Multispecific Biotherapeutics: Comparison
Please note this is a comparison between Version 2 by Vivi Li and Version 1 by Xiaotian Zhong.

Multispecific biotherapeutic drugs are protein-based therapeutic molecules that can engage multiple drug-target binding interfaces concurrently.

  • multispecific biotherapeutics
  • immune cells engagers
  • antibody-drug conjugates
  • multispecific tetherbodies
  • biologic matchmakers
  • small-scaffold multispecific modalities
Please wait, diff process is still running!

References

  1. Hodgson, J. Refreshing the biologic pipeline 2020. Nat. Biotechnol. 2021, 39, 135–143.
  2. Carter, P.J.; Lazar, G.A. Next generation antibody drugs: Pursuit of the ‘high-hanging fruit’. Nat. Rev. Drug Discov. 2018, 17, 197–223.
  3. Deshaies, R.J. Multispecific drugs herald a new era of biopharmaceutical innovation. Nature 2020, 580, 329–338.
  4. Labrijn, A.F.; Janmaat, M.L.; Reichert, J.M.; Parren, P.W. Bispecific antibodies: A mechanistic review of the pipeline. Nat. Rev. Drug Discov. 2019, 18, 585–608.
  5. Goebeler, M.E.; Bargou, R.C. T cell-engaging therapies—BiTEs and beyond. Nat. Rev. Clin. Oncol. 2020, 17, 418–434.
  6. Beck, A.; Goetsch, L.; Dumontet, C.; Corvaïa, N. Strategies and challenges for the next generation of antibody-drug conjugates. Nat. Rev. Drug Discov. 2017, 16, 315–337.
  7. Nisonoff, A.; Rivers, M.M. Recombination of a mixture of univalent antibody fragments of different specificity. Arch. Biochem. Biophys. 1961, 93, 460–462.
  8. Aalberse, R.C.; van der Gaag, R.; van Leeuwen, J. Serologic aspects of IgG4 antibodies. I. Prolonged immunization results in an IgG4-restricted response. J. Immunol. 1983, 130, 722–726.
  9. Spiess, C.; Zhai, Q.; Carter, P.J. Alternative molecular formats and therapeutic applications for bispecific antibodies. Mol. Immunol. 2015, 67, 95–106.
  10. Brinkmann, U.; Kontermann, R.E. The making of bispecific antibodies. MAbs 2017, 9, 182–212.
  11. Thakur, A.; Huang, M.; Lum, L.G. Bispecific antibody based therapeutics: Strengths and challenges. Blood Rev. 2018, 32, 339–347.
  12. Staerz, U.D.; Kanagawa, O.; Bevan, M.J. Hybrid antibodies can target sites for attack by T cells. Nature 1985, 314, 628–631.
  13. Perez, P.; Hoffman, R.W.; Shaw, S.; Bluestone, J.A.; Segal, D.M. Specific targeting of cytotoxic T cells by anti-T3 linked to anti-target cell antibody. Nature 1985, 316, 354–356.
  14. Nagorsen, D.; Kufer, P.; Baeuerle, P.A.; Bargou, R. Blinatumomab: A historical perspective. Pharmacol. Ther. 2012, 136, 334–342.
  15. Jost, C.; Pluckthun, A. Engineered proteins with desired specificity: DARPins, other alternative scaffolds and bispecific IgGs. Curr. Opin. Struct. Biol. 2014, 27, 102–112.
  16. Gilbreth, R.N.; Koide, S. Structural insights for engineering binding proteins based on non-antibody scaffolds. Curr. Opin. Struct. Biol. 2012, 22, 413–420.
  17. Dimitrov, D.S. Engineered CH2 domains (nanoantibodies). MAbs 2009, 1, 26–28.
  18. Raghavan, M.; Bonagura, V.R.; Morrison, S.L.; Bjorkman, P.J. Analysis of the pH dependence of the neonatal Fc receptor/immunoglobulin G interaction using antibody and receptor variants. Biochemistry 1995, 34, 14649–14657.
  19. Raghavan, M.; Bjorkman, P.J. Fc receptors and their interactions with immunoglobulins. Annu. Rev. Cell Dev. Biol. 1996, 12, 181–220.
  20. Krah, S.; Kolmar, H.; Becker, S.; Zielonka, S. Engineering IgG-Like Bispecific Antibodies-An Overview. Antibodies 2018, 7, 28.
  21. Santich, B.H.; Park, J.A.; Tran, H.; Guo, H.F.; Huse, M.; Cheung, N.K. Interdomain spacing and spatial configuration drive the potency of IgG-[L]-scFv T cell bispecific antibodies. Sci. Transl. Med. 2020, 12, eaax1315.
  22. Krah, S.; Sellmann, C.; Rhiel, L.; Schröter, C.; Dickgiesser, S.; Beck, J.; Zielonka, S.; Toleikis, L.; Hock, B.; Kolmar, H.; et al. Engineering bispecific antibodies with defined chain pairing. New Biotechnol. 2017, 39, 167–173.
  23. Sun, W.; Khare, P.; Wang, X.; Challa, D.K.; Greenberg, B.M.; Ober, R.J.; Ward, E.S. Selective Depletion of Antigen-Specific Antibodies for the Treatment of Demyelinating Disease. Mol. Ther. 2021, 29, 1312–1323.
  24. Zhong, X.; Kieras, E.; Sousa, E.; D’Antona, A.; Baber, J.C.; He, T.; Desharnais, J.; Wood, L.; Luxenberg, D.; Stahl, M.; et al. Pyroglutamate and O-Linked Glycan Determine Functional Production of Anti-IL17A and Anti-IL22 Peptide-Antibody Bispecific Genetic Fusions. J. Biol. Chem. 2013, 288, 1409–1419.
  25. Kanakaraj, P.; Puffer, B.A.; Yao, X.T.; Kankanala, S.; Boyd, E.; Shah, R.R.; Wang, G.; Patel, D.; Krishnamurthy, R.; Kaithamana, S.; et al. Simultaneous targeting of TNF and Ang2 with a novel bispecific antibody enhances efficacy in an in vivo model of arthritis. MAbs 2012, 4, 600–613.
  26. Cao, Y.; Suresh, M.R. Bispecific antibodies as novel bioconjugates. Bioconjug. Chem. 1998, 9, 635–644.
  27. Trier, N.; Hansen, P.; Houen, G. Peptides, Antibodies, Peptide Antibodies and More. Int. J. Mol. Sci. 2019, 20, 6289.
  28. Lowe, K.L.; Cole, D.; Kenefeck, R.; OKelly, I.; Lepore, M.; Jakobsen, B.K. Novel TCR-based biologics: Mobilising T cells to warm ‘cold’ tumours. Cancer Treat. Rev. 2019, 77, 35–43.
  29. Oates, J.; Hassan, N.J.; Jakobsen, B.K. ImmTACs for targeted cancer therapy: Why, what, how, and which. Mol. Immunol. 2015, 67, 67–74.
  30. Wu, X.; Sereno, A.J.; Huang, F.; Zhang, K.; Batt, M.; Fitchett, J.R.; He, D.; Rick, H.L.; Conner, E.M.; Demarest, S.J. Protein design of IgG/TCR chimeras for the co-expression of Fab-like moieties within bispecific antibodies. MAbs 2015, 7, 364–376.
  31. Guo, G.; Han, J.; Wang, Y.; Li, Y. A potential downstream platform approach for WuXiBody-based IgG-like bispecific antibodies. Protein Expr. Purif. 2020, 173, 105647.
  32. Dai, Z.; Zhang, X.N.; Nasertorabi, F.; Cheng, Q.; Li, J.; Katz, B.B.; Smbatyan, G.; Pei, H.; Louie, S.G.; Lenz, H.J.; et al. Synthesis of site-specific antibody-drug conjugates by ADP-ribosyl cyclases. Sci. Adv. 2020, 6, eaba6752.
  33. Sha, F.; Salzman, G.; Gupta, A.; Koide, S. Monobodies and other synthetic binding proteins for expanding protein science. Protein Sci. 2017, 26, 910–924.
  34. Ståhl, S.; Gräslund, T.; Karlström, A.E.; Frejd, F.Y.; Nygren, P.Å.; Löfblom, J. Affibody Molecules in Biotechnological and Medical Applications. Trends Biotechnol. 2017, 35, 691–712.
  35. Lipovšek, D.; Carvajal, I.; Allentoff, A.J.; Barros, A., Jr.; Brailsford, J.; Cong, Q.; Cotter, P.; Gangwar, S.; Hollander, C.; Lafont, V.; et al. Adnectin-drug conjugates for Glypican-3-specific delivery of a cytotoxic payload to tumors. Protein Eng. Des. Sel. 2018, 31, 159–171.
  36. Alder, M.N.; Rogozin, I.B.; Iyer, L.M.; Glazko, G.V.; Cooper, M.D.; Pancer, Z. Diversity and function of adaptive immune receptors in a jawless vertebrate. Science 2005, 310, 1970–1973.
  37. Kintzing, J.R.; Cochran, J.R. Engineered knottin peptides as diagnostics, therapeutics, and drug delivery vehicles. Curr. Opin. Chem. Biol. 2016, 34, 143–150.
  38. Brack, S.; Attinger-Toller, I.; Schade, B.; Mourlane, F.; Klupsch, K.; Woods, R.; Hachemi, H.; von der Bey, U.; Koenig-Friedrich, S.; Bertschinger, J.; et al. A bispecific HER2-targeting FynomAb with superior antitumor activity and novel mode of action. Mol. Cancer Ther. 2014, 13, 2030–2039.
  39. Gong, R.; Wang, Y.; Ying, T.; Dimitrov, D.S. Bispecific engineered antibody domains (nanoantibodies) that interact noncompetitively with an HIV-1 neutralizing epitope and FcRn. PLoS ONE 2012, 7, e42288.
  40. Abdollahpour-Alitappeh, M.; Lotfinia, M.; Gharibi, T.; Mardaneh, J.; Farhadihosseinabadi, B.; Larki, P.; Faghfourian, B.; Sepehr, K.S.; Abbaszadeh-Goudarzi, K.; Abbaszadeh-Goudarzi, G.; et al. Antibody-drug conjugates (ADCs) for cancer therapy: Strategies, challenges, and successes. J. Cell. Physiol. 2019, 234, 5628–5642.
  41. Leung, D.; Wurst, J.M.; Liu, T.; Martinez, R.M.; Datta-Mannan, A.; Feng, Y. Antibody Conjugates-Recent Advances and Future Innovations. Antibodies 2020, 9, 2.
  42. Benizri, S.; Gissot, A.; Martin, A.; Vialet, B.; Grinstaff, M.W.; Barthélémy, P. Bioconjugated Oligonucleotides: Recent Developments and Therapeutic Applications. Bioconjug. Chem. 2019, 30, 366–383.
  43. Atwell, S.; Ridgway, J.B.; Wells, J.A.; Carter, P. Stable heterodimers from remodeling the domain interface of a homodimer using a phage display library. J. Mol. Biol. 1997, 270, 26–35.
  44. Ridgway, J.B.; Presta, L.G.; Carter, P. ‘Knobs-into-holes’ engineering of antibody CH3 domains for heavy chain heterodimerization. Protein Eng. 1996, 9, 617–621.
  45. Gunasekaran, K.; Pentony, M.; Shen, M.; Garrett, L.; Forte, C.; Woodward, A.; Ng, S.B.; Born, T.; Retter, M.; Manchulenko, K.; et al. Enhancing antibody Fc heterodimer formation through electrostatic steering effects: Applications to bispecific molecules and monovalent IgG. J. Biol. Chem. 2010, 285, 19637–19646.
  46. Strop, P.; Ho, W.H.; Boustany, L.M.; Abdiche, Y.N.; Lindquist, K.C.; Farias, S.E.; Rickert, M.; Appah, C.T.; Pascua, E.; Radcliffe, T.; et al. Generating bispecific human IgG1 and IgG2 antibodies from any antibody pair. J. Mol. Biol. 2012, 420, 204–219.
  47. De Nardis, C.; Hendriks, L.J.; Poirier, E.; Arvinte, T.; Gros, P.; Bakker, A.B.; de Kruif, J. A new approach for generating bispecific antibodies based on a common light chain format and the stable architecture of human immunoglobulin G1. J. Biol. Chem. 2017, 292, 14706–14717.
  48. Labrijn, A.F.; Rispens, T.; Meesters, J.; Rose, R.J.; den Bleker, T.H.; Loverix, S.; van den Bremer, E.T.; Neijssen, J.; Vink, T.; Lasters, I.; et al. Species-specific determinants in the IgG CH3 domain enable Fab-arm exchange by affecting the noncovalent CH3-CH3 interaction strength. J. Immunol. 2011, 187, 3238–3246.
  49. Labrijn, A.F.; Meesters, J.I.; de Goeij, B.E.; van den Bremer, E.T.; Neijssen, J.; van Kampen, M.D.; Strumane, K.; Verploegen, S.; Kundu, A.; Gramer, M.J.; et al. Efficient generation of stable bispecific IgG1 by controlled Fab-arm exchange. Proc. Natl. Acad. Sci. USA 2013, 110, 5145–5150.
  50. Davis, J.H.; Aperlo, C.; Li, Y.; Kurosawa, E.; Lan, Y.; Lo, K.M.; Huston, J.S. SEEDbodies: Fusion proteins based on strand-exchange engineered domain (SEED) CH3 heterodimers in an Fc analogue platform for asymmetric binders or immunofusions and bispecific antibodies. Protein Eng. Des. Sel. 2010, 23, 195–202.
  51. Wranik, B.J.; Christensen, E.L.; Schaefer, G.; Jackman, J.K.; Vendel, A.C.; Eaton, D. LUZ-Y, a novel platform for the mammalian cell production of full-length IgG-bispecific antibodies. J. Biol. Chem. 2012, 287, 43331–43339.
  52. Moore, G.L.; Bautista, C.; Pong, E.; Nguyen, D.H.; Jacinto, J.; Eivazi, A.; Muchhal, U.S.; Karki, S.; Chu, S.Y.; Lazar, G.A. A novel bispecific antibody format enables simultaneous bivalent and monovalent co-engagement of distinct target antigens. MAbs 2011, 3, 546–557.
  53. Choi, H.J.; Kim, Y.J.; Lee, S.; Kim, Y.S. A heterodimeric Fc-based bispecific antibody simultaneously targeting VEGFR-2 and Met exhibits potent antitumor activity. Mol. Cancer Ther. 2013, 12, 2748–2759.
  54. Choi, H.J.; Seok, S.H.; Kim, Y.J.; Seo, M.D.; Kim, Y.S. Crystal structures of immunoglobulin Fc heterodimers reveal the molecular basis for heterodimer formation. Mol. Immunol. 2015, 65, 377–383.
  55. Von Kreudenstein, T.S.; Escobar-Carbrera, E.; Lario, P.I.; D’Angelo, I.; Brault, K.; Kelly, J.F.; Durocher, Y.; Baardsnes, J.; Woods, R.J.; Xie, M.H.; et al. Improving biophysical properties of a bispecific antibody scaffold to aid developability: Quality by molecular design. MAbs 2013, 5, 646–654.
  56. Leaver-Fay, A.; Froning, K.J.; Atwell, S.; Aldaz, H.; Pustilnik, A.; Lu, F.; Huang, F.; Yuan, R.; Hassanali, S.; Chamberlain, A.K.; et al. Computationally Designed Bispecific Antibodies using Negative State Repertoires. Structure 2016, 24, 641–651.
  57. Smith, E.J.; Olson, K.; Haber, L.J.; Varghese, B.; Duramad, P.; Tustian, A.D.; Oyejide, A.; Kirshner, J.R.; Canova, L.; Menon, J.; et al. A novel, native-format bispecific antibody triggering T-cell killing of B-cells is robustly active in mouse tumor models and cynomolgus monkeys. Sci. Rep. 2015, 5, 17943.
  58. Ollier, R.; Wassmann, P.; Monney, T.; Ries Fecourt, C.; Gn, S.; CA, V.; Ayoub, D.; Stutz, C.; Gudi, G.S.; Blein, S. Single-step Protein A and Protein G avidity purification methods to support bispecific antibody discovery and development. MAbs 2019, 11, 1464–1478.
  59. Schaefer, W.; Regula, J.T.; Bähner, M.; Schanzer, J.; Croasdale, R.; Dürr, H.; Gassner, C.; Georges, G.; Kettenberger, H.; Imhof-Jung, S.; et al. Immunoglobulin domain crossover as a generic approach for the production of bispecific IgG antibodies. Proc. Natl. Acad. Sci. USA 2011, 108, 11187–11192.
  60. Lewis, S.M.; Wu, X.; Pustilnik, A.; Sereno, A.; Huang, F.; Rick, H.L.; Guntas, G.; Leaver-Fay, A.; Smith, E.M.; Ho, C.; et al. Generation of bispecific IgG antibodies by structure-based design of an orthogonal Fab interface. Nat. Biotechnol. 2014, 32, 191–198.
  61. Liu, Z.; Leng, E.C.; Gunasekaran, K.; Pentony, M.; Shen, M.; Howard, M.; Stoops, J.; Manchulenko, K.; Razinkov, V.; Liu, H.; et al. A novel antibody engineering strategy for making monovalent bispecific heterodimeric IgG antibodies by electrostatic steering mechanism. J. Biol. Chem. 2015, 290, 7535–7562.
  62. Fischer, N.; Elson, G.; Magistrelli, G.; Dheilly, E.; Fouque, N.; Laurendon, A.; Gueneau, F.; Ravn, U.; Depoisier, J.F.; Moine, V.; et al. Exploiting light chains for the scalable generation and platform purification of native human bispecific IgG. Nat. Commun. 2015, 6, 6113.
  63. Jackman, J.; Chen, Y.; Huang, A.; Moffat, B.; Scheer, J.M.; Leong, S.R.; Lee, W.P.; Zhang, J.; Sharma, N.; Lu, Y.; et al. Development of a two-part strategy to identify a therapeutic human bispecific antibody that inhibits IgE receptor signaling. J. Biol. Chem. 2010, 285, 20850–20859.
  64. Krah, S.; Schröter, C.; Eller, C.; Rhiel, L.; Rasche, N.; Beck, J.; Sellmann, C.; Günther, R.; Toleikis, L.; Hock, B.; et al. Generation of human bispecific common light chain antibodies by combining animal immunization and yeast display. Protein Eng. Des. Sel. 2017, 30, 291–301.
  65. Golay, J.; Choblet, S.; Iwaszkiewicz, J.; Cérutti, P.; Ozil, A.; Loisel, S.; Pugnière, M.; Ubiali, G.; Zoete, V.; Michielin, O.; et al. Design and Validation of a Novel Generic Platform for the Production of Tetravalent IgG1-like Bispecific Antibodies. J. Immunol. 2016, 196, 3199–3211.
  66. Gong, S.; Ren, F.; Wu, D.; Wu, X.; Wu, C. Fabs-in-tandem immunoglobulin is a novel and versatile bispecific design for engaging multiple therapeutic targets. MAbs 2017, 9, 1118–1128.
  67. Mazor, Y.; Oganesyan, V.; Yang, C.; Hansen, A.; Wang, J.; Liu, H.; Sachsenmeier, K.; Carlson, M.; Gadre, D.V.; Borrok, M.J.; et al. Improving target cell specificity using a novel monovalent bispecific IgG design. MAbs 2015, 7, 377–389.
  68. Mazor, Y.; Hansen, A.; Yang, C.; Chowdhury, P.S.; Wang, J.; Stephens, G.; Wu, H.; Dall’Acqua, W.F. Insights into the molecular basis of a bispecific antibody’s target selectivity. MAbs 2015, 7, 461–469.
  69. Moretti, P.; Skegro, D.; Ollier, R.; Wassmann, P.; Aebischer, C.; Laurent, T.; Schmid-Printz, M.; Giovannini, R.; Blein, S.; Bertschinger, M. BEAT the bispecific challenge: A novel and efficient platform for the expression of bispecific IgGs. BMC Proc. 2013, 7 (Suppl. 6), O9.
  70. Shim, H. Bispecific Antibodies and Antibody-Drug Conjugates for Cancer Therapy: Technological Considerations. Biomolecules 2020, 10, 360.
  71. Gauzy-Lazo, L.; Sassoon, I.; Brun, M.P. Advances in Antibody-Drug Conjugate Design: Current Clinical Landscape and Future Innovations. SLAS Discov. 2020, 25, 843–868.
  72. Voynov, V.; Adam, P.J.; Nixon, A.E.; Scheer, J.M. Discovery Strategies to Maximize the Clinical Potential of T-Cell Engaging Antibodies for the Treatment of Solid Tumors. Antibodies 2020, 9, 65.
  73. Caputi, A.P.; Navarra, P. Beyond antibodies: Ankyrins and DARPins. From basic research to drug approval. Curr. Opin. Pharmacol. 2020, 51, 93–101.
  74. Strohl, W.R.; Naso, M. Bispecific T-Cell Redirection versus Chimeric Antigen Receptor (CAR)-T Cells as Approaches to Kill Cancer Cells. Antibodies 2019, 8, 41.
  75. Clynes, R.A.; Desjarlais, J.R. Redirected T Cell Cytotoxicity in Cancer Therapy. Annu. Rev. Med. 2019, 70, 437–450.
  76. Ellerman, D. Bispecific T-cell engagers: Towards understanding variables influencing the in vitro potency and tumor selectivity and their modulation to enhance their efficacy and safety. Methods 2019, 154, 102–117.
  77. Wu, Z.; Cheung, N.V. T cell engaging bispecific antibody (T-BsAb): From technology to therapeutics. Pharmacol. Ther. 2018, 182, 161–175.
  78. Middelburg, J.; Kemper, K.; Engelberts, P.; Labrijn, A.F.; Schuurman, J.; van Hall, T. Overcoming Challenges for CD3-Bispecific Antibody Therapy in Solid Tumors. Cancers 2021, 13, 287.
  79. Nie, S.; Wang, Z.; Moscoso-Castro, M.; D’Souza, P.; Lei, C.; Xu, J.; Gu, J. Biology drives the discovery of bispecific antibodies as innovative therapeutics. Antib. Ther. 2020, 3, 18–62.
  80. Seimetz, D.; Lindhofer, H.; Bokemeyer, C. Development and approval of the trifunctional antibody catumaxomab (anti-EpCAM x anti-CD3) as a targeted cancer immunotherapy. Cancer Treat. Rev. 2010, 36, 458–467.
  81. Root, A.R.; Cao, W.; Li, B.; LaPan, P.; Meade, C.; Sanford, J.; Jin, M.; O’Sullivan, C.; Cummins, E.; Lambert, M.; et al. Development of PF-06671008, a Highly Potent Anti-P-cadherin/Anti-CD3 Bispecific DART Molecule with Extended Half-Life for the Treatment of Cancer. Antibodies 2016, 5, 6.
  82. Mathur, D.; Root, A.R.; Bugaj-Gaweda, B.; Bisulco, S.; Tan, X.; Fang, W.; Kearney, J.C.; Lucas, J.; Guffroy, M.; Golas, J.; et al. A Novel GUCY2C-CD3 T-Cell Engaging Bispecific Construct (PF-07062119) for the Treatment of Gastrointestinal Cancers. Clin. Cancer Res. 2020, 26, 2188–2202.
  83. Root, A.R.; Guntas, G.; Katragadda, M.; Apgar, J.R.; Narula, J.; Chang, C.S.; Hanscom, S.; McKenna, M.; Wade, J.; Meade, C.; et al. Discovery and optimization of a novel anti-GUCY2c x CD3 bispecific antibody for the treatment of solid tumors. MAbs 2021, 13, 1850395.
  84. Zhu, G.; Foletti, D.; Liu, X.; Ding, S.; Witt, J.M.; Hasa-Moreno, A.; Rickert, M.; Holz, C.; Aschenbrenner, L.; Yang, A.H.; et al. Targeting CLDN18.2 by CD3 Bispecific and ADC Modalities for the Treatments of Gastric and Pancreatic Cancer. Sci. Rep. 2019, 9, 8420.
  85. Yeung, Y.A.; Krishnamoorthy, V.; Dettling, D.; Sommer, C.; Poulsen, K.; Ni, I.; Pham, A.; Chen, W.; Liao-Chan, S.; Lindquist, K.; et al. An Optimized Full-Length FLT3/CD3 Bispecific Antibody Demonstrates Potent Anti-leukemia Activity and Reversible Hematological Toxicity. Mol. Ther. 2020, 28, 889–900.
  86. Wu, C.; Ying, H.; Grinnell, C.; Bryant, S.; Miller, R.; Clabbers, A.; Bose, S.; McCarthy, D.; Zhu, R.R.; Santora, L.; et al. Simultaneous targeting of multiple disease mediators by a dual-variable-domain immunoglobulin. Nat. Biotechnol. 2007, 25, 1290–1297.
  87. Bardwell, P.D.; Staron, M.M.; Liu, J.; Tao, Q.; Scesney, S.; Bukofzer, G.; Rodriguez, L.E.; Choi, C.H.; Wang, J.; Chang, Q.; et al. Potent and conditional redirected T cell killing of tumor cells using Half DVD-Ig. Protein Cell 2018, 9, 121–129.
  88. Correnti, C.E.; Laszlo, G.S.; de van der Schueren, W.J.; Godwin, C.D.; Bandaranayake, A.; Busch, M.A.; Gudgeon, C.J.; Bates, O.M.; Olson, J.M.; Mehlin, C.; et al. Simultaneous multiple interaction T-cell engaging (SMITE) bispecific antibodies overcome bispecific T-cell engager (BiTE) resistance via CD28 co-stimulation. Leukemia 2018, 32, 1239–1243.
  89. Steinmetz, A.; Vallée, F.; Beil, C.; Lange, C.; Baurin, N.; Beninga, J.; Capdevila, C.; Corvey, C.; Dupuy, A.; Ferrari, P.; et al. CODV-Ig, a universal bispecific tetravalent and multifunctional immunoglobulin format for medical applications. MAbs 2016, 8, 867–878.
  90. Joubert, N.; Beck, A.; Dumontet, C.; Denevault-Sabourin, C. Antibody-Drug Conjugates: The Last Decade. Pharmaceuticals 2020, 13, 245.
  91. Lambert, J.M.; Berkenblit, A. Antibody-Drug Conjugates for Cancer Treatment. Annu. Rev. Med. 2018, 69, 191–207.
  92. Neri, D. Antibody-Cytokine Fusions: Versatile Products for the Modulation of Anticancer Immunity. Cancer Immunol. Res. 2019, 7, 348–354.
  93. Pardridge, W.M. Delivery of Biologics across the Blood-Brain Barrier with Molecular Trojan Horse Technology. BioDrugs 2017, 31, 503–519.
  94. Yu, Y.J.; Atwal, J.K.; Zhang, Y.; Tong, R.K.; Wildsmith, K.R.; Tan, C.; Bien-Ly, N.; Hersom, M.; Maloney, J.A.; Meilandt, W.J.; et al. Therapeutic bispecific antibodies cross the blood-brain barrier in nonhuman primates. Sci. Transl. Med. 2014, 6, 261ra154.
  95. Yu, Y.J.; Zhang, Y.; Kenrick, M.; Hoyte, K.; Luk, W.; Lu, Y.; Atwal, J.; Elliott, J.M.; Prabhu, S.; Watts, R.J.; et al. Boosting brain uptake of a therapeutic antibody by reducing its affinity for a transcytosis target. Sci. Transl. Med. 2011, 3, 84ra44.
  96. Sampei, Z.; Igawa, T.; Soeda, T.; Okuyama-Nishida, Y.; Moriyama, C.; Wakabayashi, T.; Tanaka, E.; Muto, A.; Kojima, T.; Kitazawa, T.; et al. Identification and multidimensional optimization of an asymmetric bispecific IgG antibody mimicking the function of factor VIII cofactor activity. PLoS ONE 2013, 8, e57479.
  97. Kitazawa, T.; Shima, M. Emicizumab, a humanized bispecific antibody to coagulation factors IXa and X with a factor VIIIa-cofactor activity. Int. J. Hematol. 2020, 111, 20–30.
  98. Neiveyans, M.; Melhem, R.; Arnoult, C.; Bourquard, T.; Jarlier, M.; Busson, M.; Laroche, A.; Cerutti, M.; Pugnière, M.; Ternant, D.; et al. A recycling anti-transferrin receptor-1 monoclonal antibody as an efficient therapy for erythroleukemia through target up-regulation and antibody-dependent cytotoxic effector functions. MAbs 2019, 11, 593–605.
  99. Baruch, A.; Wong, C.; Chinn, L.W.; Vaze, A.; Sonoda, J.; Gelzleichter, T.; Chen, S.; Lewin-Koh, N.; Morrow, L.; Dheerendra, S.; et al. Antibody-mediated activation of the FGFR1/Klothobeta complex corrects metabolic dysfunction and alters food preference in obese humans. Proc. Natl. Acad. Sci. USA 2020, 117, 28992–29000.
  100. Shi, S.Y.; Lu, Y.W.; Liu, Z.; Stevens, J.; Murawsky, C.M.; Wilson, V.; Hu, Z.; Richards, W.G.; Michaels, M.L.; Zhang, J.; et al. A biparatopic agonistic antibody that mimics fibroblast growth factor 21 ligand activity. J. Biol. Chem. 2018, 293, 5909–5919.
  101. Gumlaw, N.; Sevigny, L.M.; Zhao, H.; Luo, Z.; Bangari, D.S.; Masterjohn, E.; Chen, Y.; McDonald, B.; Magnay, M.; Travaline, T.; et al. biAb Mediated Restoration of the Linkage between Dystroglycan and Laminin-211 as a Therapeutic Approach for alpha-Dystroglycanopathies. Mol. Ther. 2020, 28, 664–676.
  102. Fernandes, R.A.; Su, L.; Nishiga, Y.; Ren, J.; Bhuiyan, A.M.; Cheng, N.; Kuo, C.J.; Picton, L.K.; Ohtsuki, S.; Majzner, R.G.; et al. Immune receptor inhibition through enforced phosphatase recruitment. Nature 2020, 586, 779–784.
  103. Kontermann, R.E. Half-life extended biotherapeutics. Expert Opin. Biol. Ther. 2016, 16, 903–915.
  104. Schellenberger, V.; Wang, C.W.; Geething, N.C.; Spink, B.J.; Campbell, A.; To, W.; Scholle, M.D.; Yin, Y.; Yao, Y.; Bogin, O.; et al. A recombinant polypeptide extends the in vivo half-life of peptides and proteins in a tunable manner. Nat. Biotechnol. 2009, 27, 1186–1190.
  105. Hutt, M.; Färber-Schwarz, A.; Unverdorben, F.; Richter, F.; Kontermann, R.E. Plasma half-life extension of small recombinant antibodies by fusion to immunoglobulin-binding domains. J. Biol. Chem. 2012, 287, 4462–4469.
  106. Kontermann, R.E. Strategies for extended serum half-life of protein therapeutics. Curr. Opin. Biotechnol. 2011, 22, 868–876.
  107. Fan, G.; Wang, Z.; Hao, M.; Li, J. Bispecific antibodies and their applications. J. Hematol. Oncol. 2015, 8, 130.
  108. Chari, R.V.; Miller, M.L.; Widdison, W.C. Antibody-drug conjugates: An emerging concept in cancer therapy. Angew. Chem. Int. Ed. Engl. 2014, 53, 3796–3827.
  109. Sellmann, C.; Doerner, A.; Knuehl, C.; Rasche, N.; Sood, V.; Krah, S.; Rhiel, L.; Messemer, A.; Wesolowski, J.; Schuette, M.; et al. Balancing Selectivity and Efficacy of Bispecific Epidermal Growth Factor Receptor (EGFR) x c-MET Antibodies and Antibody-Drug Conjugates. J. Biol. Chem. 2016, 291, 25106–25119.
  110. Wagner-Rousset, E.; Janin-Bussat, M.C.; Colas, O.; Excoffier, M.; Ayoub, D.; Haeuw, J.F.; Rilatt, I.; Perez, M.; Corvaïa, N.; Beck, A. Antibody-drug conjugate model fast characterization by LC-MS following IdeS proteolytic digestion. MAbs 2014, 6, 273–285.
  111. Dumontet, C.; Jordan, M.A. Microtubule-binding agents: A dynamic field of cancer therapeutics. Nat. Rev. Drug Discov. 2010, 9, 790–803.
  112. Levin, A.A. Treating Disease at the RNA Level with Oligonucleotides. N. Engl. J. Med. 2019, 380, 57–70.
  113. Sugo, T.; Terada, M.; Oikawa, T.; Miyata, K.; Nishimura, S.; Kenjo, E.; Ogasawara-Shimizu, M.; Makita, Y.; Imaichi, S.; Murata, S.; et al. Development of antibody-siRNA conjugate targeted to cardiac and skeletal muscles. J. Control. Release 2016, 237, 1–13.
  114. Satake, N.; Duong, C.; Yoshida, S.; Oestergaard, M.; Chen, C.; Peralta, R.; Guo, S.; Seth, P.P.; Li, Y.; Beckett, L.; et al. Novel Targeted Therapy for Precursor B Cell Acute Lymphoblastic Leukemia: Anti-CD22 Antibody-MXD3 Antisense Oligonucleotide Conjugate. Mol. Med. 2016, 22, 632–642.
  115. Arnold, A.E.; Malek-Adamian, E.; Le, P.U.; Meng, A.; Martínez-Montero, S.; Petrecca, K.; Damha, M.J.; Shoichet, M.S. Antibody-Antisense Oligonucleotide Conjugate Downregulates a Key Gene in Glioblastoma Stem Cells. Mol. Ther. Nucleic Acids 2018, 11, 518–527.
  116. Erickson, H.K.; Park, P.U.; Widdison, W.C.; Kovtun, Y.V.; Garrett, L.M.; Hoffman, K.; Lutz, R.J.; Goldmacher, V.S.; Blättler, W.A. Antibody-maytansinoid conjugates are activated in targeted cancer cells by lysosomal degradation and linker-dependent intracellular processing. Cancer Res. 2006, 66, 4426–4433.
  117. Bargh, J.D.; Isidro-Llobet, A.; Parker, J.S.; Spring, D.R. Cleavable linkers in antibody-drug conjugates. Chem. Soc. Rev. 2019, 48, 4361–4374.
  118. Acchione, M.; Kwon, H.; Jochheim, C.M.; Atkins, W.M. Impact of linker and conjugation chemistry on antigen binding, Fc receptor binding and thermal stability of model antibody-drug conjugates. MAbs 2012, 4, 362–372.
  119. Sun, M.M.; Beam, K.S.; Cerveny, C.G.; Hamblett, K.J.; Blackmore, R.S.; Torgov, M.Y.; Handley, F.G.; Ihle, N.C.; Senter, P.D.; Alley, S.C. Reduction-alkylation strategies for the modification of specific monoclonal antibody disulfides. Bioconjug. Chem. 2005, 16, 1282–1290.
  120. Junutula, J.R.; Gerber, H.P. Next-Generation Antibody-Drug Conjugates (ADCs) for Cancer Therapy. ACS Med. Chem. Lett. 2016, 7, 972–973.
  121. Panowski, S.; Bhakta, S.; Raab, H.; Polakis, P.; Junutula, J.R. Site-specific antibody drug conjugates for cancer therapy. MAbs 2014, 6, 34–45.
  122. Junutula, J.R.; Raab, H.; Clark, S.; Bhakta, S.; Leipold, D.D.; Weir, S.; Chen, Y.; Simpson, M.; Tsai, S.P.; Dennis, M.S.; et al. Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nat. Biotechnol. 2008, 26, 925–932.
  123. Shen, B.Q.; Xu, K.; Liu, L.; Raab, H.; Bhakta, S.; Kenrick, M.; Parsons-Reponte, K.L.; Tien, J.; Yu, S.F.; Mai, E.; et al. Conjugation site modulates the in vivo stability and therapeutic activity of antibody-drug conjugates. Nat. Biotechnol. 2012, 30, 184–189.
  124. Kung Sutherland, M.S.; Walter, R.B.; Jeffrey, S.C.; Burke, P.J.; Yu, C.; Kostner, H.; Stone, I.; Ryan, M.C.; Sussman, D.; Lyon, R.P.; et al. SGN-CD33A: A novel CD33-targeting antibody-drug conjugate using a pyrrolobenzodiazepine dimer is active in models of drug-resistant AML. Blood 2013, 122, 1455–1463.
  125. Zhong, X.; He, T.; Prashad, A.S.; Wang, W.; Cohen, J.; Ferguson, D.; Tam, A.S.; Sousa, E.; Lin, L.; Tchistiakova, L.; et al. Mechanistic understanding of the cysteine capping modifications of antibodies enables selective chemical engineering in live mammalian cells. J. Biotechnol. 2017, 248, 48–58.
  126. Axup, J.Y.; Bajjuri, K.M.; Ritland, M.; Hutchins, B.M.; Kim, C.H.; Kazane, S.A.; Halder, R.; Forsyth, J.S.; Santidrian, A.F.; Stafin, K.; et al. Synthesis of site-specific antibody-drug conjugates using unnatural amino acids. Proc. Natl. Acad. Sci. USA 2012, 109, 16101–16106.
  127. Strop, P.; Liu, S.H.; Dorywalska, M.; Delaria, K.; Dushin, R.G.; Tran, T.T.; Ho, W.H.; Farias, S.; Casas, M.G.; Abdiche, Y.; et al. Location matters: Site of conjugation modulates stability and pharmacokinetics of antibody drug conjugates. Chem. Biol. 2013, 20, 161–167.
  128. Lhospice, F.; Brégeon, D.; Belmant, C.; Dennler, P.; Chiotellis, A.; Fischer, E.; Gauthier, L.; Boëdec, A.; Rispaud, H.; Savard-Chambard, S.; et al. Site-Specific Conjugation of Monomethyl Auristatin E to Anti-CD30 Antibodies Improves Their Pharmacokinetics and Therapeutic Index in Rodent Models. Mol. Pharm. 2015, 12, 1863–1871.
  129. Beerli, R.R.; Hell, T.; Merkel, A.S.; Grawunder, U. Sortase Enzyme-Mediated Generation of Site-Specifically Conjugated Antibody Drug Conjugates with High In Vitro and In Vivo Potency. PLoS ONE 2015, 10, e0131177.
  130. van Geel, R.; Wijdeven, M.A.; Heesbeen, R.; Verkade, J.M.; Wasiel, A.A.; van Berkel, S.S.; van Delft, F.L. Chemoenzymatic Conjugation of Toxic Payloads to the Globally Conserved N-Glycan of Native mAbs Provides Homogeneous and Highly Efficacious Antibody-Drug Conjugates. Bioconjug. Chem. 2015, 26, 2233–2242.
  131. Rabuka, D.; Rush, J.S.; Gregory, W.D.; Wu, P.; Bertozzi, C.R. Site-specific chemical protein conjugation using genetically encoded aldehyde tags. Nat. Protoc. 2012, 7, 1052–1067.
  132. Lin, S.; Yang, X.; Jia, S.; Weeks, A.M.; Hornsby, M.; Lee, P.S.; Nichiporuk, R.V.; Iavarone, A.T.; Wells, J.A.; Toste, F.D.; et al. Redox-based reagents for chemoselective methionine bioconjugation. Science 2017, 355, 597–602.
  133. Elledge, S.K.; Tran, H.L.; Christian, A.H.; Steri, V.; Hann, B.; Toste, F.D.; Chang, C.J.; Wells, J.A. Systematic identification of engineered methionines and oxaziridines for efficient, stable, and site-specific antibody bioconjugation. Proc. Natl. Acad. Sci. USA 2020, 117, 5733–5740.
  134. Mäger, I.; Meyer, A.H.; Li, J.; Lenter, M.; Hildebrandt, T.; Leparc, G.; Wood, M.J. Targeting blood-brain-barrier transcytosis—Perspectives for drug delivery. Neuropharmacology 2017, 120, 4–7.
  135. Niewoehner, J.; Bohrmann, B.; Collin, L.; Urich, E.; Sade, H.; Maier, P.; Rueger, P.; Stracke, J.O.; Lau, W.; Tissot, A.C.; et al. Increased brain penetration and potency of a therapeutic antibody using a monovalent molecular shuttle. Neuron 2014, 81, 49–60.
  136. Bezabeh, B.; Fleming, R.; Fazenbaker, C.; Zhong, H.; Coffman, K.; Yu, X.Q.; Leow, C.C.; Gibson, N.; Wilson, S.; Stover, C.K.; et al. Insertion of scFv into the hinge domain of full-length IgG1 monoclonal antibody results in tetravalent bispecific molecule with robust properties. MAbs 2017, 9, 240–256.
  137. Wec, A.Z.; Nyakatura, E.K.; Herbert, A.S.; Howell, K.A.; Holtsberg, F.W.; Bakken, R.R.; Mittler, E.; Christin, J.R.; Shulenin, S.; Jangra, R.K.; et al. A “Trojan horse” bispecific-antibody strategy for broad protection against ebolaviruses. Science 2016, 354, 350–354.
  138. Desnoyers, L.R.; Vasiljeva, O.; Richardson, J.H.; Yang, A.; Menendez, E.E.; Liang, T.W.; Wong, C.; Bessette, P.H.; Kamath, K.; Moore, S.J.; et al. Tumor-specific activation of an EGFR-targeting probody enhances therapeutic index. Sci. Transl. Med. 2013, 5, 207ra144.
  139. Mimoto, F.; Tatsumi, K.; Shimizu, S.; Kadono, S.; Haraya, K.; Nagayasu, M.; Suzuki, Y.; Fujii, E.; Kamimura, M.; Hayasaka, A.; et al. Exploitation of Elevated Extracellular ATP to Specifically Direct Antibody to Tumor Microenvironment. Cell Rep. 2020, 33, 108542.
  140. Kamata-Sakurai, M.; Narita, Y.; Hori, Y.; Nemoto, T.; Uchikawa, R.; Honda, M.; Hironiwa, N.; Taniguchi, K.; Shida-Kawazoe, M.; Metsugi, S.; et al. Antibody to CD137 Activated by Extracellular Adenosine Triphosphate Is Tumor Selective and Broadly Effective In Vivo without Systemic Immune Activation. Cancer Discov. 2021, 11, 158–175.
  141. Polu, K.R.; Lowman, H.B. Probody therapeutics for targeting antibodies to diseased tissue. Expert Opin. Biol. Ther. 2014, 14, 1049–1053.
  142. Maniaci, C.; Ciulli, A. Bifunctional chemical probes inducing protein-protein interactions. Curr. Opin. Chem. Biol. 2019, 52, 145–156.
  143. Pettersson, M.; Crews, C.M. PROteolysis TArgeting Chimeras (PROTACs)—Past, present and future. Drug Discov. Today Technol. 2019, 31, 15–27.
  144. Verma, R.; Mohl, D.; Deshaies, R.J. Harnessing the Power of Proteolysis for Targeted Protein Inactivation. Mol. Cell. 2020, 77, 446–460.
  145. Kanner, S.A.; Morgenstern, T.; Colecraft, H.M. Sculpting ion channel functional expression with engineered ubiquitin ligases. Elife 2017, 6, e29744.
  146. Kanner, S.A.; Shuja, Z.; Choudhury, P.; Jain, A.; Colecraft, H.M. Targeted deubiquitination rescues distinct trafficking-deficient ion channelopathies. Nat. Methods 2020, 17, 1245–1253.
  147. Grabulovski, D.; Kaspar, M.; Neri, D. A novel, non-immunogenic Fyn SH3-derived binding protein with tumor vascular targeting properties. J. Biol. Chem. 2007, 282, 3196–3204.
  148. Silacci, M.; Lembke, W.; Woods, R.; Attinger-Toller, I.; Baenziger-Tobler, N.; Batey, S.; Santimaria, R.; von der Bey, U.; Koenig-Friedrich, S.; Zha, W.; et al. Discovery and characterization of COVA322, a clinical-stage bispecific TNF/IL-17A inhibitor for the treatment of inflammatory diseases. MAbs 2016, 8, 141–149.
  149. Bloom, L.; Calabro, V. FN3: A new protein scaffold reaches the clinic. Drug Discov. Today 2009, 14, 949–955.
  150. Koide, A.; Wojcik, J.; Gilbreth, R.N.; Hoey, R.J.; Koide, S. Teaching an old scaffold new tricks: Monobodies constructed using alternative surfaces of the FN3 scaffold. J. Mol. Biol. 2012, 415, 393–405.
  151. Koide, A.; Bailey, C.W.; Huang, X.; Koide, S. The fibronectin type III domain as a scaffold for novel binding proteins. J. Mol. Biol. 1998, 284, 1141–1151.
  152. Diem, M.D.; Hyun, L.; Yi, F.; Hippensteel, R.; Kuhar, E.; Lowenstein, C.; Swift, E.J.; O’Neil, K.T.; Jacobs, S.A. Selection of high-affinity Centyrin FN3 domains from a simple library diversified at a combination of strand and loop positions. Protein Eng. Des. Sel. 2014, 27, 419–429.
  153. Wojcik, J.; Hantschel, O.; Grebien, F.; Kaupe, I.; Bennett, K.L.; Barkinge, J.; Jones, R.B.; Koide, A.; Superti-Furga, G.; Koide, S. A potent and highly specific FN3 monobody inhibitor of the Abl SH2 domain. Nat. Struct. Mol. Biol. 2010, 17, 519–527.
  154. Ludwicki, M.B.; Li, J.; Stephens, E.A.; Roberts, R.W.; Koide, S.; Hammond, P.T.; DeLisa, M.P. Broad-Spectrum Proteome Editing with an Engineered Bacterial Ubiquitin Ligase Mimic. ACS Cent. Sci. 2019, 5, 852–866.
  155. Goldberg, S.D.; Cardoso, R.M.; Lin, T.; Spinka-Doms, T.; Klein, D.; Jacobs, S.A.; Dudkin, V.; Gilliland, G.; O’Neil, K.T. Engineering a targeted delivery platform using Centyrins. Protein Eng. Des. Sel. 2016, 29, 563–572.
  156. Feldwisch, J.; Tolmachev, V. Engineering of affibody molecules for therapy and diagnostics. Methods Mol. Biol. 2012, 899, 103–126.
  157. Löfblom, J.; Feldwisch, J.; Tolmachev, V.; Carlsson, J.; Ståhl, S.; Frejd, F.Y. Affibody molecules: Engineered proteins for therapeutic, diagnostic and biotechnological applications. FEBS Lett. 2010, 584, 2670–2680.
  158. Frejd, F.Y.; Kim, K.T. Affibody molecules as engineered protein drugs. Exp. Mol. Med. 2017, 49, e306.
  159. Akhtari, J.; Rezayat, S.M.; Teymouri, M.; Alavizadeh, S.H.; Gheybi, F.; Badiee, A.; Jaafari, M.R. Targeting, bio distributive and tumor growth inhibiting characterization of anti-HER2 affibody coupling to liposomal doxorubicin using BALB/c mice bearing TUBO tumors. Int. J. Pharm. 2016, 505, 89–95.
  160. Meister, S.W.; Hjelm, L.C.; Dannemeyer, M.; Tegel, H.; Lindberg, H.; Ståhl, S.; Löfblom, J. An Affibody Molecule Is Actively Transported into the Cerebrospinal Fluid via Binding to the Transferrin Receptor. Int. J. Mol. Sci. 2020, 21, 2999.
  161. Pluckthun, A. Designed ankyrin repeat proteins (DARPins): Binding proteins for research, diagnostics, and therapy. Annu. Rev. Pharmacol. Toxicol. 2015, 55, 489–511.
  162. Simon, M.; Stefan, N.; Borsig, L.; Plückthun, A.; Zangemeister-Wittke, U. Increasing the antitumor effect of an EpCAM-targeting fusion toxin by facile click PEGylation. Mol. Cancer Ther. 2014, 13, 375–385.
  163. Sokolova, E.; Proshkina, G.; Kutova, O.; Shilova, O.; Ryabova, A.; Schulga, A.; Stremovskiy, O.; Zdobnova, T.; Balalaeva, I.; Deyev, S. Recombinant targeted toxin based on HER2-specific DARPin possesses a strong selective cytotoxic effect in vitro and a potent antitumor activity in vivo. J. Control. Release 2016, 233, 48–56.
  164. Soto-Gamez, A.; Chen, D.; Nabuurs, A.G.; Quax, W.J.; Demaria, M.; Boersma, Y.L. A Bispecific Inhibitor of the EGFR/ADAM17 Axis Decreases Cell Proliferation and Migration of EGFR-Dependent Cancer Cells. Cancers 2020, 12, 411.
  165. Baird, R.; Linossi, C.; Middleton, M.; Lord, S.; Harris, A.; Rodón, J.; Zitt, C.; Fiedler, U.; Dawson, K.M.; Leupin, N.; et al. First-in-Human Phase I Study of MP0250, a First-in-Class DARPin Drug Candidate Targeting VEGF and HGF, in Patients With Advanced Solid Tumors. J. Clin. Oncol. 2020, JCO2000596.
  166. Babaee, N.; Garoosi, Y.T.; Karimipoor, M.; Davami, F.; Bayat, E.; Safarpour, H.; Mahboudi, F.; Barkhordari, F. DARPin Ec1-LMWP protein scaffold in targeted delivery of siRNA molecules through EpCAM cancer stem cell marker. Mol. Biol. Rep. 2020, 47, 7323–7331.
  167. Cox, N.; Kintzing, J.R.; Smith, M.; Grant, G.A.; Cochran, J.R. Integrin-Targeting Knottin Peptide-Drug Conjugates Are Potent Inhibitors of Tumor Cell Proliferation. Angew. Chem. Int. Ed. Engl. 2016, 55, 9894–9897.
  168. Lee, S.C.; Park, K.; Han, J.; Lee, J.J.; Kim, H.J.; Hong, S.; Heu, W.; Kim, Y.J.; Ha, J.S.; Lee, S.G.; et al. Design of a binding scaffold based on variable lymphocyte receptors of jawless vertebrates by module engineering. Proc. Natl. Acad. Sci. USA 2012, 109, 3299–3304.
  169. Richter, A.; Eggenstein, E.; Skerra, A. Anticalins: Exploiting a non-Ig scaffold with hypervariable loops for the engineering of binding proteins. FEBS Lett. 2014, 588, 213–218.
  170. Rothe, C.; Skerra, A. Anticalin((R)) Proteins as Therapeutic Agents in Human Diseases. BioDrugs 2018, 32, 233–243.
  171. Thakur, A.; Lum, L.G. Cancer therapy with bispecific antibodies: Clinical experience. Curr. Opin. Mol. Ther. 2010, 12, 340–349.
  172. Doppalapudi, V.R.; Huang, J.; Liu, D.; Jin, P.; Liu, B.; Li, L.; Desharnais, J.; Hagen, C.; Levin, N.J.; Shields, M.J.; et al. Chemical generation of bispecific antibodies. Proc. Natl. Acad. Sci. USA 2010, 107, 22611–22616.
  173. White, B.H.; Whalen, K.; Kriksciukaite, K.; Alargova, R.; Au Yeung, T.; Bazinet, P.; Brockman, A.; DuPont, M.; Oller, H.; Lemelin, C.A.; et al. Discovery of an SSTR2-Targeting Maytansinoid Conjugate (PEN-221) with Potent Activity in Vitro and in Vivo. J. Med. Chem. 2019, 62, 2708–2719.
  174. Wang, L.; Xu, J.; Kong, Y.; Liang, R.; Li, W.; Li, J.; Lu, J.; Dimitrov, D.S.; Yu, F.; Wu, Y.; et al. Engineering a Novel Antibody-Peptide Bispecific Fusion Protein against MERS-CoV. Antibodies 2019, 8, 53.
More
Video Production Service