Nanobodies as Versatile Tool for Multiscale Imaging Modalities
Abstract
:1. Introduction
2. Nanobodies: Characteristic and Structure
3. Generation of Nanobody Libraries
4. Nanobodies as Imaging Tools
4.1. Imaging Tumor
4.1.1. Imaging Tumor Cells
4.1.2. Imaging Tumor Microenvironment
4.1.3. Imaging Tumor Immune Checkpoint
4.1.4. Theranostic Approach Using Nbs
4.2. Imaging Inflammatory Diseases
Nanobody | Inflammatory Disease Model | Imaging Modality | Reference |
---|---|---|---|
Anti-VCAM-1 | Atherosclerosis | SPECT, PET/CT, Ultrasound | [121,122,123] |
Anti-MMR | Atherosclerosis | PET/CT, SPECT | [125,126] |
Anti-MMR | Rheumatoid Arthritis | SPECT/CT | [128] |
Anti-CRIg | Rheumatoid Arthritis, Sterile Inflammatory Arthritis | SPECT/CT | [129,130] |
Anti-Vsig4 | Rheumatoid Arthritis | NIRF-optical imaging | [131] |
Anti-Vsig4 | Acute Hepatitis, Steatohepatitis | SPECT | [132,133] |
Anti-Clec4F | Acute Hepatitis, Steatohepatitis | SPECT | [132,133] |
Anti-DPP6 | Diabetes | SPECT/CT, PET | [134,135] |
4.3. Imaging Pathologies of the Central Nervous System (CNS)
5. Nanobodies in Fluorescence-Guided Surgery
6. Nanobodies in Microscopy
7. Conclusive Remarks
Author Contributions
Funding
Conflicts of Interest
References
- Mezzanotte, L.; van’t Root, M.; Karatas, H.; Goun, E.A.; Lowik, C. In Vivo Molecular Bioluminescence Imaging: New Tools and Applications. Trends Biotechnol. 2017, 35, 640–652. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Z.; Lu, Z.R. Molecular imaging of the tumor microenvironment. Adv. Drug Deliv. Rev. 2017, 113, 24–48. [Google Scholar] [CrossRef] [PubMed]
- Power, R.M.; Huisken, J. A guide to light-sheet fluorescence microscopy for multiscale imaging. Nat. Methods 2017, 14, 360–373. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.; Wang, Z.; Zhao, T.; Li, Y.; Huang, G.; Sumer, B.D.; Gao, J. Optical molecular imaging for tumor detection and image-guided surgery. Biomaterials 2018, 157, 62–75. [Google Scholar] [CrossRef] [PubMed]
- Ebrahim, S.; Weigert, R. Intravital microscopy in mammalian multicellular organisms. Curr. Opin. Cell Biol. 2019, 59, 97–103. [Google Scholar] [CrossRef] [PubMed]
- Wessels, J.T.; Busse, A.C.; Mahrt, J.; Dullin, C.; Grabbe, E.; Mueller, G.A. In vivo imaging in experimental preclinical tumor research—A review. Cytom. A 2007, 71, 542–549. [Google Scholar] [CrossRef] [PubMed]
- Marelli, G.; Avigni, R.; Allavena, P.; Garlanda, C.; Mantovani, A.; Doni, A.; Erreni, M. Optical in vivo imaging detection of preclinical models of gut tumors through the expression of integrin alphaVbeta3. Oncotarget 2018, 9, 31380–31396. [Google Scholar] [CrossRef] [Green Version]
- Wu, M.; Shu, J. Multimodal Molecular Imaging: Current Status and Future Directions. Contrast Media Mol. Imaging. 2018, 2018, 1382183. [Google Scholar] [CrossRef] [Green Version]
- Ward, E.S.; Gussow, D.; Griffiths, A.D.; Jones, P.T.; Winter, G. Binding activities of a repertoire of single immunoglobulin variable domains secreted from Escherichia coli. Nature 1989, 341, 544–546. [Google Scholar] [CrossRef]
- Hamers-Casterman, C.; Atarhouch, T.; Muyldermans, S.; Robinson, G.; Hamers, C.; Songa, E.B.; Bendahman, N.; Hamers, R. Naturally occurring antibodies devoid of light chains. Nature 1993, 363, 446–448. [Google Scholar] [CrossRef]
- Arbabi-Ghahroudi, M.; Tanha, J.; MacKenzie, R. Prokaryotic expression of antibodies. Cancer Metastasis Rev. 2005, 24, 501–519. [Google Scholar] [CrossRef] [PubMed]
- Muyldermans, S.; Atarhouch, T.; Saldanha, J.; Barbosa, J.A.; Hamers, R. Sequence and structure of VH domain from naturally occurring camel heavy chain immunoglobulins lacking light chains. Protein Eng. 1994, 7, 1129–1135. [Google Scholar] [CrossRef] [PubMed]
- Chakravarty, R.; Goel, S.; Cai, W. Nanobody: The “magic bullet” for molecular imaging? Theranostics 2014, 4, 386–398. [Google Scholar] [CrossRef] [PubMed]
- Patel, Y.C.; Srikant, C.B. Somatostatin receptors. Trends Endocrinol. Metab. 1997, 8, 398–405. [Google Scholar] [CrossRef]
- Reynolds, F.; Kelly, K.A. Techniques for molecular imaging probe design. Mol. Imaging 2011, 10, 407–419. [Google Scholar] [CrossRef]
- Wu, A.M. Engineered antibodies for molecular imaging of cancer. Methods 2014, 65, 139–147. [Google Scholar] [CrossRef] [Green Version]
- Marqus, S.; Pirogova, E.; Piva, T.J. Evaluation of the use of therapeutic peptides for cancer treatment. J. Biomed. Sci. 2017, 24, 21. [Google Scholar] [CrossRef] [Green Version]
- Capozzi, E.; Aureli, S.; Minicozzi, V.; Rossi, G.C.; Stellato, F.; Morante, S. Designing effective anticancer-radiopeptides. A Molecular Dynamics study of their interaction with model tumor and healthy cell membranes. Biochim. Biophys. Acta Biomembr. 2018, 1860, 2348–2355. [Google Scholar] [CrossRef]
- Mandleywala, K.; Shmuel, S.; Pereira, P.M.R.; Lewis, J.S. Antibody-Targeted Imaging of Gastric Cancer. Molecules 2020, 25, 4621. [Google Scholar] [CrossRef]
- Chiangjong, W.; Chutipongtanate, S.; Hongeng, S. Anticancer peptide: Physicochemical property, functional aspect and trend in clinical application (Review). Int. J. Oncol. 2020, 57, 678–696. [Google Scholar] [CrossRef]
- Liu, J.K. The history of monoclonal antibody development—Progress, remaining challenges and future innovations. Ann. Med. Surg. 2014, 3, 113–116. [Google Scholar] [CrossRef] [PubMed]
- Huston, J.S.; Levinson, D.; Mudgett-Hunter, M.; Tai, M.S.; Novotny, J.; Margolies, M.N.; Ridge, R.J.; Bruccoleri, R.E.; Haber, E.; Crea, R.; et al. Protein engineering of antibody binding sites: Recovery of specific activity in an anti-digoxin single-chain Fv analogue produced in Escherichia coli. Proc. Natl. Acad. Sci. USA 1988, 85, 5879–5883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ewert, S.; Huber, T.; Honegger, A.; Pluckthun, A. Biophysical properties of human antibody variable domains. J. Mol. Biol. 2003, 325, 531–553. [Google Scholar] [CrossRef]
- Olafsen, T.; Wu, A.M. Antibody vectors for imaging. Semin Nucl. Med. 2010, 40, 167–181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kobayashi, H.; Choyke, P.L.; Ogawa, M. Monoclonal antibody-based optical molecular imaging probes; considerations and caveats in chemistry, biology and pharmacology. Curr. Opin. Chem. Biol. 2016, 33, 32–38. [Google Scholar] [CrossRef] [Green Version]
- Muyldermans, S. Nanobodies: Natural single-domain antibodies. Annu. Rev. Biochem. 2013, 82, 775–797. [Google Scholar] [CrossRef] [Green Version]
- Arbabi Ghahroudi, M.; Desmyter, A.; Wyns, L.; Hamers, R.; Muyldermans, S. Selection and identification of single domain antibody fragments from camel heavy-chain antibodies. FEBS Lett. 1997, 414, 521–526. [Google Scholar] [CrossRef] [Green Version]
- De Meyer, T.; Muyldermans, S.; Depicker, A. Nanobody-based products as research and diagnostic tools. Trends Biotechnol. 2014, 32, 263–270. [Google Scholar] [CrossRef]
- Skerra, A. Alternative non-antibody scaffolds for molecular recognition. Curr. Opin. Biotechnol. 2007, 18, 295–304. [Google Scholar] [CrossRef]
- Vincke, C.; Loris, R.; Saerens, D.; Martinez-Rodriguez, S.; Muyldermans, S.; Conrath, K. General strategy to humanize a camelid single-domain antibody and identification of a universal humanized nanobody scaffold. J. Biol. Chem. 2009, 284, 3273–3284. [Google Scholar] [CrossRef] [Green Version]
- Muruganandam, A.; Tanha, J.; Narang, S.; Stanimirovic, D. Selection of phage-displayed llama single-domain antibodies that transmigrate across human blood-brain barrier endothelium. FASEB J. 2002, 16, 240–242. [Google Scholar] [CrossRef]
- Wolak, D.J.; Thorne, R.G. Diffusion of macromolecules in the brain: Implications for drug delivery. Mol. Pharm. 2013, 10, 1492–1504. [Google Scholar] [CrossRef] [PubMed]
- Cartwright, A.N.; Griggs, J.; Davis, D.M. The immune synapse clears and excludes molecules above a size threshold. Nat. Commun. 2014, 5, 5479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vaneycken, I.; D’Huyvetter, M.; Hernot, S.; De Vos, J.; Xavier, C.; Devoogdt, N.; Caveliers, V.; Lahoutte, T. Immuno-imaging using nanobodies. Curr. Opin. Biotechnol. 2011, 22, 877–881. [Google Scholar] [CrossRef]
- Debie, P.; Lafont, C.; Defrise, M.; Hansen, I.; van Willigen, D.M.; van Leeuwen, F.W.B.; Gijsbers, R.; D’Huyvetter, M.; Devoogdt, N.; Lahoutte, T.; et al. Size and affinity kinetics of nanobodies influence targeting and penetration of solid tumours. J. Control. Release 2020, 317, 34–42. [Google Scholar] [CrossRef] [PubMed]
- Jovcevska, I.; Muyldermans, S. The Therapeutic Potential of Nanobodies. BioDrugs 2020, 34, 11–26. [Google Scholar] [CrossRef] [Green Version]
- Stanfield, R.L.; Dooley, H.; Flajnik, M.F.; Wilson, I.A. Crystal structure of a shark single-domain antibody V region in complex with lysozyme. Science 2004, 305, 1770–1773. [Google Scholar] [CrossRef]
- De Vos, J.; Devoogdt, N.; Lahoutte, T.; Muyldermans, S. Camelid single-domain antibody-fragment engineering for (pre)clinical in vivo molecular imaging applications: Adjusting the bullet to its target. Expert Opin. Biol. Ther. 2013, 13, 1149–1160. [Google Scholar] [CrossRef]
- Kunz, P.; Zinner, K.; Mucke, N.; Bartoschik, T.; Muyldermans, S.; Hoheisel, J.D. The structural basis of nanobody unfolding reversibility and thermoresistance. Sci. Rep. 2018, 8, 7934. [Google Scholar] [CrossRef]
- Desmyter, A.; Decanniere, K.; Muyldermans, S.; Wyns, L. Antigen specificity and high affinity binding provided by one single loop of a camel single-domain antibody. J. Biol. Chem. 2001, 276, 26285–26290. [Google Scholar] [CrossRef] [Green Version]
- De Genst, E.; Silence, K.; Decanniere, K.; Conrath, K.; Loris, R.; Kinne, J.; Muyldermans, S.; Wyns, L. Molecular basis for the preferential cleft recognition by dromedary heavy-chain antibodies. Proc. Natl. Acad. Sci. USA 2006, 103, 4586–4591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bradley, M.E.; Dombrecht, B.; Manini, J.; Willis, J.; Vlerick, D.; De Taeye, S.; Van den Heede, K.; Roobrouck, A.; Grot, E.; Kent, T.C.; et al. Potent and efficacious inhibition of CXCR2 signaling by biparatopic nanobodies combining two distinct modes of action. Mol. Pharmacol. 2015, 87, 251–262. [Google Scholar] [CrossRef] [PubMed]
- Danquah, W.; Meyer-Schwesinger, C.; Rissiek, B.; Pinto, C.; Serracant-Prat, A.; Amadi, M.; Iacenda, D.; Knop, J.H.; Hammel, A.; Bergmann, P.; et al. Nanobodies that block gating of the P2X7 ion channel ameliorate inflammation. Sci. Transl. Med. 2016, 8, 366ra162. [Google Scholar] [CrossRef] [PubMed]
- Al Qaraghuli, M.M.; Ferro, V.A. Analysis of the binding loops configuration and surface adaptation of different crystallized single-domain antibodies in response to various antigens. J. Mol. Recognit 2017, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Groof, T.W.M.; Bobkov, V.; Heukers, R.; Smit, M.J. Nanobodies: New avenues for imaging, stabilizing and modulating GPCRs. Mol. Cell Endocrinol. 2019, 484, 15–24. [Google Scholar] [CrossRef] [PubMed]
- Steeland, S.; Vandenbroucke, R.E.; Libert, C. Nanobodies as therapeutics: Big opportunities for small antibodies. Drug Discov. Today 2016, 21, 1076–1113. [Google Scholar] [CrossRef] [PubMed]
- Iezzi, M.E.; Policastro, L.; Werbajh, S.; Podhajcer, O.; Canziani, G.A. Single-Domain Antibodies and the Promise of Modular Targeting in Cancer Imaging and Treatment. Front. Immunol. 2018, 9, 273. [Google Scholar] [CrossRef] [Green Version]
- Ubah, O.C.; Buschhaus, M.J.; Ferguson, L.; Kovaleva, M.; Steven, J.; Porter, A.J.; Barelle, C.J. Next-generation flexible formats of VNAR domains expand the drug platform’s utility and developability. Biochem. Soc. Trans. 2018, 46, 1559–1565. [Google Scholar] [CrossRef]
- Harmsen, M.M.; De Haard, H.J. Properties, production, and applications of camelid single-domain antibody fragments. Appl. Microbiol. Biotechnol. 2007, 77, 13–22. [Google Scholar] [CrossRef] [Green Version]
- Muyldermans, S. A guide to: Generation and design of nanobodies. FEBS J. 2020. [Google Scholar] [CrossRef]
- Makvandi-Nejad, S.; Fjallman, T.; Arbabi-Ghahroudi, M.; MacKenzie, C.R.; Hall, J.C. Selection and expression of recombinant single domain antibodies from a hyper-immunized library against the hapten azoxystrobin. J. Immunol. Methods 2011, 373, 8–18. [Google Scholar] [CrossRef] [PubMed]
- Ezzine, A.; M’Hirsi El Adab, S.; Bouhaouala-Zahar, B.; Hmila, I.; Baciou, L.; Marzouki, M.N. Efficient expression of the anti-AahI’ scorpion toxin nanobody under a new functional form in a Pichia pastoris system. Biotechnol. Appl. Biochem. 2012, 59, 15–21. [Google Scholar] [CrossRef] [PubMed]
- Gorlani, A.; de Haard, H.; Verrips, T. Expression of VHHs in Saccharomyces cerevisiae. Methods Mol. Biol. 2012, 911, 277–286. [Google Scholar] [CrossRef] [PubMed]
- Lecocq, Q.; De Vlaeminck, Y.; Hanssens, H.; D’Huyvetter, M.; Raes, G.; Goyvaerts, C.; Keyaerts, M.; Devoogdt, N.; Breckpot, K. Theranostics in immuno-oncology using nanobody derivatives. Theranostics 2019, 9, 7772–7791. [Google Scholar] [CrossRef]
- Liu, Y.; Huang, H. Expression of single-domain antibody in different systems. Appl. Microbiol. Biotechnol. 2018, 102, 539–551. [Google Scholar] [CrossRef]
- Olichon, A.; de Marco, A. Preparation of a naive library of camelid single domain antibodies. Methods Mol. Biol. 2012, 911, 65–78. [Google Scholar] [CrossRef]
- Yan, J.; Li, G.; Hu, Y.; Ou, W.; Wan, Y. Construction of a synthetic phage-displayed Nanobody library with CDR3 regions randomized by trinucleotide cassettes for diagnostic applications. J. Transl. Med. 2014, 12, 343. [Google Scholar] [CrossRef] [Green Version]
- Woods, J. Selection of Functional Intracellular Nanobodies. SLAS Discov. 2019, 24, 703–713. [Google Scholar] [CrossRef]
- Sabir, J.S.; Atef, A.; El-Domyati, F.M.; Edris, S.; Hajrah, N.; Alzohairy, A.M.; Bahieldin, A. Construction of naive camelids VHH repertoire in phage display-based library. C. R. Biol. 2014, 337, 244–249. [Google Scholar] [CrossRef]
- Yan, J.; Wang, P.; Zhu, M.; Li, G.; Romao, E.; Xiong, S.; Wan, Y. Characterization and applications of Nanobodies against human procalcitonin selected from a novel naive Nanobody phage display library. J. Nanobiotechnol. 2015, 13, 33. [Google Scholar] [CrossRef] [Green Version]
- Zimmermann, I.; Egloff, P.; Hutter, C.A.J.; Kuhn, B.T.; Brauer, P.; Newstead, S.; Dawson, R.J.P.; Geertsma, E.R.; Seeger, M.A. Generation of synthetic nanobodies against delicate proteins. Nat. Protoc. 2020, 15, 1707–1741. [Google Scholar] [CrossRef] [PubMed]
- Hoogenboom, H.R. Selecting and screening recombinant antibody libraries. Nat. Biotechnol. 2005, 23, 1105–1116. [Google Scholar] [CrossRef] [PubMed]
- Hussack, G.; Riazi, A.; Ryan, S.; van Faassen, H.; MacKenzie, R.; Tanha, J.; Arbabi-Ghahroudi, M. Protease-resistant single-domain antibodies inhibit Campylobacter jejuni motility. Protein Eng. Des. Sel. 2014, 27, 191–198. [Google Scholar] [CrossRef] [PubMed]
- Janssens, R.; Dekker, S.; Hendriks, R.W.; Panayotou, G.; van Remoortere, A.; San, J.K.; Grosveld, F.; Drabek, D. Generation of heavy-chain-only antibodies in mice. Proc. Natl. Acad. Sci. USA 2006, 103, 15130–15135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Teng, Y.; Young, J.L.; Edwards, B.; Hayes, P.; Thompson, L.; Johnston, C.; Edwards, C.; Sanders, Y.; Writer, M.; Pinto, D.; et al. Diverse human VH antibody fragments with bio-therapeutic properties from the Crescendo Mouse. New Biotechnol. 2020, 55, 65–76. [Google Scholar] [CrossRef]
- Chanier, T.; Chames, P. Nanobody Engineering: Toward Next Generation Immunotherapies and Immunoimaging of Cancer. Antibodies 2019, 8, 13. [Google Scholar] [CrossRef] [Green Version]
- D’Huyvetter, M.; Xavier, C.; Caveliers, V.; Lahoutte, T.; Muyldermans, S.; Devoogdt, N. Radiolabeled nanobodies as theranostic tools in targeted radionuclide therapy of cancer. Expert Opin. Drug Deliv. 2014, 11, 1939–1954. [Google Scholar] [CrossRef]
- Debie, P.; Devoogdt, N.; Hernot, S. Targeted Nanobody-Based Molecular Tracers for Nuclear Imaging and Image-Guided Surgery. Antibodies 2019, 8, 12. [Google Scholar] [CrossRef] [Green Version]
- Hernot, S.; Unnikrishnan, S.; Du, Z.; Shevchenko, T.; Cosyns, B.; Broisat, A.; Toczek, J.; Caveliers, V.; Muyldermans, S.; Lahoutte, T.; et al. Nanobody-coupled microbubbles as novel molecular tracer. J. Control. Release 2012, 158, 346–353. [Google Scholar] [CrossRef] [Green Version]
- Rothbauer, U.; Zolghadr, K.; Tillib, S.; Nowak, D.; Schermelleh, L.; Gahl, A.; Backmann, N.; Conrath, K.; Muyldermans, S.; Cardoso, M.C.; et al. Targeting and tracing antigens in live cells with fluorescent nanobodies. Nat. Methods 2006, 3, 887–889. [Google Scholar] [CrossRef]
- Beghein, E.; Gettemans, J. Nanobody Technology: A Versatile Toolkit for Microscopic Imaging, Protein-Protein Interaction Analysis, and Protein Function Exploration. Front. Immunol 2017, 8, 771. [Google Scholar] [CrossRef] [PubMed]
- Schoonooghe, S.; Laoui, D.; Van Ginderachter, J.A.; Devoogdt, N.; Lahoutte, T.; De Baetselier, P.; Raes, G. Novel applications of nanobodies for in vivo bio-imaging of inflamed tissues in inflammatory diseases and cancer. Immunobiology 2012, 217, 1266–1272. [Google Scholar] [CrossRef] [PubMed]
- Hu, Y.; Liu, C.; Muyldermans, S. Nanobody-Based Delivery Systems for Diagnosis and Targeted Tumor Therapy. Front. Immunol. 2017, 8, 1442. [Google Scholar] [CrossRef]
- Belanger, K.; Iqbal, U.; Tanha, J.; MacKenzie, R.; Moreno, M.; Stanimirovic, D. Single-Domain Antibodies as Therapeutic and Imaging Agents for the Treatment of CNS Diseases. Antibodies 2019, 8, 27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, E.Y.; Shah, K. Nanobodies: Next Generation of Cancer Diagnostics and Therapeutics. Front. Oncol. 2020, 10, 1182. [Google Scholar] [CrossRef] [PubMed]
- Xavier, C.; Vaneycken, I.; D’Huyvetter, M.; Heemskerk, J.; Keyaerts, M.; Vincke, C.; Devoogdt, N.; Muyldermans, S.; Lahoutte, T.; Caveliers, V. Synthesis, preclinical validation, dosimetry, and toxicity of 68Ga-NOTA-anti-HER2 Nanobodies for iPET imaging of HER2 receptor expression in cancer. J. Nucl. Med. 2013, 54, 776–784. [Google Scholar] [CrossRef] [Green Version]
- Keyaerts, M.; Xavier, C.; Heemskerk, J.; Devoogdt, N.; Everaert, H.; Ackaert, C.; Vanhoeij, M.; Duhoux, F.P.; Gevaert, T.; Simon, P.; et al. Phase I Study of 68Ga-HER2-Nanobody for PET/CT Assessment of HER2 Expression in Breast Carcinoma. J. Nucl. Med. 2016, 57, 27–33. [Google Scholar] [CrossRef] [Green Version]
- Puttemans, J.; Dekempeneer, Y.; Eersels, J.L.; Hanssens, H.; Debie, P.; Keyaerts, M.; Windhorst, A.D.; van der Aa, F.; Lecocq, Q.; Breckpot, K.; et al. Preclinical Targeted alpha- and beta(-)-Radionuclide Therapy in HER2-Positive Brain Metastasis Using Camelid Single-Domain Antibodies. Cancers 2020, 12, 17. [Google Scholar] [CrossRef] [Green Version]
- Kijanka, M.M.; van Brussel, A.S.; van der Wall, E.; Mali, W.P.; van Diest, P.J.; van Bergen En Henegouwen, P.M.; Oliveira, S. Optical imaging of pre-invasive breast cancer with a combination of VHHs targeting CAIX and HER2 increases contrast and facilitates tumour characterization. EJNMMI Res. 2016, 6, 14. [Google Scholar] [CrossRef] [Green Version]
- Cai, W.; Niu, G.; Chen, X. Multimodality imaging of the HER-kinase axis in cancer. Eur. J. Nucl. Med. Mol. Imaging 2008, 35, 186–208. [Google Scholar] [CrossRef]
- Gainkam, L.O.; Keyaerts, M.; Caveliers, V.; Devoogdt, N.; Vanhove, C.; Van Grunsven, L.; Muyldermans, S.; Lahoutte, T. Correlation between epidermal growth factor receptor-specific nanobody uptake and tumor burden: A tool for noninvasive monitoring of tumor response to therapy. Mol. Imaging. Biol. 2011, 13, 940–948. [Google Scholar] [CrossRef] [PubMed]
- Gainkam, L.O.; Caveliers, V.; Devoogdt, N.; Vanhove, C.; Xavier, C.; Boerman, O.; Muyldermans, S.; Bossuyt, A.; Lahoutte, T. Localization, mechanism and reduction of renal retention of technetium-99m labeled epidermal growth factor receptor-specific nanobody in mice. Contrast Media Mol. Imaging. 2011, 6, 85–92. [Google Scholar] [CrossRef] [Green Version]
- Oliveira, S.; van Dongen, G.A.; Stigter-van Walsum, M.; Roovers, R.C.; Stam, J.C.; Mali, W.; van Diest, P.J.; van Bergen en Henegouwen, P.M. Rapid visualization of human tumor xenografts through optical imaging with a near-infrared fluorescent anti-epidermal growth factor receptor nanobody. Mol. Imaging. 2012, 11, 33–46. [Google Scholar] [CrossRef] [PubMed]
- Oliveira, S.; Heukers, R.; Sornkom, J.; Kok, R.J.; van Bergen En Henegouwen, P.M. Targeting tumors with nanobodies for cancer imaging and therapy. J. Control. Release 2013, 172, 607–617. [Google Scholar] [CrossRef]
- Hong, H.; Sun, J.; Cai, W. Radionuclide-Based Cancer Imaging Targeting the Carcinoembryonic Antigen. Biomark. Insights 2008, 3, 435–451. [Google Scholar] [CrossRef] [PubMed]
- Lwin, T.M.; Hernot, S.; Hollandsworth, H.; Amirfakhri, S.; Filemoni, F.; Debie, P.; Hoffman, R.M.; Bouvet, M. Tumor-specific near-infrared nanobody probe rapidly labels tumors in an orthotopic mouse model of pancreatic cancer. Surgery 2020, 168, 85–91. [Google Scholar] [CrossRef]
- Wu, T.; Dai, Y. Tumor microenvironment and therapeutic response. Cancer Lett. 2017, 387, 61–68. [Google Scholar] [CrossRef]
- Hui, L.; Chen, Y. Tumor microenvironment: Sanctuary of the devil. Cancer Lett. 2015, 368, 7–13. [Google Scholar] [CrossRef]
- Eble, J.A.; Niland, S. The extracellular matrix in tumor progression and metastasis. Clin. Exp. Metastasis 2019, 36, 171–198. [Google Scholar] [CrossRef]
- Jailkhani, N.; Ingram, J.R.; Rashidian, M.; Rickelt, S.; Tian, C.; Mak, H.; Jiang, Z.; Ploegh, H.L.; Hynes, R.O. Noninvasive imaging of tumor progression, metastasis, and fibrosis using a nanobody targeting the extracellular matrix. Proc. Natl. Acad. Sci. USA 2019, 116, 14181–14190. [Google Scholar] [CrossRef] [Green Version]
- Astrof, S.; Crowley, D.; George, E.L.; Fukuda, T.; Sekiguchi, K.; Hanahan, D.; Hynes, R.O. Direct test of potential roles of EIIIA and EIIIB alternatively spliced segments of fibronectin in physiological and tumor angiogenesis. Mol. Cell Biol. 2004, 24, 8662–8670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Uldry, E.; Faes, S.; Demartines, N.; Dormond, O. Fine-Tuning Tumor Endothelial Cells to Selectively Kill Cancer. Int. J. Mol. Sci. 2017, 18, 1401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kong, D.H.; Kim, Y.K.; Kim, M.R.; Jang, J.H.; Lee, S. Emerging Roles of Vascular Cell Adhesion Molecule-1 (VCAM-1) in Immunological Disorders and Cancer. Int. J. Mol. Sci. 2018, 19, 1057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Balkwill, F.R.; Mantovani, A. Cancer-related inflammation: Common themes and therapeutic opportunities. Semin Cancer Biol. 2012, 22, 33–40. [Google Scholar] [CrossRef]
- Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399–416. [Google Scholar] [CrossRef]
- Movahedi, K.; Laoui, D.; Gysemans, C.; Baeten, M.; Stange, G.; Van den Bossche, J.; Mack, M.; Pipeleers, D.; In’t Veld, P.; De Baetselier, P.; et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 2010, 70, 5728–5739. [Google Scholar] [CrossRef] [Green Version]
- Movahedi, K.; Schoonooghe, S.; Laoui, D.; Houbracken, I.; Waelput, W.; Breckpot, K.; Bouwens, L.; Lahoutte, T.; De Baetselier, P.; Raes, G.; et al. Nanobody-based targeting of the macrophage mannose receptor for effective in vivo imaging of tumor-associated macrophages. Cancer Res. 2012, 72, 4165–4177. [Google Scholar] [CrossRef] [Green Version]
- Blykers, A.; Schoonooghe, S.; Xavier, C.; D’Hoe, K.; Laoui, D.; D’Huyvetter, M.; Vaneycken, I.; Cleeren, F.; Bormans, G.; Heemskerk, J.; et al. PET Imaging of Macrophage Mannose Receptor-Expressing Macrophages in Tumor Stroma Using 18F-Radiolabeled Camelid Single-Domain Antibody Fragments. J. Nucl. Med. 2015, 56, 1265–1271. [Google Scholar] [CrossRef] [Green Version]
- Xavier, C.; Blykers, A.; Laoui, D.; Bolli, E.; Vaneyken, I.; Bridoux, J.; Baudhuin, H.; Raes, G.; Everaert, H.; Movahedi, K.; et al. Clinical Translation of [(68)Ga]Ga-NOTA-anti-MMR-sdAb for PET/CT Imaging of Protumorigenic Macrophages. Mol. Imaging. Biol. 2019, 21, 898–906. [Google Scholar] [CrossRef] [Green Version]
- Rashidian, M.; Keliher, E.; Dougan, M.; Juras, P.K.; Cavallari, M.; Wojtkiewicz, G.R.; Jacobsen, J.; Edens, J.G.; Tas, J.M.; Victora, G.; et al. The use of (18)F-2-fluorodeoxyglucose (FDG) to label antibody fragments for immuno-PET of pancreatic cancer. ACS Cent. Sci. 2015, 1, 142–147. [Google Scholar] [CrossRef] [Green Version]
- De Groeve, K.; Deschacht, N.; De Koninck, C.; Caveliers, V.; Lahoutte, T.; Devoogdt, N.; Muyldermans, S.; De Baetselier, P.; Raes, G. Nanobodies as tools for in vivo imaging of specific immune cell types. J. Nucl. Med. 2010, 51, 782–789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef] [PubMed]
- Zou, W.; Wolchok, J.D.; Chen, L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci. Transl. Med. 2016, 8, 328rv324. [Google Scholar] [CrossRef] [Green Version]
- Broos, K.; Keyaerts, M.; Lecocq, Q.; Renmans, D.; Nguyen, T.; Escors, D.; Liston, A.; Raes, G.; Breckpot, K.; Devoogdt, N. Non-invasive assessment of murine PD-L1 levels in syngeneic tumor models by nuclear imaging with nanobody tracers. Oncotarget 2017, 8, 41932–41946. [Google Scholar] [CrossRef] [Green Version]
- Broos, K.; Lecocq, Q.; Xavier, C.; Bridoux, J.; Nguyen, T.T.; Corthals, J.; Schoonooghe, S.; Lion, E.; Raes, G.; Keyaerts, M.; et al. Evaluating a Single Domain Antibody Targeting Human PD-L1 as a Nuclear Imaging and Therapeutic Agent. Cancers 2019, 11, 872. [Google Scholar] [CrossRef] [Green Version]
- Xing, Y.; Chand, G.; Liu, C.; Cook, G.J.R.; O’Doherty, J.; Zhao, L.; Wong, N.C.L.; Meszaros, L.K.; Ting, H.H.; Zhao, J. Early Phase I Study of a (99m)Tc-Labeled Anti-Programmed Death Ligand-1 (PD-L1) Single-Domain Antibody in SPECT/CT Assessment of PD-L1 Expression in Non-Small Cell Lung Cancer. J. Nucl. Med. 2019, 60, 1213–1220. [Google Scholar] [CrossRef] [Green Version]
- Bridoux, J.; Broos, K.; Lecocq, Q.; Debie, P.; Martin, C.; Ballet, S.; Raes, G.; Neyt, S.; Vanhove, C.; Breckpot, K.; et al. Anti-human PD-L1 Nanobody for Immuno-PET Imaging: Validation of a Conjugation Strategy for Clinical Translation. Biomolecules 2020, 10, 1388. [Google Scholar] [CrossRef]
- Ingram, J.R.; Blomberg, O.S.; Rashidian, M.; Ali, L.; Garforth, S.; Fedorov, E.; Fedorov, A.A.; Bonanno, J.B.; Le Gall, C.; Crowley, S.; et al. Anti-CTLA-4 therapy requires an Fc domain for efficacy. Proc. Natl. Acad. Sci. USA 2018, 115, 3912–3917. [Google Scholar] [CrossRef] [Green Version]
- Lecocq, Q.; Zeven, K.; De Vlaeminck, Y.; Martens, S.; Massa, S.; Goyvaerts, C.; Raes, G.; Keyaerts, M.; Breckpot, K.; Devoogdt, N. Noninvasive Imaging of the Immune Checkpoint LAG-3 Using Nanobodies, from Development to Pre-Clinical Use. Biomolecules 2019, 9, 548. [Google Scholar] [CrossRef] [Green Version]
- Datar, I.; Sanmamed, M.F.; Wang, J.; Henick, B.S.; Choi, J.; Badri, T.; Dong, W.; Mani, N.; Toki, M.; Mejias, L.D.; et al. Expression Analysis and Significance of PD-1, LAG-3, and TIM-3 in Human Non-Small Cell Lung Cancer Using Spatially Resolved and Multiparametric Single-Cell Analysis. Clin. Cancer Res. 2019, 25, 4663–4673. [Google Scholar] [CrossRef] [PubMed]
- Krasniqi, A.; D’Huyvetter, M.; Xavier, C.; Van der Jeught, K.; Muyldermans, S.; Van Der Heyden, J.; Lahoutte, T.; Tavernier, J.; Devoogdt, N. Theranostic Radiolabeled Anti-CD20 sdAb for Targeted Radionuclide Therapy of Non-Hodgkin Lymphoma. Mol. Cancer Ther. 2017, 16, 2828–2839. [Google Scholar] [CrossRef] [Green Version]
- Lemaire, M.; D’Huyvetter, M.; Lahoutte, T.; Van Valckenborgh, E.; Menu, E.; De Bruyne, E.; Kronenberger, P.; Wernery, U.; Muyldermans, S.; Devoogdt, N.; et al. Imaging and radioimmunotherapy of multiple myeloma with anti-idiotypic Nanobodies. Leukemia 2014, 28, 444–447. [Google Scholar] [CrossRef] [PubMed]
- D’Huyvetter, M.; De Vos, J.; Xavier, C.; Pruszynski, M.; Sterckx, Y.G.J.; Massa, S.; Raes, G.; Caveliers, V.; Zalutsky, M.R.; Lahoutte, T.; et al. (131)I-labeled Anti-HER2 Camelid sdAb as a Theranostic Tool in Cancer Treatment. Clin. Cancer Res. 2017, 23, 6616–6628. [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]
- Zhang, F.; Wei, H.; Wang, X.; Bai, Y.; Wang, P.; Wu, J.; Jiang, X.; Wang, Y.; Cai, H.; Xu, T.; et al. Structural basis of a novel PD-L1 nanobody for immune checkpoint blockade. Cell Discov. 2017, 3, 17004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dougan, M.; Ingram, J.R.; Jeong, H.J.; Mosaheb, M.M.; Bruck, P.T.; Ali, L.; Pishesha, N.; Blomberg, O.; Tyler, P.M.; Servos, M.M.; et al. Targeting Cytokine Therapy to the Pancreatic Tumor Microenvironment Using PD-L1-Specific VHHs. Cancer Immunol. Res. 2018, 6, 389–401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Broos, K.; Lecocq, Q.; Keersmaecker, B.; Raes, G.; Corthals, J.; Lion, E.; Thielemans, K.; Devoogdt, N.; Keyaerts, M.; Breckpot, K. Single Domain Antibody-Mediated Blockade of Programmed Death-Ligand 1 on Dendritic Cells Enhances CD8 T-cell Activation and Cytokine Production. Vaccines 2019, 7, 85. [Google Scholar] [CrossRef] [Green Version]
- Fang, T.; Li, R.; Li, Z.; Cho, J.; Guzman, J.S.; Kamm, R.D.; Ploegh, H.L. Remodeling of the Tumor Microenvironment by a Chemokine/Anti-PD-L1 Nanobody Fusion Protein. Mol. Pharm. 2019, 16, 2838–2844. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Cybulsky, M.I.; Gimbrone, M.A., Jr.; Libby, P. An atherogenic diet rapidly induces VCAM-1, a cytokine-regulatable mononuclear leukocyte adhesion molecule, in rabbit aortic endothelium. Arterioscler. Thromb. 1993, 13, 197–204. [Google Scholar] [CrossRef] [Green Version]
- Broisat, A.; Hernot, S.; Toczek, J.; De Vos, J.; Riou, L.M.; Martin, S.; Ahmadi, M.; Thielens, N.; Wernery, U.; Caveliers, V.; et al. Nanobodies targeting mouse/human VCAM1 for the nuclear imaging of atherosclerotic lesions. Circ. Res. 2012, 110, 927–937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Punjabi, M.; Xu, L.; Ochoa-Espinosa, A.; Kosareva, A.; Wolff, T.; Murtaja, A.; Broisat, A.; Devoogdt, N.; Kaufmann, B.A. Ultrasound Molecular Imaging of Atherosclerosis With Nanobodies: Translatable Microbubble Targeting Murine and Human VCAM (Vascular Cell Adhesion Molecule) 1. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 2520–2530. [Google Scholar] [CrossRef] [PubMed]
- Bridoux, J.; Neyt, S.; Debie, P.; Descamps, B.; Devoogdt, N.; Cleeren, F.; Bormans, G.; Broisat, A.; Caveliers, V.; Xavier, C.; et al. Improved Detection of Molecular Markers of Atherosclerotic Plaques Using Sub-Millimeter PET Imaging. Molecules 2020, 25, 1838. [Google Scholar] [CrossRef] [Green Version]
- Finn, A.V.; Nakano, M.; Polavarapu, R.; Karmali, V.; Saeed, O.; Zhao, X.; Yazdani, S.; Otsuka, F.; Davis, T.; Habib, A.; et al. Hemoglobin directs macrophage differentiation and prevents foam cell formation in human atherosclerotic plaques. J. Am. Coll Cardiol. 2012, 59, 166–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bala, G.; Baudhuin, H.; Remory, I.; Gillis, K.; Debie, P.; Krasniqi, A.; Lahoutte, T.; Raes, G.; Devoogdt, N.; Cosyns, B.; et al. Evaluation of [(99m)Tc]Radiolabeled Macrophage Mannose Receptor-Specific Nanobodies for Targeting of Atherosclerotic Lesions in Mice. Mol. Imaging. Biol. 2018, 20, 260–267. [Google Scholar] [CrossRef]
- Varasteh, Z.; Mohanta, S.; Li, Y.; Lopez Armbruster, N.; Braeuer, M.; Nekolla, S.G.; Habenicht, A.; Sager, H.B.; Raes, G.; Weber, W.; et al. Targeting mannose receptor expression on macrophages in atherosclerotic plaques of apolipoprotein E-knockout mice using (68)Ga-NOTA-anti-MMR nanobody: Non-invasive imaging of atherosclerotic plaques. EJNMMI Res. 2019, 9, 5. [Google Scholar] [CrossRef]
- Klareskog, L.; Malmstrom, V.; Lundberg, K.; Padyukov, L.; Alfredsson, L. Smoking, citrullination and genetic variability in the immunopathogenesis of rheumatoid arthritis. Semin. Immunol. 2011, 23, 92–98. [Google Scholar] [CrossRef]
- Put, S.; Schoonooghe, S.; Devoogdt, N.; Schurgers, E.; Avau, A.; Mitera, T.; D’Huyvetter, M.; De Baetselier, P.; Raes, G.; Lahoutte, T.; et al. SPECT imaging of joint inflammation with Nanobodies targeting the macrophage mannose receptor in a mouse model for rheumatoid arthritis. J. Nucl. Med. 2013, 54, 807–814. [Google Scholar] [CrossRef] [Green Version]
- Zheng, F.; Put, S.; Bouwens, L.; Lahoutte, T.; Matthys, P.; Muyldermans, S.; De Baetselier, P.; Devoogdt, N.; Raes, G.; Schoonooghe, S. Molecular imaging with macrophage CRIg-targeting nanobodies for early and preclinical diagnosis in a mouse model of rheumatoid arthritis. J. Nucl. Med. 2014, 55, 824–829. [Google Scholar] [CrossRef] [Green Version]
- Zheng, F.; Perlman, H.; Matthys, P.; Wen, Y.; Lahoutte, T.; Muyldermans, S.; Lu, S.; De Baetselier, P.; Schoonooghe, S.; Devoogdt, N.; et al. Specificity Evaluation and Disease Monitoring in Arthritis Imaging with Complement Receptor of the Ig superfamily targeting Nanobodies. Sci. Rep. 2016, 6, 35966. [Google Scholar] [CrossRef] [Green Version]
- Zheng, F.; Luo, S.; Ouyang, Z.; Zhou, J.; Mo, H.; Schoonooghe, S.; Muyldermans, S.; De Baetselier, P.; Raes, G.; Wen, Y. NIRF-Molecular Imaging with Synovial Macrophages-Targeting Vsig4 Nanobody for Disease Monitoring in a Mouse Model of Arthritis. Int. J. Mol. Sci. 2019, 20, 3347. [Google Scholar] [CrossRef] [Green Version]
- Zheng, F.; Devoogdt, N.; Sparkes, A.; Morias, Y.; Abels, C.; Stijlemans, B.; Lahoutte, T.; Muyldermans, S.; De Baetselier, P.; Schoonooghe, S.; et al. Monitoring liver macrophages using nanobodies targeting Vsig4: Concanavalin A induced acute hepatitis as paradigm. Immunobiology 2015, 220, 200–209. [Google Scholar] [CrossRef] [PubMed]
- Zheng, F.; Sparkes, A.; De Baetselier, P.; Schoonooghe, S.; Stijlemans, B.; Muyldermans, S.; Flamand, V.; Van Ginderachter, J.A.; Devoogdt, N.; Raes, G.; et al. Molecular Imaging with Kupffer Cell-Targeting Nanobodies for Diagnosis and Prognosis in Mouse Models of Liver Pathogenesis. Mol. Imaging. Biol. 2017, 19, 49–58. [Google Scholar] [CrossRef] [PubMed]
- Balhuizen, A.; Massa, S.; Mathijs, I.; Turatsinze, J.V.; De Vos, J.; Demine, S.; Xavier, C.; Villate, O.; Millard, I.; Egrise, D.; et al. A nanobody-based tracer targeting DPP6 for non-invasive imaging of human pancreatic endocrine cells. Sci. Rep. 2017, 7, 15130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Demine, S.; Garcia Ribeiro, R.; Thevenet, J.; Marselli, L.; Marchetti, P.; Pattou, F.; Kerr-Conte, J.; Devoogdt, N.; Eizirik, D.L. A nanobody-based nuclear imaging tracer targeting dipeptidyl peptidase 6 to determine the mass of human beta cell grafts in mice. Diabetologia 2020, 63, 825–836. [Google Scholar] [CrossRef] [PubMed]
- Abbott, N.J.; Patabendige, A.A.; Dolman, D.E.; Yusof, S.R.; Begley, D.J. Structure and function of the blood-brain barrier. Neurobiol. Dis. 2010, 37, 13–25. [Google Scholar] [CrossRef] [PubMed]
- Jones, A.R.; Shusta, E.V. Blood-brain barrier transport of therapeutics via receptor-mediation. Pharm. Res. 2007, 24, 1759–1771. [Google Scholar] [CrossRef] [Green Version]
- Iqbal, U.; Abulrob, A.; Stanimirovic, D.B. Integrated platform for brain imaging and drug delivery across the blood-brain barrier. Methods Mol. Biol. 2011, 686, 465–481. [Google Scholar] [CrossRef]
- Herve, F.; Ghinea, N.; Scherrmann, J.M. CNS delivery via adsorptive transcytosis. AAPS J. 2008, 10, 455–472. [Google Scholar] [CrossRef] [Green Version]
- Li, T.; Bourgeois, J.P.; Celli, S.; Glacial, F.; Le Sourd, A.M.; Mecheri, S.; Weksler, B.; Romero, I.; Couraud, P.O.; Rougeon, F.; et al. Cell-penetrating anti-GFAP VHH and corresponding fluorescent fusion protein VHH-GFP spontaneously cross the blood-brain barrier and specifically recognize astrocytes: Application to brain imaging. FASEB J. 2012, 26, 3969–3979. [Google Scholar] [CrossRef]
- Chen, G.F.; Xu, T.H.; Yan, Y.; Zhou, Y.R.; Jiang, Y.; Melcher, K.; Xu, H.E. Amyloid beta: Structure, biology and structure-based therapeutic development. Acta Pharmacol. Sin. 2017, 38, 1205–1235. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Vandesquille, M.; Koukouli, F.; Dudeffant, C.; Youssef, I.; Lenormand, P.; Ganneau, C.; Maskos, U.; Czech, C.; Grueninger, F.; et al. Camelid single-domain antibodies: A versatile tool for in vivo imaging of extracellular and intracellular brain targets. J. Control. Release 2016, 243, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Verhelle, A.; Van Overbeke, W.; Peleman, C.; De Smet, R.; Zwaenepoel, O.; Lahoutte, T.; Van Dorpe, J.; Devoogdt, N.; Gettemans, J. Non-Invasive Imaging of Amyloid Deposits in a Mouse Model of AGel Using (99m)Tc-Modified Nanobodies and SPECT/CT. Mol. Imaging. Biol. 2016, 18, 887–897. [Google Scholar] [CrossRef] [PubMed]
- Das, S.; Marsden, P.A. Angiogenesis in glioblastoma. N. Engl. J. Med. 2013, 369, 1561–1563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Iqbal, U.; Trojahn, U.; Albaghdadi, H.; Zhang, J.; O’Connor-McCourt, M.; Stanimirovic, D.; Tomanek, B.; Sutherland, G.; Abulrob, A. Kinetic analysis of novel mono- and multivalent VHH-fragments and their application for molecular imaging of brain tumours. Br. J. Pharmacol. 2010, 160, 1016–1028. [Google Scholar] [CrossRef]
- Olsson, A.K.; Dimberg, A.; Kreuger, J.; Claesson-Welsh, L. VEGF receptor signalling—In control of vascular function. Nat. Rev. Mol. Cell Biol. 2006, 7, 359–371. [Google Scholar] [CrossRef]
- Varner, J.A.; Cheresh, D.A. Integrins and cancer. Curr. Opin. Cell Biol. 1996, 8, 724–730. [Google Scholar] [CrossRef]
- Iqbal, U.; Albaghdadi, H.; Luo, Y.; Arbabi, M.; Desvaux, C.; Veres, T.; Stanimirovic, D.; Abulrob, A. Molecular imaging of glioblastoma multiforme using anti-insulin-like growth factor-binding protein-7 single-domain antibodies. Br. J. Cancer 2010, 103, 1606–1616. [Google Scholar] [CrossRef] [Green Version]
- Iqbal, U.; Albaghdadi, H.; Nieh, M.P.; Tuor, U.I.; Mester, Z.; Stanimirovic, D.; Katsaras, J.; Abulrob, A. Small unilamellar vesicles: A platform technology for molecular imaging of brain tumors. Nanotechnology 2011, 22, 195102. [Google Scholar] [CrossRef]
- Tomanek, B.; Iqbal, U.; Blasiak, B.; Abulrob, A.; Albaghdadi, H.; Matyas, J.R.; Ponjevic, D.; Sutherland, G.R. Evaluation of brain tumor vessels specific contrast agents for glioblastoma imaging. Neuro Oncol. 2012, 14, 53–63. [Google Scholar] [CrossRef] [Green Version]
- Debie, P.; Hernot, S. Emerging Fluorescent Molecular Tracers to Guide Intra-Operative Surgical Decision-Making. Front. Pharmacol. 2019, 10, 510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- van Dam, G.M.; Themelis, G.; Crane, L.M.; Harlaar, N.J.; Pleijhuis, R.G.; Kelder, W.; Sarantopoulos, A.; de Jong, J.S.; Arts, H.J.; van der Zee, A.G.; et al. Intraoperative tumor-specific fluorescence imaging in ovarian cancer by folate receptor-alpha targeting: First in-human results. Nat. Med. 2011, 17, 1315–1319. [Google Scholar] [CrossRef] [PubMed]
- Hong, G.; Antaris, A.L.; Dai, H. Near-infrared fluorophores for biomedical imaging. Nat. Biomed. Eng. 2017, 1. [Google Scholar] [CrossRef]
- Cilliers, C.; Nessler, I.; Christodolu, N.; Thurber, G.M. Tracking Antibody Distribution with Near-Infrared Fluorescent Dyes: Impact of Dye Structure and Degree of Labeling on Plasma Clearance. Mol. Pharm. 2017, 14, 1623–1633. [Google Scholar] [CrossRef] [PubMed]
- Morone, D.; Autilia, F.; Schorn, T.; Erreni, M.; Doni, A. Evaluation of cell metabolic adaptation in wound and tumour by Fluorescence Lifetime Imaging Microscopy. Sci. Rep. 2020, 10, 6289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Debie, P.; Van Quathem, J.; Hansen, I.; Bala, G.; Massa, S.; Devoogdt, N.; Xavier, C.; Hernot, S. Effect of Dye and Conjugation Chemistry on the Biodistribution Profile of Near-Infrared-Labeled Nanobodies as Tracers for Image-Guided Surgery. Mol. Pharm. 2017, 14, 1145–1153. [Google Scholar] [CrossRef]
- Bannas, P.; Lenz, A.; Kunick, V.; Well, L.; Fumey, W.; Rissiek, B.; Haag, F.; Schmid, J.; Schutze, K.; Eichhoff, A.; et al. Molecular imaging of tumors with nanobodies and antibodies: Timing and dosage are crucial factors for improved in vivo detection. Contrast Media Mol. Imaging 2015, 10, 367–378. [Google Scholar] [CrossRef] [Green Version]
- van Driel, P.B.; van der Vorst, J.R.; Verbeek, F.P.; Oliveira, S.; Snoeks, T.J.; Keereweer, S.; Chan, B.; Boonstra, M.C.; Frangioni, J.V.; van Bergen en Henegouwen, P.M.; et al. Intraoperative fluorescence delineation of head and neck cancer with a fluorescent anti-epidermal growth factor receptor nanobody. Int. J. Cancer 2014, 134, 2663–2673. [Google Scholar] [CrossRef] [Green Version]
- Van Brussel, A.S.; Adams, A.; Oliveira, S.; Dorresteijn, B.; El Khattabi, M.; Vermeulen, J.F.; van der Wall, E.; Mali, W.P.; Derksen, P.W.; van Diest, P.J.; et al. Hypoxia-Targeting Fluorescent Nanobodies for Optical Molecular Imaging of Pre-Invasive Breast Cancer. Mol. Imaging. Biol. 2016, 18, 535–544. [Google Scholar] [CrossRef] [Green Version]
- Kijanka, M.; Warnders, F.J.; El Khattabi, M.; Lub-de Hooge, M.; van Dam, G.M.; Ntziachristos, V.; de Vries, L.; Oliveira, S.; van Bergen En Henegouwen, P.M. Rapid optical imaging of human breast tumour xenografts using anti-HER2 VHHs site-directly conjugated to IRDye 800CW for image-guided surgery. Eur. J. Nucl. Med. Mol. Imaging 2013, 40, 1718–1729. [Google Scholar] [CrossRef]
- Debie, P.; Vanhoeij, M.; Poortmans, N.; Puttemans, J.; Gillis, K.; Devoogdt, N.; Lahoutte, T.; Hernot, S. Improved Debulking of Peritoneal Tumor Implants by Near-Infrared Fluorescent Nanobody Image Guidance in an Experimental Mouse Model. Mol. Imaging. Biol. 2018, 20, 361–367. [Google Scholar] [CrossRef] [PubMed]
- Hell, S.W.; Wichmann, J. Breaking the diffraction resolution limit by stimulated emission: Stimulated-emission-depletion fluorescence microscopy. Opt. Lett. 1994, 19, 780–782. [Google Scholar] [CrossRef] [PubMed]
- Gustafsson, M.G. Surpassing the lateral resolution limit by a factor of two using structured illumination microscopy. J. Microsc. 2000, 198, 82–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Betzig, E.; Patterson, G.H.; Sougrat, R.; Lindwasser, O.W.; Olenych, S.; Bonifacino, J.S.; Davidson, M.W.; Lippincott-Schwartz, J.; Hess, H.F. Imaging intracellular fluorescent proteins at nanometer resolution. Science 2006, 313, 1642–1645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rust, M.J.; Bates, M.; Zhuang, X. Sub-diffraction-limit imaging by stochastic optical reconstruction microscopy (STORM). Nat. Methods 2006, 3, 793–795. [Google Scholar] [CrossRef] [Green Version]
- Schnitzbauer, J.; Strauss, M.T.; Schlichthaerle, T.; Schueder, F.; Jungmann, R. Super-resolution microscopy with DNA-PAINT. Nat. Protoc. 2017, 12, 1198–1228. [Google Scholar] [CrossRef]
- Schermelleh, L.; Ferrand, A.; Huser, T.; Eggeling, C.; Sauer, M.; Biehlmaier, O.; Drummen, G.P.C. Super-resolution microscopy demystified. Nat. Cell Biol. 2019, 21, 72–84. [Google Scholar] [CrossRef]
- Traenkle, B.; Rothbauer, U. Under the Microscope: Single-Domain Antibodies for Live-Cell Imaging and Super-Resolution Microscopy. Front. Immunol 2017, 8, 1030. [Google Scholar] [CrossRef] [Green Version]
- Riedl, J.; Crevenna, A.H.; Kessenbrock, K.; Yu, J.H.; Neukirchen, D.; Bista, M.; Bradke, F.; Jenne, D.; Holak, T.A.; Werb, Z.; et al. Lifeact: A versatile marker to visualize F-actin. Nat. Methods 2008, 5, 605–607. [Google Scholar] [CrossRef]
- Gross, G.G.; Junge, J.A.; Mora, R.J.; Kwon, H.B.; Olson, C.A.; Takahashi, T.T.; Liman, E.R.; Ellis-Davies, G.C.; McGee, A.W.; Sabatini, B.L.; et al. Recombinant probes for visualizing endogenous synaptic proteins in living neurons. Neuron 2013, 78, 971–985. [Google Scholar] [CrossRef] [Green Version]
- Biocca, S.; Neuberger, M.S.; Cattaneo, A. Expression and targeting of intracellular antibodies in mammalian cells. EMBO J. 1990, 9, 101–108. [Google Scholar] [CrossRef] [PubMed]
- Freund, G.; Desplancq, D.; Stoessel, A.; Weinsanto, R.; Sibler, A.P.; Robin, G.; Martineau, P.; Didier, P.; Wagner, J.; Weiss, E. Generation of an intrabody-based reagent suitable for imaging endogenous proliferating cell nuclear antigen in living cancer cells. J. Mol. Recognit. 2014, 27, 549–558. [Google Scholar] [CrossRef] [PubMed]
- Helma, J.; Cardoso, M.C.; Muyldermans, S.; Leonhardt, H. Nanobodies and recombinant binders in cell biology. J. Cell Biol. 2015, 209, 633–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kirchhofer, A.; Helma, J.; Schmidthals, K.; Frauer, C.; Cui, S.; Karcher, A.; Pellis, M.; Muyldermans, S.; Casas-Delucchi, C.S.; Cardoso, M.C.; et al. Modulation of protein properties in living cells using nanobodies. Nat. Struct. Mol. Biol. 2010, 17, 133–138. [Google Scholar] [CrossRef]
- Caussinus, E.; Kanca, O.; Affolter, M. Fluorescent fusion protein knockout mediated by anti-GFP nanobody. Nat. Struct. Mol. Biol. 2011, 19, 117–121. [Google Scholar] [CrossRef]
- Herce, H.D.; Deng, W.; Helma, J.; Leonhardt, H.; Cardoso, M.C. Visualization and targeted disruption of protein interactions in living cells. Nat. Commun. 2013, 4, 2660. [Google Scholar] [CrossRef] [Green Version]
- Maier, J.; Traenkle, B.; Rothbauer, U. Real-time analysis of epithelial-mesenchymal transition using fluorescent single-domain antibodies. Sci. Rep. 2015, 5, 13402. [Google Scholar] [CrossRef] [Green Version]
- Maier, J.; Traenkle, B.; Rothbauer, U. Visualizing Epithelial-Mesenchymal Transition Using the Chromobody Technology. Cancer Res. 2016, 76, 5592–5596. [Google Scholar] [CrossRef] [Green Version]
- Burgess, A.; Lorca, T.; Castro, A. Quantitative live imaging of endogenous DNA replication in mammalian cells. PLoS ONE 2012, 7, e45726. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Y.; Araki, S.; Wu, J.; Teramoto, T.; Chang, Y.F.; Nakano, M.; Abdelfattah, A.S.; Fujiwara, M.; Ishihara, T.; Nagai, T.; et al. An expanded palette of genetically encoded Ca(2)(+) indicators. Science 2011, 333, 1888–1891. [Google Scholar] [CrossRef] [Green Version]
- Prole, D.L.; Taylor, C.W. A genetically encoded toolkit of functionalized nanobodies against fluorescent proteins for visualizing and manipulating intracellular signalling. BMC Biol. 2019, 17, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, J.C.; Drokhlyansky, E.; Etemad, B.; Rudolph, S.; Guo, B.; Wang, S.; Ellis, E.G.; Li, J.Z.; Cepko, C.L. Detection and manipulation of live antigen-expressing cells using conditionally stable nanobodies. Elife 2016, 5. [Google Scholar] [CrossRef] [PubMed]
- Roebroek, T.; Duwe, S.; Vandenberg, W.; Dedecker, P. Reduced Fluorescent Protein Switching Fatigue by Binding-Induced Emissive State Stabilization. Int. J. Mol. Sci. 2017, 18, 2015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de Beer, M.A.; Giepmans, B.N.G. Nanobody-Based Probes for Subcellular Protein Identification and Visualization. Front. Cell. Neurosci. 2020, 14, 3278. [Google Scholar] [CrossRef]
- Zhou, X.; Hao, R.; Chen, C.; Su, Z.; Zhao, L.; Luo, Z.; Xie, W. Rapid Delivery of Nanobodies/VHHs into Living Cells via Expressing In Vitro-Transcribed mRNA. Mol. Ther. Methods Clin. Dev. 2020, 17, 401–408. [Google Scholar] [CrossRef]
- Wegner, W.; Ilgen, P.; Gregor, C.; van Dort, J.; Mott, A.C.; Steffens, H.; Willig, K.I. In vivo mouse and live cell STED microscopy of neuronal actin plasticity using far-red emitting fluorescent proteins. Sci. Rep. 2017, 7. [Google Scholar] [CrossRef] [Green Version]
- Guizetti, J.; Schermelleh, L.; Mantler, J.; Maar, S.; Poser, I.; Leonhardt, H.; Muller-Reichert, T.; Gerlich, D.W. Cortical constriction during abscission involves helices of ESCRT-III-dependent filaments. Science 2011, 331, 1616–1620. [Google Scholar] [CrossRef] [Green Version]
- Ries, J.; Kaplan, C.; Platonova, E.; Eghlidi, H.; Ewers, H. A simple, versatile method for GFP-based super-resolution microscopy via nanobodies. Nat. Methods 2012, 9, 582–584. [Google Scholar] [CrossRef]
- Mikhaylova, M.; Cloin, B.M.; Finan, K.; van den Berg, R.; Teeuw, J.; Kijanka, M.M.; Sokolowski, M.; Katrukha, E.A.; Maidorn, M.; Opazo, F.; et al. Resolving bundled microtubules using anti-tubulin nanobodies. Nat. Commun. 2015, 6, 7933. [Google Scholar] [CrossRef]
- Platonova, E.; Winterflood, C.M.; Junemann, A.; Albrecht, D.; Faix, J.; Ewers, H. Single-molecule microscopy of molecules tagged with GFP or RFP derivatives in mammalian cells using nanobody binders. Methods 2015, 88, 89–97. [Google Scholar] [CrossRef]
- Sograte-Idrissi, S.; Oleksiievets, N.; Isbaner, S.; Eggert-Martinez, M.; Enderlein, J.; Tsukanov, R.; Opazo, F. Nanobody Detection of Standard Fluorescent Proteins Enables Multi-Target DNA-PAINT with High Resolution and Minimal Displacement Errors. Cells 2019, 8, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cramer, K.; Bolender, A.L.; Stockmar, I.; Jungmann, R.; Kasper, R.; Shin, J.Y. Visualization of Bacterial Protein Complexes Labeled with Fluorescent Proteins and Nanobody Binders for STED Microscopy. Int. J. Mol. Sci. 2019, 20, 3376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Virant, D.; Traenkle, B.; Maier, J.; Kaiser, P.D.; Bodenhofer, M.; Schmees, C.; Vojnovic, I.; Pisak-Lukats, B.; Endesfelder, U.; Rothbauer, U. A peptide tag-specific nanobody enables high-quality labeling for dSTORM imaging. Nat. Commun. 2018, 9, 930. [Google Scholar] [CrossRef] [PubMed]
- Gotzke, H.; Kilisch, M.; Martinez-Carranza, M.; Sograte-Idrissi, S.; Rajavel, A.; Schlichthaerle, T.; Engels, N.; Jungmann, R.; Stenmark, P.; Opazo, F.; et al. The ALFA-tag is a highly versatile tool for nanobody-based bioscience applications. Nat. Commun. 2019, 10, 4403. [Google Scholar] [CrossRef] [Green Version]
- Traenkle, B.; Segan, S.; Fagbadebo, F.O.; Kaiser, P.D.; Rothbauer, U. A novel epitope tagging system to visualize and monitor antigens in live cells with chromobodies. Sci. Rep. 2020, 10, 14267. [Google Scholar] [CrossRef] [PubMed]
- Gerdes, C.; Waal, N.; Offner, T.; Fornasiero, E.F.; Wender, N.; Verbarg, H.; Manzini, I.; Trenkwalder, C.; Mollenhauer, B.; Strohaker, T.; et al. A nanobody-based fluorescent reporter reveals human alpha-synuclein in the cell cytosol. Nat. Commun. 2020, 11, 2729. [Google Scholar] [CrossRef]
- Maidorn, M.; Olichon, A.; Rizzoli, S.O.; Opazo, F. Nanobodies reveal an extra-synaptic population of SNAP-25 and Syntaxin 1A in hippocampal neurons. MAbs 2019, 11, 305–321. [Google Scholar] [CrossRef] [Green Version]
- Bates, M.; Huang, B.; Dempsey, G.T.; Zhuang, X. Multicolor super-resolution imaging with photo-switchable fluorescent probes. Science 2007, 317, 1749–1753. [Google Scholar] [CrossRef] [Green Version]
- Pleiner, T.; Bates, M.; Gorlich, D. A toolbox of anti-mouse and anti-rabbit IgG secondary nanobodies. J. Cell Biol. 2018, 217, 1143–1154. [Google Scholar] [CrossRef] [Green Version]
- Sograte-Idrissi, S.; Schlichthaerle, T.; Duque-Afonso, C.J.; Alevra, M.; Strauss, S.; Moser, T.; Jungmann, R.; Rizzoli, S.O.; Opazo, F. Circumvention of common labelling artefacts using secondary nanobodies. Nanoscale 2020, 12, 10226–10239. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Erreni, M.; Schorn, T.; D’Autilia, F.; Doni, A. Nanobodies as Versatile Tool for Multiscale Imaging Modalities. Biomolecules 2020, 10, 1695. https://doi.org/10.3390/biom10121695
Erreni M, Schorn T, D’Autilia F, Doni A. Nanobodies as Versatile Tool for Multiscale Imaging Modalities. Biomolecules. 2020; 10(12):1695. https://doi.org/10.3390/biom10121695
Chicago/Turabian StyleErreni, Marco, Tilo Schorn, Francesca D’Autilia, and Andrea Doni. 2020. "Nanobodies as Versatile Tool for Multiscale Imaging Modalities" Biomolecules 10, no. 12: 1695. https://doi.org/10.3390/biom10121695