Therapeutic Potential of Intrabodies for Cancer Immunotherapy: Current Status and Future Directions
Abstract
:1. Introduction
2. Tumor-Associated Antigens
3. Intrabodies against Oncogenic Cell Surface Receptors
4. Intrabodies against Cytoplasmic or Nucleus Located TAAS
5. Neoantigens
Identification of Neoantigens for Adoptive T-Cell Therapy, Cancer Vaccines and Intrabodies
6. Intrabodies against Intracellular Neoantigens
7. Bringing Intrabodies into Cancer Patients: Delivery of Intrabodies with Nanoparticles or AAV
7.1. Nanoparticles Embedded with Intrabody mRNA
7.2. Delivery of Intrabody Genes by AAV
7.2.1. Direct Evolution
7.2.2. Rational Design
Transcriptional Targeting
Transductional Targeting
8. Conclusions
Funding
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Gerber, H.-P.; Sibener, L.V.; Lee, L.J.; Gee, M. Intracellular targets as source for cleaner targets for the treatment of solid tumors. Biochem. Pharmacol. 2019, 168, 275–284. [Google Scholar] [CrossRef] [PubMed]
- Biernacki, M.A.; Bleakley, M. Neoantigens in Hematologic Malignancies. Front. Immunol. 2020, 11, 121. [Google Scholar] [CrossRef] [PubMed]
- Garcia-Garijo, A.; Fajardo, C.A.; Gros, A. Determinants for Neoantigen Identification. Front. Immunol. 2019, 10, 1392. [Google Scholar] [CrossRef] [Green Version]
- Naran, K.; Nundalall, T.; Chetty, S.; Barth, S. Principles of Immunotherapy: Implications for Treatment Strategies in Cancer and Infectious Diseases. Front. Microbiol. 2018, 9, 3158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yarmarkovich, M.; Marshall, Q.F.; Warrington, J.M.; Premaratne, R.; Farrel, A.; Groff, D.; Li, W.; di Marco, M.; Runbeck, E.; Truong, H.; et al. Cross-HLA targeting of intracellular oncoproteins with peptide-centric CARs. Nature 2021, 599, 477–484. [Google Scholar] [CrossRef]
- Duan, Z.; Ho, M. T-Cell Receptor Mimic Antibodies for Cancer Immunotherapy. Mol. Cancer Ther. 2021, 20, 1533–1541. [Google Scholar] [CrossRef]
- Zhang, C.; Ötjengerdes, R.M.; Roewe, J.; Mejias-Estevez, R.; Marschall, A.L.J. Applying Antibodies Inside Cells: Principles and Recent Advances in Neurobiology, Virology and Oncology. BioDrugs 2020, 34, 435–462. [Google Scholar] [CrossRef] [Green Version]
- Marschall, A.L.; Dubel, S.; Boldicke, T. Specific in vivo knockdown of protein function by intrabodies. MAbs 2015, 7, 1010–1035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Akatsuka, Y. TCR-Like CAR-T Cells Targeting MHC-Bound Minor Histocompatibility Antigens. Front. Immunol. 2020, 11, 257. [Google Scholar] [CrossRef]
- Wei, F.; Cheng, X.-X.; Xue, J.Z.; Xue, S.-A. Emerging Strategies in TCR-Engineered T Cells. Front. Immunol. 2022, 13, 358. [Google Scholar] [CrossRef]
- Sterner, R.C.; Sterner, R.M. CAR-T cell therapy: Current limitations and potential strategies. Blood Cancer J. 2021, 11, 69. [Google Scholar] [CrossRef] [PubMed]
- Terlikowska, K.; Dobrzycka, B.; Terlikowski, S. Chimeric Antigen Receptor Design and Efficacy in Ovarian Cancer Treatment. Int. J. Mol. Sci. 2021, 22, 3495. [Google Scholar] [CrossRef] [PubMed]
- Rohaan, M.W.; Wilgenhof, S.; Haanen, J.B.A.G. Adoptive cellular therapies: The current landscape. Virchows Arch. 2018, 474, 449–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morotti, M.; Albukhari, A.; Alsaadi, A.; Artibani, M.; Brenton, J.D.; Curbishley, S.M.; Dong, T.; Dustin, M.L.; Hu, Z.; McGranahan, N.; et al. Promises and challenges of adoptive T-cell therapies for solid tumours. Br. J. Cancer 2021, 124, 1759–1776. [Google Scholar] [CrossRef] [PubMed]
- Guo, Y.; Lei, K.; Tang, L. Neoantigen Vaccine Delivery for Personalized Anticancer Immunotherapy. Front. Immunol. 2018, 9, 1499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Valdés-Tresanco, M.S.; Molina-Zapata, A.; Pose, A.G.; Moreno, E. Structural Insights into the Design of Synthetic Nanobody Libraries. Molecules 2022, 27, 2198. [Google Scholar] [CrossRef]
- English, H.; Hong, J.; Ho, M. Ancient species offers contemporary therapeutics: An update on shark VNAR single domain antibody sequences, phage libraries and potential clinical applications. Antib. Ther. 2020, 3, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Zimmermann, I.; Egloff, P.; Hutter, C.A.J.; Kuhn, B.T.; Bräuer, 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]
- Böldicke, T. Single domain antibodies for the knockdown of cytosolic and nuclear proteins. Protein Sci. 2017, 26, 925–945. [Google Scholar] [CrossRef] [Green Version]
- Verdon, D.J.; Jenkins, M.R. Identification and Targeting of Mutant Peptide Neoantigens in Cancer Immunotherapy. Cancers 2021, 13, 4245. [Google Scholar] [CrossRef]
- Peng, M.; Mo, Y.; Wang, Y.; Wu, P.; Zhang, Y.; Xiong, F.; Guo, C.; Wu, X.; Li, Y.; Li, X.; et al. Neoantigen vaccine: An emerging tumor immunotherapy. Mol. Cancer 2019, 18, 128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef] [PubMed]
- Marschall, A.L.; Dubel, S.; Boldicke, T. Recent Advances with ER Targeted Intrabodies. Adv. Exp. Med. Biol. 2016, 917, 77–93. [Google Scholar] [PubMed]
- Kang, E.A.G.; Hu, M.; Ren, H.; Wang, J.; Cheng, X.; Li, R.; Yuan, B.; Balan, Y.; Bai, Z.; Huang, H. VHH212 nanobody targeting the hypoxia-inducible factor 1α suppresses angiogenesis and potentiates gemcitabine therapy in pancreatic cancer in vivo. Cancer Biol. Med. 2021, 18, 772–787. [Google Scholar] [CrossRef] [PubMed]
- Merckaert, T.; Zwaenepoel, O.; Gevaert, K.; Gettemans, J. An AKT2-specific nanobody that targets the hydrophobic motif induces cell cycle arrest, autophagy and loss of focal adhesions in MDA-MB-231 cells. Biomed. Pharmacother. 2021, 133, 111055. [Google Scholar] [CrossRef] [PubMed]
- D’Agostino, S.; Mazzega, E.; Praček, K.; Piccinin, S.; Pivetta, F.; Armellin, M.; Fortuna, S.; Maestro, R.; de Marco, A. Interference of p53:Twist1 interaction through competing nanobodies. Int. J. Biol. Macromol. 2022, 194, 24–31. [Google Scholar] [CrossRef]
- De Groof, T.W.M.; Bergkamp, N.D.; Heukers, R.; Giap, T.; Bebelman, M.P.; Haas, R.G.-D.; Piersma, S.R.; Jimenez, C.R.; Garcia, K.C.; Ploegh, H.L.; et al. Selective targeting of ligand-dependent and -independent signaling by GPCR conformation-specific anti-US28 intrabodies. Nat. Commun. 2021, 12, 4357. [Google Scholar] [CrossRef]
- Tanaka, T.; Rabbitts, T.H. Intrabodies based on intracellular capture frameworks that bind the RAS protein with high af-finity and impair oncogenic transformation. EMBO J. 2003, 22, 1025–1035. [Google Scholar] [CrossRef] [Green Version]
- Tanaka, T.; Williams, R.L.; Rabbitts, T.H. Tumour prevention by a single antibody domain targeting the interaction of signal transduction proteins with RAS. EMBO J. 2007, 26, 3250–3259. [Google Scholar] [CrossRef] [Green Version]
- Cetin, M.; Evenson, W.E.; Gross, G.G.; Jalali-Yazdi, F.; Krieger, D.; Arnold, D.; Takahashi, T.T.; Roberts, R.W. RasIns: Genetically Encoded Intrabodies of Activated Ras Proteins. J. Mol. Biol. 2017, 429, 562–573. [Google Scholar] [CrossRef] [Green Version]
- Shin, S.-M.; Choi, D.-K.; Bae, J.; Kim, J.-S.; Park, S.-W.; Kim, Y.-S. Antibody targeting intracellular oncogenic Ras mutants exerts anti-tumour effects after systemic admin-istration. Nat. Commun. 2017, 8, 15090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shin, S.-M.; Kim, J.-S.; Park, S.-W.; Jun, S.-Y.; Kweon, H.-J.; Choi, D.-K.; Lee, D.; Cho, Y.B.; Kim, Y.-S. Direct targeting of oncogenic RAS mutants with a tumor-specific cytosol-penetrating antibody inhibits RAS mutant–driven tumor growth. Sci. Adv. 2020, 6, eaay2174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, J.-L.; Pan, X.-Y.; Zhao, W.-X.; Hu, Q.-C.; Ding, F.; Feng, Q.; Li, G.-Y.; Luo, Y. The antitumor efficacy of a novel adenovirus-mediated anti-p21Ras single chain fragment variable antibody on human cancers in vitro and in vivo. Int. J. Oncol. 2016, 48, 1218–1228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bery, N.; Legg, S.; Debreczeni, J.; Breed, J.; Embrey, K.; Stubbs, C.; Kolasinska-Zwierz, P.; Barrett, N.; Marwood, R.; Watson, J.; et al. KRAS-specific inhibition using a DARPin binding to a site in the allosteric lobe. Nat. Commun. 2019, 10, 2607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Somplatzki, S.; Mühlenhoff, M.; Kröger, A.; Gerardy-Schahn, R.; Böldicke, T. Intrabodies against the Polysialyltransferases ST8SiaII and ST8SiaIV inhibit Polysialylation of NCAM in rhabdomyosarcoma tumor cells. BMC Biotechnol. 2017, 17, 42. [Google Scholar] [CrossRef] [Green Version]
- Van Impe, K.; Bethuyne, J.; Cool, S.; Impens, F.; Ruano-Gallego, D.; De Wever, O.; Vanloo, B.; Van Troys, M.; Lambein, K.; Boucherie, C.; et al. A nanobody targeting the F-actin capping protein CapG restrains breast cancer metastasis. Breast Cancer Res. 2013, 15, R116. [Google Scholar] [CrossRef] [Green Version]
- Deshane, J.; Siegal, G.P.; Wang, M.; Wright, M.; Bucy, R.P.; Alvarez, R.D.; Curiel, D.T. Transductional efficacy and safety of an intraperitoneally delivered adenovirus encoding an anti-erbB-2 intracellular single-chain antibody for ovarian cancer gene therapy. Gynecol. Oncol. 1997, 64, 378–385. [Google Scholar] [CrossRef]
- Popkov, M.; Jendreyko, N.; McGavern, D.B.; Rader, C.; Barbas, C.F., 3rd. Targeting tumor angiogenesis with adenovi-rus-delivered anti-Tie-2 intrabody. Cancer Res. 2005, 65, 972–981. [Google Scholar] [CrossRef]
- Jendreyko, N.; Popkov, M.; Rader, C.; Barbas, C.F. Phenotypic knockout of VEGF-R2 and Tie-2 with an intradiabody reduces tumor growth and angiogenesis in vivo. Proc. Natl. Acad. Sci. USA 2005, 102, 8293–8298. [Google Scholar] [CrossRef] [Green Version]
- Chen, J.; Guo, H.; Jiang, H.; Namusamba, M.; Wang, C.; Lan, T.; Wang, T.; Wang, B. A BAP31 intrabody induces gastric cancer cell death by inhibiting p27(kip1) proteasome degradation. Int. J. Cancer 2019, 144, 2051–2062. [Google Scholar] [CrossRef]
- Zhang, W.; Shan, H.; Jiang, K.; Huang, W.; Li, S. A novel intracellular nanobody against HPV16 E6 oncoprotein. Clin. Immunol. 2021, 225, 108684. [Google Scholar] [CrossRef] [PubMed]
- Koo, M.Y.; Park, J.; Lim, J.M.; Joo, S.Y.; Shin, S.-P.; Shim, H.B.; Chung, J.; Kang, D.; Woo, H.A.; Rhee, S.G. Selective inhibition of the function of tyrosine-phosphorylated STAT3 with a phosphorylation site-specific intrabody. Proc. Natl. Acad. Sci. USA 2014, 111, 6269–6274. [Google Scholar] [CrossRef] [Green Version]
- Png, Y.T.; Vinanica, N.; Kamiya, T.; Shimasaki, N.; Coustan-Smith, E.; Campana, D. Blockade of CD7 expression in T cells for effective chimeric antigen receptor targeting of T-cell malignancies. Blood Adv. 2017, 1, 2348–2360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nidetz, N.F.; McGee, M.C.; Tse, L.V.; Li, C.; Cong, L.; Li, Y.; Huang, W. Adeno-associated viral vector-mediated immune responses: Understanding barriers to gene delivery. Pharmacol. Ther. 2020, 207, 107453. [Google Scholar] [CrossRef] [PubMed]
- Hacker, U.T.; Bentler, M.; Kaniowska, D.; Morgan, M.; Büning, H. Towards Clinical Implementation of Adeno-Associated Virus (AAV) Vectors for Cancer Gene Therapy: Current Status and Future Perspectives. Cancers 2020, 12, 1889. [Google Scholar] [CrossRef] [PubMed]
- Münch, R.C.; Muth, A.; Muik, A.; Friedel, T.; Schmatz, J.; Dreier, B.; Trkola, A.; Plückthun, A.; Büning, H.; Buchholz, C.J. Off-target-free gene delivery by affinity-purified receptor-targeted viral vectors. Nat. Commun. 2015, 6, 6246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Colón-Thillet, R.; Jerome, K.R.; Stone, D. Optimization of AAV vectors to target persistent viral reservoirs. Virol. J. 2021, 18, 85. [Google Scholar] [CrossRef]
- Xie, W.; Chen, B.; Wong, J. Evolution of the market for mRNA technology. Nat. Rev. Drug Discov. 2021, 20, 735–736. [Google Scholar] [CrossRef]
- Guevara, M.L.; Persano, F.; Persano, S. Advances in Lipid Nanoparticles for mRNA-Based Cancer Immunotherapy. Front. Chem. 2020, 8, 589959. [Google Scholar] [CrossRef] [PubMed]
- Sanz, L.; Álvarez-Vallina, L. Engineered mRNA and the Rise of Next-Generation Antibodies. Antibodies 2021, 10, 37. [Google Scholar] [CrossRef]
- Deal, C.; Carfi, A.; Plante, O. Advancements in mRNA Encoded Antibodies for Passive Immunotherapy. Vaccines 2021, 9, 108. [Google Scholar] [CrossRef] [PubMed]
- Sahin, U.; Muik, A.; Derhovanessian, E.; Vogler, I.; Kranz, L.M.; Vormehr, M.; Baum, A.; Pascal, K.; Quandt, J.; Maurus, D.; et al. COVID-19 vaccine BNT162b1 elicits human antibody and TH1 T-cell responses. Nature 2020, 586, 594–599. [Google Scholar] [CrossRef] [PubMed]
- Alibakhshi, A.; Kahaki, F.A.; Ahangarzadeh, S.; Yaghoobi, H.; Yarian, F.; Arezumand, R.; Ranjbari, J.; Mokhtarzadeh, A.; de la Guardia, M. Targeted cancer therapy through antibody fragments-decorated nanomedicines. J. Control. Release 2017, 268, 323–334. [Google Scholar] [CrossRef] [PubMed]
- Lang, F.; Schrörs, B.; Löwer, M.; Türeci, Ö.; Sahin, U. Identification of neoantigens for individualized therapeutic cancer vaccines. Nat. Rev. Drug Discov. 2022, 21, 261–282. [Google Scholar] [CrossRef] [PubMed]
- Kamta, J.; Chaar, M.; Ande, A.; Altomare, D.A.; Ait-Oudhia, S. Advancing Cancer Therapy with Present and Emerging Immuno-Oncology Approaches. Front. Oncol. 2017, 7, 64. [Google Scholar] [CrossRef] [PubMed]
- Jin, K.-T.; Chen, B.; Liu, Y.-Y.; Lan, H.U.-R.; Yan, J.-P. Monoclonal antibodies and chimeric antigen receptor (CAR) T cells in the treatment of colorectal cancer. Cancer Cell Int. 2021, 21, 83. [Google Scholar] [CrossRef]
- Liu, C.-C.; Yang, H.; Zhang, R.; Zhao, J.-J.; Hao, D.-J. Tumour-associated antigens and their anti-cancer applications. Eur. J. Cancer Care 2017, 26, e12446. [Google Scholar] [CrossRef]
- Hakomori, S.-I. Tumor-Associated Carbohydrate Antigens Defining Tumor Malignancy: Basis for Development of Anti-Cancer Vaccines; Springer: Berlin/Heidelberg, Germany, 2001; Volume 491, pp. 369–402. [Google Scholar]
- Asaadi, Y.; Jouneghani, F.F.; Janani, S.; Rahbarizadeh, F. A comprehensive comparison between camelid nanobodies and single chain variable fragments. Biomark. Res. 2021, 9, 87. [Google Scholar] [CrossRef]
- Böldicke, T.; Somplatzki, S.; Sergeev, G.; Mueller, P.P. Functional inhibition of transitory proteins by intrabody-mediated retention in the endoplasmatic reticulum. Methods 2012, 56, 338–350. [Google Scholar] [CrossRef]
- Boldicke, T. Blocking translocation of cell surface molecules from the ER to the cell surface by intracellular antibodies tar-geted to the ER. J. Cell Mol. Med. 2007, 11, 54–70. [Google Scholar] [CrossRef]
- Kirschning, C.J.; Dreher, S.; Maaß, B.; Fichte, S.; Schade, J.; Köster, M.; Noack, A.; Lindenmaier, W.; Wagner, H.; Böldicke, T. Generation of anti-TLR2 intrabody mediating inhibition of macrophage surface TLR2 expression and TLR2-driven cell activation. BMC Biotechnol. 2010, 10, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Böldicke, T.; Tesar, M.; Griesel, C.; Rohde, M.; Gröne, H.J.; Waltenberger, J.; Kollet, O.; Lapidot, T.; Yayon, A.; Weich, H. Anti-VEGFR-2 scFvs for cell isolation. Single-chain antibodies recognizing the human vascular endothelial growth factor receptor-2 (VEGFR-2/flk-1) on the surface of primary endothelial cells and preselected CD34+ cells from cord blood. Stem Cells 2001, 19, 24–36. [Google Scholar] [CrossRef]
- Paolini, F.; Amici, C.; Carosi, M.; Bonomo, C.; Di Bonito, P.; Venuti, A.; Accardi, L. Intrabodies targeting human papillomavirus 16 E6 and E7 oncoproteins for therapy of established HPV-associated tumors. J. Exp. Clin. Cancer Res. 2021, 40, 37. [Google Scholar] [CrossRef] [PubMed]
- Grimmig, T.; Moench, R.; Kreckel, J.; Haack, S.; Rueckert, F.; Rehder, R.; Tripathi, S.; Ribas, C.; Chandraker, A.; Germer, C.T.; et al. Toll Like Receptor 2, 4, and 9 Signaling Promotes Autoregulative Tumor Cell Growth and VEGF/PDGF Expression in Human Pancreatic Cancer. Int. J. Mol. Sci. 2016, 17, 2060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Al-Saraireh, Y.M.J.; Sutherland, M.; Springett, B.R.; Freiberger, F.; Morais, G.R.; Loadman, P.M.; Errington, R.J.; Smith, P.J.; Fukuda, M.; Gerardy-Schahn, R.; et al. Pharmacological Inhibition of polysialyltransferase ST8SiaII Modulates Tumour Cell Migration. PLoS ONE 2013, 8, e73366. [Google Scholar] [CrossRef] [Green Version]
- Reimer, E.; Somplatzki, S.; Zegenhagen, D.; Hänel, S.; Fels, A.; Bollhorst, T.; Hovest, L.G.; Bauer, S.; Kirschning, C.J.; Böldicke, T. Molecular cloning and characterization of a novel anti-TLR9 intrabody. Cell. Mol. Biol. Lett. 2013, 18, 433–446. [Google Scholar] [CrossRef] [Green Version]
- Shah, V.; Sheppard, B.; Sears, R.; Alani, A.W. Hypoxia: Friend or Foe for drug delivery in Pancreatic Cancer. Cancer Lett. 2020, 492, 63–70. [Google Scholar] [CrossRef]
- Manning, B.D.; Toker, A. AKT/PKB Signaling: Navigating the Network. Cell 2017, 169, 381–405. [Google Scholar] [CrossRef] [Green Version]
- Brown, A.L.; Li, M.; Goncearenco, A.; Panchenko, A.R. Finding driver mutations in cancer: Elucidating the role of back-ground mutational processes. PLoS Comput. Biol. 2019, 15, e1006981. [Google Scholar] [CrossRef] [Green Version]
- Alspach, E.; Lussier, D.M.; Miceli, A.P.; Kizhvatov, I.; DuPage, M.; Luoma, A.M.; Meng, W.; Lichti, C.F.; Esaulova, E.; Vomund, A.N.; et al. MHC-II neoantigens shape tumour immunity and response to immunotherapy. Nature 2019, 574, 696–701. [Google Scholar] [CrossRef]
- van Dongen, J.J.; Macintyre, E.A.; Gabert, J.A.; Delabesse, E.; Rossi, V.; Saglio, G.; Gottardi, E.; Rambaldi, A.; Dotti, G.; Griesinger, F.; et al. Standardized RT-PCR analysis of fusion gene transcripts from chromosome aberrations in acute leukemia for detection of minimal residual disease. Report of the BIOMED-1 Concerted Action: Investigation of minimal residual disease in acute leukemia. Leukemia 1999, 13, 1901–1928. [Google Scholar] [CrossRef] [PubMed]
- Falini, B.; Martelli, M.P.; Bolli, N.; Sportoletti, P.; Liso, A.; Tiacci, E.; Haferlach, T. Acute myeloid leukemia with mutated nucleophosmin (NPM1): Is it a distinct entity? Blood 2011, 117, 1109–1120. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.; Yin, Q.; Huang, H.; Lu, J.; Qin, H.; Chen, S.; Zhang, W.; Su, X.; Sun, W.; Dong, Y.; et al. Personal Neoantigens From Patients With NSCLC Induce Efficient Antitumor Responses. Front. Oncol. 2021, 11, 628456. [Google Scholar] [CrossRef] [PubMed]
- Bykov, V.J.N.; Eriksson, S.E.; Bianchi, J.; Wiman, K.G. Targeting mutant p53 for efficient cancer therapy. Nat. Rev. Cancer 2018, 18, 89–102. [Google Scholar] [CrossRef]
- Hobbs, A.; Der, C.J.; Rossman, K.L. RAS isoforms and mutations in cancer at a glance. J. Cell Sci. 2016, 129, 1287–1292. [Google Scholar] [CrossRef] [Green Version]
- Forbes, S.A.; Beare, D.; Gunasekaran, P.; Leung, K.; Bindal, N.; Boutselakis, H.; Ding, M.; Bamford, S.; Cole, C.; Ward, S.; et al. COSMIC: Exploring the world’s knowledge of somatic mutations in human cancer. Nucleic. Acids Res. 2015, 43, D805–D811. [Google Scholar] [CrossRef]
- Hundal, J.; Kiwala, S.; McMichael, J.; Miller, C.A.; Xia, H.; Wollam, A.T.; Liu, C.J.; Zhao, S.; Feng, Y.-Y.; Graubert, A.P.; et al. pVACtools: A Computational Toolkit to Identify and Visualize Cancer Neoantigens. Cancer Immunol. Res. 2020, 8, 409–420. [Google Scholar] [CrossRef] [Green Version]
- Richters, M.M.; Xia, H.; Campbell, K.M.; Gillanders, W.E.; Griffith, O.L.; Griffith, M. Best practices for bioinformatic characterization of neoantigens for clinical utility. Genome Med. 2019, 11, 56. [Google Scholar] [CrossRef]
- Boegel, S.; Castle, J.C.; Kodysh, J.; O’Donnell, T.; Rubinsteyn, A. Bioinformatic methods for cancer neoantigen prediction. Prog. Mol. Biol. Transl. Sci. 2019, 164, 25–60. [Google Scholar]
- Gros, A.; Parkhurst, M.R.; Tran, E.; Pasetto, A.; Robbins, P.F.; Ilyas, S.; Prickett, T.D.; Gartner, J.J.; Crystal, J.S.; Roberts, I.M.; et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat. Med. 2016, 22, 433–438. [Google Scholar] [CrossRef]
- Kashima, D.; Kageoka, M.; Kimura, Y.; Horikawa, M.; Miura, M.; Nakakido, M.; Tsumoto, K.; Nagamune, T.; Kawahara, M. A Novel Cell-Based Intracellular Protein-Protein Interaction Detection Platform (SOLIS) for Multimo-dality Screening. ACS Synth. Biol. 2021, 10, 990–999. [Google Scholar] [CrossRef]
- Milburn, M.V.; Tong, L.; Devos, A.M.; Brünger, A.; Yamaizumi, Z.; Nishimura, S.; Kim, S.-H. Molecular Switch for Signal Transduction: Structural Differences Between Active and Inactive Forms of Protooncogenic ras Proteins. Science 1990, 247, 939–945. [Google Scholar] [CrossRef] [Green Version]
- Prior, I.A.; Lewis, P.D.; Mattos, C. A Comprehensive Survey of Ras Mutations in Cancer. Cancer Res. 2012, 72, 2457–2467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Visintin, M.; Quondam, M.; Cattaneo, A. The intracellular antibody capture technology: Towards the high-throughput se-lection of functional intracellular antibodies for target validation. Methods 2004, 34, 200–214. [Google Scholar] [CrossRef] [PubMed]
- Choi, D.-K.; Bae, J.; Shin, S.-M.; Shin, J.-Y.; Kim, S.; Kim, Y.-S. A general strategy for generating intact, full-length IgG antibodies that penetrate into the cytosol of living cells. mAbs 2014, 6, 1402–1414. [Google Scholar] [CrossRef] [Green Version]
- Huang, C.C.; Liu, F.R.; Feng, Q.; Pan, X.Y.; Song, S.L.; Yang, J.L. RGD4C peptide mediates anti-p21Ras scFv entry into tumor cells and produces an inhibitory effect on the human colon cancer cell line SW480. BMC Cancer 2021, 21, 321. [Google Scholar]
- Kawe, M.; Forrer, P.; Amstutz, P.; Plückthun, A. Isolation of Intracellular Proteinase Inhibitors Derived from Designed Ankyrin Repeat Proteins by Genetic Screening. J. Biol. Chem. 2006, 281, 40252–40263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Burns, T.F.; Borghaei, H.; Ramalingam, S.S.; Mok, T.S.; Peters, S. Targeting KRAS-Mutant Non-Small-Cell Lung Cancer: One Mutation at a Time, with a Focus on KRAS G12C Mutations. J. Clin. Oncol. 2020, 38, 4208–4218. [Google Scholar] [CrossRef] [PubMed]
- Tang, D.; Kroemer, G.; Kang, R. Oncogenic KRAS blockade therapy: Renewed enthusiasm and persistent challenges. Mol. Cancer 2021, 20, 128. [Google Scholar] [CrossRef]
- Jamal-Hanjani, M.; Thanopoulou, E.; Peggs, K.S.; Quezada, S.A.; Swanton, C. Tumour heterogeneity and im-mune-modulation. Curr. Opin. Pharmacol. 2013, 13, 497–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chan, T.; Yarchoan, M.; Jaffee, E.; Swanton, C.; Quezada, S.; Stenzinger, A.; Peters, S. Development of tumor mutation burden as an immunotherapy biomarker: Utility for the oncology clinic. Ann. Oncol. 2019, 30, 44–56. [Google Scholar] [CrossRef] [PubMed]
- McGranahan, N.; Furness, A.J.S.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463–1469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Samstein, R.M.; Lee, C.-H.; Shoushtari, A.N.; Hellmann, M.D.; Shen, R.; Janjigian, Y.Y.; Barron, D.A.; Zehir, A.; Jordan, E.J.; Omuro, A.; et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 2019, 51, 202–206. [Google Scholar] [CrossRef] [PubMed]
- Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124. [Google Scholar] [CrossRef] [PubMed]
- Miao, L.; Zhang, Y.; Huang, L. mRNA vaccine for cancer immunotherapy. Mol. Cancer 2021, 20, 41. [Google Scholar] [CrossRef]
- Tenchov, R.; Bird, R.; Curtze, A.E.; Zhou, Q. Lipid Nanoparticles—From Liposomes to mRNA Vaccine Delivery, a Landscape of Research Diversity and Advancement. ACS Nano 2021, 15, 16982–17015. [Google Scholar] [CrossRef]
- Slastnikova, T.A.; Ulasov, A.V.; Rosenkranz, A.A.; Sobolev, A.S. Targeted Intracellular Delivery of Antibodies: The State of the Art. Front. Pharmacol. 2018, 9, 1208. [Google Scholar] [CrossRef] [Green Version]
- Collén, A.; Bergenhem, N.; Carlsson, L.; Chien, K.R.; Hoge, S.; Gan, L.-M.; Fritsche-Danielson, R. VEGFA mRNA for regenerative treatment of heart failure. Nat. Rev. Drug Discov. 2022, 21, 79–80. [Google Scholar] [CrossRef]
- Stadler, C.R.; Bähr-Mahmud, H.; Celik, L.; Hebich, B.; Roth, A.S.; Roth, R.P.; Karikó, K.; Türeci, Ö.; Sahin, U. Elimination of large tumors in mice by mRNA-encoded bispecific antibodies. Nat. Med. 2017, 23, 815–817. [Google Scholar] [CrossRef]
- August, A.; Attarwala, H.Z.; Himansu, S.; Kalidindi, S.; Lu, S.; Pajon, R.; Han, S.; Lecerf, J.-M.; Tomassini, J.E.; Hard, M.; et al. A phase 1 trial of lipid-encapsulated mRNA encoding a monoclonal antibody with neutralizing activity against Chikungunya virus. Nat. Med. 2021, 27, 2224–2233. [Google Scholar] [CrossRef]
- Oh, S.; Kessler, J.A. Design, Assembly, Production, and Transfection of Synthetic Modified mRNA. Methods 2018, 133, 29–43. [Google Scholar] [CrossRef] [PubMed]
- Ramanathan, A.; Robb, G.B.; Chan, S.-H. mRNA capping: Biological functions and applications. Nucleic Acids Res. 2016, 44, 7511–7526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Holtkamp, S.; Kreiter, S.; Selmi, A.; Simon, P.; Koslowski, M.; Huber, C.; Türeci, O.; Sahin, U. Modification of antigen-encoding RNA increases stability, translational efficacy, and T-cell stimulatory capacity of dendritic cells. Blood 2006, 108, 4009–4017. [Google Scholar] [CrossRef] [PubMed]
- Guhaniyogi, J.; Brewer, G. Regulation of mRNA stability in mammalian cells. Gene 2001, 265, 11–23. [Google Scholar] [CrossRef] [Green Version]
- Mauger, D.M.; Cabral, B.J.; Presnyak, V.; Su, S.V.; Reid, D.W.; Goodman, B.; Link, K.; Khatwani, N.; Reynders, J.; Moore, M.J.; et al. mRNA structure regulates protein expression through changes in functional half-life. Proc. Natl. Acad. Sci. USA 2019, 116, 24075–24083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Triana-Alonso, F.J.; Dabrowski, M.; Wadzack, J.; Nierhaus, K.H. Self-coded 3′-Extension of Run-off Transcripts Produces Aberrant Products during in Vitro Transcription with T7 RNA Polymerase. J. Biol. Chem. 1995, 270, 6298–6307. [Google Scholar] [CrossRef] [Green Version]
- Gómez-Aguado, I.; Rodríguez-Castejón, J.; Beraza-Millor, M.; Vicente-Pascual, M.; Rodríguez-Gascón, A.; Garelli, S.; Battaglia, L.; Del Pozo-Rodríguez, A.; Solinís, M.Á. mRNA-Based Nanomedicinal Products to Address Corneal Inflammation by Interleukin-10 Sup-plementation. Pharmaceutics 2021, 13, 1472. [Google Scholar] [CrossRef]
- Kadkhoda, J.; Akrami-Hasan-Kohal, M.; Tohidkia, M.R.; Khaledi, S.; Davaran, S.; Aghanejad, A. Advances in antibody nanoconjugates for diagnosis and therapy: A review of recent studies and trends. Int. J. Biol. Macromol. 2021, 185, 664–678. [Google Scholar] [CrossRef]
- Juan, A.; Cimas, F.J.; Bravo, I.; Pandiella, A.; Ocaña, A.; Alonso-Moreno, C. An Overview of Antibody Conjugated Polymeric Nanoparticles for Breast Cancer Therapy. Pharmaceutics 2020, 12, 802. [Google Scholar] [CrossRef]
- Sofou, S.; Sgouros, G. Antibody-targeted liposomes in cancer therapy and imaging. Expert Opin. Drug Deliv. 2008, 5, 189–204. [Google Scholar] [CrossRef]
- van der Meel, R.; Oliveira, S.; Altintas, I.; Haselberg, R.; van der Veeken, J.; Roovers, R.C.; Henegouwen PM PV, B.E.; Storm, G.; Hennink, W.E.; Schiffelers, R.M.; et al. Tumor-targeted Nanobullets: Anti-EGFR nanobody-liposomes loaded with anti-IGF-1R kinase in-hibitor for cancer treatment. J. Control. Release 2012, 159, 281–289. [Google Scholar] [CrossRef] [PubMed]
- Zou, T.; Dembele, F.; Beugnet, A.; Sengmanivong, L.; Trepout, S.; Marco, S.; de Marco, A.; Li, M.-H. Nanobody-functionalized PEG-b-PCL polymersomes and their targeting study. J. Biotechnol. 2015, 214, 147–155. [Google Scholar] [CrossRef] [PubMed]
- Moffett, H.F.; Coon, M.E.; Radtke, S.; Stephan, S.B.; McKnight, L.; Lambert, A.; Stoddard, B.L.; Kiem, H.P.; Stephan, M.T. Hit-and-run programming of therapeutic cytoreagents using mRNA nanocarriers. Nat. Commun. 2017, 8, 389. [Google Scholar] [CrossRef] [PubMed]
- Mendell, J.R.; Al-Zaidy, S.A.; Rodino-Klapac, L.R.; Goodspeed, K.; Gray, S.J.; Kay, C.N.; Boye, S.L.; Boye, S.E.; George, L.A.; Salabarria, S.; et al. Current Clinical Applications of In Vivo Gene Therapy with AAVs. Mol. Ther. 2021, 29, 464–488. [Google Scholar] [CrossRef] [PubMed]
- Kuzmin, D.A.; Shutova, M.V.; Johnston, N.R.; Smith, O.P.; Fedorin, V.V.; Kukushkin, Y.S.; van der Loo, J.C.; Johnstone, E.C. The clinical landscape for AAV gene therapies. Nat. Rev. Drug Discov. 2021, 20, 173–174. [Google Scholar] [CrossRef] [PubMed]
- Ogden, P.J.; Kelsic, E.D.; Sinai, S.; Church, G.M. Comprehensive AAV capsid fitness landscape reveals a viral gene and enables machine-guided design. Science 2019, 366, 1139–1143. [Google Scholar] [CrossRef]
- Börner, K.; Kienle, E.; Huang, L.-Y.; Weinmann, J.; Sacher, A.; Bayer, P.; Stüllein, C.; Fakhiri, J.; Zimmermann, L.; Westhaus, A.; et al. Pre-arrayed Pan-AAV Peptide Display Libraries for Rapid Single-Round Screening. Mol. Ther. 2020, 28, 1016–1032. [Google Scholar] [CrossRef]
- Büning, H.; Srivastava, A. Capsid Modifications for Targeting and Improving the Efficacy of AAV Vectors. Mol. Ther. Methods Clin. Dev. 2019, 12, 248–265. [Google Scholar] [CrossRef]
- Michelfelder, S.; Varadi, K.; Raupp, C.; Hunger, A.; Körbelin, J.; Pahrmann, C.; Schrepfer, S.; Müller, O.J.; Kleinschmidt, J.A.; Trepel, M. Peptide Ligands Incorporated into the Threefold Spike Capsid Domain to Re-Direct Gene Transduction of AAV8 and AAV9 In Vivo. PLoS ONE 2011, 6, e23101. [Google Scholar] [CrossRef] [Green Version]
- Huttner, N.A.; Girod, A.; Perabo, L.; Edbauer, D.; Kleinschmidt, J.A.; Büning, H.; Hallek, M. Genetic modifications of the adeno-associated virus type 2 capsid reduce the affinity and the neu-tralizing effects of human serum antibodies. Gene Ther. 2003, 10, 2139–2147. [Google Scholar] [CrossRef] [Green Version]
- Perabo, L.; Büning, H.; Kofler, D.M.; Ried, M.U.; Girod, A.; Wendtner, C.M.; Enssle, J.; Hallek, M. In vitro selection of viral vectors with modified tropism: The adeno-associated virus display. Mol. Ther. 2003, 8, 151–157. [Google Scholar] [CrossRef]
- Michelfelder, S.; Kohlschütter, J.; Skorupa, A.; Pfennings, S.; Müller, O.; Kleinschmidt, J.A.; Trepel, M. Successful Expansion but Not Complete Restriction of Tropism of Adeno-Associated Virus by In Vivo Biopanning of Random Virus Display Peptide Libraries. PLoS ONE 2009, 4, e5122. [Google Scholar] [CrossRef] [PubMed]
- Geisler, A.; Fechner, H. MicroRNA-regulated viral vectors for gene therapy. World J. Exp. Med. 2016, 6, 37–54. [Google Scholar] [CrossRef] [PubMed]
- Shamsabadi, F.; Eidgahi, M.R.A.; Mehrbod, P.; Daneshvar, N.; Allaudin, Z.N.; Yamchi, A.; Shahbazi, M. Survivin, a Promising Gene for Targeted Cancer Treatment. Asian Pac. J. Cancer Prev. 2016, 17, 3711–3719. [Google Scholar] [PubMed]
- Yuan, X.; Larsson, C.; Xu, D. Mechanisms underlying the activation of TERT transcription and telomerase activity in human cancer: Old actors and new players. Oncogene 2019, 38, 6172–6183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Javan, B.; Shahbazi, M. Hypoxia-inducible tumour-specific promoters as a dual-targeting transcriptional regulation system for cancer gene therapy. Ecancermedicalscience 2017, 11, 751. [Google Scholar] [CrossRef]
- Li, C.-F.; Chen, L.-B.; Li, D.-D.; Yang, L.; Zhang, B.-G.; Jin, J.-P.; Zhang, Y.; Zhang, B. Dual-sensitive HRE/Egr1 promoter regulates Smac overexpression and enhances radiation-induced A549 human lung adenocarcinoma cell death under hypoxia. Mol. Med. Rep. 2014, 10, 1108–1116. [Google Scholar] [CrossRef]
- Ho, S.-Y.; Chang, B.-H.; Chung, C.-H.; Lin, Y.-L.; Chuang, C.-H.; Hsieh, P.-J.; Huang, W.-C.; Tsai, N.-M.; Huang, S.-C.; Liu, Y.-K.; et al. Development of a computational promoter with highly efficient expression in tumors. BMC Cancer 2018, 18, 480. [Google Scholar] [CrossRef]
- Dai, W.; Wu, J.; Wang, D.; Wang, J. Cancer gene therapy by NF-kappaB-activated cancer cell-specific expression of CRISPR/Cas9 targeting telomeres. Gene Ther. 2020, 27, 266–280. [Google Scholar] [CrossRef]
- Kuklik, J.; Michelfelder, S.; Schiele, F.; Kreuz, S.; Lamla, T.; Müller, P.; Park, J. Development of a Bispecific Antibody-Based Platform for Retargeting of Capsid Modified AAV Vectors. Int. J. Mol. Sci. 2021, 22, 8355. [Google Scholar] [CrossRef]
- Ponnazhagan, S.; Mahendra, G.; Kumar, S.; Thompson, J.A.; Castillas, J.M. Conjugate-Based Targeting of Recombinant Adeno-Associated Virus Type 2 Vectors by Using Avidin-Linked Ligands. J. Virol. 2002, 76, 12900–12907. [Google Scholar] [CrossRef] [Green Version]
- Eichhoff, A.M.; Börner, K.; Albrecht, B.; Schäfer, W.; Baum, N.; Haag, F.; Körbelin, J.; Trepel, M.; Braren, I.; Grimm, D.; et al. Nanobody-Enhanced Targeting of AAV Gene Therapy Vectors. Mol. Ther. Methods Clin. Dev. 2019, 15, 211–220. [Google Scholar] [CrossRef] [Green Version]
- Hamann, M.V.; Beschorner, N.; Vu, X.-K.; Hauber, I.; Lange, U.C.; Traenkle, B.; Kaiser, P.D.; Foth, D.; Schneider, C.; Büning, H.; et al. Improved targeting of human CD4+ T cells by nanobody-modified AAV2 gene therapy vectors. PLoS ONE 2021, 16, e0261269. [Google Scholar] [CrossRef] [PubMed]
- Shi, W.; Bartlett, J.S. RGD inclusion in VP3 provides adeno-associated virus type 2 (AAV2)-based vectors with a heparan sulfate-independent cell entry mechanism. Mol. Ther. 2003, 7, 515–525. [Google Scholar] [CrossRef]
- Stachler, M.D.; Bartlett, J.S. Mosaic vectors comprised of modified AAV1 capsid proteins for efficient vector purification and targeting to vascular endothelial cells. Gene Ther. 2006, 13, 926–931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, C.; Narkbunnam, N.; Samulski, R.J.; Asokan, A.; Hu, G.; Jacobson, L.J.; Manco-Johnson, M.J.; Monahan, P.E.; Joint Outcome Study Investigators. Neutralizing antibodies against adeno-associated virus examined prospectively in pediatric patients with he-mophilia. Gene Ther. 2012, 19, 288–294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Colella, P.; Ronzitti, G.; Mingozzi, F. Emerging Issues in AAV-Mediated In Vivo Gene Therapy. Mol. Ther. Methods Clin. Dev. 2018, 8, 87–104. [Google Scholar] [CrossRef] [Green Version]
- Meliani, A.; Boisgerault, F.; Fitzpatrick, Z.; Marmier, S.; Leborgne, C.; Collaud, F.; Sola, M.S.; Charles, S.; Ronzitti, G.; Vignaud, A.; et al. Enhanced liver gene transfer and evasion of preexisting humoral immunity with exosome-enveloped AAV vectors. Blood Adv. 2017, 1, 2019–2031. [Google Scholar] [CrossRef] [Green Version]
- Liu, Q.; Zhang, X.; Zhang, J. Exosome-Based Nanoplatforms: The Emerging Tools for Breast Cancer Therapy. Front. Oncol. 2022, 12, 1835. [Google Scholar] [CrossRef]
- Kamerkar, S.; LeBleu, V.S.; Sugimoto, H.; Yang, S.; Ruivo, C.; Melo, S.; Lee, J.J.; Kalluri, R. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature 2017, 546, 498–503. [Google Scholar] [CrossRef] [PubMed]
- Barker, S.D.; Dmitriev, I.P.; Nettelbeck, D.M.; Liu, B.; Rivera, A.A.; Alvarez, R.D.; Curiel, D.T.; Hemminki, A. Combined transcriptional and transductional targeting improves the specificity and efficacy of adenoviral gene delivery to ovarian carcinoma. Gene Ther. 2003, 10, 1198–1204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wei, S.C.; Duffy, C.R.; Allison, J.P. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018, 8, 1069–1086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koerner, J.; Horvath, D.; Herrmann, V.L.; MacKerracher, A.; Gander, B.; Yagita, H.; Rohayem, J.; Groettrup, M. PLGA-particle vaccine carrying TLR3/RIG-I ligand Riboxxim synergizes with immune checkpoint blockade for effective anti-cancer immunotherapy. Nat. Commun. 2021, 12, 2935. [Google Scholar] [CrossRef]
- Bartoszewski, R.; Sikorski, A.F. Editorial focus: Understanding off-target effects as the key to successful RNAi therapy. Cell. Mol. Biol. Lett. 2019, 24, 69. [Google Scholar] [CrossRef] [Green Version]
- Kleinstiver, B.P.; Pattanayak, V.; Prew, M.S.; Tsai, S.Q.; Nguyen, N.T.; Zheng, Z.; Joung, J.K. High-fidelity CRISPR–Cas9 nucleases with no detectable genome-wide off-target effects. Nature 2016, 529, 490–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Katti, A.; Diaz, B.J.; Caragine, C.M.; Sanjana, N.E.; Dow, L.E. CRISPR in cancer biology and therapy. Nat. Cancer 2022, 22, 259–279. [Google Scholar] [CrossRef]
- Steiner, D.; Forrer, P.; Stumpp, M.T.; Plückthun, A. Signal sequences directing cotranslational translocation expand the range of proteins amenable to phage display. Nat. Biotechnol. 2006, 24, 823–831. [Google Scholar] [CrossRef]
- Hartmann, J.; Münch, R.C.; Freiling, R.-T.; Schneider, I.C.; Dreier, B.; Samukange, W.; Koch, J.; Seeger, M.; Plückthun, A.; Buchholz, C.J. A Library-Based Screening Strategy for the Identification of DARPins as Ligands for Receptor-Targeted AAV and Lentiviral Vectors. Mol. Ther. Methods Clin. Dev. 2018, 10, 128–143. [Google Scholar] [CrossRef]
- Ciucci, A.; Buttarelli, M.; Fagotti, A.; Scambia, G.; Gallo, D. Preclinical models of epithelial ovarian cancer: Practical consid-erations and challenges for a meaningful application. Cell Mol. Life Sci. 2022, 79, 364. [Google Scholar] [CrossRef]
Target | Selection of Intrabody | Physiological Knockdown Readout | Ref. |
---|---|---|---|
HRASG12V | A disulfide free scFv was selected with the intracellular antibody capture (IAC) technology [85]. | When NIH 3T3 cells were co-transfected with the disulfide-free scFv and RAS mutant-dependent luciferase construct the number of transformed foci was reduced to 30%. | [28] |
HRASG12V | A human VH domain was selected from two synthetic VH domain libraries with fully randomized complementarity determining regions (CDRs) introduced into a single stable intrabody framework. One human VH was selected in yeast. | The VH binds to activated GTP-bound wild-type HRAS and HRAS (G12V). Nude mice were injected subcutaneously with mouse (NIH3T3-EJ) or human (HT-1080 or DLD-1) tumor cells transduced with the anti-Ras intrabody. Tumors were not developed in mice when the sdAb was expressed in the tumor cells compared to cells with an empty vector or an irrelevant intrabody. | [29] |
GTP-bound K- and H-Ras and the corresponding G12V mutants | Antibody-like ligands as intrabodies were developed (RasIn1 and RasIn2). They were selected by mRNA display using an antigen K-Ras(G12V)-GTPγS. | Detailed binding analysis demonstrated that RasIn1 and RasIn2 recognized the binding domain of the Raf kinase in activated H-RasG12V. | [30] |
Ras mutants | This anti-Ras mutant antibody was engineered from a human antibody previously generated and is internalized through clathrin-mediated endocytosis using heparan sulfate proteoglycan (HSPG) as a receptor and escapes from early endosomes into the cytosol [86]. | The intrabody (RT11-i) recognizes the GTP-bound active forms of wild-type (WT) KRas, NRas and HRas and their oncogenic mutants with mutations at positions 12, 13 or 61, such as KRasG12D, KRasG12V, KRasG13D, KRasQ61H, HRasG12V and NRasQ61R. RT11-i significantly inhibits the tumor growth of oncogenic Ras mutant tumor xenografts in mice. | [31] |
Ras mutants | A new human IgG intrabody (inRas37) binding to activated GTP-bound Ras mutants with two-fold stronger activity was engineered from RT11-i. | Inhibition of tumor growth was seen in several xenograft tumor mice bearing different preestablished colorectal tumors. | [32] |
p21 Ras | scFv antibody was generated from a hybridoma. | The scFv recognizes wild-type H-p21Ras, K-p21Ras and N-p21Ras [32] and their mutated variants. The intrabody significantly inhibited the tumor growth of nude mice with established tumors derived from human colon cancer cell line SW480 or human liver cancer cell line BEL-7402. | [33] |
KRASG12V | DARPins were selected from a phage display library by biopanning using biotinylated KRASG12V. | DARPins bound to an allosteric site of GDP or GTP-bound KRASWT and KRASG12V inhibiting KRAS nucleotide exchange and dimerization. | [34] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the author. 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
Böldicke, T. Therapeutic Potential of Intrabodies for Cancer Immunotherapy: Current Status and Future Directions. Antibodies 2022, 11, 49. https://doi.org/10.3390/antib11030049
Böldicke T. Therapeutic Potential of Intrabodies for Cancer Immunotherapy: Current Status and Future Directions. Antibodies. 2022; 11(3):49. https://doi.org/10.3390/antib11030049
Chicago/Turabian StyleBöldicke, Thomas. 2022. "Therapeutic Potential of Intrabodies for Cancer Immunotherapy: Current Status and Future Directions" Antibodies 11, no. 3: 49. https://doi.org/10.3390/antib11030049
APA StyleBöldicke, T. (2022). Therapeutic Potential of Intrabodies for Cancer Immunotherapy: Current Status and Future Directions. Antibodies, 11(3), 49. https://doi.org/10.3390/antib11030049