liu, J.; Xu, Q.; et al. Indometacin ameliorates high glucose-induced proliferation and invasion via modulation of e-cadherin in pancreatic cancer cells. Curr. Med. Chem. 2013, 20, 4142–4152. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.; Wu, Q.; Liu, Z.; Luo, X.; Fan, Y.; Liu, Y.; Zhang, Y.; Hua, S.; Fu, Q.; Zhao, M.; et al. Downregulation of FAP suppresses cell proliferation and metastasis through PTEN/PI3K/AKT and Ras-ERK signaling in oral squamous cell carcinoma. Cell Death Dis. 2014, 5. [Google Scholar] [CrossRef]
- Jia, J.; Martin, T.A.; Ye, L.; Jiang, W.G. FAP-α (fibroblast activation protein-α) is involved in the control of human breast cancer cell line growth and motility via the FAK pathway. BMC Cell Biol. 2014, 15. [Google Scholar] [CrossRef] [Green Version]
- Zhao, H.F.; Wang, J.; Shao, W.; Wu, C.P.; Chen, Z.P.; To, S.T.; Li, W.P. Recent advances in the use of PI3K inhibitors for glioblastoma multiforme: Current preclinical and clinical development. Mol. Cancer 2017, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alvarez-Garcia, V.; Tawil, Y.; Wise, H.M.; Leslie, N.R. Mechanisms of PTEN loss in cancer: It’s all about diversity. Semin. Cancer Biol. 2019, 59, 66–79. [Google Scholar] [CrossRef] [PubMed]
- Verhaak, R.G.; Hoadley, K.A.; Purdom, E.; Wang, V.; Qi, Y.; Wilkerson, M.D.; Miller, C.R.; Ding, L.; Golub, T.; Mesirov, J.P.; et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010, 17, 98–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jia, J.; Martin, T.A.; Ye, L.; Meng, L.; Xia, N.; Jiang, W.G.; Zhang, X. Fibroblast activation protein-alpha promotes the growth and migration of lung cancer cells via the PI3K and sonic hedgehog pathways. Int. J. Mol. Med. 2018, 41, 275–283. [Google Scholar] [CrossRef] [Green Version]
- Yue, D.; Li, H.; Che, J.; Zhang, Y.; Tseng, H.H.; Jin, J.Q.; Luh, T.M.; Giroux-Leprieur, E.; Mo, M.; Zheng, Q.; et al. Hedgehog/Gli promotes epithelial-mesenchymal transition in lung squamous cell carcinomas. J. Exp. Clin. Cancer Res. 2014, 33. [Google Scholar] [CrossRef] [Green Version]
- Doheny, D.; Manore, S.G.; Wong, G.L.; Lo, H.-W. Hedgehog signaling and truncated GLI1 in cancer. Cells 2020, 9, 2114. [Google Scholar] [CrossRef] [PubMed]
- Lo, H.W.; Zhu, H.; Cao, X.; Aldrich, A.; Ali-Osman, F. A novel splice variant of GLI1 that promotes glioblastoma cell migration and invasion. Cancer Res. 2009, 69, 6790–6798. [Google Scholar] [CrossRef] [Green Version]
- Neoh, C.A.; Wu, W.T.; Dai, G.F.; Su, J.H.; Liu, C.I.; Su, T.R.; Wu, Y.J. Flaccidoxide-13-acetate extracted from the soft coral cladiella kashmani reduces human bladder cancer cell migration and invasion through reducing activation of the FAK/PI3K/AKT/mTOR signaling pathway. Molecules 2018, 23, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santos, A.M.; Jung, J.; Aziz, N.; Kissil, J.L.; Pure, E. Targeting fibroblast activation protein inhibits tumor stromagenesis and growth in mice. J. Clin. Investig. 2009, 119, 3613–3625. [Google Scholar] [CrossRef] [PubMed]
- Yang, X.; Lin, Y.; Shi, Y.; Li, B.; Liu, W.; Yin, W.; Dang, Y.; Chu, Y.; Fan, J.; He, R. FAP promotes immunosuppression by cancer-associated fibroblasts in the tumor microenvironment via STAT3-CCL2 signaling. Cancer Res. 2016, 76, 4124–4135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Natarajan, M.; Hecker, T.P.; Gladson, C.L. FAK signaling in anaplastic astrocytoma and glioblastoma tumors. Cancer J. 2003, 9, 126–133. [Google Scholar] [CrossRef]
- Park, A.K.; Kim, P.; Ballester, L.Y.; Esquenazi, Y.; Zhao, Z. Subtype-specific signaling pathways and genomic aberrations associated with prognosis of glioblastoma. Neuro Oncol. 2019, 21, 59–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rao, J.S. Molecular mechanisms of glioma invasiveness: The role of proteases. Nat. Rev. Cancer 2003, 3, 489–501. [Google Scholar] [CrossRef] [PubMed]
- Lettau, I.; Hattermann, K.; Held-Feindt, J.; Brauer, R.; Sedlacek, R.; Mentlein, R. Matrix metalloproteinase-19 is highly expressed in astroglial tumors and promotes invasion of glioma cells. J. Neuropathol. Exp. Neurol. 2010, 69, 215–223. [Google Scholar] [CrossRef] [Green Version]
- Mentlein, R.; Hattermann, K.; Hemion, C.; Jungbluth, A.A.; Held-Feindt, J. Expression and role of the cell surface protease seprase/fibroblast activation protein-alpha (FAP-alpha) in astroglial tumors. Biol. Chem. 2011, 392, 199–207. [Google Scholar] [CrossRef] [PubMed]
- He, J.; Liu, Y.; Zhu, T.; Zhu, J.; Dimeco, F.; Vescovi, A.L.; Heth, J.A.; Muraszko, K.M.; Fan, X.; Lubman, D.M. CD90 is identified as a candidate marker for cancer stem cells in primary high-grade gliomas using tissue microarrays. Mol. Cell Proteom. 2012, 11, M111.010744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Calabrese, C.; Poppleton, H.; Kocak, M.; Hogg, T.L.; Fuller, C.; Hamner, B.; Oh, E.Y.; Gaber, M.W.; Finklestein, D.; Allen, M.; et al. A perivascular niche for brain tumor stem cells. Cancer Cell 2007, 11, 69–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Turley, S.J.; Cremasco, V.; Astarita, J.L. Immunological hallmarks of stromal cells in the tumour microenvironment. Nat. Rev. Immunol. 2015, 15, 669–682. [Google Scholar] [CrossRef]
- Costa, A.; Kieffer, Y.; Scholer-Dahirel, A.; Pelon, F.; Bourachot, B.; Cardon, M.; Sirven, P.; Magagna, I.; Fuhrmann, L.; Bernard, C.; et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell 2018, 33, 463–479. [Google Scholar] [CrossRef] [Green Version]
- Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Canellas, A.; Hernando-Momblona, X.; et al. TGFbeta drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [Green Version]
- Korner, M.; Reubi, J.C. Neuropeptide Y receptors in primary human brain tumors- overexpression in high-grade tumors. J. Neuropathol. Exp. Neurol. 2008, 67, 741–749. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Du, Y.; Jiang, Z.; Tian, Y.; Qiu, N.; Wang, Y.; Lqbal, M.Z.; Hu, M.; Zou, R.; Luo, L.; et al. Y1 receptor ligand-based nanomicelle as a novel nanoprobe for glioma-targeted imaging and therapy. Nanoscale 2018, 10, 5845–5851. [Google Scholar] [CrossRef]
- Siebzehnrubl, F.A.; Silver, D.J.; Tugertimur, B.; Deleyrolle, L.P.; Siebzehnrubl, D.; Sarkisian, M.R.; Devers, K.G.; Yachnis, A.T.; Kupper, M.D.; Neal, D.; et al. The ZEB1 pathway links glioblastoma initiation, invasion and chemoresistance. EMBO Mol. Med. 2013, 5, 1196–1212. [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]
- Yang, Y.N.; Zhang, X.H.; Wang, Y.M.; Zhang, X.; Gu, Z. miR-204 reverses temozolomide resistance and inhibits cancer initiating cells phenotypes by degrading FAP-alpha in glioblastoma. Oncol. Lett. 2018, 15, 7563–7570. [Google Scholar] [CrossRef] [PubMed]
- Kratochwil, C.; Flechsig, P.; Lindner, T.; Abderrahim, L.; Altmann, A.; Mier, W.; Adeberg, S.; Rathke, H.; Rohrich, M.; Winter, H.; et al. 68Ga-FAPI PET/CT: Tracer uptake in 28 different kinds of cancer. J. Nucl. Med. 2019, 60, 801–805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rohrich, M.; Floca, R.; Loi, L.; Adeberg, S.; Windisch, P.; Giesel, F.L.; Kratochwil, C.; Flechsig, P.; Rathke, H.; Lindner, T.; et al. FAP-specific PET signaling shows a moderately positive correlation with relative CBV and no correlation with ADC in 13 IDH wildtype glioblastomas. Eur. J. Radiol. 2020, 127. [Google Scholar] [CrossRef] [PubMed]
- Fedele, M.; Cerchia, L.; Pegoraro, S.; Sgarra, R.; Manfioletti, G. Proneural-mesenchymal transition: Phenotypic plasticity to acquire multitherapy resistance in glioblastoma. Int. J. Mol. Sci. 2019, 20, 2746. [Google Scholar] [CrossRef] [Green Version]
- Meany, H.; Balis, F.M.; Aikin, A.; Whitcomb, P.; Murphy, R.F.; Steinberg, S.M.; Widemann, B.C.; Fox, E. Pediatric phase I trial design using maximum target inhibition as the primary endpoint. J. Natl. Cancer Inst. 2010, 102, 909–912. [Google Scholar] [CrossRef]
- Henry, L.R.; Lee, H.O.; Lee, J.S.; Klein-Szanto, A.; Watts, P.; Ross, E.A.; Chen, W.T.; Cheng, J.D. Clinical implications of fibroblast activation protein in patients with colon cancer. Clin. Cancer Res. 2007, 13, 1736–1741. [Google Scholar] [CrossRef] [Green Version]
- Eager, R.M.; Cunningham, C.C.; Senzer, N.N.; Stephenson, J., Jr.; Anthony, S.P.; O’Day, S.J.; Frenette, G.; Pavlick, A.C.; Jones, B.; Uprichard, M.; et al. Phase II assessment of talabostat and cisplatin in second-line stage IV melanoma. BMC Cancer 2009, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fitzgerald, A.A.; Weiner, L.M. The role of fibroblast activation protein in health and malignancy. Cancer Metastasis Rev. 2020, 39, 783–803. [Google Scholar] [CrossRef]
- Wen, Y.; Wang, C.T.; Ma, T.T.; Li, Z.Y.; Zhou, L.N.; Mu, B.; Leng, F.; Shi, H.S.; Li, Y.O.; Wei, Y.Q. Immunotherapy targeting fibroblast activation protein inhibits tumor growth and increases survival in a murine colon cancer model. Cancer Sci. 2010, 101, 2325–2332. [Google Scholar] [CrossRef]
- Chen, M.; Xiang, R.; Wen, Y.; Xu, G.; Wang, C.; Luo, S.; Yin, T.; Wei, X.; Shao, B.; Liu, N.; et al. A whole-cell tumor vaccine modified to express fibroblast activation protein induces antitumor immunity against both tumor cells and cancer-associated fibroblasts. Sci. Rep. 2015, 5. [Google Scholar] [CrossRef]
- Schuberth, P.C.; Hagedorn, C.; Jensen, S.M.; Gulati, P.; van der Broek, M.; Mischo, A.; Soltermann, A.; Jüngel, A.; Belaunzaran, O.M.; Stahel, R.; et al. Treatment of malignant pleural mesothelioma by fibroblast activation protein-specific re-directed T cells. J. Transl. Med. 2013, 11, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kakarla, S.; Chow, K.K.; Mata, M.; Shaffer, D.R.; Song, X.T.; Wu, M.F.; Liu, H.; Wang, L.L.; Rowley, D.R.; Pfizenmaier, K.; et al. Antitumor effects of chimeric receptor engineered human T cells directed to tumor stroma. Mol. Ther. 2013, 21, 1611–1620. [Google Scholar] [CrossRef]
- Tran, E.; Chinnasamy, D.; Yu, Z.; Morgan, R.A.; Lee, C.C.; Restifo, N.P.; Rosenberg, S.A. Immune targeting of fibroblast activation protein triggers recognition of multipotent bone marrow stromal cells and cachexia. J. Exp. Med. 2013, 210, 1125–1135. [Google Scholar] [CrossRef] [PubMed]
- Krepela, E.; Busek, P.; Hilser, M.; Vanickova, Z.; Sedo, A. Species-specific real-time RT-PCR analysis of expression of stromal cell genes in a tumor xenotransplantation model in mice. Biochem. Biophys. Res. Commun. 2017, 491, 126–133. [Google Scholar] [CrossRef]
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
Shi, Y.; Kong, Z.; Liu, P.; Hou, G.; Wu, J.; Ma, W.; Cheng, X.; Wang, Y. Oncogenesis, Microenvironment Modulation and Clinical Potentiality of FAP in Glioblastoma: Lessons Learned from Other Solid Tumors. Cells 2021, 10, 1142. https://doi.org/10.3390/cells10051142
Shi Y, Kong Z, Liu P, Hou G, Wu J, Ma W, Cheng X, Wang Y. Oncogenesis, Microenvironment Modulation and Clinical Potentiality of FAP in Glioblastoma: Lessons Learned from Other Solid Tumors. Cells. 2021; 10(5):1142. https://doi.org/10.3390/cells10051142
Chicago/Turabian StyleShi, Yixin, Ziren Kong, Penghao Liu, Guozhu Hou, Jiaming Wu, Wenbin Ma, Xin Cheng, and Yu Wang. 2021. "Oncogenesis, Microenvironment Modulation and Clinical Potentiality of FAP in Glioblastoma: Lessons Learned from Other Solid Tumors" Cells 10, no. 5: 1142. https://doi.org/10.3390/cells10051142
APA StyleShi, Y., Kong, Z., Liu, P., Hou, G., Wu, J., Ma, W., Cheng, X., & Wang, Y. (2021). Oncogenesis, Microenvironment Modulation and Clinical Potentiality of FAP in Glioblastoma: Lessons Learned from Other Solid Tumors. Cells, 10(5), 1142. https://doi.org/10.3390/cells10051142