RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field
Abstract
:Simple Summary
Abstract
1. Introduction
2. Integrin/ECM Cell Adhesion and Signaling Receptors
3. The Role of RGD Binding Integrins in Cancer and Tumor Neovascularization
3.1. RGD Binding Integrin Receptors
3.2. Integrin Functions in Angiogenesis and Tumor Vessel Phenotype
3.2.1. The Role of Integrins in Angiogenesis
3.2.2. The Therapeutic Benefit of Vascular Normalization Therapy in Cancer
3.2.3. Integrins in Tumor Cell Extravasation
3.3. The Cellular Fate of Tumor Cell-Expressed Integrins
3.3.1. Cellular Internalization of Integrins
3.3.2. Intracellular Endosomal Integrin Trafficking and Sorting
3.3.3. Exosomal Integrins in Cancer
3.3.4. Exosomes as Therapeutic and/or Diagnostic Tools
4. Design and Development of RGD-Based Integrin Ligands for Biomedical Applications and Translation
4.1. From Natural ECM Proteins to Synthethic RGD-Based Integrin Ligands: The Generation of Highly Selective and Affine Integrin Ligands
4.2. Design of Synthetic RGD Binding Integrin Ligands
5. Integrin Targeted Molecular Imaging and Radiotherapy
5.1. Nuclear Imaging (PET/SPECT)
5.2. Targeted Radiotherapy (TRT)
5.3. Non-Nuclear Imaging of Integrins
6. Integrin Targeting in Non-Cancer Diseases
6.1. Fatal Role of Integrins in Promoting Sepsis
6.2. Integrins in Fibrosis
6.3. Integrins and SARS-COV-2
7. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Ruoslahti, E.; Pierschbacher, M.D. New perspectives in cell adhesion: RGD and integrins. Science 1987, 238, 491. [Google Scholar] [CrossRef] [PubMed]
- Hynes, R.O. Integrins: Versatility, modulation, and signaling in cell adhesion. Cell 1992, 69, 11–25. [Google Scholar] [CrossRef]
- Travis, J. Biotech gets a grip on cell adhesion. Science 1993, 260, 906. [Google Scholar] [CrossRef] [PubMed]
- Temming, K.; Schiffelers, R.M.; Molema, G.; Kok, R.J. RGD-based strategies for selective delivery of therapeutics and imaging agents to the tumour vasculature. Drug Resist. Updates 2005, 8, 381–402. [Google Scholar] [CrossRef] [PubMed]
- Barczyk, M.; Carracedo, S.; Gullberg, D. Integrins. Cell Tissue Res. 2010, 339, 269–280. [Google Scholar] [CrossRef] [Green Version]
- Kechagia, J.Z.; Ivaska, J.; Roca-Cusachs, P. Integrins as biomechanical sensors of the microenvironment. Nat. Rev. Mol. Cell Biol. 2019, 20, 457–473. [Google Scholar] [CrossRef]
- Reynolds, L.E.; Wyder, L.; Lively, J.C.; Taverna, D.; Robinson, S.D.; Huang, X.Z.; Sheppard, D.; Hynes, O.; Hodivala-Dilke, K.M. Enhanced pathological angiogenesis in mice lacking β(3) integrin or β(3) and β(5) integrins. Nat. Med. 2002, 8, 27–34. [Google Scholar] [CrossRef]
- Takada, Y.; Ye, X.; Simon, S. The integrins. Genome Biol. 2007, 8, 215. [Google Scholar] [CrossRef] [Green Version]
- Ley, K.; Rivera-Nieves, J.; Sandborn, W.J.; Shattil, S. Integrin-based therapeutics: Biological basis, clinical use and new drugs. Nat. Rev. Drug Discov. 2016, 15, 173–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nieberler, M.; Reuning, U.; Reichart, F.; Notni, J.; Wester, H.-J.; Schwaiger, M.; Weinmüller, M.; Räder, A.; Steiger, K.; Kessler, H. Exploring the Role of RGD-Recognizing Integrins in Cancer. Cancers 2017, 9, 116. [Google Scholar] [CrossRef]
- Beer, A.J.; Kessler, H.; Wester, H.-J.; Schwaiger, M. PET Imaging of Integrin αVβ3 Expression. Theranostics 2011, 1, 48–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiong, J.P.; Stehle, T.; Diefenbach, B.; Zhang, R.; Dunker, R.; Scott, D.L.; Joachimiak, A.; Goodman, S.L.; Arnaout, M.A. Crystal structure of the extracellular segment of integrin alpha Vbeta3. Science 2001, 294, 339–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiong, J.P.; Stehle, T.; Zhang, R.; Joachimiak, A.; Frech, M.; Goodman, S.L.; Arnaout, M.A. Crystal structure of the extracellular segment of integrin alpha Vbeta3 in complex with an Arg-Gly-Asp ligand. Science 2002, 296, 151–155. [Google Scholar] [CrossRef] [PubMed]
- Meyer, A.; Auernheimer, J.; Modlinger, A.; Kessler, H. Targeting RGD recognizing integrins: Drug development, biomaterial research, tumor imaging and targeting. Curr. Pharm. Des. 2006, 12, 2723–2747. [Google Scholar] [CrossRef] [PubMed]
- Heckmann, D.; Meyer, A.; Marinelli, L.; Zahn, G.; Stragies, R.; Kessler, H. Probing integrin selectivity: Rational design of highly active and selective ligands for the alpha5beta1 and alphavbeta3 integrin receptor. Angew. Chem. Int. Ed. Engl. 2007, 46, 3571–3574. [Google Scholar] [CrossRef] [PubMed]
- Schottelius, M.; Laufer, B.; Kessler, H.; Wester, H.-J. Ligands for mapping αvβ3-integrin expression in vivo. Acc. Chem. Res. 2009, 42, 969–980. [Google Scholar] [CrossRef]
- Nagae, M.; Re, S.; Mihara, E.; Nogi, T.; Sugita, Y.; Takagi, J. Crystal structure of α5β1 integrin ectodomain: Atomic details of the fibronectin receptor. J. Cell Biol. 2012, 197, 131–140. [Google Scholar] [CrossRef] [Green Version]
- Neubauer, S.; Rechenmacher, F.; Beer, A.J.; Curnis, F.; Pohle, K.; D’Alessandria, C.; Wester, H.-J.; Reuning, U.; Corti, A.; Schwaiger, M.; et al. Selective imaging of the angiogenic relevant integrins α5β1 and αvβ3. Angew. Chem. Int. Ed. Engl. 2013, 52, 11656–11659. [Google Scholar] [CrossRef]
- Arosio, D.; Casagrande, C. Advancement in integrin facilitated drug delivery. Adv. Drug Deliv. Rev. 2016, 97, 111–143. [Google Scholar] [CrossRef] [PubMed]
- Mas-Moruno, C.; Fraioli, R.; Rechenmacher, F.; Neubauer, S.; Kapp, T.G.; Kessler, H. αvβ3- or α5β1-Integrin-Selective Peptidomimetics for Surface Coating. Angew. Chem. Int. Ed. Engl. 2016, 55, 7048–7067. [Google Scholar] [CrossRef]
- Jarvelainen, H.; Botta, G.P.; Mendoza, T.H.D.; Ruoslahti, E. Abstract 1106: Co-administration of the iRGD tumor-penetrating peptide improves the tumor immunostimulatory effects of low-dose IL-2. Cancer Res. 2019, 79, 1106. [Google Scholar] [CrossRef]
- Zuo, H. iRGD: A Promising Peptide for Cancer Imaging and a Potential Therapeutic Agent for Various Cancers. J. Oncol. 2019, 2019, 9367845. [Google Scholar] [CrossRef]
- Pierschbacher, M.D.; Ruoslahti, E. Variants of the cell recognition site of fibronectin that retain attachment-promoting activity. Proc. Natl. Acad. Sci. USA 1984, 81, 5985. [Google Scholar] [CrossRef] [Green Version]
- Gottschalk, K.E.; Kessler, H. The structures of integrins and integrin-ligand complexes: Implications for drug design and signal transduction. Angew. Chem. Int. Ed. Engl. 2002, 41, 3767–3774. [Google Scholar] [CrossRef]
- Marinelli, L.; Meyer, A.; Heckmann, D.; Lavecchia, A.; Novellino, E.; Kessler, H. Ligand binding analysis for human α5β1 integrin: Strategies for designing new alpha5beta1 integrin antagonists. J. Med. Chem. 2005, 48, 4204–4207. [Google Scholar] [CrossRef] [PubMed]
- Rocha, L.A.; Learmonth, D.A.; Sousa, R.A.; Salgado, A.J. αvβ3 and α5β1 integrin-specific ligands: From tumor angiogenesis inhibitors to vascularization promoters in regenerative medicine? Biotechnol. Adv. 2018, 36, 208–227. [Google Scholar] [CrossRef]
- Wong, P.-P.; Demircioglu, F.; Ghazaly, E.; Alrawashdeh, W.; Stratford, M.R.L.; Scudamore, C.L.; Cereser, B.; Crnogorac-Jurcevic, T.; McDonald, S.; Elia, G.; et al. Dual-Action Combination Therapy Enhances Angiogenesis while Reducing Tumor Growth and Spread. Cancer Cell 2015, 27, 123–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Misra, A.; Sheikh, A.Q.; Kumar, A.; Luo, J.; Zhang, J.; Hinton, R.B.; Smoot, L.; Kaplan, P.; Urban, Z.; Qyang, Y.; et al. Integrin β3 inhibition is a therapeutic strategy for supravalvular aortic stenosis. J. Exp. Med. 2016, 213, 451–463. [Google Scholar] [CrossRef]
- Dukinfield, M.; Maniati, E.; Reynolds, L.E.; Aubdool, A.; Baliga, R.S.; D’Amico, G.; Maiques, O.; Wang, J.; Bedi, K.C.; Margulies, K.B.; et al. Repurposing an anti-cancer agent for the treatment of hypertrophic heart disease. J. Pathol. 2019, 249, 523–535. [Google Scholar] [CrossRef] [Green Version]
- Giancotti, F.G.; Ruoslahti, E. Integrin Signaling. Science 1999, 285, 1028. [Google Scholar] [CrossRef]
- Desgrosellier, J.S.; Cheresh, D.A. Integrins in cancer: Biological implications and therapeutic opportunities. Nat. Rev. Cancer 2010, 10, 9–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- White, D.J.; Puranen, S.; Johnson, M.S.; Heino, J. The collagen receptor subfamily of the integrins. Int. J. Biochem. Cell Biol. 2004, 36, 1405–1410. [Google Scholar] [CrossRef] [PubMed]
- Harris, E.S.; McIntyre, T.M.; Prescott, S.M.; Zimmerman, G.A. The leukocyte integrins. J. Biol. Chem. 2000, 275, 23409–23412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koivisto, L.; Heino, J.; Hakkinen, L.; Larjava, H. Integrins in Wound Healing. Adv. Wound Care 2014, 3, 762–783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kishimoto, T.K.; Larson, R.S.; Corbi, A.L.; Dustin, M.L.; Staunton, D.E.; Springer, T.A. The Leukocyte Integrins. In Advances in Immunology; Dixon, F.J., Ed.; Academic Press: Cambridge, MA, USA, 1989; Volume 46, pp. 149–182. [Google Scholar]
- Wu, X.; Reddy, D.S. Integrins as receptor targets for neurological disorders. Pharmacol. Ther. 2012, 134, 68–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nader, D.; Curley, G.F.; Kerrigan, S.W. A new perspective in sepsis treatment: Could RGD-dependent integrins be novel targets? Drug Discov. Today 2020, 25, 2317–2325. [Google Scholar] [CrossRef] [PubMed]
- Conroy, K.P.; Kitto, L.J.; Henderson, N.C. αv integrins: Key regulators of tissue fibrosis. Cell Tissue Res. 2016, 365, 511–519. [Google Scholar] [CrossRef] [Green Version]
- Chen, C.; Li, R.; Ross, R.S.; Manso, A.M. Integrins and integrin-related proteins in cardiac fibrosis. J. Mol. Cell Cardiol. 2016, 93, 162–174. [Google Scholar] [CrossRef] [Green Version]
- Hussein, H.A.; Walker, L.R.; Abdel-Raouf, U.M.; Desouky, S.A.; Montasser, A.K.; Akula, S.M. Beyond RGD: Virus interactions with integrins. Arch. Virol. 2015, 160, 2669–2681. [Google Scholar] [CrossRef]
- Belkin, A.M.; Stepp, M.A. Integrins as receptors for laminins. Microsc. Res. Tech. 2000, 51, 280–301. [Google Scholar] [CrossRef]
- Mitroulis, I.; Alexaki, V.I.; Kourtzelis, I.; Ziogas, A.; Hajishengallis, G.; Chavakis, T. Leukocyte integrins: Role in leukocyte recruitment and as therapeutic targets in inflammatory disease. Pharmacol. Ther. 2015, 147, 123–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pierschbacher, M.D.; Ruoslahti, E. Cell attachment activity of fibronectin can be duplicated by small synthetic fragments of the molecule. Nature 1984, 309, 30–33. [Google Scholar] [CrossRef]
- Suzuki, S.; Oldberg, A.; Hayman, E.G.; Pierschbacher, M.D.; Ruoslahti, E. Complete amino acid sequence of human vitronectin deduced from cDNA. Similarity of cell attachment sites in vitronectin and fibronectin. Embo J. 1985, 4, 2519–2524. [Google Scholar] [CrossRef] [PubMed]
- Plow, E.F.; Pierschbacher, M.D.; Ruoslahti, E.; Marguerie, G.A.; Ginsberg, M.H. The effect of Arg-Gly-Asp-containing peptides on fibrinogen and von Willebrand factor binding to platelets. Proc. Natl. Acad. Sci. USA 1985, 82, 8057–8061. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hantgan, R.R.; Stahle, M.C.; Lord, S.T. Dynamic regulation of fibrinogen: Integrin αIIbβ3 binding. Biochemistry 2010, 49, 9217–9225. [Google Scholar] [CrossRef] [Green Version]
- Oldberg, A.; Franzén, A.; Heinegård, D. Cloning and sequence analysis of rat bone sialoprotein (osteopontin) cDNA reveals an Arg-Gly-Asp cell-binding sequence. Proc. Natl. Acad. Sci. USA 1986, 83, 8819–8823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Humphries, J.D.; Byron, A.; Humphries, M.J. Integrin ligands at a glance. J. Cell Sci. 2006, 119, 3901. [Google Scholar] [CrossRef] [Green Version]
- Schnittert, J.; Bansal, R.; Storm, G.; Prakash, J. Integrins in wound healing, fibrosis and tumor stroma: High potential targets for therapeutics and drug delivery. Adv. Drug Deliv. Rev. 2018, 129, 37–53. [Google Scholar] [CrossRef]
- Humphries, M.J. Integrin structure. Biochem. Soc. Trans. 2000, 28, 311–339. [Google Scholar] [CrossRef]
- Humphries, M.J.; McEwan, P.A.; Barton, S.J.; Buckley, P.A.; Bella, J.; Mould, A.P. Integrin structure: Heady advances in ligand binding, but activation still makes the knees wobble. Trends Biochem. Sci. 2003, 28, 313–320. [Google Scholar] [CrossRef]
- Berman, A.E.; Kozlova, N.I.; Morozevich, G.E. Integrins: Structure and signaling. Biochemistry 2003, 68, 1284–1299. [Google Scholar] [CrossRef] [PubMed]
- Gailit, J.; Ruoslahti, E. Regulation of the fibronectin receptor affinity by divalent cations. J. Biol. Chem. 1988, 263, 12927–12932. [Google Scholar] [CrossRef]
- D’Souza, S.E.; Haas, T.A.; Piotrowicz, R.S.; Byers-Ward, V.; McGrath, D.E.; Soule, H.R.; Cierniewski, C.; Plow, E.F.; Smith, J.W. Ligand and cation binding are dual functions of a discrete segment of the integrin beta 3 subunit: Cation displacement is involved in ligand binding. Cell 1994, 79, 659–667. [Google Scholar] [CrossRef]
- Smith, J.W.; Piotrowicz, R.S.; Mathis, D. A mechanism for divalent cation regulation of beta 3-integrins. J. Biol. Chem. 1994, 269, 960–967. [Google Scholar] [CrossRef]
- Smallridge, R. The MIDAS touch. Nat. Rev. Mol. Cell Biol. 2002, 3, 313. [Google Scholar] [CrossRef]
- Heckmann, D.; Laufer, B.; Marinelli, L.; Limongelli, V.; Novellino, E.; Zahn, G.; Stragies, R.; Kessler, H. Breaking the Dogma of the Metal-Coordinating Carboxylate Group in Integrin Ligands: Introducing Hydroxamic Acids to the MIDAS To Tune Potency and Selectivity. Angew. Chem. Int. Ed. 2009, 48, 4436–4440. [Google Scholar] [CrossRef]
- Bollinger, M.; Manzenrieder, F.; Kolb, R.; Bochen, A.; Neubauer, S.; Marinelli, L.; Limongelli, V.; Novellino, E.; Moessmer, G.; Pell, R.; et al. Tailoring of Integrin Ligands: Probing the Charge Capability of the Metal Ion-Dependent Adhesion Site. J. Med. Chem. 2012, 55, 871–882. [Google Scholar] [CrossRef]
- Zhang, K.; Chen, J. The regulation of integrin function by divalent cations. Cell Adh. Migr. 2012, 6, 20–29. [Google Scholar] [CrossRef] [Green Version]
- Shimaoka, M.; Springer, T.A. Therapeutic antagonists and conformational regulation of integrin function. Nat. Rev. Drug Discov. 2003, 2, 703–716. [Google Scholar] [CrossRef] [PubMed]
- Harburger, D.S.; Calderwood, D.A. Integrin signalling at a glance. J. Cell Sci. 2009, 122, 159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mas-Moruno, C.; Rechenmacher, F.; Kessler, H. Cilengitide: The first anti-angiogenic small molecule drug candidate design, synthesis and clinical evaluation. Anticancer Agents Med. Chem. 2010, 10, 753–768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, J.; Zhu, J.; Springer, T.A. Complete integrin headpiece opening in eight steps. J. Cell Biol. 2013, 201, 1053–1068. [Google Scholar] [CrossRef] [Green Version]
- Luo, B.-H.; Springer, T.A. Integrin structures and conformational signaling. Curr. Opin. Cell Biol. 2006, 18, 579–586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beglova, N.; Blacklow, S.C.; Takagi, J.; Springer, T.A. Cysteine-rich module structure reveals a fulcrum for integrin rearrangement upon activation. Nat. Struct. Biol. 2002, 9, 282–287. [Google Scholar] [CrossRef] [PubMed]
- Takagi, J.; Petre, B.M.; Walz, T.; Springer, T.A. Global conformational rearrangements in integrin extracellular domains in outside-in and inside-out signaling. Cell 2002, 110, 599–611. [Google Scholar] [CrossRef] [Green Version]
- Kim, C.; Ye, F.; Ginsberg, M.H. Regulation of Integrin Activation. Annu. Rev. Cell Dev. Biol. 2011, 27, 321–345. [Google Scholar] [CrossRef] [PubMed]
- Mould, A.P.; Humphries, M.J. Cell biology: Adhesion articulated. Nature 2004, 432, 27–28. [Google Scholar] [CrossRef]
- Shattil, S.J.; Kim, C.; Ginsberg, M.H. The final steps of integrin activation: The end game. Nat. Rev. Mol. Cell Biol. 2010, 11, 288–300. [Google Scholar] [CrossRef] [Green Version]
- Campbell, I.D.; Humphries, M.J. Integrin structure, activation, and interactions. Cold Spring Harb. Perspect. Biol. 2011, 3. [Google Scholar] [CrossRef] [Green Version]
- Xiao, T.; Takagi, J.; Coller, B.S.; Wang, J.H.; Springer, T.A. Structural basis for allostery in integrins and binding to fibrinogen-mimetic therapeutics. Nature 2004, 432, 59–67. [Google Scholar] [CrossRef] [Green Version]
- Arnaout, M.A.; Goodman, S.L.; Xiong, J.P. Structure and mechanics of integrin-based cell adhesion. Curr. Opin. Cell Biol. 2007, 19, 495–507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gottschalk, K.E.; Adams, P.D.; Brunger, A.T.; Kessler, H. Transmembrane signal transduction of the alpha(IIb)beta(3) integrin. Protein Sci. 2002, 11, 1800–1812. [Google Scholar] [CrossRef]
- Gottschalk, K.E.; Kessler, H. Evidence for hetero-association of transmembrane helices of integrins. FEBS Lett. 2004, 557, 253–258. [Google Scholar] [CrossRef] [Green Version]
- Hoefling, M.; Kessler, H.; Gottschalk, K.E. The transmembrane structure of integrin alphaIIbbeta3: Significance for signal transduction. Angew. Chem. Int. Ed. Engl. 2009, 48, 6590–6593. [Google Scholar] [CrossRef]
- Müller, M.A.; Opfer, J.; Brunie, L.; Volkhardt, L.A.; Sinner, E.K.; Boettiger, D.; Bochen, A.; Kessler, H.; Gottschalk, K.E.; Reuning, U. The glycophorin A transmembrane sequence within integrin αvβ3 creates a non-signaling integrin with low basal affinity that is strongly adhesive under force. J. Mol. Biol. 2013, 425, 2988–3006. [Google Scholar] [CrossRef]
- Gottschalk, K.E.; Günther, R.; Kessler, H. A Three-State Mechanism of Integrin Activation and Signal Transduction for Integrin αvβ3. ChemBioChem 2002, 3, 470–473. [Google Scholar] [CrossRef]
- Tadokoro, S.; Shattil, S.J.; Eto, K.; Tai, V.; Liddington, R.C.; de Pereda, J.M.; Ginsberg, M.H.; Calderwood, D.A. Talin binding to integrin beta tails: A final common step in integrin activation. Science 2003, 302, 103–106. [Google Scholar] [CrossRef] [PubMed]
- Moser, M.; Legate, K.R.; Zent, R.; Fässler, R. The tail of integrins, talin, and kindlins. Science 2009, 324, 895–899. [Google Scholar] [CrossRef]
- Eilken, H.M.; Adams, R.H. Turning on the angiogenic microswitch. Nat. Med. 2010, 16, 853–854. [Google Scholar] [CrossRef]
- Müller, M.A.; Brunie, L.; Bächer, A.S.; Kessler, H.; Gottschalk, K.E.; Reuning, U. Cytoplasmic salt bridge formation in integrin αvß3 stabilizes its inactive state affecting integrin-mediated cell biological effects. Cell Signal. 2014, 26, 2493–2503. [Google Scholar] [CrossRef] [PubMed]
- Takagi, J.; Erickson, H.P.; Springer, T.A. C-terminal opening mimics ‘inside-out’ activation of integrin α5β1. Nat. Struct. Biol. 2001, 8, 412–416. [Google Scholar] [CrossRef]
- Xiong, Y.M.; Chen, J.; Zhang, L. Modulation of CD11b/CD18 adhesive activity by its extracellular, membrane-proximal regions. J. Immunol. 2003, 171, 1042–1050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, B.H.; Springer, T.A.; Takagi, J. A specific interface between integrin transmembrane helices and affinity for ligand. PLoS Biol. 2004, 2, e153. [Google Scholar] [CrossRef]
- Gottschalk, K.E.; Kessler, H. A computational model of transmembrane integrin clustering. Structure 2004, 12, 1109–1116. [Google Scholar] [CrossRef] [Green Version]
- Fong, K.P.; Zhu, H.; Span, L.M.; Moore, D.T.; Yoon, K.; Tamura, R.; Yin, H.; DeGrado, W.F.; Bennett, J.S. Directly Activating the Integrin αIIbβ3 Initiates Outside-In Signaling by Causing αIIbβ3 Clustering. J. Biol. Chem. 2016, 291, 11706–11716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schaller, M.D.; Parsons, J.T. Focal adhesion kinase and associated proteins. Curr. Opin Cell Biol. 1994, 6, 705–710. [Google Scholar] [CrossRef]
- Stupack, D.G.; Cheresh, D.A. Get a ligand, get a life: Integrins, signaling and cell survival. J. Cell Sci. 2002, 115, 3729–3738. [Google Scholar] [CrossRef] [Green Version]
- Henning Stumpf, B.; Ambriović-Ristov, A.; Radenovic, A.; Smith, A.-S. Recent Advances and Prospects in the Research of Nascent Adhesions. Front. Physiol. 2020, 11. [Google Scholar] [CrossRef]
- Shattil, S.J.; Kashiwagi, H.; Pampori, N. Integrin signaling: The platelet paradigm. Blood 1998, 91, 2645–2657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Askari, J.A.; Buckley, P.A.; Mould, A.P.; Humphries, M.J. Linking integrin conformation to function. J. Cell Sci. 2009, 122, 165–170. [Google Scholar] [CrossRef] [Green Version]
- Arnold, M.; Cavalcanti-Adam, E.A.; Glass, R.; Blümmel, J.; Eck, W.; Kantlehner, M.; Kessler, H.; Spatz, J.P. Activation of integrin function by nanopatterned adhesive interfaces. Chemphyschem 2004, 5, 383–388. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.; Wang, F.; Chen, X. Integrin αvβ3-Targeted Cancer Therapy. Drug Dev. Res. 2008, 69, 329–339. [Google Scholar] [CrossRef] [Green Version]
- Avraamides, C.J.; Garmy-Susini, B.; Varner, J.A. Integrins in angiogenesis and lymphangiogenesis. Nat. Rev. Cancer 2008, 8, 604–617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Demircioglu, F.; Hodivala-Dilke, K. αvβ3 Integrin and tumour blood vessels-learning from the past to shape the future. Curr. Opin. Cell Biol. 2016, 42, 121–127. [Google Scholar] [CrossRef] [PubMed]
- Kerr, J.S.; Slee, A.M.; Mousa, S.A. The alpha v integrin antagonists as novel anticancer agents: An update. Expert Opin. Investig. Drugs 2002, 11, 1765–1774. [Google Scholar] [CrossRef] [PubMed]
- Weis, S.M.; Cheresh, D.A. αV integrins in angiogenesis and cancer. Cold Spring Harb. Perspect. Med. 2011, 1, a006478. [Google Scholar] [CrossRef] [Green Version]
- Ria, R.; Vacca, A.; Ribatti, D.; Di Raimondo, F.; Merchionne, F.; Dammacco, F. αvβ3 integrin engagement enhances cell invasiveness in human multiple myeloma. Haematologica 2002, 87, 836–845. [Google Scholar]
- Felding-Habermann, B.; Toole, T.E.; Smith, J.W.; Fransvea, E.; Ruggeri, Z.M.; Ginsberg, M.H.; Hughes, P.E.; Pampori, N.; Shattil, S.J.; Saven, A.; et al. Integrin activation controls metastasis in human breast cancer. Proc. Natl. Acad. Sci. USA 2001, 98, 1853. [Google Scholar] [CrossRef] [Green Version]
- Böger, C.; Warneke, V.S.; Behrens, H.M.; Kalthoff, H.; Goodman, S.L.; Becker, T.; Röcken, C. Integrins αvβ3 and αvβ5 as prognostic, diagnostic, and therapeutic targets in gastric cancer. Gastric Cancer 2015, 18, 784–795. [Google Scholar] [CrossRef] [Green Version]
- Schnell, O.; Krebs, B.; Wagner, E.; Romagna, A.; Beer, A.J.; Grau, S.J.; Thon, N.; Goetz, C.; Kretzschmar, H.A.; Tonn, J.C.; et al. Expression of integrin αvβ3 in gliomas correlates with tumor grade and is not restricted to tumor vasculature. Brain Pathol. 2008, 18, 378–386. [Google Scholar] [CrossRef] [Green Version]
- Berghoff, A.S.; Kovanda, A.K.; Melchardt, T.; Bartsch, R.; Hainfellner, J.A.; Sipos, B.; Schittenhelm, J.; Zielinski, C.C.; Widhalm, G.; Dieckmann, K.; et al. αvβ3, αvβ5 and αvβ6 integrins in brain metastases of lung cancer. Clin. Exp. Metastasis 2014, 31, 841–851. [Google Scholar] [CrossRef] [PubMed]
- Cooper, C.R.; Chay, C.H.; Pienta, K.J. The role of αvβ3 in prostate cancer progression. Neoplasia 2002, 4, 191–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Heß, K.; Böger, C.; Behrens, H.M.; Röcken, C. Correlation between the expression of integrins in prostate cancer and clinical outcome in 1284 patients. Ann. Diagn Pathol. 2014, 18, 343–350. [Google Scholar] [CrossRef] [Green Version]
- Hosotani, R.; Kawaguchi, M.; Masui, T.; Koshiba, T.; Ida, J.; Fujimoto, K.; Wada, M.; Doi, R.; Imamura, M. Expression of integrin alphaVbeta3 in pancreatic carcinoma: Relation to MMP-2 activation and lymph node metastasis. Pancreas 2002, 25, e30–e35. [Google Scholar] [CrossRef] [PubMed]
- Fabricius, E.M.; Wildner, G.P.; Kruse-Boitschenko, U.; Hoffmeister, B.; Goodman, S.L.; Raguse, J.D. Immunohistochemical analysis of integrins αvβ3, αvβ5 and α5β1, and their ligands, fibrinogen, fibronectin, osteopontin and vitronectin, in frozen sections of human oral head and neck squamous cell carcinomas. Exp. Med. 2011, 2, 9–19. [Google Scholar] [CrossRef]
- Caccavari, F.; Valdembri, D.; Sandri, C.; Bussolino, F.; Serini, G. Integrin signaling and lung cancer. Cell Adh. Migr. 2010, 4, 124–129. [Google Scholar] [CrossRef] [Green Version]
- Böger, C.; Kalthoff, H.; Goodman, S.L.; Behrens, H.M.; Röcken, C. Integrins and their ligands are expressed in non-small cell lung cancer but not correlated with parameters of disease progression. Virchows Arch. 2014, 464, 69–78. [Google Scholar] [CrossRef]
- Ermolayev, V.; Mohajerani, P.; Ale, A.; Sarantopoulos, A.; Aichler, M.; Kayser, G.; Walch, A.; Ntziachristos, V. Early recognition of lung cancer by integrin targeted imaging in K-ras mouse model. Int. J. Cancer 2015, 137, 1107–1118. [Google Scholar] [CrossRef]
- Hatley, R.J.D.; Macdonald, S.J.F.; Slack, R.J.; Le, J.; Ludbrook, S.B.; Lukey, P.T. An αv-RGD Integrin Inhibitor Toolbox: Drug Discovery Insight, Challenges and Opportunities. Angew. Chem. Int. Ed. Engl. 2018, 57, 3298–3321. [Google Scholar] [CrossRef]
- Bai, S.Y.; Xu, N.; Chen, C.; Song, Y.-l.; Hu, J.; Bai, C.-X. Integrin αvβ5 as a biomarker for the assessment of non-small cell lung cancer metastasis and overall survival. Clin. Respir. J. 2015, 9, 457–467. [Google Scholar] [CrossRef]
- Klemke, R.L.; Yebra, M.; Bayna, E.M.; Cheresh, D.A. Receptor tyrosine kinase signaling required for integrin alpha v beta 5-directed cell motility but not adhesion on vitronectin. J. Cell Biol. 1994, 127, 859–866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ricono, J.M.; Huang, M.; Barnes, L.A.; Lau, S.K.; Weis, S.M.; Schlaepfer, D.D.; Hanks, S.K.; Cheresh, D.A. Specific cross-talk between epidermal growth factor receptor and integrin alphavbeta5 promotes carcinoma cell invasion and metastasis. Cancer Res. 2009, 69, 1383–1391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Friedlander, M.; Brooks, P.C.; Shaffer, R.W.; Kincaid, C.M.; Varner, J.A.; Cheresh, D.A. Definition of two angiogenic pathways by distinct alpha v integrins. Science 1995, 270, 1500–1502. [Google Scholar] [CrossRef]
- Alavi, A.; Hood, J.D.; Frausto, R.; Stupack, D.G.; Cheresh, D.A. Role of Raf in vascular protection from distinct apoptotic stimuli. Science 2003, 301, 94–96. [Google Scholar] [CrossRef] [PubMed]
- Caswell, P.T.; Chan, M.; Lindsay, A.J.; McCaffrey, M.W.; Boettiger, D.; Norman, J.C. Rab-coupling protein coordinates recycling of alpha5beta1 integrin and EGFR1 to promote cell migration in 3D microenvironments. J. Cell Biol. 2008, 183, 143–155. [Google Scholar] [CrossRef] [Green Version]
- Sawada, K.; Mitra, A.K.; Radjabi, A.R.; Bhaskar, V.; Kistner, E.O.; Tretiakova, M.; Jagadeeswaran, S.; Montag, A.; Becker, A.; Kenny, H.A.; et al. Loss of E-cadherin promotes ovarian cancer metastasis via alpha 5-integrin, which is a therapeutic target. Cancer Res. 2008, 68, 2329–2339. [Google Scholar] [CrossRef] [Green Version]
- Mierke, C.T.; Frey, B.; Fellner, M.; Herrmann, M.; Fabry, B. Integrin α5β1 facilitates cancer cell invasion through enhanced contractile forces. J. Cell Sci. 2011, 124, 369–383. [Google Scholar] [CrossRef] [Green Version]
- Diaz, C.; Neubauer, S.; Rechenmacher, F.; Kessler, H.; Missirlis, D. Recruitment of αvβ3 integrin to α5β1 integrin-induced clusters enables focal adhesion maturation and cell spreading. J. Cell Sci. 2020, 133. [Google Scholar] [CrossRef]
- Morozevich, G.E.; Kozlova, N.I.; Ushakova, N.A.; Preobrazhenskaya, M.E.; Berman, A.E. Integrin α5β1 simultaneously controls EGFR-dependent proliferation and Akt-dependent pro-survival signaling in epidermoid carcinoma cells. Aging 2012, 4, 368–374. [Google Scholar] [CrossRef]
- Impola, U.; Uitto, V.J.; Hietanen, J.; Hakkinen, L.; Zhang, L.; Larjava, H.; Isaka, K.; Saarialho-Kere, U. Differential expression of matrilysin-1 (MMP-7), 92 kD gelatinase (MMP-9), and metalloelastase (MMP-12) in oral verrucous and squamous cell cancer. J. Pathol. 2004, 202, 14–22. [Google Scholar] [CrossRef]
- Moore, K.M.; Thomas, G.J.; Duffy, S.W.; Warwick, J.; Gabe, R.; Chou, P.; Ellis, I.O.; Green, A.R.; Haider, S.; Brouilette, K.; et al. Therapeutic targeting of integrin αvβ6 in breast cancer. J. Natl. Cancer Inst. 2014, 106. [Google Scholar] [CrossRef] [Green Version]
- Yang, G.Y.; Guo, S.; Dong, C.Y.; Wang, X.Q.; Hu, B.Y.; Liu, Y.F.; Chen, Y.W.; Niu, J.; Dong, J.H. Integrin αvβ6 sustains and promotes tumor invasive growth in colon cancer progression. World J. Gastroenterol. 2015, 21, 7457–7467. [Google Scholar] [CrossRef]
- Patsenker, E.; Wilkens, L.; Banz, V.; Osterreicher, C.H.; Weimann, R.; Eisele, S.; Keogh, A.; Stroka, D.; Zimmermann, A.; Stickel, F. The alphavbeta6 integrin is a highly specific immunohistochemical marker for cholangiocarcinoma. J. Hepatol. 2010, 52, 362–369. [Google Scholar] [CrossRef]
- Ahmed, N.; Riley, C.; Rice, G.E.; Quinn, M.A.; Baker, M.S. αvβ6 integrin-A marker for the malignant potential of epithelial ovarian cancer. J. Histochem. Cytochem. 2002, 50, 1371–1380. [Google Scholar] [CrossRef] [Green Version]
- Reader, C.S.; Vallath, S.; Steele, C.W.; Haider, S.; Brentnall, A.; Desai, A.; Moore, K.M.; Jamieson, N.B.; Chang, D.; Bailey, P.; et al. The integrin αvβ6 drives pancreatic cancer through diverse mechanisms and represents an effective target for therapy. J. Pathol. 2019, 249, 332–342. [Google Scholar] [CrossRef]
- Moore, K.M.; Desai, A.; Delgado, B.d.L.; Trabulo, S.M.D.; Reader, C.; Brown, N.F.; Murray, E.R.; Brentnall, A.; Howard, P.; Masterson, L.; et al. Integrin αvβ6-specific therapy for pancreatic cancer developed from foot-and-mouth-disease virus. Theranostics 2020, 10, 2930–2942. [Google Scholar] [CrossRef] [PubMed]
- Niu, J.; Li, Z. The roles of integrin αvβ6 in cancer. Cancer Lett. 2017, 403, 128–137. [Google Scholar] [CrossRef]
- Lawrence, D.A.; Pircher, R.; Krycevemartinerie, C.; Jullien, P. Normal Embryo Fibroblasts Release Transforming Growth-Factors in a Latent Form. J. Cell Physiol. 1984, 121, 184–188. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Wang, J.; Niu, W.; Liu, E.; Wang, J.; Peng, C.; Lin, P.; Wang, B.; Khan, A.Q.; Gao, H.; et al. The β6-integrin-ERK/MAP kinase pathway contributes to chemo resistance in colon cancer. Cancer Lett. 2013, 328, 325–334. [Google Scholar] [CrossRef] [PubMed]
- Desnoyers, A.; González, C.; Pérez-Segura, P.; Pandiella, A.; Amir, E.; Ocaña, A. Integrin ανβ6 Protein Expression and Prognosis in Solid Tumors: A Meta-Analysis. Mol. Diagn. Ther. 2020, 24, 143–151. [Google Scholar] [CrossRef] [PubMed]
- Mu, D.; Cambier, S.; Fjellbirkeland, L.; Baron, J.L.; Munger, J.S.; Kawakatsu, H.; Sheppard, D.; Broaddus, V.C.; Nishimura, S.L. The integrin αvβ8 mediates epithelial homeostasis through MT1-MMP-dependent activation of TGF-beta1. J. Cell Biol. 2002, 157, 493–507. [Google Scholar] [CrossRef] [Green Version]
- Khan, Z.; Marshall, J.F. The role of integrins in TGFβ activation in the tumour stroma. Cell Tissue Res. 2016, 365, 657–673. [Google Scholar] [CrossRef] [Green Version]
- Robertson, I.B.; Rifkin, D.B. Regulation of the Bioavailability of TGF-β and TGF-β-Related Proteins. Cold Spring Harb. Perspect. Biol. 2016, 8, a021907. [Google Scholar] [CrossRef] [PubMed]
- Zargham, R.; Thibault, G. Alpha 8 integrin expression is required for maintenance of the smooth muscle cell differentiated phenotype. Cardiovasc. Res. 2006, 71, 170–178. [Google Scholar] [CrossRef]
- Zargham, R.; Wamhoff, B.R.; Thibault, G. RNA interference targeting alpha8 integrin attenuates smooth muscle cell growth. FEBS Lett. 2007, 581, 939–943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Müller, U.; Wang, D.; Denda, S.; Meneses, J.J.; Pedersen, R.A.; Reichardt, L.F. Integrin alpha8beta1 is critically important for epithelial-mesenchymal interactions during kidney morphogenesis. Cell 1997, 88, 603–613. [Google Scholar] [CrossRef] [Green Version]
- Scherberich, A.; Tucker, R.P.; Degen, M.; Brown-Luedi, M.; Andres, A.-C.; Chiquet-Ehrismann, R. Tenascin-W is found in malignant mammary tumors, promotes alpha8 integrin-dependent motility and requires p38MAPK activity for BMP-2 and TNF-alpha induced expression in vitro. Oncogene 2005, 24, 1525–1532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bledzka, K.; Smyth, S.S.; Plow, E.F. Integrin αIIbβ3: From discovery to efficacious therapeutic target. Circ. Res. 2013, 112, 1189–1200. [Google Scholar] [CrossRef] [Green Version]
- Hodivala-Dilke, K.M.; McHugh, K.P.; Tsakiris, D.A.; Rayburn, H.; Crowley, D.; Ullman-Culleré, M.; Ross, F.P.; Coller, B.S.; Teitelbaum, S.; Hynes, R.O. Beta3-integrin-deficient mice are a model for Glanzmann thrombasthenia showing placental defects and reduced survival. J. Clin. Investig. 1999, 103, 229–238. [Google Scholar] [CrossRef] [Green Version]
- Reed, N.I.; Jo, H.; Chen, C.; Tsujino, K.; Arnold, T.D.; DeGrado, W.F.; Sheppard, D. The αvβ1 integrin plays a critical in vivo role in tissue fibrosis. Sci. Transl. Med. 2015, 7, 288ra279. [Google Scholar] [CrossRef] [Green Version]
- Wilkinson, A.L.; Barrett, J.W.; Slack, R.J. Pharmacological characterisation of a tool αvβ1 integrin small molecule RGD-mimetic inhibitor. Eur. J. Pharm. 2019, 842, 239–247. [Google Scholar] [CrossRef] [PubMed]
- Folkman, J. Tumor angiogenesis: Therapeutic implications. N. Engl. J. Med. 1971, 285, 1182–1186. [Google Scholar] [CrossRef]
- Carmeliet, P.; Jain, R.K. Angiogenesis in cancer and other diseases. Nature 2000, 407, 249–257. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hodivala-Dilke, K.M.; Reynolds, A.R.; Reynolds, L.E. Integrins in angiogenesis: Multitalented molecules in a balancing act. Cell Tissue Res. 2003, 314, 131–144. [Google Scholar] [CrossRef]
- Brooks, P.C.; Montgomery, A.M.; Rosenfeld, M.; Reisfeld, R.A.; Hu, T.; Klier, G.; Cheresh, D.A. Integrin αvβ3 antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels. Cell 1994, 79, 1157–1164. [Google Scholar] [CrossRef]
- Caswell, P.; Norman, J. Endocytic transport of integrins during cell migration and invasion. Trends Cell Biol. 2008, 18, 257–263. [Google Scholar] [CrossRef]
- Rüegg, C.; Alghisi, G.C. Vascular integrins: Therapeutic and imaging targets of tumor angiogenesis. Recent Results Cancer Res. 2010, 180, 83–101. [Google Scholar] [CrossRef]
- Mas-Moruno, C.; Beck, J.G.; Doedens, L.; Frank, A.O.; Marinelli, L.; Cosconati, S.; Novellino, E.; Kessler, H. Increasing αvβ3 selectivity of the anti-angiogenic drug cilengitide by N-methylation. Angew. Chem. (Int. Ed. Engl.) 2011, 50, 9496–9500. [Google Scholar] [CrossRef] [Green Version]
- Łasiñska, I.; Mackiewicz, J. Integrins as A New Target for Cancer Treatment. Anticancer Agents Med. Chem. 2019, 19, 580–586. [Google Scholar] [CrossRef]
- Lombardi, G.; Pambuku, A.; Bellu, L.; Farina, M.; Della Puppa, A.; Denaro, L.; Zagonel, V. Effectiveness of antiangiogenic drugs in glioblastoma patients: A systematic review and meta-analysis of randomized clinical trials. Crit. Rev. Oncol. Hematol. 2017, 111, 94–102. [Google Scholar] [CrossRef]
- Weller, M.; Nabors, L.B.; Gorlia, T.; Leske, H.; Rushing, E.; Bady, P.; Hicking, C.; Perry, J.; Hong, Y.K.; Roth, P.; et al. Cilengitide in newly diagnosed glioblastoma: Biomarker expression and outcome. Oncotarget 2016, 7, 15018–15032. [Google Scholar] [CrossRef] [PubMed]
- Nabors, L.B.; Fink, K.L.; Mikkelsen, T.; Grujicic, D.; Tarnawski, R.; Nam, D.H.; Mazurkiewicz, M.; Salacz, M.; Ashby, L.; Zagonel, V.; et al. Two cilengitide regimens in combination with standard treatment for patients with newly diagnosed glioblastoma and unmethylated MGMT gene promoter: Results of the open-label, controlled, randomized phase II CORE study. Neuro Oncol. 2015, 17, 708–717. [Google Scholar] [CrossRef]
- MacDonald, T.J.; Taga, T.; Shimada, H.; Tabrizi, P.; Zlokovic, B.V.; Cheresh, D.A.; Laug, W.E. Preferential susceptibility of brain tumors to the antiangiogenic effects of an alpha(v) integrin antagonist. Neurosurgery 2001, 48, 151–157. [Google Scholar] [CrossRef] [PubMed]
- Yamada, S.; Bu, X.Y.; Khankaldyyan, V.; Gonzales-Gomez, I.; McComb, J.G.; Laug, W.E. Effect of the angiogenesis inhibitor Cilengitide (EMD 121974) on glioblastoma growth in nude mice. Neurosurgery 2006, 59, 1304–1312; discussion 1312. [Google Scholar] [CrossRef] [PubMed]
- Reynolds, A.R.; Hart, I.R.; Watson, A.R.; Welti, J.C.; Silva, R.G.; Robinson, S.D.; Da Violante, G.; Gourlaouen, M.; Salih, M.; Jones, M.C.; et al. Stimulation of tumor growth and angiogenesis by low concentrations of RGD-mimetic integrin inhibitors. Nat. Med. 2009, 15, 392–400. [Google Scholar] [CrossRef]
- Mahabeleshwar, G.H.; Feng, W.Y.; Reddy, K.; Plow, E.F.; Byzova, T.V. Mechanisms of integrin-vascular endothelial growth factor receptor cross-activation in angiogenesis. Circ. Res. 2007, 101, 570–580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eskens, F.A.; Dumez, H.; Hoekstra, R.; Perschl, A.; Brindley, C.; Böttcher, S.; Wynendaele, W.; Drevs, J.; Verweij, J.; van Oosterom, A.T. Phase I and pharmacokinetic study of continuous twice weekly intravenous administration of Cilengitide (EMD 121974), a novel inhibitor of the integrins alphavbeta3 and alphavbeta5 in patients with advanced solid tumours. Eur. J. Cancer 2003, 39, 917–926. [Google Scholar] [CrossRef]
- Stupp, R.; Hegi, M.E.; Gorlia, T.; Erridge, S.C.; Perry, J.; Hong, Y.K.; Aldape, K.D.; Lhermitte, B.; Pietsch, T.; Grujicic, D.; et al. Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma with methylated MGMT promoter (CENTRIC EORTC 26071-22072 study): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2014, 15, 1100–1108. [Google Scholar] [CrossRef] [Green Version]
- Davis, G.E. Affinity of integrins for damaged extracellular matrix: αvβ3 binds to denatured collagen type I through RGD sites. Biochem. Biophys. Res. Commun. 1992, 182, 1025–1031. [Google Scholar] [CrossRef]
- Tilki, D.; Kilic, N.; Sevinc, S.; Zywietz, F.; Stief, C.G.; Ergun, S. Zone-specific remodeling of tumor blood vessels affects tumor growth. Cancer 2007, 110, 2347–2362. [Google Scholar] [CrossRef]
- Gaengel, K.; Genové, G.; Armulik, A.; Betsholtz, C. Endothelial-mural cell signaling in vascular development and angiogenesis. Arter. Thromb. Vasc. Biol. 2009, 29, 630–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rahbari, N.N.; Kedrin, D.; Incio, J.; Liu, H.; Ho, W.W.; Nia, H.T.; Edrich, C.M.; Jung, K.; Daubriac, J.; Chen, I.; et al. Anti-VEGF therapy induces ECM remodeling and mechanical barriers to therapy in colorectal cancer liver metastases. Sci. Transl. Med. 2016, 8, 360ra135. [Google Scholar] [CrossRef] [Green Version]
- Mai, J.; Virtue, A.; Shen, J.; Wang, H.; Yang, X.F. An evolving new paradigm: Endothelial cells--conditional innate immune cells. J. Hematol. Oncol. 2013, 6, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Klein, D. Vascular Wall-Resident Multipotent Stem Cells of Mesenchymal Nature within the Process of Vascular Remodeling: Cellular Basis, Clinical Relevance, and Implications for Stem Cell Therapy. Stem Cells Int. 2016, 2016, 1905846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guipaud, O.; Jaillet, C.; Clément-Colmou, K.; François, A.; Supiot, S.; Milliat, F. The importance of the vascular endothelial barrier in the immune-inflammatory response induced by radiotherapy. Br. J. Radiol. 2018, 91, 20170762. [Google Scholar] [CrossRef] [PubMed]
- Maeda, H.; Bharate, G.Y.; Daruwalla, J. Polymeric drugs for efficient tumor-targeted drug delivery based on EPR-effect. Eur. J. Pharm. Biopharm. 2009, 71, 409–419. [Google Scholar] [CrossRef]
- Weis, S.M.; Cheresh, D.A. Tumor angiogenesis: Molecular pathways and therapeutic targets. Nat. Med. 2011, 17, 1359–1370. [Google Scholar] [CrossRef]
- Torsvik, A.; Bjerkvig, R. Mesenchymal stem cell signaling in cancer progression. Cancer Treat. Rev. 2013, 39, 180–188. [Google Scholar] [CrossRef]
- Fukumura, D.; Jain, R.K. Tumor microvasculature and microenvironment: Targets for anti-angiogenesis and normalization. Microvasc. Res. 2007, 74, 72–84. [Google Scholar] [CrossRef] [Green Version]
- Greenberg, J.I.; Shields, D.J.; Barillas, S.G.; Acevedo, L.M.; Murphy, E.; Huang, J.; Scheppke, L.; Stockmann, C.; Johnson, R.S.; Angle, N.; et al. A role for VEGF as a negative regulator of pericyte function and vessel maturation. Nature 2008, 456, 809–813. [Google Scholar] [CrossRef]
- Carmeliet, P.; Jain, R.K. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat. Rev. Drug Discov. 2011, 10, 417–427. [Google Scholar] [CrossRef] [PubMed]
- Van der Veldt, A.A.; Lubberink, M.; Bahce, I.; Walraven, M.; de Boer, M.P.; Greuter, H.N.; Hendrikse, N.H.; Eriksson, J.; Windhorst, A.D.; Postmus, P.E.; et al. Rapid decrease in delivery of chemotherapy to tumors after anti-VEGF therapy: Implications for scheduling of anti-angiogenic drugs. Cancer Cell 2012, 21, 82–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jain, R.K. Antiangiogenesis strategies revisited: From starving tumors to alleviating hypoxia. Cancer Cell 2014, 26, 605–622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Z.; Dabrosin, C.; Yin, X.; Fuster, M.M.; Arreola, A.; Rathmell, W.K.; Generali, D.; Nagaraju, G.P.; El-Rayes, B.; Ribatti, D.; et al. Broad targeting of angiogenesis for cancer prevention and therapy. Semin Cancer Biol. 2015, 35, S224–S243. [Google Scholar] [CrossRef] [PubMed]
- Ye, W. The Complexity of Translating Anti-angiogenesis Therapy from Basic Science to the Clinic. Dev. Cell 2016, 37, 114–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [Green Version]
- Lambert, A.W.; Pattabiraman, D.R.; Weinberg, R.A. Emerging Biological Principles of Metastasis. Cell 2017, 168, 670–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Niland, S.; Eble, J.A. Neuropilins in the Context of Tumor Vasculature. Int. J. Mol. Sci. 2019, 20, 639. [Google Scholar] [CrossRef] [Green Version]
- Mecollari, V.; Nieuwenhuis, B.; Verhaagen, J. A perspective on the role of class III semaphorin signaling in central nervous system trauma. Front. Cell Neuro Sci. 2014, 8, 328. [Google Scholar] [CrossRef] [Green Version]
- Soker, S.; Miao, H.Q.; Nomi, M.; Takashima, S.; Klagsbrun, M. VEGF165 mediates formation of complexes containing VEGFR-2 and neuropilin-1 that enhance VEGF165-receptor binding. J. Cell Biochem. 2002, 85, 357–368. [Google Scholar] [CrossRef]
- Hamerlik, P.; Lathia, J.D.; Rasmussen, R.; Wu, Q.; Bartkova, J.; Lee, M.; Moudry, P.; Bartek, J., Jr.; Fischer, W.; Lukas, J.; et al. Autocrine VEGF-VEGFR2-Neuropilin-1 signaling promotes glioma stem-like cell viability and tumor growth. J. Exp. Med. 2012, 209, 507–520. [Google Scholar] [CrossRef] [Green Version]
- Bagci, T.; Wu, J.K.; Pfannl, R.; Ilag, L.L.; Jay, D.G. Autocrine semaphorin 3A signaling promotes glioblastoma dispersal. Oncogene 2009, 28, 3537–3550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, Z.; Tessier-Lavigne, M. Neuropilin is a receptor for the axonal chemorepellent Semaphorin III. Cell 1997, 90, 739–751. [Google Scholar] [CrossRef] [Green Version]
- Kolodkin, A.L.; Levengood, D.V.; Rowe, E.G.; Tai, Y.T.; Giger, R.J.; Ginty, D.D. Neuropilin is a semaphorin III receptor. Cell 1997, 90, 753–762. [Google Scholar] [CrossRef] [Green Version]
- Valdembri, D.; Caswell, P.T.; Anderson, K.I.; Schwarz, J.P.; König, I.; Astanina, E.; Caccavari, F.; Norman, J.C.; Humphries, M.J.; Bussolino, F.; et al. Neuropilin-1/GIPC1 signaling regulates alpha5beta1 integrin traffic and function in endothelial cells. PLoS Biol. 2009, 7, e25. [Google Scholar] [CrossRef]
- Robinson, S.D.; Reynolds, L.E.; Kostourou, V.; Reynolds, A.R.; da Silva, R.G.; Tavora, B.; Baker, M.; Marshall, J.F.; Hodivala-Dilke, K.M. αvβ3 integrin limits the contribution of neuropilin-1 to vascular endothelial growth factor-induced angiogenesis. J. Biol. Chem. 2009, 284, 33966–33981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fukasawa, M.; Matsushita, A.; Korc, M. Neuropilin-1 interacts with integrin beta1 and modulates pancreatic cancer cell growth, survival and invasion. Cancer Biol. 2007, 6, 1173–1180. [Google Scholar] [CrossRef] [Green Version]
- Byzova, T.V.; Goldman, C.K.; Pampori, N.; Thomas, K.A.; Bett, A.; Shattil, S.J.; Plow, E.F. A mechanism for modulation of cellular responses to VEGF: Activation of the integrins. Mol. Cell 2000, 6, 851–860. [Google Scholar] [CrossRef]
- Cao, Y.; Hoeppner, L.H.; Bach, S.E.G.; Guo, Y.; Wang, E.; Wu, J.; Cowley, M.J.; Chang, D.K.; Waddell, N.; Grimmond, S.M. Neuropilin-2 promotes extravasation and metastasis by interacting with endothelial α5 integrin. Cancer Res. 2013, 73, 4579–4590. [Google Scholar] [CrossRef] [Green Version]
- Ou, J.J.; Wei, X.; Peng, Y.; Zha, L.; Zhou, R.B.; Shi, H.; Zhou, Q.; Liang, H.J. Neuropilin-2 mediates lymphangiogenesis of colorectal carcinoma via a VEGFC/VEGFR3 independent signaling. Cancer Lett. 2015, 358, 200–209. [Google Scholar] [CrossRef] [PubMed]
- Koch, S.; Tugues, S.; Li, X.; Gualandi, L.; Claesson-Welsh, L. Signal transduction by vascular endothelial growth factor receptors. Biochem. J. 2011, 437, 169–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perrot-Applanat, M.; Di Benedetto, M. Autocrine functions of VEGF in breast tumor cells: Adhesion, survival, migration and invasion. Cell Adh. Migr. 2012, 6, 547–553. [Google Scholar] [CrossRef] [Green Version]
- Goel, H.L.; Pursell, B.; Chang, C.; Shaw, L.M.; Mao, J.; Simin, K.; Kumar, P.; Vander Kooi, C.W.; Shultz, L.D.; Greiner, D.L.; et al. GLI1 regulates a novel neuropilin-2/α6β1 integrin based autocrine pathway that contributes to breast cancer initiation. Embo Mol. Med. 2013, 5, 488–508. [Google Scholar] [CrossRef] [PubMed]
- Goel, H.L.; Mercurio, A.M. Enhancing integrin function by VEGF/neuropilin signaling: Implications for tumor biology. Cell Adh. Migr. 2012, 6, 554–560. [Google Scholar] [CrossRef] [Green Version]
- Moreno-Layseca, P.; Icha, J.; Hamidi, H.; Ivaska, J. Integrin trafficking in cells and tissues. Nat. Cell Biol. 2019, 21, 122–132. [Google Scholar] [CrossRef]
- Kawauchi, T. Cell adhesion and its endocytic regulation in cell migration during neural development and cancer metastasis. Int. J. Mol. Sci. 2012, 13, 4564–4590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gu, Z.; Noss, E.H.; Hsu, V.W.; Brenner, M.B. Integrins traffic rapidly via circular dorsal ruffles and macropinocytosis during stimulated cell migration. J. Cell Biol. 2011, 193, 61–70. [Google Scholar] [CrossRef]
- Lakshminarayan, R.; Wunder, C.; Becken, U.; Howes, M.T.; Benzing, C.; Arumugam, S.; Sales, S.; Ariotti, N.; Chambon, V.; Lamaze, C.; et al. Galectin-3 drives glycosphingolipid-dependent biogenesis of clathrin-independent carriers. Nat. Cell Biol. 2014, 16, 595–606. [Google Scholar] [CrossRef]
- Arias-Salgado, E.G.; Lizano, S.; Sarkar, S.; Brugge, J.S.; Ginsberg, M.H.; Shattil, S.J. Src kinase activation by direct interaction with the integrin beta cytoplasmic domain. Proc. Natl. Acad. Sci. USA 2003, 100, 13298–13302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chao, W.T.; Kunz, J. Focal adhesion disassembly requires clathrin-dependent endocytosis of integrins. FEBS Lett. 2009, 583, 1337–1343. [Google Scholar] [CrossRef] [Green Version]
- Ezratty, E.J.; Bertaux, C.; Marcantonio, E.E.; Gundersen, G.G. Clathrin mediates integrin endocytosis for focal adhesion disassembly in migrating cells. J. Cell Biol. 2009, 187, 733–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, F.; Sottile, J. Caveolin-1-dependent beta1 integrin endocytosis is a critical regulator of fibronectin turnover. J. Cell Sci. 2008, 121, 2360–2371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bridgewater, R.E.; Norman, J.C.; Caswell, P.T. Integrin trafficking at a glance. J. Cell Sci. 2012, 125, 3695–3701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dozynkiewicz, M.A.; Jamieson, N.B.; Macpherson, I.; Grindlay, J.; van den Berghe, P.V.; von Thun, A.; Morton, J.P.; Gourley, C.; Timpson, P.; Nixon, C.; et al. Rab25 and CLIC3 collaborate to promote integrin recycling from late endosomes/lysosomes and drive cancer progression. Dev. Cell 2012, 22, 131–145. [Google Scholar] [CrossRef] [Green Version]
- Tiwari, S.; Askari, J.A.; Humphries, M.J.; Bulleid, N.J. Divalent cations regulate the folding and activation status of integrins during their intracellular trafficking. J. Cell Sci. 2011, 124, 1672–1680. [Google Scholar] [CrossRef] [Green Version]
- Böttcher, R.T.; Stremmel, C.; Meves, A.; Meyer, H.; Widmaier, M.; Tseng, H.Y.; Fässler, R. Sorting nexin 17 prevents lysosomal degradation of β1 integrins by binding to the β1-integrin tail. Nat. Cell Biol. 2012, 14, 584–592. [Google Scholar] [CrossRef]
- Margadant, C.; Kreft, M.; de Groot, D.J.; Norman, J.C.; Sonnenberg, A. Distinct roles of talin and kindlin in regulating integrin α5β1 function and trafficking. Curr. Biol. 2012, 22, 1554–1563. [Google Scholar] [CrossRef] [Green Version]
- Steinberg, F.; Heesom, K.J.; Bass, M.D.; Cullen, P.J. SNX17 protects integrins from degradation by sorting between lysosomal and recycling pathways. J. Cell Biol. 2012, 197, 219–230. [Google Scholar] [CrossRef]
- Caswell, P.T.; Norman, J.C. Integrin trafficking and the control of cell migration. Traffic 2006, 7, 14–21. [Google Scholar] [CrossRef] [Green Version]
- Morgan, M.R.; Byron, A.; Humphries, M.J.; Bass, M.D. Giving off mixed signals--distinct functions of alpha5beta1 and alphavbeta3 integrins in regulating cell behaviour. IUBMB Life 2009, 61, 731–738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scita, G.; Di Fiore, P.P. The endocytic matrix. Nature 2010, 463, 464–473. [Google Scholar] [CrossRef]
- Roberts, M.; Barry, S.; Woods, A.; van der Sluijs, P.; Norman, J. PDGF-regulated rab4-dependent recycling of αvβ3 integrin from early endosomes is necessary for cell adhesion and spreading. Curr. Biol. 2001, 11, 1392–1402. [Google Scholar] [CrossRef] [Green Version]
- Powelka, A.M.; Sun, J.; Li, J.; Gao, M.; Shaw, L.M.; Sonnenberg, A.; Hsu, V.W. Stimulation-dependent recycling of integrin beta1 regulated by ARF6 and Rab11. Traffic 2004, 5, 20–36. [Google Scholar] [CrossRef] [PubMed]
- Jones, M.C.; Caswell, P.T.; Moran-Jones, K.; Roberts, M.; Barry, S.T.; Gampel, A.; Mellor, H.; Norman, J.C. VEGFR1 (Flt1) regulates Rab4 recycling to control fibronectin polymerization and endothelial vessel branching. Traffic 2009, 10, 754–766. [Google Scholar] [CrossRef] [PubMed]
- Grant, B.D.; Donaldson, J.G. Pathways and mechanisms of endocytic recycling. Nat. Rev. Mol. Cell Biol. 2009, 10, 597–608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roberts, M.S.; Woods, A.J.; Dale, T.C.; Van Der Sluijs, P.; Norman, J.C. Protein kinase B/Akt acts via glycogen synthase kinase 3 to regulate recycling of alpha v beta 3 and alpha 5 beta 1 integrins. Mol. Cell Biol. 2004, 24, 1505–1515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yoon, S.O.; Shin, S.; Mercurio, A.M. Hypoxia stimulates carcinoma invasion by stabilizing microtubules and promoting the Rab11 trafficking of the alpha6beta4 integrin. Cancer Res. 2005, 65, 2761–2769. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Ballif, B.A.; Powelka, A.M.; Dai, J.; Gygi, S.P.; Hsu, V.W. Phosphorylation of ACAP1 by Akt regulates the stimulation-dependent recycling of integrin beta1 to control cell migration. Dev. Cell 2005, 9, 663–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, J.; Peters, P.J.; Bai, M.; Dai, J.; Bos, E.; Kirchhausen, T.; Kandror, K.V.; Hsu, V.W. An ACAP1-containing clathrin coat complex for endocytic recycling. J. Cell Biol. 2007, 178, 453–464. [Google Scholar] [CrossRef]
- Fang, Z.; Takizawa, N.; Wilson, K.A.; Smith, T.C.; Delprato, A.; Davidson, M.W.; Lambright, D.G.; Luna, E.J. The membrane-associated protein, supervillin, accelerates F-actin-dependent rapid integrin recycling and cell motility. Traffic 2010, 11, 782–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- White, D.P.; Caswell, P.T.; Norman, J.C. αvβ3 and α5β1 integrin recycling pathways dictate downstream Rho kinase signaling to regulate persistent cell migration. J. Cell Biol. 2007, 177, 515–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Danen, E.H.; van Rheenen, J.; Franken, W.; Huveneers, S.; Sonneveld, P.; Jalink, K.; Sonnenberg, A. Integrins control motile strategy through a Rho-cofilin pathway. J. Cell Biol. 2005, 169, 515–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morgan, M.R.; Hamidi, H.; Bass, M.D.; Warwood, S.; Ballestrem, C.; Humphries, M.J. Syndecan-4 phosphorylation is a control point for integrin recycling. Dev. Cell 2013, 24, 472–485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jacquemet, G.; Green, D.M.; Bridgewater, R.E.; von Kriegsheim, A.; Humphries, M.J.; Norman, J.C.; Caswell, P.T. RCP-driven α5β1 recycling suppresses Rac and promotes RhoA activity via the RacGAP1-IQGAP1 complex. J. Cell Biol. 2013, 202, 917–935. [Google Scholar] [CrossRef] [Green Version]
- Rainero, E.; Caswell, P.T.; Muller, P.A.; Grindlay, J.; McCaffrey, M.W.; Zhang, Q.; Wakelam, M.J.; Vousden, K.H.; Graziani, A.; Norman, J.C. Diacylglycerol kinase α controls RCP-dependent integrin trafficking to promote invasive migration. J. Cell Biol. 2012, 196, 277–295. [Google Scholar] [CrossRef]
- Sottile, J.; Chandler, J. Fibronectin matrix turnover occurs through a caveolin-1-dependent process. Mol. Biol. Cell 2005, 16, 757–768. [Google Scholar] [CrossRef] [Green Version]
- Arjonen, A.; Alanko, J.; Veltel, S.; Ivaska, J. Distinct recycling of active and inactive β1 integrins. Traffic 2012, 13, 610–625. [Google Scholar] [CrossRef] [Green Version]
- Lobert, V.H.; Stenmark, H. The ESCRT machinery mediates polarization of fibroblasts through regulation of myosin light chain. J. Cell Sci. 2012, 125, 29–36. [Google Scholar] [CrossRef] [Green Version]
- Lobert, V.H.; Brech, A.; Pedersen, N.M.; Wesche, J.; Oppelt, A.; Malerød, L.; Stenmark, H. Ubiquitination of alpha 5 beta 1 integrin controls fibroblast migration through lysosomal degradation of fibronectin-integrin complexes. Dev. Cell 2010, 19, 148–159. [Google Scholar] [CrossRef] [Green Version]
- Zhou, X.; Rowe, R.G.; Hiraoka, N.; George, J.P.; Wirtz, D.; Mosher, D.F.; Virtanen, I.; Chernousov, M.A.; Weiss, S.J. Fibronectin fibrillogenesis regulates three-dimensional neovessel formation. Genes Dev. 2008, 22, 1231–1243. [Google Scholar] [CrossRef] [Green Version]
- Astrof, S.; Hynes, R.O. Fibronectins in vascular morphogenesis. Angiogenesis 2009, 12, 165–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schwarzbauer, J.E.; DeSimone, D.W. Fibronectins, their fibrillogenesis, and in vivo functions. Cold Spring Harb. Perspect. Biol. 2011, 3. [Google Scholar] [CrossRef] [Green Version]
- George, E.L.; Baldwin, H.S.; Hynes, R.O. Fibronectins are essential for heart and blood vessel morphogenesis but are dispensable for initial specification of precursor cells. Blood 1997, 90, 3073–3081. [Google Scholar] [CrossRef] [PubMed]
- Shi, F.; Sottile, J. MT1-MMP regulates the turnover and endocytosis of extracellular matrix fibronectin. J. Cell Sci. 2011, 124, 4039–4050. [Google Scholar] [CrossRef] [Green Version]
- Novo, D.; Heath, N.; Mitchell, L.; Caligiuri, G.; MacFarlane, A.; Reijmer, D.; Charlton, L.; Knight, J.; Calka, M.; McGhee, E.; et al. Mutant p53s generate pro-invasive niches by influencing exosome podocalyxin levels. Nat. Commun. 2018, 9, 5069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Caswell, P.T.; Vadrevu, S.; Norman, J.C. Integrins: Masters and slaves of endocytic transport. Nat. Rev. Mol. Cell Biol. 2009, 10, 843–853. [Google Scholar] [CrossRef]
- Chung, Y.C.; Wei, W.C.; Huang, S.H.; Shih, C.M.; Hsu, C.P.; Chang, K.J.; Chao, W.T. Rab11 regulates E-cadherin expression and induces cell transformation in colorectal carcinoma. BMC Cancer 2014, 14, 587. [Google Scholar] [CrossRef] [Green Version]
- Palmieri, D.; Bouadis, A.; Ronchetti, R.; Merino, M.J.; Steeg, P.S. Rab11a differentially modulates epidermal growth factor-induced proliferation and motility in immortal breast cells. Breast Cancer Res. Treat. 2006, 100, 127–137. [Google Scholar] [CrossRef]
- Pellinen, T.; Arjonen, A.; Vuoriluoto, K.; Kallio, K.; Fransen, J.A.; Ivaska, J. Small GTPase Rab21 regulates cell adhesion and controls endosomal traffic of beta1-integrins. J. Cell Biol. 2006, 173, 767–780. [Google Scholar] [CrossRef]
- Mai, A.; Veltel, S.; Pellinen, T.; Padzik, A.; Coffey, E.; Marjomäki, V.; Ivaska, J. Competitive binding of Rab21 and p120RasGAP to integrins regulates receptor traffic and migration. J. Cell Biol. 2011, 194, 291–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, S.; Hu, C.; Wu, F.; He, S. Rab25 GTPase: Functional roles in cancer. Oncotarget 2017, 8, 64591–64599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Caswell, P.T.; Spence, H.J.; Parsons, M.; White, D.P.; Clark, K.; Cheng, K.W.; Mills, G.B.; Humphries, M.J.; Messent, A.J.; Anderson, K.I.; et al. Rab25 associates with α5vβ1 integrin to promote invasive migration in 3D microenvironments. Dev. Cell 2007, 13, 496–510. [Google Scholar] [CrossRef] [PubMed]
- Rink, J.; Ghigo, E.; Kalaidzidis, Y.; Zerial, M. Rab conversion as a mechanism of progression from early to late endosomes. Cell 2005, 122, 735–749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Z.Y.; Cai, L.; Zhu, J.; Chen, M.; Chen, J.; Li, Z.H.; Liu, X.D.; Wang, S.G.; Bie, P.; Jiang, P.; et al. Fyn requires HnRNPA2B1 and Sam68 to synergistically regulate apoptosis in pancreatic cancer. Carcinogenesis 2011, 32, 1419–1426. [Google Scholar] [CrossRef] [Green Version]
- Goldenring, J.R. Recycling endosomes. Curr. Opin. Cell Biol. 2015, 35, 117–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Das, L.; Anderson, T.A.; Gard, J.M.; Sroka, I.C.; Strautman, S.R.; Nagle, R.B.; Morrissey, C.; Knudsen, B.S.; Cress, A.E. Characterization of Laminin Binding Integrin Internalization in Prostate Cancer Cells. J. Cell Biochem. 2017, 118, 1038–1049. [Google Scholar] [CrossRef] [Green Version]
- Bretscher, M.S. Circulating integrins: α5β1, α6β4 and Mac-1, but not α3β1, α4β1 or LFA-1. Embo J. 1992, 11, 405–410. [Google Scholar] [CrossRef]
- Harryman, W.L.; Hinton, J.P.; Rubenstein, C.P.; Singh, P.; Nagle, R.B.; Parker, S.J.; Knudsen, B.S.; Cress, A.E. The Cohesive Metastasis Phenotype in Human Prostate Cancer. Biochim. Biophys. Acta 2016, 1866, 221–231. [Google Scholar] [CrossRef] [Green Version]
- Harryman, W.L.; Gard, J.M.C.; Pond, K.W.; Simpson, S.J.; Heppner, L.H.; Hernandez-Cortes, D.; Little, A.S.; Eschbacher, J.M.; Cress, A.E. Targeting the Cohesive Cluster Phenotype in Chordoma via β1 Integrin Increases Ionizing Radiation Efficacy. Neoplasia 2017, 19, 919–927. [Google Scholar] [CrossRef]
- Willis, S.; Villalobos, V.M.; Gevaert, O.; Abramovitz, M.; Williams, C.; Sikic, B.I.; Leyland-Jones, B. Single Gene Prognostic Biomarkers in Ovarian Cancer: A Meta-Analysis. PLoS ONE 2016, 11, e0149183. [Google Scholar] [CrossRef]
- Das, L.; Gard, J.M.C.; Prekeris, R.; Nagle, R.B.; Morrissey, C.; Knudsen, B.S.; Miranti, C.K.; Cress, A.E. Novel Regulation of Integrin Trafficking by Rab11-FIP5 in Aggressive Prostate Cancer. Mol. Cancer Res. 2018, 16, 1319–1331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Howe, E.N.; Burnette, M.D.; Justice, M.E.; Schnepp, P.M.; Hedrick, V.; Clancy, J.W.; Guldner, I.H.; Lamere, A.T.; Li, J.; Aryal, U.K.; et al. Rab11b-mediated integrin recycling promotes brain metastatic adaptation and outgrowth. Nat. Commun. 2020, 11, 3017. [Google Scholar] [CrossRef] [PubMed]
- Mosesson, Y.; Mills, G.B.; Yarden, Y. Derailed endocytosis: An emerging feature of cancer. Nat. Rev. Cancer 2008, 8, 835–850. [Google Scholar] [CrossRef] [PubMed]
- Boman, A.L.; Zhang, C.; Zhu, X.; Kahn, R.A. A family of ADP-ribosylation factor effectors that can alter membrane transport through the trans-Golgi. Mol. Biol. Cell 2000, 11, 1241–1255. [Google Scholar] [CrossRef] [Green Version]
- Dell’Angelica, E.C.; Puertollano, R.; Mullins, C.; Aguilar, R.C.; Vargas, J.D.; Hartnell, L.M.; Bonifacino, J.S. GGAs: A family of ADP ribosylation factor-binding proteins related to adaptors and associated with the Golgi complex. J. Cell Biol. 2000, 149, 81–94. [Google Scholar] [CrossRef] [Green Version]
- Puertollano, R.; Randazzo, P.A.; Presley, J.F.; Hartnell, L.M.; Bonifacino, J.S. The GGAs promote ARF-dependent recruitment of clathrin to the TGN. Cell 2001, 105, 93–102. [Google Scholar] [CrossRef] [Green Version]
- Puertollano, R.; Bonifacino, J.S. Interactions of GGA3 with the ubiquitin sorting machinery. Nat. Cell Biol. 2004, 6, 244–251. [Google Scholar] [CrossRef]
- Sahgal, P.; Alanko, J.; Icha, J.; Paatero, I.; Hamidi, H.; Arjonen, A.; Pietilä, M.; Rokka, A.; Ivaska, J. GGA2 and RAB13 promote activity-dependent β1-integrin recycling. J. Cell Sci. 2019, 132. [Google Scholar] [CrossRef] [Green Version]
- McNally, K.E.; Faulkner, R.; Steinberg, F.; Gallon, M.; Ghai, R.; Pim, D.; Langton, P.; Pearson, N.; Danson, C.M.; Nägele, H.; et al. Retriever is a multiprotein complex for retromer-independent endosomal cargo recycling. Nat. Cell Biol. 2017, 19, 1214–1225. [Google Scholar] [CrossRef]
- Jonker, C.T.H.; Galmes, R.; Veenendaal, T.; Ten Brink, C.; van der Welle, R.E.N.; Liv, N.; de Rooij, J.; Peden, A.A.; van der Sluijs, P.; Margadant, C.; et al. Vps3 and Vps8 control integrin trafficking from early to recycling endosomes and regulate integrin-dependent functions. Nat. Commun. 2018, 9, 792. [Google Scholar] [CrossRef]
- Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef] [PubMed]
- Théry, C.; Zitvogel, L.; Amigorena, S. Exosomes: Composition, biogenesis and function. Nat. Rev. Immunol. 2002, 2, 569–579. [Google Scholar] [CrossRef] [PubMed]
- Simons, M.; Raposo, G. Exosomes--vesicular carriers for intercellular communication. Curr. Opin. Cell Biol. 2009, 21, 575–581. [Google Scholar] [CrossRef] [PubMed]
- Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.; et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef] [Green Version]
- Skog, J.; Würdinger, T.; van Rijn, S.; Meijer, D.H.; Gainche, L.; Sena-Esteves, M.; Curry, W.T., Jr.; Carter, B.S.; Krichevsky, A.M.; Breakefield, X.O. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell Biol. 2008, 10, 1470–1476. [Google Scholar] [CrossRef] [PubMed]
- Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [Green Version]
- Kahlert, C.; Melo, S.A.; Protopopov, A.; Tang, J.; Seth, S.; Koch, M.; Zhang, J.; Weitz, J.; Chin, L.; Futreal, A.; et al. Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J. Biol. Chem. 2014, 289, 3869–3875. [Google Scholar] [CrossRef] [Green Version]
- Nilendu, P.; Sarode, S.C.; Jahagirdar, D.; Tandon, I.; Patil, S.; Sarode, G.S.; Pal, J.K.; Sharma, N.K. Mutual concessions and compromises between stromal cells and cancer cells: Driving tumor development and drug resistance. Cell Oncol. 2018, 41, 353–367. [Google Scholar] [CrossRef]
- Sun, W.; Luo, J.D.; Jiang, H.; Duan, D.D. Tumor exosomes: A double-edged sword in cancer therapy. Acta Pharm. Sin. 2018, 39, 534–541. [Google Scholar] [CrossRef] [Green Version]
- Qiu, X.; Li, Z.; Han, X.; Zhen, L.; Luo, C.; Liu, M.; Yu, K.; Ren, Y. Tumor-derived nanovesicles promote lung distribution of the therapeutic nanovector through repression of Kupffer cell-mediated phagocytosis. Theranostics 2019, 9, 2618–2636. [Google Scholar] [CrossRef]
- Mirzaei, H.; Sahebkar, A.; Jaafari, M.R.; Goodarzi, M.; Mirzaei, H.R. Diagnostic and Therapeutic Potential of Exosomes in Cancer: The Beginning of a New Tale? J. Cell Physiol. 2017, 232, 3251–3260. [Google Scholar] [CrossRef] [PubMed]
- Carney, R.P.; Hazari, S.; Rojalin, T.; Knudson, A.; Gao, T.; Tang, Y.; Liu, R.; Viitala, T.; Yliperttula, M.; Lam, K.S. Targeting Tumor-Associated Exosomes with Integrin-Binding Peptides. Adv. Biosyst. 2017, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wortzel, I.; Dror, S.; Kenific, C.M.; Lyden, D. Exosome-Mediated Metastasis: Communication from a Distance. Dev. Cell 2019, 49, 347–360. [Google Scholar] [CrossRef]
- Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H.; et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 2015, 17, 816–826. [Google Scholar] [CrossRef]
- Tominaga, N.; Kosaka, N.; Ono, M.; Katsuda, T.; Yoshioka, Y.; Tamura, K.; Lötvall, J.; Nakagama, H.; Ochiya, T. Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood-brain barrier. Nat. Commun. 2015, 6, 6716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, X.; Tang, M.; Zhu, Q.; Wang, X.; Lin, Y.; Wang, X. The exosomal integrin α5β1/AEP complex derived from epithelial ovarian cancer cells promotes peritoneal metastasis through regulating mesothelial cell proliferation and migration. Cell Oncol. 2020, 43, 263–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hurwitz, S.N.; Meckes, D.G., Jr. Extracellular Vesicle Integrins Distinguish Unique Cancers. Proteomes 2019, 7, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, X.; Kang, Y. Organotropism of breast cancer metastasis. J. Mammary Gland Biol. Neoplasia 2007, 12, 153–162. [Google Scholar] [CrossRef]
- Singh, A.; Fedele, C.; Lu, H.; Nevalainen, M.T.; Keen, J.H.; Languino, L.R. Exosome-mediated Transfer of αvβ3 Integrin from Tumorigenic to Nontumorigenic Cells Promotes a Migratory Phenotype. Mol. Cancer Res. 2016, 14, 1136–1146. [Google Scholar] [CrossRef] [Green Version]
- Fedele, C.; Singh, A.; Zerlanko, B.J.; Iozzo, R.V.; Languino, L.R. The αvβ6 integrin is transferred intercellularly via exosomes. J. Biol. Chem. 2015, 290, 4545–4551. [Google Scholar] [CrossRef] [Green Version]
- Sun, D.; Zhuang, X.; Zhang, S.; Deng, Z.B.; Grizzle, W.; Miller, D.; Zhang, H.G. Exosomes are endogenous nanoparticles that can deliver biological information between cells. Adv. Drug Deliv. Rev. 2013, 65, 342–347. [Google Scholar] [CrossRef]
- Vader, P.; Mol, E.A.; Pasterkamp, G.; Schiffelers, R.M. Extracellular vesicles for drug delivery. Adv. Drug Deliv. Rev. 2016, 106, 148–156. [Google Scholar] [CrossRef] [PubMed]
- Pegtel, D.M.; Gould, S.J. Exosomes. Annu. Rev. Biochem. 2019, 88, 487–514. [Google Scholar] [CrossRef] [PubMed]
- Rayamajhi, S.; Nguyen, T.D.T.; Marasini, R.; Aryal, S. Macrophage-derived exosome-mimetic hybrid vesicles for tumor targeted drug delivery. Acta Biomater. 2019, 94, 482–494. [Google Scholar] [CrossRef]
- Sato, Y.T.; Umezaki, K.; Sawada, S.; Mukai, S.A.; Sasaki, Y.; Harada, N.; Shiku, H.; Akiyoshi, K. Engineering hybrid exosomes by membrane fusion with liposomes. Sci. Rep. 2016, 6, 21933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nogueira, E.; Gomes, A.C.; Preto, A.; Cavaco-Paulo, A. Design of liposomal formulations for cell targeting. Colloids Surf. B Biointerfaces 2015, 136, 514–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tian, Y.; Li, S.; Song, J.; Ji, T.; Zhu, M.; Anderson, G.J.; Wei, J.; Nie, G. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 2014, 35, 2383–2390. [Google Scholar] [CrossRef] [PubMed]
- Tian, T.; Zhang, H.X.; He, C.P.; Fan, S.; Zhu, Y.L.; Qi, C.; Huang, N.P.; Xiao, Z.D.; Lu, Z.H.; Tannous, B.A.; et al. Surface functionalized exosomes as targeted drug delivery vehicles for cerebral ischemia therapy. Biomaterials 2018, 150, 137–149. [Google Scholar] [CrossRef]
- Cheruiyot, C.; Pataki, Z.; Ramratnam, B.; Li, M. Proteomic Analysis of Exosomes and Its Application in HIV-1 Infection. Proteom. Clin. Appl. 2018, 12, e1700142. [Google Scholar] [CrossRef]
- Sesé, M.; Fuentes, P.; Esteve-Codina, A.; Béjar, E.; McGrail, K.; Thomas, G.; Aasen, T.; Ramón, Y.C.S. Hypoxia-mediated translational activation of ITGB3 in breast cancer cells enhances TGF-β signaling and malignant features in vitro and in vivo. Oncotarget 2017, 8, 114856–114876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fuentes, P.; Sesé, M.; Guijarro, P.J.; Emperador, M.; Sánchez-Redondo, S.; Peinado, H.; Hümmer, S.; Ramón, Y.C.S. ITGB3-mediated uptake of small extracellular vesicles facilitates intercellular communication in breast cancer cells. Nat. Commun. 2020, 11, 4261. [Google Scholar] [CrossRef] [PubMed]
- Afratis, N.A.; Nikitovic, D.; Multhaupt, H.A.; Theocharis, A.D.; Couchman, J.R.; Karamanos, N.K. Syndecans-key regulators of cell signaling and biological functions. FEBS J. 2017, 284, 27–41. [Google Scholar] [CrossRef] [Green Version]
- Haubner, R.; Finsinger, D.; Kessler, H. Stereoisomeric Peptide Libraries and Peptidomimetics for Designing Selective Inhibitors of the αvβ3 Integrin for a New Cancer Therapy. Angew. Chem. Int. Ed. Engl. 1997, 36, 1374–1389. [Google Scholar] [CrossRef]
- Brooks, P.C.; Strömblad, S.; Klemke, R.; Visscher, D.; Sarkar, F.H.; Cheresh, D.A. Antiintegrin αvβ3 blocks human breast cancer growth and angiogenesis in human skin. J. Clin. Investig. 1995, 96, 1815–1822. [Google Scholar] [CrossRef] [Green Version]
- Goodman, S.L.; Hölzemann, G.; Sulyok, G.A.; Kessler, H. Nanomolar small molecule inhibitors for alphav(beta)6, alphav(beta)5, and alphav(beta)3 integrins. J. Med. Chem. 2002, 45, 1045–1051. [Google Scholar] [CrossRef]
- Sulyok, G.A.G.; Gibson, C.; Goodman, S.L.; Hölzemann, G.; Wiesner, M.; Kessler, H. Solid-Phase Synthesis of a Nonpeptide RGD Mimetic Library: New Selective αvβ3 Integrin Antagonists. J. Med. Chem. 2001, 44, 1938–1950. [Google Scholar] [CrossRef]
- Wu, H.; Beuerlein, G.; Nie, Y.; Smith, H.; Lee, B.A.; Hensler, M.; Huse, W.D.; Watkins, J.D. Stepwise in vitro affinity maturation of Vitaxin, an αvβ3-specific humanized mAb. Proc. Natl. Acad. Sci. USA 1998, 95, 6037–6042. [Google Scholar] [CrossRef] [Green Version]
- Ovadia, O.; Greenberg, S.; Chatterjee, J.; Laufer, B.; Opperer, F.; Kessler, H.; Gilon, C.; Hoffman, A. The effect of multiple N-methylation on intestinal permeability of cyclic hexapeptides. Mol. Pharm. 2011, 8, 479–487. [Google Scholar] [CrossRef]
- Kessler, H. Conformation and Biological Activity of Cyclic Peptides. Angew. Chem. Int. Ed. Engl. 1982, 21, 512–523. [Google Scholar] [CrossRef]
- Hruby, V.J. Conformational restrictions of biologically active peptides via amino acid side chain groups. Life Sci. 1982, 31, 189–199. [Google Scholar] [CrossRef]
- Gilon, C.; Halle, D.; Chorev, M.; Selinger, Z.; Byk, G. Backbone cyclization: A new method for conferring conformational constraint on peptides. Biopolymers 1991, 31, 745–750. [Google Scholar] [CrossRef]
- Chatterjee, J.; Ovadia, O.; Zahn, G.; Marinelli, L.; Hoffman, A.; Gilon, C.; Kessler, H. Multiple N-methylation by a designed approach enhances receptor selectivity. J. Med. Chem. 2007, 50, 5878–5881. [Google Scholar] [CrossRef]
- Chatterjee, J.; Gilon, C.; Hoffman, A.; Kessler, H. N-methylation of peptides: A new perspective in medicinal chemistry. Acc. Chem. Res. 2008, 41, 1331–1342. [Google Scholar] [CrossRef] [PubMed]
- Haviv, F.; Fitzpatrick, T.D.; Swenson, R.E.; Nichols, C.J.; Mort, N.A.; Bush, E.N.; Diaz, G.; Bammert, G.; Nguyen, A. Effect of N-methyl substitution of the peptide bonds in luteinizing hormone-releasing hormone agonists. J. Med. Chem. 1993, 36, 363–369. [Google Scholar] [CrossRef]
- Cody, W.L.; He, J.X.; Reily, M.D.; Haleen, S.J.; Walker, D.M.; Reyner, E.L.; Stewart, B.H.; Doherty, A.M. Design of a potent combined pseudopeptide endothelin-A/endothelin-B receptor antagonist, Ac-DBhg16-Leu-Asp-Ile-[NMe]Ile-Trp21 (PD 156252): Examination of its pharmacokinetic and spectral properties. J. Med. Chem. 1997, 40, 2228–2240. [Google Scholar] [CrossRef] [PubMed]
- Weinmüller, M.; Rechenmacher, F.; Kiran Marelli, U.; Reichart, F.; Kapp, T.G.; Räder, A.F.B.; Di Leva, F.S.; Marinelli, L.; Novellino, E.; Muñoz-Félix, J.M.; et al. Overcoming the Lack of Oral Availability of Cyclic Hexapeptides: Design of a Selective and Orally Available Ligand for the Integrin αvβ3. Angew. Chem. Int. Ed. Engl. 2017, 56, 16405–16409. [Google Scholar] [CrossRef]
- Armstrong, C.T.; Mason, P.E.; Anderson, J.L.R.; Dempsey, C.E. Arginine side chain interactions and the role of arginine as a gating charge carrier in voltage sensitive ion channels. Sci. Rep. UK 2016, 6, 21759. [Google Scholar] [CrossRef] [Green Version]
- Peterlin Masic, L. Arginine Mimetic Structures in Biologically Active Antagonists and Inhibitors. Curr. Med. Chem. 2006, 13, 3627–3648. [Google Scholar] [CrossRef]
- Duggan, M.E.; Duong, L.T.; Fisher, J.E.; Hamill, T.G.; Hoffman, W.F.; Huff, J.R.; Ihle, N.C.; Leu, C.T.; Nagy, R.M.; Perkins, J.J.; et al. Nonpeptide αvβ3 antagonists. 1. Transformation of a potent, integrin-selective αIIbβ3 antagonist into a potent αvβ3 antagonist. J. Med. Chem. 2000, 43, 3736–3745. [Google Scholar] [CrossRef]
- Ghosh, S.; Santulli, R.J.; Kinney, W.A.; DeCorte, B.L.; Liu, L.; Lewis, J.M.; Proost, J.C.; Leo, G.C.; Masucci, J.; Hageman, W.E.; et al. 1,2,3,4-Tetrahydroquinoline-containing αVβ3 integrin antagonists with enhanced oral bioavailability. Bioorganic Med. Chem. Lett. 2004, 14, 5937–5941. [Google Scholar] [CrossRef] [PubMed]
- Kapp, T.G.; Fottner, M.; Maltsev, O.V.; Kessler, H. Small Cause, Great Impact: Modification of the Guanidine Group in the RGD Motif Controls Integrin Subtype Selectivity. Angew. Chem. Int. Ed. 2016, 55, 1540–1543. [Google Scholar] [CrossRef]
- Bauer, W.; Briner, U.; Doepfner, W.; Haller, R.; Huguenin, R.; Marbach, P.; Petcher, T.J.; Pless, J. SMS 201-995: A very potent and selective octapeptide analogue of somatostatin with prolonged action. Life Sci. 1982, 31, 1133–1140. [Google Scholar] [CrossRef]
- Powell, M.F.; Stewart, T.; Jr Otvos, L.; Urge, L.; Gaeta, F.C.A.; Sette, A.; Arrhenius, T.; Thomson, D.; Soda, K.; Colon, S.M. Peptide Stability in Drug Development. II. Effect of Single Amino Acid Substitution and Glycosylation on Peptide Reactivity in Human Serum. Pharm. Res. 1993, 10, 1268–1273. [Google Scholar] [CrossRef] [PubMed]
- Werle, M.; Bernkop-Schnürch, A. Strategies to improve plasma half life time of peptide and protein drugs. Amino Acids 2006, 30, 351–367. [Google Scholar] [CrossRef] [PubMed]
- Bochen, A.; Marelli, U.K.; Otto, E.; Pallarola, D.; Mas-Moruno, C.; Di Leva, F.S.; Boehm, H.; Spatz, J.P.; Novellino, E.; Kessler, H.; et al. Biselectivity of isoDGR Peptides for Fibronectin Binding Integrin Subtypes α5β1 and αvβ6: Conformational Control through Flanking Amino Acids. J. Med. Chem. 2013, 56, 1509–1519. [Google Scholar] [CrossRef]
- Kapp, T.G.; Rechenmacher, F.; Neubauer, S.; Maltsev, O.V.; Cavalcanti-Adam, E.A.; Zarka, R.; Reuning, U.; Notni, J.; Wester, H.-J.; Mas-Moruno, C.; et al. A Comprehensive Evaluation of the Activity and Selectivity Profile of Ligands for RGD-binding Integrins. Sci. Rep. 2017, 7, 39805. [Google Scholar] [CrossRef] [Green Version]
- Murphy, M.G.; Cerchio, K.; Stoch, S.A.; Gottesdiener, K.; Wu, M.; Recker, R. Effect of L-000845704, an αvβ3 integrin antagonist, on markers of bone turnover and bone mineral density in postmenopausal osteoporotic women. J. Clin. Endocrinol. Metab. 2005, 90, 2022–2028. [Google Scholar] [CrossRef]
- Hutchinson, J.H.; Halczenko, W.; Brashear, K.M.; Breslin, M.J.; Coleman, P.J.; Duong, L.T.; Fernandez-Metzler, C.; Gentile, M.A.; Fisher, J.E.; Hartman, G.D.; et al. Nonpeptide αvβ3 antagonists. In vitro and in vivo evaluation of a potent alphavbeta3 antagonist for the prevention and treatment of osteoporosis. J. Med. Chem. 2003, 46, 4790–4798. [Google Scholar] [CrossRef]
- Raab-Westphal, S.; Marshall, J.F.; Goodman, S.L. Integrins as Therapeutic Targets: Successes and Cancers. Cancers 2017, 9, 110. [Google Scholar] [CrossRef]
- Alday-Parejo, B.; Stupp, R.; Ruegg, C. Are Integrins Still Practicable Targets for Anti-Cancer Therapy? Cancers 2019, 11, 978. [Google Scholar] [CrossRef] [Green Version]
- Sani, S.; Messe, M.; Fuchs, Q.; Pierrevelcin, M.; Laquerriere, P.; Entz-Werle, N.; Reita, D.; Etienne-Selloum, N.; Bruban, V.; Choulier, L.; et al. Biological relevance of RGD-integrin subtype-specific ligands in cancer. ChemBioChem 2020. [Google Scholar] [CrossRef] [PubMed]
- Reardon, D.A.; Fink, K.L.; Mikkelsen, T.; Cloughesy, T.F.; O’Neill, A.; Plotkin, S.; Glantz, M.; Ravin, P.; Raizer, J.J.; Rich, K.M.; et al. Randomized phase II study of cilengitide, an integrin-targeting arginine-glycine-aspartic acid peptide, in recurrent glioblastoma multiforme. J. Clin. Oncol. 2008, 26, 5610–5617. [Google Scholar] [CrossRef]
- Nabors, L.B.; Mikkelsen, T.; Hegi, M.E.; Ye, X.; Batchelor, T.; Lesser, G.; Peereboom, D.; Rosenfeld, M.R.; Olsen, J.; Brem, S.; et al. A safety run-in and randomized phase 2 study of cilengitide combined with chemoradiation for newly diagnosed glioblastoma (NABTT 0306). Cancer 2012, 118, 5601–5607. [Google Scholar] [CrossRef] [PubMed]
- Stupp, R.; Hegi, M.E.; Neyns, B.; Goldbrunner, R.; Schlegel, U.; Clement, P.M.; Grabenbauer, G.G.; Ochsenbein, A.F.; Simon, M.; Dietrich, P.Y.; et al. Phase I/IIa study of cilengitide and temozolomide with concomitant radiotherapy followed by cilengitide and temozolomide maintenance therapy in patients with newly diagnosed glioblastoma. J. Clin. Oncol. 2010, 28, 2712–2718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Legler, D.F.; Wiedle, G.; Ross, F.P.; Imhof, B.A. Superactivation of integrin alphavbeta3 by low antagonist concentrations. J. Cell Sci. 2001, 114, 1545–1553. [Google Scholar]
- Li, J.; Fukase, Y.; Shang, Y.; Zou, W.; Muñoz-Félix, J.M.; Buitrago, L.; van Agthoven, J.; Zhang, Y.; Hara, R.; Tanaka, Y.; et al. Novel Pure αVβ3 Integrin Antagonists That Do Not Induce Receptor Extension, Prime the Receptor, or Enhance Angiogenesis at Low Concentrations. ACS Pharmacol. Transl. Sci. 2019, 2, 387–401. [Google Scholar] [CrossRef] [PubMed]
- O’Donnell, P.H.; Undevia, S.D.; Stadler, W.M.; Karrison, T.M.; Nicholas, M.K.; Janisch, L.; Ratain, M.J. A phase I study of continuous infusion cilengitide in patients with solid tumors. Investig. New Drugs 2012, 30, 604–610. [Google Scholar] [CrossRef]
- Wong, P.P.; Bodrug, N.; Hodivala-Dilke, K.M. Exploring Novel Methods for Modulating Tumor Blood Vessels in Cancer Treatment. Curr. Biol. 2016, 26, R1161–R1166. [Google Scholar] [CrossRef]
- Dechantsreiter, M.A.; Planker, E.; Mathä, B.; Lohof, E.; Hölzemann, G.; Jonczyk, A.; Goodman, S.L.; Kessler, H. N-Methylated cyclic RGD peptides as highly active and selective αvβ3 integrin antagonists. J. Med. Chem. 1999, 42, 3033–3040. [Google Scholar] [CrossRef]
- Pickarski, M.; Gleason, A.; Bednar, B.; Duong, L.T. Orally active αvβ3 integrin inhibitor MK-0429 reduces melanoma metastasis. Oncol. Rep. 2015, 33, 2737–2745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, X.; Zhang, J.; Haimbach, R.; Zhu, W.; Mayer-Ezell, R.; Garcia-Calvo, M.; Smith, E.; Price, O.; Kan, Y.; Zycband, E.; et al. An integrin antagonist (MK-0429) decreases proteinuria and renal fibrosis in the ZSF1 rat diabetic nephropathy model. Pharmacol. Res. Perspect. 2017, 5, e00354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Agthoven, J.F.; Xiong, J.P.; Alonso, J.L.; Rui, X.; Adair, B.D.; Goodman, S.L.; Arnaout, M.A. Structural basis for pure antagonism of integrin αVβ3 by a high-affinity form of fibronectin. Nat. Struct. Mol. Biol. 2014, 21, 383–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kessler, H.; Gratias, R.; Hessler, G.; Gurrath, M.; Müller, G. Conformation of cyclic peptides. Principle concepts and the design of selectivity and superactivity in bioactive sequences by ‘spatial screening’. Pure Appl. Chem. 1996, 68, 1201–1205. [Google Scholar] [CrossRef]
- Räder, A.F.B.; Weinmüller, M.; Reichart, F.; Schumacher-Klinger, A.; Merzbach, S.; Gilon, C.; Hoffman, A.; Kessler, H. Orally Active Peptides: Is There a Magic Bullet? Angew. Chem. Int. Ed. 2018, 57, 14414–14438. [Google Scholar] [CrossRef]
- Harris, I.S.; Treloar, A.E.; Inoue, S.; Sasaki, M.; Gorrini, C.; Lee, K.C.; Yung, K.Y.; Brenner, D.; Knobbe-Thomsen, C.B.; Cox, M.A.; et al. Glutathione and thioredoxin antioxidant pathways synergize to drive cancer initiation and progression. Cancer Cell 2015, 27, 211–222. [Google Scholar] [CrossRef] [Green Version]
- Sugahara, K.N.; Teesalu, T.; Karmali, P.P.; Kotamraju, V.R.; Agemy, L.; Girard, O.M.; Hanahan, D.; Mattrey, R.F.; Ruoslahti, E. Tissue-penetrating delivery of compounds and nanoparticles into tumors. Cancer Cell 2009, 16, 510–520. [Google Scholar] [CrossRef] [Green Version]
- Kang, S.; Lee, S.; Park, S. iRGD Peptide as a Tumor-Penetrating Enhancer for Tumor-Targeted Drug Delivery. Polymers 2020, 12, 1906. [Google Scholar] [CrossRef]
- Sugahara, K.N.; Teesalu, T.; Karmali, P.P.; Kotamraju, V.R.; Agemy, L.; Greenwald, D.R.; Ruoslahti, E. Coadministration of a tumor-penetrating peptide enhances the efficacy of cancer drugs. Science 2010, 328, 1031–1035. [Google Scholar] [CrossRef] [Green Version]
- Liu, X.; Jiang, J.; Ji, Y.; Lu, J.; Chan, R.; Meng, H. Targeted drug delivery using iRGD peptide for solid cancer treatment. Mol. Syst. Des. Eng. 2017, 2, 370–379. [Google Scholar] [CrossRef]
- Liu, X.; Lin, P.; Perrett, I.; Lin, J.; Liao, Y.P.; Chang, C.H.; Jiang, J.; Wu, N.; Donahue, T.; Wainberg, Z.; et al. Tumor-penetrating peptide enhances transcytosis of silicasome-based chemotherapy for pancreatic cancer. J. Clin. Investig. 2017, 127, 2007–2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marinelli, L.; Gottschalk, K.-E.; Meyer, A.; Novellino, E.; Kessler, H. Human Integrin αvβ5: Homology Modeling and Ligand Binding. J. Med. Chem. 2004, 47, 4166–4177. [Google Scholar] [CrossRef] [PubMed]
- Lippa, R.A.; Barrett, J.; Pal, S.; Rowedder, J.E.; Murphy, J.A.; Barrett, T.N. Discovery of the first potent and selective αvβ5 integrin inhibitor based on an amide-containing core. Eur. J. Med. Chem. 2020, 208, 112719. [Google Scholar] [CrossRef] [PubMed]
- Rechenmacher, F.; Neubauer, S.; Polleux, J.; Mas-Moruno, C.; De Simone, M.; Cavalcanti-Adam, E.A.; Spatz, J.P.; Fässler, R.; Kessler, H. Functionalizing αvβ3- or α5β1-selective integrin antagonists for surface coating: A method to discriminate integrin subtypes in vitro. Angew. Chem. Int. Ed. Engl. 2013, 52, 1572–1575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Höltke, C. isoDGR-Peptides for Integrin Targeting: Is the Time Up for RGD? J. Med. Chem. 2018, 61, 7471–7473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Curnis, F.; Longhi, R.; Crippa, L.; Cattaneo, A.; Dondossola, E.; Bachi, A.; Corti, A. Spontaneous formation of L-isoaspartate and gain of function in fibronectin. J. Biol. Chem. 2006, 281, 36466–36476. [Google Scholar] [CrossRef] [Green Version]
- Takahashi, S.; Leiss, M.; Moser, M.; Ohashi, T.; Kitao, T.; Heckmann, D.; Pfeifer, A.; Kessler, H.; Takagi, J.; Erickson, H.P.; et al. The RGD motif in fibronectin is essential for development but dispensable for fibril assembly. J. Cell Biol. 2007, 178, 167–178. [Google Scholar] [CrossRef] [Green Version]
- Ghitti, M.; Spitaleri, A.; Valentinis, B.; Mari, S.; Asperti, C.; Traversari, C.; Rizzardi, G.P.; Musco, G. Molecular Dynamics Reveal that isoDGR-Containing Cyclopeptides Are True αvβ3 Antagonists Unable To Promote Integrin Allostery and Activation. Angew. Chem. Int. Ed. 2012, 51, 7702–7705. [Google Scholar] [CrossRef]
- Curnis, F.; Sacchi, A.; Longhi, R.; Colombo, B.; Gasparri, A.; Corti, A. IsoDGR-tagged albumin: A new αvβ3 selective carrier for nanodrug delivery to tumors. Small 2013, 9, 673–678. [Google Scholar] [CrossRef] [PubMed]
- Gao, H.; Luo, C.; Yang, G.; Du, S.; Li, X.; Zhao, H.; Shi, J.; Wang, F. Improved in Vivo Targeting Capability and Pharmacokinetics of 99mTc-Labeled isoDGR by Dimerization and Albumin-Binding for Glioma Imaging. Bioconjugate Chem. 2019, 30, 2038–2048. [Google Scholar] [CrossRef]
- Zhao, H.; Gao, H.; Zhai, L.; Liu, X.; Jia, B.; Shi, J.; Wang, F. 99mTc-HisoDGR as a Potential SPECT Probe for Orthotopic Glioma Detection via Targeting of Integrin α5β1. Bioconjugate Chem. 2016, 27, 1259–1266. [Google Scholar] [CrossRef]
- Altmann, A.; Sauter, M.; Roesch, S.; Mier, W.; Warta, R.; Debus, J.; Dyckhoff, G.; Herold-Mende, C.; Haberkorn, U. Identification of a Novel ITGαvβ6-Binding Peptide Using Protein Separation and Phage Display. Clin. Cancer Res. 2017, 23, 4170. [Google Scholar] [CrossRef] [Green Version]
- Roesch, S.; Lindner, T.; Sauter, M.; Loktev, A.; Flechsig, P.; Müller, M.; Mier, W.; Warta, R.; Dyckhoff, G.; Herold-Mende, C.; et al. Comparison of the RGD Motif-Containing αvβ6 Integrin-Binding Peptides SFLAP3 and SFITGv6 for Diagnostic Application in HNSCC. J. Nucl. Med. 2018, 59, 1679–1685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hausner, S.H.; DiCara, D.; Marik, J.; Marshall, J.F.; Sutcliffe, J.L. Use of a peptide derived from foot-and-mouth disease virus for the Noninvasive Imaging of human cancer: Generation and evaluation of 4-[F-18]fluorobenzoyl A20FMDV2 for in vivo imaging of integrin alpha(v)beta(6) expression with positron emission tomography. Cancer Res. 2007, 67, 7833–7840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jackson, T.; Sheppard, D.; Denyer, M.; Blakemore, W.; King, A.M. The epithelial integrin alphavbeta6 is a receptor for foot-and-mouth disease virus. J. Virol. 2000, 74, 4949–4956. [Google Scholar] [CrossRef] [PubMed]
- Burman, A.; Clark, S.; Abrescia, N.G.; Fry, E.E.; Stuart, D.I.; Jackson, T. Specificity of the VP1 GH loop of Foot-and-Mouth Disease virus for alphav integrins. J. Virol. 2006, 80, 9798–9810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maltsev, O.V.; Marelli, U.K.; Kapp, T.G.; Di Leva, F.S.; Di Maro, S.; Nieberler, M.; Reuning, U.; Schwaiger, M.; Novellino, E.; Marinelli, L.; et al. Stable Peptides Instead of Stapled Peptides: Highly Potent αvβ6-Selective Integrin Ligands. Angew. Chem. Int. Ed. 2016, 55, 1535–1539. [Google Scholar] [CrossRef] [PubMed]
- Di’Leva, F.S.; Tomassi, S.; Di Maro, S.; Reichart, F.; Notni, J.; Dangi, A.; Marelli, U.K.; Brancaccio, D.; Merlino, F.; Wester, H.-J.; et al. From a Helix to a Small Cycle: Metadynamics-Inspired αvβ6 Integrin Selective Ligands. Angew. Chem. Int. Ed. 2018, 57, 14645–14649. [Google Scholar] [CrossRef] [PubMed]
- Reichart, F.; Maltsev, O.V.; Kapp, T.G.; Räder, A.F.B.; Weinmüller, M.; Marelli, U.K.; Notni, J.; Wurzer, A.; Beck, R.; Wester, H.J.; et al. Selective Targeting of Integrin αvβ8 by a Highly Active Cyclic Peptide. J. Med. Chem. 2019, 62, 2024–2037. [Google Scholar] [CrossRef]
- Nadrah, K.; Dolenc, M.S. Dual Antagonists of Integrins. Curr. Med. Chem. 2005, 12, 1449–1466. [Google Scholar] [CrossRef]
- Sheldrake, H.M.; Patterson, L.H. Strategies To Inhibit Tumor Associated Integrin Receptors: Rationale for Dual and Multi-Antagonists. J. Med. Chem. 2014, 57, 6301–6315. [Google Scholar] [CrossRef] [PubMed]
- Henderson, N.C.; Arnold, T.D.; Katamura, Y.; Giacomini, M.M.; Rodriguez, J.D.; McCarty, J.H.; Pellicoro, A.; Raschperger, E.; Betsholtz, C.; Ruminski, P.G.; et al. Targeting of αv integrin identifies a core molecular pathway that regulates fibrosis in several organs. Nat. Med. 2013, 19, 1617–1624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eo, J.S.; Jeong, J.M. Angiogenesis Imaging Using Ga-68-RGD PET/CT: Therapeutic Implications. Semin Nucl. Med. 2016, 46, 419–427. [Google Scholar] [CrossRef]
- Jia, B.; Shi, J.Y.; Yang, Z.; Xu, B.; Liu, Z.F.; Zhao, H.Y.; Liu, S.; Wang, F. Tc-99m-labeled cyclic RGDfK dimer: Initial evaluation for SPECT imaging of glioma integrin alpha(v)beta(3) expression. Bioconjugate Chem. 2006, 17, 1069–1076. [Google Scholar] [CrossRef]
- Li, Z.B.; Cai, W.B.; Cao, Q.Z.; Chen, K.; Wu, Z.H.; He, L.N.; Chen, X.Y. 64Cu-Labeled “Tetrameric and octameric RGD peptides for small-animal PET of Tumor alpha(v)beta(3) integrin expression. J. Nucl. Med. 2007, 48, 1162–1171. [Google Scholar] [CrossRef] [PubMed]
- Haubner, R.; Weber, W.A.; Beer, A.J.; Vabuliene, E.; Reim, D.; Sarbia, M.; Becker, K.F.; Goebel, M.; Hein, R.; Wester, H.J.; et al. Noninvasive visualization of the activated alpha v beta 3 integrin in cancer patients by positron emission tomography and [F-18]Galacto-RGD. PLoS Med. 2005, 2, 244–252. [Google Scholar] [CrossRef] [Green Version]
- Atkinson, S.J.; Ellison, T.S.; Steri, V.; Gould, E.; Robinson, S.D. Redefining the role(s) of endothelial αvβ3-integrin in angiogenesis. Biochem. Soc. Trans. 2014, 42, 1590–1595. [Google Scholar] [CrossRef]
- Beer, A.J.; Haubner, R.; Sarbia, M.; Goebel, M.; Luderschmidt, S.; Grosu, A.L.; Schnell, O.; Niemeyer, M.; Kessler, H.; Wester, H.J.; et al. Positron emission tomography using [F-18]Galacto-RGD identifies the level of integrin alpha(v)beta(3) expression in man. Clin. Cancer Res. 2006, 12, 3942–3949. [Google Scholar] [CrossRef] [Green Version]
- Chen, H.; Niu, G.; Wu, H.; Chen, X. Clinical Application of Radiolabeled RGD Peptides for PET Imaging of Integrin αvβ3. Theranostics 2016, 6, 78–92. [Google Scholar] [CrossRef] [Green Version]
- Haubner, R.; Maschauer, S.; Prante, O. PET Radiopharmaceuticals for Imaging Integrin Expression: Tracers in Clinical Studies and Recent Developments. Biomed. Res. Int. 2014, 2014. [Google Scholar] [CrossRef] [Green Version]
- Debordeaux, F.; Chansel-Debordeaux, L.; Pinaquy, J.B.; Fernandez, P.; Schulz, J. What about alpha(v)beta(3) integrins in molecular imaging in oncology? Nucl. Med. Biol. 2018, 62–63, 31–46. [Google Scholar] [CrossRef] [PubMed]
- Dijkgraaf, I.; Kruijtzer, J.A.; Liu, S.; Soede, A.C.; Oyen, W.J.; Corstens, F.H.; Liskamp, R.M.; Boerman, O.C. Improved targeting of the αvβ3 integrin by multimerisation of RGD peptides. Eur. J. Nucl. Med. Mol. Imaging 2007, 34, 267–273. [Google Scholar] [CrossRef] [PubMed]
- Liu, S. Radiolabeled multimeric cyclic RGD peptides as integrin αvβ3-targeted radiotracers for tumor imaging. Mol. Pharm. 2006, 3, 472–487. [Google Scholar] [CrossRef] [PubMed]
- Maschauer, S.; Einsiedel, J.; Reich, D.; Hubner, H.; Gmeiner, P.; Wester, H.J.; Prante, O.; Notni, J. Theranostic Value of Multimers: Lessons Learned from Trimerization of Neurotensin Receptor Ligands and Other Targeting Vectors. Pharm. Base 2017, 10, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Janssen, M.; Oyen, W.J.G.; Massuger, L.F.A.G.; Frielink, C.; Dijkgraaf, I.; Edwards, D.S.; Radjopadhye, M.; Corstens, F.H.M.; Boerman, O.C. Comparison of a monomeric and dimeric radiolabeled RGD-peptide for tumor targeting. Cancer Biother. Radiopharm. 2002, 17, 641–646. [Google Scholar] [CrossRef]
- Liu, Z.F.; Niu, G.; Shi, J.Y.; Liu, S.L.; Wang, F.; Liu, S.; Chen, X.Y. Ga-68-labeled cyclic RGD dimers with Gly(3) and PEG(4) linkers: Promising agents for tumor integrin alpha(v)beta(3) PET imaging. Eur. J. Nucl. Med. Mol. Imaging 2009, 36, 947–957. [Google Scholar] [CrossRef]
- Gao, S.; Wu, H.H.; Li, W.W.; Zhao, S.Q.; Teng, X.P.; Lu, H.; Hu, X.D.; Wang, S.Z.; Yu, J.M.; Yuan, S.H. A pilot study imaging integrin αvβ3 with RGD PET/CT in suspected lung cancer patients. Eur. J. Nucl. Med. Mol. Imaging 2015, 42, 2029–2037. [Google Scholar] [CrossRef]
- Mi, B.M.; Yu, C.J.; Pan, D.H.; Yang, M.; Wan, W.X.; Niu, G.; Chen, X.Y. Pilot Prospective Evaluation of F-18-Alfatide II for Detection of Skeletal Metastases. Theranostics 2015, 5, 1115–1121. [Google Scholar] [CrossRef] [Green Version]
- Mittra, E.S.; Goris, M.L.; Iagaru, A.H.; Kardan, A.; Burton, L.; Berganos, R.; Chang, E.; Liu, S.L.; Shen, B.; Chin, F.T.; et al. Pilot Pharmacokinetic and Dosimetric Studies of F-18-FPPRGD2: A PET Radiopharmaceutical Agent for Imaging alpha(v)beta(3) Integrin Levels. Radiology 2011, 260, 182–191. [Google Scholar] [CrossRef] [Green Version]
- Dong, Y.J.; Wei, Y.C.; Chen, G.X.; Huang, Y.; Song, P.P.; Liu, S.G.; Zheng, J.S.; Cheng, M.; Yuan, S.H. Relationship Between Clinicopathological Characteristics and PET/CT Uptake in Esophageal Squamous Cell Carcinoma: [F-18]Alfatide versus [F-18]FDG. Mol. Imaging Biol. 2019, 21, 175–182. [Google Scholar] [CrossRef]
- Wu, J.; Wang, S.H.; Zhang, X.Z.; Teng, Z.G.; Wang, J.J.; Yung, B.C.; Niu, G.; Zhu, H.; Lu, G.M.; Chen, X.Y. F-18-Alfatide II PET/CT for Identification of Breast Cancer: A Preliminary Clinical Study. J. Nucl. Med. 2018, 59, 1809–1816. [Google Scholar] [CrossRef] [Green Version]
- Li, L.; Zhao, W.; Sun, X.R.; Liu, N.; Zhou, Y.; Luan, X.H.; Gao, S.; Zhao, S.Q.; Yu, J.M.; Yuan, S.H. F-18-RGD PET/CT imaging reveals characteristics of angiogenesis in non-small cell lung cancer. Transl. Lung Cancer Res. 2020, 9, 1324–1332. [Google Scholar] [CrossRef] [PubMed]
- Kang, F.; Wang, Z.; Li, G.Q.; Wang, S.J.; Liu, D.L.; Zhang, M.R.; Zhao, M.X.; Yang, W.D.; Wang, J. Inter-heterogeneity and intra-heterogeneity of alpha(v)beta(3) in non-small cell lung cancer and small cell lung cancer patients as revealed by Ga-68-RGD2 PET imaging. Eur. J. Nucl. Med. Mol. Imaging 2017, 44, 1520–1528. [Google Scholar] [CrossRef] [PubMed]
- Kang, F.; Wang, S.J.; Tian, F.; Zhao, M.X.; Zhang, M.R.; Wang, Z.; Li, G.Q.; Liu, C.L.; Yang, W.D.; Li, X.F.; et al. Comparing the Diagnostic Potential of Ga-68-Alfatide II and F-18-FDG in Differentiating Between Non Small Cell Lung Cancer and Tuberculosis. J. Nucl. Med. 2016, 57, 672–677. [Google Scholar] [CrossRef] [Green Version]
- Wei, Y.C.; Qin, X.T.; Luan, X.H.; Zhou, Y.; Yuan, S.H.; Yu, J.M. F-18-alfatide PET/CT may predict the survival of patients with stage II-IV lung cancer. J. Nucl. Med. 2020, 61, 1334. [Google Scholar]
- Zhou, Y.; Gao, S.; Huang, Y.; Zheng, J.S.; Dong, Y.J.; Zhang, B.J.; Zhao, S.Q.; Lu, H.; Liu, Z.B.; Yu, J.M.; et al. A Pilot Study of F-18-Alfatide PET/CT Imaging for Detecting Lymph Node Metastases in Patients with Non-Small Cell Lung Cancer. Sci. Rep. UK 2017, 7. [Google Scholar] [CrossRef] [Green Version]
- Yu, C.J.; Pan, D.H.; Mi, B.M.; Xu, Y.P.; Lang, L.X.; Niu, G.; Yang, M.; Wan, W.X.; Chen, X.Y. F-18-Alfatide II PET/CT in healthy human volunteers and patients with brain metastases. Eur. J. Nucl. Med. Mol. Imaging 2015, 42, 2021–2028. [Google Scholar] [CrossRef] [Green Version]
- Parihar, A.S.; Mittal, B.R.; Kumar, R.; Shukla, J.; Bhattacharya, A. Ga-68-DOTA-RGD(2) Positron Emission Tomography/Computed Tomography in Radioiodine Refractory Thyroid Cancer: Prospective Comparison of Diagnostic Accuracy with F-18-FDG Positron Emission Tomography/Computed Tomography and Evaluation Toward Potential Theranostics. Thyroid 2020, 30, 557–567. [Google Scholar] [CrossRef]
- Liu, Z.F.; Liu, S.L.; Wang, F.; Liu, S.; Chen, X.Y. Noninvasive imaging of tumor integrin expression using F-18-labeled RGD dimer peptide with PEG(4) linkers. Eur. J. Nucl. Med. Mol. Imaging 2009, 36, 1296–1307. [Google Scholar] [CrossRef] [PubMed]
- Notni, J.; Pohle, K.; Wester, H.J. Be spoilt for choice with radiolabelled RGD peptides: Preclinical evaluation of Ga-68-TRAP(RGD)(3). Nucl. Med. Biol. 2013, 40, 33–41. [Google Scholar] [CrossRef]
- Zhai, C.Y.; Franssen, G.M.; Petrik, M.; Laverman, P.; Summer, D.; Rangger, C.; Haubner, R.; Haas, H.; Decristoforo, C. Comparison of Ga-68-Labeled Fusarinine C-Based Multivalent RGD Conjugates and [Ga-68]NODAGA-RGD-In Vivo Imaging Studies in Human Xenograft Tumors. Mol. Imaging Biol. 2016, 18, 758–767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Imberti, C.; Terry, S.Y.A.; Cullinane, C.; Clarke, F.; Cornish, G.H.; Ramakrishnan, N.K.; Roselt, P.; Cope, A.P.; Hicks, R.J.; Blower, P.J.; et al. Enhancing PET Signal at Target Tissue in Vivo: Dendritic and Multimeric Tris(hydroxypyridinone) Conjugates for Molecular Imaging of alpha(v)beta(3) Integrin Expression with Gallium-68. Bioconjugate Chem. 2017, 28, 481–495. [Google Scholar] [CrossRef] [PubMed]
- Lobeek, D.; Franssen, G.M.; Ma, M.T.; Wester, H.J.; Decristoforo, C.; Oyen, W.J.G.; Boerman, O.C.; Terry, S.Y.A.; Rijpkema, M. In Vivo Characterization of 4 Ga-68-Labeled Multimeric RGD Peptides to Image alpha(v)beta(3) Integrin Expression in 2 Human Tumor Xenograft Mouse Models. J. Nucl. Med. 2018, 59, 1296–1301. [Google Scholar] [CrossRef] [Green Version]
- Yan, Y.J.; Chen, X.Y. Peptide heterodimers for molecular imaging. Amino Acids 2011, 41, 1081–1092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Judmann, B.; Braun, D.; Wangler, B.; Schirrmacher, R.; Fricker, G.; Wangler, C. Current State of Radiolabeled Heterobivalent Peptidic Ligands in Tumor Imaging and Therapy. Pharm. Base 2020, 13, 173. [Google Scholar] [CrossRef] [PubMed]
- Wu, H.; Chen, H.J.; Pan, D.F.; Ma, Y.F.; Liang, S.; Wan, Y.; Fang, Y. Imaging Integrin αvβ3 and NRP-1 Positive Gliomas with a Novel Fluorine-18 Labeled RGD-ATWLPPR Heterodimeric Peptide Probe. Mol. Imaging Biol. 2014, 16, 781–792. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.J.; Mao, F.; Niu, G.; Peng, L.; Lang, L.X.; Li, F.; Ying, H.Y.; Wu, H.W.; Zhu, Z.H.; Chen, X.Y. Dual Gastrin-Releasing Peptide Receptor and Integrin alpha(v)beta(3) targeting PET/CT using Ga-68-BBN-RGD in Patients with Breast Cancer. J. Nucl. Med. 2018, 59, 55. [Google Scholar]
- Zhang, J.; Mao, F.; Niu, G.; Peng, L.; Lang, L.; Li, F.; Ying, H.; Wu, H.; Pan, B.; Zhu, Z.; et al. 68Ga-BBN-RGD PET/CT for GRPR and Integrin αvβ3 Imaging in Patients with Breast Cancer. Theranostics 2018, 8, 1121–1130. [Google Scholar] [CrossRef]
- Zheng, Y.; Wang, H.; Tan, H.; Cui, X.; Yao, S.; Zang, J.; Zhang, L.; Zhu, Z. Evaluation of Lung Cancer and Neuroendocrine Neoplasm in a Single Scan by Targeting Both Somatostatin Receptor and Integrin αvβ3. Clin. Nucl. Med. 2019, 44, 687–694. [Google Scholar] [CrossRef]
- Shallal, H.M.; Minn, I.; Banerjee, S.R.; Lisok, A.; Mease, R.C.; Pomper, M.G. Heterobivalent Agents Targeting PSMA and Integrin-αvβ3. Bioconjugate Chem. 2014, 25, 393–405. [Google Scholar] [CrossRef] [PubMed]
- Reubi, J.C.; Maecke, H.R. Approaches to Multireceptor Targeting: Hybrid Radioligands, Radioligand Cocktails, and Sequential Radioligand Applications. J. Nucl. Med. 2017, 58, 10s–16s. [Google Scholar] [CrossRef] [Green Version]
- Lee, M.S.; Park, H.S.; Lee, B.C.; Jung, J.H.; Yoo, J.S.; Kim, S.E. Identification of Angiogenesis Rich-Viable Myocardium using RGD Dimer based SPECT after Myocardial Infarction. Sci. Rep. UK 2016, 6. [Google Scholar] [CrossRef] [PubMed]
- Grönman, M.; Tarkia, M.; Kiviniemi, T.; Halonen, P.; Kuivanen, A.; Savunen, T.; Tolvanen, T.; Teuho, J.; Käkelä, M.; Metsälä, O.; et al. Imaging of αvβ3 integrin expression in experimental myocardial ischemia with [68Ga]NODAGA-RGD positron emission tomography. J. Transl. Med. 2017, 15. [Google Scholar] [CrossRef] [PubMed]
- Makowski, M.R.; Rischpler, C.; Ebersberger, U.; Keithahn, A.; Kasel, M.; Hoffmann, E.; Rassaf, T.; Kessler, H.; Wester, H.-J.; Nekolla, S.G.; et al. Multiparametric PET and MRI of myocardial damage after myocardial infarction: Correlation of integrin αvβ3 expression and myocardial blood flow. Eur. J. Nucl. Med. Mol. Imaging 2020. [Google Scholar] [CrossRef] [PubMed]
- Higuchi, T.; Bengel, F.M.; Seidl, S.; Watzlowik, P.; Kessler, H.; Hegenloh, R.; Reder, S.; Nekolla, S.G.; Wester, H.J.; Schwaiger, M. Assessment of alpha(v)beta(3) integrin expression after myocardial infarction by positron emission tomography. Cardiovasc. Res. 2008, 78, 395–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hendrikx, G.; Vöö, S.; Bauwens, M.; Post, M.J.; Mottaghy, F.M. SPECT and PET imaging of angiogenesis and arteriogenesis in pre-clinical models of myocardial ischemia and peripheral vascular disease. Eur. J. Nucl. Med. Mol. Imaging 2016, 43, 2433–2447. [Google Scholar] [CrossRef] [Green Version]
- Liu, Z.F.; Liu, H.; Ma, T.; Sun, X.L.; Shi, J.Y.; Jia, B.; Sun, Y.; Zhan, J.; Zhang, H.Q.; Zhu, Z.H.; et al. Integrin αvβ6-Targeted SPECT Imaging for Pancreatic Cancer Detection. J. Nucl. Med. 2014, 55, 989–994. [Google Scholar] [CrossRef] [Green Version]
- Hackel, B.J.; Kimura, R.H.; Miao, Z.; Liu, H.G.; Sathirachinda, A.; Cheng, Z.; Chin, F.T.; Gambhir, S.S. F-18-Fluorobenzoate-Labeled Cystine Knot Peptides for PET Imaging of Integrin alpha(v)beta(6). J. Nucl. Med. 2013, 54, 1101–1105. [Google Scholar] [CrossRef] [Green Version]
- Kimura, R.H.; Cheng, Z.; Gambhir, S.S.; Cochran, J.R. Engineered Knottin Peptides: A New Class of Agents for Imaging Integrin Expression in Living Subjects. Cancer Res. 2009, 69, 2435–2442. [Google Scholar] [CrossRef] [Green Version]
- Kimura, R.H.; Teed, R.; Hackel, B.J.; Pysz, M.A.; Chuang, C.Z.; Sathirachinda, A.; Willmann, J.K.; Gambhir, S.S. Pharmacokinetically Stabilized Cystine Knot Peptides That Bind Alpha-v-Beta-6 Integrin with Single-Digit Nanomolar Affinities for Detection of Pancreatic Cancer. Clin. Cancer Res. 2012, 18, 839–849. [Google Scholar] [CrossRef] [Green Version]
- Notni, J.; Steiger, K.; Mendler, C.; Reich, D.; Maltsev, O.V.; Kapp, T.G.; Hoffmann, F.; Weichert, W.; Kessler, H.; Schwaiger, M.; et al. Preclinical Characterization of Ga-68-Avebehexin, a Specific Probe for the “Cancer Integrin” αvβ6. Eur. J. Nucl. Med. Mol. Imaging 2016, 43, S107. [Google Scholar]
- Notni, J.; Reich, D.; Maltsev, O.V.; Kapp, T.G.; Steiger, K.; Hoffmann, F.; Esposito, I.; Weichert, W.; Kessler, H.; Wester, H.J. In Vivo PET Imaging of the Cancer Integrin αvβ6 Using (68)Ga-Labeled Cyclic RGD Nonapeptides. J. Nucl. Med. 2017, 58, 671–677. [Google Scholar] [CrossRef] [Green Version]
- Färber, S.F.; Wurzer, A.; Reichart, F.; Beck, R.; Kessler, H.; Wester, H.-J.; Notni, J. Therapeutic Radiopharmaceuticals Targeting Integrin αvβ6. ACS Omega 2018, 3, 2428–2436. [Google Scholar] [CrossRef] [Green Version]
- Flechsig, P.; Lindner, T.; Loktev, A.; Roesch, S.; Mier, W.; Sauter, M.; Meister, M.; Herold-Mende, C.; Haberkorn, U.; Altmann, A. PET/CT Imaging of NSCLC with a alpha(v)beta(6) Integrin-Targeting Peptide. Mol. Imaging Biol. 2019, 21, 973–983. [Google Scholar] [CrossRef] [PubMed]
- Kimura, R.H.; Wang, L.; Shen, B.; Huo, L.; Tummers, W.; Filipp, F.V.; Guo, H.W.H.; Haywood, T.; Abou-Elkacem, L.; Baratto, L.; et al. Evaluation of integrin alpha v beta(6) cystine knot PET tracers to detect cancer and idiopathic pulmonary fibrosis. Nat. Commun. 2019, 10. [Google Scholar] [CrossRef] [Green Version]
- Hausner, S.H.; Bold, R.J.; Cheuy, L.Y.; Chew, H.K.; Daly, M.E.; Davis, R.A.; Foster, C.C.; Kim, E.J.; Sutcliffe, J.L. Preclinical Development and First-in-Human Imaging of the Integrin alphavbeta6 with [18F]alphavbeta6-Binding Peptide in Metastatic Carcinoma. Clin. Cancer Res. 2019, 25, 1206–1215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hausner, S.H.; Bauer, N.; Davis, R.A.; Ganguly, T.; Tang, S.Y.C.; Sutcliffe, J.L. The Effects of an Albumin Binding Moiety on the Targeting and Pharmacokinetics of an Integrin αvβ6-Selective Peptide Labeled with Aluminum [18F]Fluoride. Mol. Imaging Biol. 2020, 22, 1543–1552. [Google Scholar] [CrossRef]
- Quigley, N.G.; Tomassi, S.; Di Leva, F.S.; Di Maro, S.; Richter, F.; Steiger, K.; Kossatz, S.; Marinelli, L.; Notni, J. Click-Chemistry (CuAAC) Trimerization of an alpha(v)beta(6) Integrin Targeting Ga-68-Peptide: Enhanced Contrast for in-Vivo PET Imaging of Human Lung Adenocarcinoma Xenografts. ChemBiochem. 2020, 21, 2836–2843. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Bell, K.; Mousa, S.A.; Varner, J.A. Regulation of angiogenesis in vivo by ligation of integrin alpha 5 beta 1 with the central cell-binding domain of fibronectin. Am. J. Pathol. 2000, 156, 1345–1362. [Google Scholar] [CrossRef]
- D’Alessandria, C.; Pohle, K.; Rechenmacher, F.; Neubauer, S.; Notni, J.; Wester, H.-J.; Schwaiger, M.; Kessler, H.; Beer, A.J. In vivo biokinetic and metabolic characterization of the 68Ga-labelled α5β1-selective peptidomimetic FR366. Eur. J. Nucl. Med. Mol. Imaging 2015, 43, 953–963. [Google Scholar] [CrossRef]
- Kapp, T.G.; Di Leva, F.S.; Notni, J.; Rader, A.F.B.; Fottner, M.; Reichart, F.; Reich, D.; Wurzer, A.; Steiger, K.; Novellino, E.; et al. N-Methylation of isoDGR Peptides: Discovery of a Selective alpha 5 beta 1-Integrin Ligand as a Potent Tumor Imaging Agent. J. Med. Chem. 2018, 61, 2490–2499. [Google Scholar] [CrossRef] [PubMed]
- Notni, J.; Steiger, K.; Hoffmann, F.; Reich, D.; Kapp, T.G.; Rechenmacher, F.; Neubauer, S.; Kessler, H.; Wester, H.J. Complementary, Selective PET Imaging of Integrin Subtypes α5β1 and αvβ3 Using 68Ga-Aquibeprin and 68Ga-Avebetrin. J. Nucl. Med. 2015, 57, 460–466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Notni, J.; Gassert, F.T.; Steiger, K.; Sommer, P.; Weichert, W.; Rummeny, E.J.; Schwaiger, M.; Kessler, H.; Meier, R.; Kimm, M.A. In vivo imaging of early stages of rheumatoid arthritis by alpha 5 beta 1-integrin-targeted positron emission tomography (vol 9, 87, 2019). Ejnmmi Res. 2019, 9. [Google Scholar] [CrossRef]
- Hayashido, Y.; Kitano, H.; Sakaue, T.; Fujii, T.; Suematsu, M.; Sakurai, S.; Okamoto, T. Overexpression of integrin αv facilitates proliferation and invasion of oral squamous cell carcinoma cells via mek/erk signaling pathway that is activated by interaction of integrin αvβ8 with type I collagen. Int. J. Oncol. 2014, 45, 1875–1882. [Google Scholar] [CrossRef] [Green Version]
- Quigley, N.G.; Steiger, K.; Richter, F.; Weichert, W.; Hoberuck, S.; Kotzerke, J.; Notni, J. Tracking a TGF-beta activator in vivo: Sensitive PET imaging of alpha v beta 8-integrin with the Ga-68-labeled cyclic RGD octapeptide trimer Ga-68-Triveoctin. Ejnmmi Res. 2020, 10. [Google Scholar] [CrossRef]
- Hennrich, U.; Kopka, K. Lutathera (R): The First FDA- and EMA-Approved Radiopharmaceutical for Peptide Receptor Radionuclide Therapy. Pharm. Base 2019, 12, 114. [Google Scholar] [CrossRef] [Green Version]
- Jones, W.; Griffiths, K.; Barata, P.C.; Paller, C.J. PSMA Theranostics: Review of the Current Status of PSMA-Targeted Imaging and Radioligand Therapy. Cancers 2020, 12, 1367. [Google Scholar] [CrossRef]
- Chen, H.; Zhao, L.; Fu, K.; Lin, Q.; Wen, X.; Jacobson, O.; Sun, L.; Wu, H.; Zhang, X.; Guo, Z.; et al. Integrin αvβ3-targeted radionuclide therapy combined with immune checkpoint blockade immunotherapy synergistically enhances anti-tumor efficacy. Theranostics 2019, 9, 7948–7960. [Google Scholar] [CrossRef]
- Zhao, L.; Chen, H.; Guo, Z.; Fu, K.; Yao, L.; Fu, L.; Guo, W.; Wen, X.; Jacobson, O.; Zhang, X.; et al. Targeted Radionuclide Therapy in Patient-Derived Xenografts Using 177Lu-EB-RGD. Mol. Cancer Ther. 2020, 19, 2034–2043. [Google Scholar] [CrossRef]
- Jin, Z.H.; Furukawa, T.; Degardin, M.; Sugyo, A.; Tsuji, A.B.; Yamasaki, T.; Kawamura, K.; Fujibayashi, Y.; Zhang, M.R.; Boturyn, D.; et al. αVβ3 Integrin-Targeted Radionuclide Therapy with 64Cu-cyclam-RAFT-c(-RGDfK-)4. Mol. Cancer Ther. 2016, 15, 2076–2085. [Google Scholar] [CrossRef] [Green Version]
- Ye, Y.P.; Chen, X.Y. Integrin Targeting for Tumor Optical Imaging. Theranostics 2011, 1, 102–126. [Google Scholar] [CrossRef] [Green Version]
- Beer, A.J.; Schwaiger, M. Imaging of integrin αvβ3 expression. Cancer Metast Rev. 2008, 27, 631–644. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.J.; Yang, Y.; Zhang, C.F. A concise review of magnetic resonance molecular imaging of tumor angiogenesis by targeting integrin alpha v beta 3 with magnetic probes. Int. J. Nanomed. 2013, 8, 1083–1093. [Google Scholar] [CrossRef] [Green Version]
- Kiessling, F.; Gaetjens, J.; Palmowski, M. Application of Molecular Ultrasound for Imaging Integrin Expression. Theranostics 2011, 1, 127–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martelli, C.; Lo Dico, A.; Diceglie, C.; Lucignani, G.; Ottobrini, L. Optical imaging probes in oncology. Oncotarget 2016, 7, 48753–48787. [Google Scholar] [CrossRef] [Green Version]
- Wu, P.H.; Opadele, A.E.; Onodera, Y.; Nam, J.M. Targeting Integrins in Cancer Nanomedicine: Applications in Cancer Diagnosis and Therapy. Cancers 2019, 11, 1783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hernot, S.; van Manen, L.; Debie, P.; Mieog, J.S.D.; Vahrmeijer, A.L. Latest developments in molecular tracers for fluorescence image-guided cancer surgery. Lancet Oncol. 2019, 20, E354–E367. [Google Scholar] [CrossRef]
- Favril, S.; Brioschi, C.; Vanderperren, K.; Abma, E.; Stock, E.; Devriendt, N.; Polis, I.; De Cock, H.; Cordaro, A.; Miragoli, L.; et al. Preliminary safety and imaging efficacy of the near-infrared fluorescent contrast agent DA364 during fluorescence-guided surgery in dogs with spontaneous superficial tumors. Oncotarget 2020, 11, 2310–2326. [Google Scholar] [CrossRef] [PubMed]
- Wenk, C.H.F.; Ponce, F.; Guillermet, S.; Tenaud, C.; Boturyn, D.; Dumy, P.; Watrelot-Virieux, D.; Carozzo, C.; Josserand, V.; Coll, J.L. Near-infrared optical guided surgery of highly infiltrative fibrosarcomas in cats using an anti-alpha(v)beta(3) integrin molecular probe. Cancer Lett. 2013, 334, 188–195. [Google Scholar] [CrossRef]
- Baart, V.M.; van Duijn, C.; van Egmond, S.L.; Dijckmeester, W.A.; Jansen, J.C.; Vahrmeijer, A.L.; Sier, C.F.M.; Cohen, D. EGFR and αvβ6 as Promising Targets for Molecular Imaging of Cutaneous and Mucosal Squamous Cell Carcinoma of the Head and Neck Region. Cancers 2020, 12, 1474. [Google Scholar] [CrossRef]
- Nieberler, M.; Reuning, U.; Kessler, H.; Reichart, F.; Weirich, G.; Wolff, K.D. Fluorescence imaging of invasive head and neck carcinoma cells with integrin αvβ6-targeting RGD-peptides: An approach to a fluorescence-assisted intraoperative cytological assessment of bony resection margins. Br. J. Oral Maxillofac. Surg. 2018, 56, 972–978. [Google Scholar] [CrossRef]
- Tummers, W.S.; Kimura, R.H.; Abou-Elkacem, L.; Beinat, C.; Vahrmeijer, A.L.; Swijnenburg, R.-J.; Willmann, J.K.; Gambhir, S.S. Development and Preclinical Validation of a Cysteine Knottin Peptide Targeting Integrin αvβ6 for Near-infrared Fluorescent-guided Surgery in Pancreatic Cancer. Clin. Cancer Res. 2018, 24, 1667–1676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ganguly, T.; Tang, S.Y.; Bauer, N.; Sutcliffe, J.L. Evaluation of Two Optical Probes for Imaging the Integrin αvβ6− In Vitro and In Vivo in Tumor-Bearing Mice. Mol. Imaging Biol. 2020, 22, 1170–1181. [Google Scholar] [CrossRef] [PubMed]
- Handgraaf, H.J.M.; Boonstra, M.C.; Prevoo, H.A.J.M.; Kuil, J.; Bordo, M.W.; Boogerd, L.S.F.; Mulder, B.G.S.; Sier, C.F.M.; Vinkenburg-van Slooten, M.L.; Valentijn, A.R.P.M.; et al. Real-time near-infrared fluorescence imaging using cRGDZW800-1 for intraoperative visualization of multiple cancer types. Oncotarget 2017, 8, 21054–21066. [Google Scholar] [CrossRef] [Green Version]
- de Valk, K.S.; Deken, M.M.; Handgraaf, H.J.M.; Bhairosingh, S.S.; Bijlstra, O.D.; van Esdonk, M.J.; Terwisscha van Scheltinga, A.G.T.; Valentijn, A.R.P.M.; March, T.L.; Vuijk, J.; et al. 1 First-in-Human Assessment of cRGD-ZW800-1, a Zwitterionic, Integrin-Targeted, Near-Infrared Fluorescent Peptide in Colon Carcinoma. Clin. Cancer Res. 2020, 26. [Google Scholar] [CrossRef]
- Chen, F.; Madajewski, B.; Ma, K.; Zanoni, D.K.; Stambuk, H.; Turker, M.Z.; Monette, S.; Zhang, L.; Yoo, B.; Chen, P.M.; et al. Molecular phenotyping and image-guided surgical treatment of melanoma using spectrally distinct ultrasmall core-shell silica nanoparticles. Sci. Adv. 2019, 5. [Google Scholar] [CrossRef] [Green Version]
- Pirovano, G.; Roberts, S.; Kossatz, S.; Reiner, T. Optical Imaging Modalities: Principles and Applications in Preclinical Research and Clinical Settings. J. Nucl. Med. 2020, 61, 1419–1427. [Google Scholar] [CrossRef]
- Haedicke, K.; Brand, C.; Omar, M.; Ntziachristos, V.; Reiner, T.; Grimm, J. Sonophore labeled RGD: A targeted contrast agent for optoacoustic imaging. Photoacoustics 2017, 6, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Manzoni, L.; Belvisi, L.; Arosio, D.; Civera, M.; Pilkington-Miksa, M.; Potenza, D.; Caprini, A.; Araldi, E.M.V.; Monferini, E.; Mancino, M.; et al. Cyclic RGD-Containing Functionalized Azabicycloalkane Peptides as Potent Integrin Antagonists for Tumor Targeting. Chemmedchem 2009, 4, 615–632. [Google Scholar] [CrossRef] [PubMed]
- Capozza, M.; Blasi, F.; Valbusa, G.; Oliva, P.; Cabella, C.; Buonsanti, F.; Cordaro, A.; Pizzuto, L.; Maiocchi, A.; Poggi, L. Photoacoustic imaging of integrin-overexpressing tumors using a novel ICG-based contrast agent in mice. Photoacoustics 2018, 11, 36–45. [Google Scholar] [CrossRef]
- Huang, R.; Harmsen, S.; Samii, J.M.; Karabeber, H.; Pitter, K.L.; Holland, E.C.; Kircher, M.F. High Precision Imaging of Microscopic Spread of Glioblastoma with a Targeted Ultrasensitive SERRS Molecular Imaging Probe. Theranostics 2016, 6, 1075–1084. [Google Scholar] [CrossRef]
- Nicolson, F.; Andreiuk, B.; Andreou, C.; Hsu, H.-T.; Rudder, S.; Kircher, M.F. Non-invasive In Vivo Imaging of Cancer Using Surface-Enhanced Spatially Offset Raman Spectroscopy (SESORS). Theranostics 2019, 9, 5899–5913. [Google Scholar] [CrossRef] [PubMed]
- Peters, K.; Unger, R.E.; Brunner, J.; Kirkpatrick, C.J. Molecular basis of endothelial dysfunction in sepsis. Cardiovasc Res. 2003, 60, 49–57. [Google Scholar] [CrossRef] [Green Version]
- Kerrigan, S.W.; Devine, T.; Fitzpatrick, G.; Thachil, J.; Cox, D. Early Host Interactions That Drive the Dysregulated Response in Sepsis. Front. Immunol. 2019, 10, 1748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ince, C.; Mayeux, P.R.; Nguyen, T.; Gomez, H.; Kellum, J.A.; Ospina-Tascón, G.A.; Hernandez, G.; Murray, P.; De Backer, D. The endothelium in sepsis. Shock 2016, 45, 259–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Noiri, E.; Gailit, J.; Sheth, D.; Magazine, H.; Gurrath, M.; Muller, G.; Kessler, H.; Goligorsky, M.S. Cyclic RGD peptides ameliorate ischemic acute renal failure in rats. Kidney Int. 1994, 46, 1050–1058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McHale, T.M.; Garciarena, C.D.; Fagan, R.P.; Smith, S.G.J.; Martin-Loches, I.; Curley, G.F.; Fitzpatrick, F.; Kerrigan, S.W. Inhibition of Vascular Endothelial Cell Leak Following Escherichia coli Attachment in an Experimental Model of Sepsis. Crit Care Med. 2018, 46, e805–e810. [Google Scholar] [CrossRef] [PubMed]
- McDonnell, C.J.; Garciarena, C.D.; Watkin, R.L.; McHale, T.M.; McLoughlin, A.; Claes, J.; Verhamme, P.; Cummins, P.M.; Kerrigan, S.W. Inhibition of major integrin αvβ3 reduces Staphylococcus aureus attachment to sheared human endothelial cells. J. Thromb. Haemost. 2016, 14, 2536–2547. [Google Scholar] [CrossRef]
- Viela, F.; Speziale, P.; Pietrocola, G.; Dufrêne, Y.F. Mechanostability of the Fibrinogen Bridge between Staphylococcal Surface Protein ClfA and Endothelial Cell Integrin αvβ3. Nano Lett. 2019, 19, 7400–7410. [Google Scholar] [CrossRef]
- Sinha, B.; François, P.P.; Nüsse, O.; Foti, M.; Hartford, O.M.; Vaudaux, P.; Foster, T.J.; Lew, D.P.; Herrmann, M.; Krause, K.H. Fibronectin-binding protein acts as Staphylococcus aureus invasin via fibronectin bridging to integrin alpha5beta1. Cell Microbiol. 1999, 1, 101–117. [Google Scholar] [CrossRef]
- Fowler, T.; Wann, E.R.; Joh, D.; Johansson, S.; Foster, T.J.; Höök, M. Cellular invasion by Staphylococcus aureus involves a fibronectin bridge between the bacterial fibronectin-binding MSCRAMMs and host cell beta1 integrins. Eur J. Cell Biol. 2000, 79, 672–679. [Google Scholar] [CrossRef]
- Grundmeier, M.; Hussain, M.; Becker, P.; Heilmann, C.; Peters, G.; Sinha, B. Truncation of fibronectin-binding proteins in Staphylococcus aureus strain Newman leads to deficient adherence and host cell invasion due to loss of the cell wall anchor function. Infect. Immun. 2004, 72, 7155–7163. [Google Scholar] [CrossRef] [Green Version]
- Bingham, R.J.; Rudiño-Piñera, E.; Meenan, N.A.; Schwarz-Linek, U.; Turkenburg, J.P.; Höök, M.; Garman, E.F.; Potts, J.R. Crystal structures of fibronectin-binding sites from Staphylococcus aureus FnBPA in complex with fibronectin domains. Proc. Natl. Acad. Sci. USA 2008, 105, 12254–12258. [Google Scholar] [CrossRef] [Green Version]
- Garciarena, C.D.; McHale, T.M.; Martin-Loeches, I.; Kerrigan, S.W. Pre-emptive and therapeutic value of blocking bacterial attachment to the endothelial alphaVbeta3 integrin with cilengitide in sepsis. Crit Care 2017, 21, 246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Frangogiannis, N. Transforming growth factor-β in tissue fibrosis. J. Exp. Med. 2020, 217, e20190103. [Google Scholar] [CrossRef]
- Munger, J.S.; Huang, X.; Kawakatsu, H.; Griffiths, M.J.; Dalton, S.L.; Wu, J.; Pittet, J.F.; Kaminski, N.; Garat, C.; Matthay, M.A.; et al. The integrin alpha v beta 6 binds and activates latent TGF beta 1: A mechanism for regulating pulmonary inflammation and fibrosis. Cell 1999, 96, 319–328. [Google Scholar] [CrossRef] [Green Version]
- Häkkinen, L.; Koivisto, L.; Gardner, H.; Saarialho-Kere, U.; Carroll, J.M.; Lakso, M.; Rauvala, H.; Laato, M.; Heino, J.; Larjava, H. Increased expression of beta6-integrin in skin leads to spontaneous development of chronic wounds. Am. J. Pathol. 2004, 164, 229–242. [Google Scholar] [CrossRef]
- Shi, M.; Zhu, J.; Wang, R.; Chen, X.; Mi, L.; Walz, T.; Springer, T.A. Latent TGF-β structure and activation. Nature 2011, 474, 343–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Froese, A.R.; Shimbori, C.; Bellaye, P.S.; Inman, M.; Obex, S.; Fatima, S.; Jenkins, G.; Gauldie, J.; Ask, K.; Kolb, M. Stretch-induced Activation of Transforming Growth Factor-β1 in Pulmonary Fibrosis. Am. J. Respir Crit Care Med. 2016, 194, 84–96. [Google Scholar] [CrossRef]
- Sarrazy, V.; Koehler, A.; Chow, M.L.; Zimina, E.; Li, C.X.; Kato, H.; Caldarone, C.A.; Hinz, B. Integrins αvβ5 and αvβ3 promote latent TGF-β1 activation by human cardiac fibroblast contraction. Cardiovasc Res. 2014, 102, 407–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, L.J.; Yang, H.; Gaspert, A.; Carlesso, G.; Barty, M.M.; Davidson, J.M.; Sheppard, D.; Fogo, A.B. Transforming growth factor-beta-dependent and -independent pathways of induction of tubulointerstitial fibrosis in beta6(-/-) mice. Am. J. Pathol. 2003, 163, 1261–1273. [Google Scholar] [CrossRef]
- Chang, Y.; Lau, W.L.; Jo, H.; Tsujino, K.; Gewin, L.; Reed, N.I.; Atakilit, A.; Nunes, A.C.F.; DeGrado, W.F.; Sheppard, D. Pharmacologic Blockade of αvβ1 Integrin Ameliorates Renal Failure and Fibrosis In Vivo. J. Am. Soc. Nephrol. 2017, 28, 1998–2005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Horan, G.S.; Wood, S.; Ona, V.; Li, D.J.; Lukashev, M.E.; Weinreb, P.H.; Simon, K.J.; Hahm, K.; Allaire, N.E.; Rinaldi, N.J.; et al. Partial inhibition of integrin alpha(v)beta6 prevents pulmonary fibrosis without exacerbating inflammation. Am. J. Respir. Crit. Care Med. 2008, 177, 56–65. [Google Scholar] [CrossRef] [PubMed]
- John, A.E.; Graves, R.H.; Pun, K.T.; Vitulli, G.; Forty, E.J.; Mercer, P.F.; Morrell, J.L.; Barrett, J.W.; Rogers, R.F.; Hafeji, M.; et al. Translational pharmacology of an inhaled small molecule alpha v beta 6 integrin inhibitor for idiopathic pulmonary fibrosis. Nat. Commun. 2020, 11. [Google Scholar] [CrossRef]
- Anderson, N.A.; Campbell, I.B.; Fallon, B.J.; Lynn, S.M.; Macdonald, S.J.F.; Pritchard, J.M.; Procopiou, P.A.; Sollis, S.L.; Thorp, L.R. Synthesis and determination of absolute configuration of a non-peptidic alpha(v)beta(6) integrin antagonist for the treatment of idiopathic pulmonary fibrosis. Org. Biomol. Chem. 2016, 14, 5992–6009. [Google Scholar] [CrossRef]
- Procopiou, P.A.; Anderson, N.A.; Barrett, J.; Barrett, T.N.; Crawford, M.H.J.; Fallon, B.J.; Hancock, A.P.; Le, J.; Lemma, S.; Marshall, R.P.; et al. Discovery of (S)-3-(3-(3,5-Dimethyl-1H-pyrazol-1-yl)phenyl)-4-((R)-3(2-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)ethyl)pyrrolidin-1-yl)butanoic Acid, a Nonpeptidic alpha(v)beta(6) Integrin Inhibitor for the Inhaled Treatment of Idiopathic Pulmonary Fibrosis. J. Med. Chem. 2018, 61, 8417–8443. [Google Scholar] [CrossRef] [PubMed]
- Maher, T.M.; Simpson, J.K.; Porter, J.C.; Wilson, F.J.; Chan, R.B.; Eames, R.; Cui, Y.; Siederer, S.; Parry, S.; Kenny, J.; et al. A positron emission tomography imaging study to confirm target engagement in the lungs of patients with idiopathic pulmonary fibrosis following a single dose of a novel inhaled alpha v beta 6 integrin inhibitor. Resp Res. 2020, 21. [Google Scholar] [CrossRef]
- Minagawa, S.; Lou, J.; Seed, R.I.; Cormier, A.; Wu, S.; Cheng, Y.; Murray, L.; Tsui, P.; Connor, J.; Herbst, R.; et al. Selective targeting of TGF-β activation to treat fibroinflammatory airway disease. Sci. Transl. Med. 2014, 6, 241ra279. [Google Scholar] [CrossRef] [Green Version]
- Sigrist, C.J.A.; Bridge, A.; Le Mercier, P. A potential role for integrins in host cell entry by SARS-CoV-2. Antivir. Res. 2020, 177. [Google Scholar] [CrossRef]
- Wrapp, D.; Wang, N.S.; Corbett, K.S.; Goldsmith, J.A.; Hsieh, C.L.; Abiona, O.; Graham, B.S.; McLellan, J.S. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 2020, 367, 1260. [Google Scholar] [CrossRef] [Green Version]
- Pirone, L.; Del Gatto, A.; Di Gaetano, S.; Saviano, M.; Capasso, D.; Zaccaro, L.; Pedone, E. A Multi-Targeting Approach to Fight SARS-CoV-2 Attachment. Front. Mol. Biosci. 2020, 7, 186. [Google Scholar] [CrossRef] [PubMed]
- Tresoldi, I.; Sangiuolo, C.F.; Manzari, V.; Modesti, A. SARS-COV-2 and infectivity: Possible increase in infectivity associated to integrin motif expression. J. Med. Virol. 2020, 92, 1741–1742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luan, J.W.; Lu, Y.; Gao, S.; Zhang, L.L. A potential inhibitory role for integrin in the receptor targeting of SARS-CoV-2. J. Infect. 2020, 81, 337–339. [Google Scholar] [CrossRef] [PubMed]
- Dakal, T.C. SARS-CoV-2 attachment to host cells is possibly mediated via RGD-integrin interaction in a calcium-dependent manner and suggests pulmonary EDTA chelation therapy as a novel treatment for COVID 19. Immunobiology 2021, 226. [Google Scholar] [CrossRef] [PubMed]
- Beddingfield, B.J.; Iwanaga, N.; Chapagain, P.P.; Zheng, W.; Roy, C.J.; Hu, T.Y.; Kolls, J.K.; Bix, G.J. The Integrin Binding Peptide, ATN-161, as a Novel Therapy for SARS-CoV-2 Infection. Jacc Basic Transl. Sci. 2020. [Google Scholar] [CrossRef]
- Bristow, M.R.; Zisman, L.S.; Altman, N.L.; Gilbert, E.M.; Lowes, B.D.; Minobe, W.A.; Slavov, D.; Schwisow, J.A.; Rodriguez, E.M.; Carroll, I.A.; et al. Dynamic Regulation of SARS-Cov-2 Binding and Cell Entry Mechanisms in Remodeled Human Ventricular Myocardium. Jacc Basic Transl. Sci. 2020, 5, 871–883. [Google Scholar] [CrossRef] [PubMed]
- Aguirre, C.; Meca-Lallana, V.; Barrios-Blandino, A.; del Río, B.; Vivancos, J. Covid-19 in a patient with multiple sclerosis treated with natalizumab: May the blockade of integrins have a protective role? Mult. Scler. Relat. Disord. 2020, 44. [Google Scholar] [CrossRef] [PubMed]
- Foster, C.C.; Davis, R.A.; Hausner, S.H.; Sutcliffe, J.L. αvβ6-Targeted Molecular PET/CT Imaging of the Lungs After SARS-CoV-2 Infection. J. Nucl. Med. 2020, 61, 1717–1719. [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
Ludwig, B.S.; Kessler, H.; Kossatz, S.; Reuning, U. RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field. Cancers 2021, 13, 1711. https://doi.org/10.3390/cancers13071711
Ludwig BS, Kessler H, Kossatz S, Reuning U. RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field. Cancers. 2021; 13(7):1711. https://doi.org/10.3390/cancers13071711
Chicago/Turabian StyleLudwig, Beatrice S., Horst Kessler, Susanne Kossatz, and Ute Reuning. 2021. "RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field" Cancers 13, no. 7: 1711. https://doi.org/10.3390/cancers13071711
APA StyleLudwig, B. S., Kessler, H., Kossatz, S., & Reuning, U. (2021). RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field. Cancers, 13(7), 1711. https://doi.org/10.3390/cancers13071711