Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Multispecific drugs herald a new era of biopharmaceutical innovation

Abstract

The modern biopharmaceutical industry traces its roots to the dawn of the twentieth century, coincident with marketing of aspirin—a signature event in the history of modern drug development. Although the archetypal discovery process did not change markedly in the first seven decades of the industry, the past fifty years have seen two successive waves of transformative innovation in the development of drug molecules: the rise of ‘rational drug discovery’ methodology in the 1970s, followed by the invention of recombinant protein-based therapeutic agents in the 1980s. An incipient fourth wave is the advent of multispecific drugs. The successful development of prospectively designed multispecific drugs has the potential to reconfigure our ideas of how target-based therapeutic molecules can work, and what it is possible to achieve with them. Here I review the two major classes of multispecific drugs: those that enrich a therapeutic agent at a particular site of action and those that link a therapeutic target to a biological effector. The latter class—being freed from the constraint of having to directly modulate the target upon binding—may enable access to components of the proteome that currently cannot be targeted by drugs.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Four transformative waves have shaped development of the biopharmaceutical industry.
Fig. 2: Tetherbodies are multispecific drugs that enrich an active agent at a particular location.
Fig. 3: Small-molecule-based concurrent obligate matchmaker multispecific drugs that induce the proximity of a target and biological effector.
Fig. 4: Biologic matchmaker drugs that induce proximity of a target with a biological effector.
Fig. 5: Potential next-generation bispecific and trispecific CD3 engagers.

Similar content being viewed by others

References

  1. Madsen, U., Krogsgaard-Larsen, P. & Liljefors, T. Textbook of Drug Design and Discovery (Taylor & Francis, 2002).

  2. Drews, J. Drug discovery: a historical perspective. Science 287, 1960–1964 (2000).

    ADS  CAS  PubMed  Google Scholar 

  3. Patlak, M. From viper’s venom to drug design: treating hypertension. FASEB J. 18, 421 (2004).

    CAS  PubMed  Google Scholar 

  4. Altman, L. K. A new insulin given approval for use in U.S. NY Times (30 October 1982).

  5. Ortho Multicenter Transplant Study Group. A randomized clinical trial of OKT3 monoclonal antibody for acute rejection of cadaveric renal transplants. N. Engl. J. Med. 313, 337–342 (1985).

    Google Scholar 

  6. Labrijn, A. F., Janmaat, M. L., Reichert, J. M. & Parren, P. W. H. I. Bispecific antibodies: a mechanistic review of the pipeline. Nat. Rev. Drug Discov. 18, 585–608 (2019).

    CAS  PubMed  Google Scholar 

  7. Nair, J. K. et al. Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J. Am. Chem. Soc. 136, 16958–16961 (2014).A key paper that established the utility of using N-acetylglucosamine to specify liver uptake of siRNAs.

    CAS  PubMed  Google Scholar 

  8. Springer, A. D. & Dowdy, S. F. GalNAc–siRNA conjugates: leading the way for delivery of RNAi therapeutics. Nucleic Acid Ther. 28, 109–118 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Alderson, R. F. et al. CAT-8015: a second-generation pseudomonas exotoxin A-based immunotherapy targeting CD22-expressing hematologic malignancies. Clin. Cancer Res. 15, 832–839 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Abdollahpour-Alitappeh, M. et al. Antibody–drug conjugates (ADCs) for cancer therapy: strategies, challenges, and successes. J. Cell. Physiol. 234, 5628–5642 (2019).

    CAS  PubMed  Google Scholar 

  11. Beck, A., Goetsch, L., Dumontet, C. & Corvaïa, N. Strategies and challenges for the next generation of antibody–drug conjugates. Nat. Rev. Drug Discov. 16, 315–337 (2017).

    CAS  PubMed  Google Scholar 

  12. Neri, D. Antibody–cytokine fusions: versatile products for the modulation of anticancer immunity. Cancer Immunol. Res. 7, 348–354 (2019).

    PubMed  PubMed Central  Google Scholar 

  13. Stanton, B. Z., Chory, E. J. & Crabtree, G. R. Chemically induced proximity in biology and medicine. Science 359, eaao5902 (2018).

    PubMed  PubMed Central  Google Scholar 

  14. Kolata, G. FDA speeds approval of cyclosporin. Science 221, 1273 (1983).

    ADS  CAS  PubMed  Google Scholar 

  15. Liu, J. et al. Calcineurin is a common target of cyclophilin–cyclosporin A and FKBP–FK506 complexes. Cell 66, 807–815 (1991). A landmark paper that established an unexpected mode of drug action via neocomplex formation.

    CAS  PubMed  Google Scholar 

  16. Maniaci, C. & Ciulli, A. Bifunctional chemical probes inducing protein–protein interactions. Curr. Opin. Chem. Biol. 52, 145–156 (2019).

    CAS  PubMed  Google Scholar 

  17. Griffith, J. P. et al. X-ray structure of calcineurin inhibited by the immunophilin-immunosuppressant FKBP12–FK506 complex. Cell 82, 507–522 (1995).

    CAS  PubMed  Google Scholar 

  18. Brown, E. J. et al. A mammalian protein targeted by G1-arresting rapamycin–receptor complex. Nature 369, 756–758 (1994).

    ADS  CAS  PubMed  Google Scholar 

  19. Gray, W. M., Kepinski, S., Rouse, D., Leyser, O. & Estelle, M. Auxin regulates SCFTIR1-dependent degradation of AUX/IAA proteins. Nature 414, 271–276 (2001).

    ADS  CAS  PubMed  Google Scholar 

  20. Tan, X. et al. Mechanism of auxin perception by the TIR1 ubiquitin ligase. Nature 446, 640–645 (2007). Auxin works by a remarkable mechanism, whereby it binds at the interface between ubiquitin ligase subunit TIR1 and a substrate protein, stabilizing the ternary complex and inducing ubiquitination and degradation of the substrate protein.

    ADS  CAS  PubMed  Google Scholar 

  21. Fujiwara, T., Oda, K., Yokota, S., Takatsuki, A. & Ikehara, Y. Brefeldin A causes disassembly of the Golgi complex and accumulation of secretory proteins in the endoplasmic reticulum. J. Biol. Chem. 263, 18545–18552 (1988).

    CAS  PubMed  Google Scholar 

  22. Lippincott-Schwartz, J., Yuan, L. C., Bonifacino, J. S. & Klausner, R. D. Rapid redistribution of Golgi proteins into the ER in cells treated with brefeldin A: evidence for membrane cycling from Golgi to ER. Cell 56, 801–813 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Peyroche, A. et al. Brefeldin A acts to stabilize an abortive ARF–GDP–Sec7 domain protein complex: involvement of specific residues of the Sec7 domain. Mol. Cell 3, 275–285 (1999).

    CAS  PubMed  Google Scholar 

  24. Mossessova, E., Corpina, R. A. & Goldberg, J. Crystal structure of ARF1*Sec7 complexed with brefeldin A and its implications for the guanine nucleotide exchange mechanism. Mol. Cell 12, 1403–1411 (2003).These two papers 23,24 establish that brefeldin A sits at the interface between ARF and SEC7 to stabilize what is normally a very dynamic protein–protein interaction, pointing to a novel mechanism for drug action.

    CAS  PubMed  Google Scholar 

  25. Chardin, P. & McCormick, F. Brefeldin A: the advantage of being uncompetitive. Cell 97, 153–155 (1999).

    CAS  PubMed  Google Scholar 

  26. Palacino, J. et al. SMN2 splice modulators enhance U1-pre-mRNA association and rescue SMA mice. Nat. Chem. Biol. 11, 511–517 (2015).

    CAS  PubMed  Google Scholar 

  27. Gandhi, A. K. et al. Immunomodulatory agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex CRL4(CRBN.). Br. J. Haematol. 164, 811–821 (2014).

    CAS  PubMed  Google Scholar 

  28. Krönke, J. et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 343, 301–305 (2014).

    ADS  PubMed  Google Scholar 

  29. Lu, G. et al. The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins. Science 343, 305–309 (2014). These three papers 27–29report the discovery of a suprising mechanism of action for one of the best-selling drugs in the world; similar to auxin, lenalidomide stabilizes an interaction between a substrate (IKZF1 and/or IKZF3) and an ubiquitin ligase (CRBN), resulting in ubiquitination and degradation of the IKZF proteins.

    ADS  CAS  PubMed  Google Scholar 

  30. Speirs, A. L. Thalidomide and congenital abnormalities. Lancet 279, 303–305 (1962).

    Google Scholar 

  31. Sheskin, J. Thalidomide in the treatment of lepra reactions. Clin. Pharmacol. Ther. 6, 303–306 (1965).

    CAS  PubMed  Google Scholar 

  32. Larkin, M. Low-dose thalidomide seems to be effective in multiple myeloma. Lancet 354, 925 (1999).

    CAS  PubMed  Google Scholar 

  33. Muller, G. W. et al. Amino-substituted thalidomide analogs: potent inhibitors of TNF-alpha production. Bioorg. Med. Chem. Lett. 9, 1625–1630 (1999).

    CAS  PubMed  Google Scholar 

  34. Ito, T. et al. Identification of a primary target of thalidomide teratogenicity. Science 327, 1345–1350 (2010). This seminal paper set in motion the series of discoveries that culminated in the unravelling of the unusual mechanism of action of lenalidomide.

    ADS  CAS  PubMed  Google Scholar 

  35. Lopez-Girona, A. et al. Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide. Leukemia 26, 2326–2335 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhu, Y. X., Kortuem, K. M. & Stewart, A. K. Molecular mechanism of action of immune-modulatory drugs thalidomide, lenalidomide and pomalidomide in multiple myeloma. Leuk. Lymphoma 54, 683–687 (2013).

    CAS  PubMed  Google Scholar 

  37. Cortés, M. & Georgopoulos, K. Aiolos is required for the generation of high affinity bone marrow plasma cells responsible for long-term immunity. J. Exp. Med. 199, 209–219 (2004).

    PubMed  PubMed Central  Google Scholar 

  38. Han, T. et al. Anticancer sulfonamides target splicing by inducing RBM39 degradation via recruitment to DCAF15. Science 356, eaal3755 (2017).

    PubMed  Google Scholar 

  39. Uehara, T. et al. Selective degradation of splicing factor CAPERα by anticancer sulfonamides. Nat. Chem. Biol. 13, 675–680 (2017).

    CAS  PubMed  Google Scholar 

  40. Sakamoto, K. M. et al. Protacs: chimeric molecules that target proteins to the Skp1–Cullin-F box complex for ubiquitination and degradation. Proc. Natl Acad. Sci. USA 98, 8554–8559 (2001). This work provided the first demonstration that it is possible to specify the degradation of a defined target by inducing its proximity to an ubiquitin ligase using a synthetic molecule.

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  41. Sakamoto, K. M. et al. Development of protacs to target cancer-promoting proteins for ubiquitination and degradation. Mol. Cell. Proteomics 2, 1350–1358 (2003).

    CAS  PubMed  Google Scholar 

  42. Schneekloth, J. S. Jr et al. Chemical genetic control of protein levels: selective in vivo targeted degradation. J. Am. Chem. Soc. 126, 3748–3754 (2004).

    CAS  PubMed  Google Scholar 

  43. Pettersson, M. & Crews, C. M. Proteolysis targeting chimeras (PROTACs) - past, present and future. Drug Discov. Today. Technol. 31, 15–27 (2019).

    PubMed  PubMed Central  Google Scholar 

  44. Verma, R., Mohl, D. & Deshaies, R. J. Harnessing the power of proteolysis for targeted protein inactivation. Mol. Cell 77, 446–460 (2020).

    PubMed  Google Scholar 

  45. Buckley, D. L. et al. Small-molecule inhibitors of the interaction between the E3 ligase VHL and HIF1α. Angew. Chem. Int. Edn Engl. 51, 11463–11467 (2012).

    CAS  Google Scholar 

  46. Bondeson, D. P. et al. Catalytic in vivo protein knockdown by small-molecule PROTACs. Nat. Chem. Biol. 11, 611–617 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Winter, G. E. et al. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 348, 1376–1381 (2015).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zengerle, M., Chan, K. H. & Ciulli, A. Selective small molecule induced degradation of the BET bromodomain protein BRD4. ACS Chem. Biol. 10, 1770–1777 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Lu, J. et al. Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4. Chem. Biol. 22, 755–763 (2015).These four papers 46–49established that it is possible to construct highly potent and selective Protacs that work in vivo using ligands that bind ubiquitin ligases VHL or CRBN.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Arvinas, Inc. Clinical trial of ARV-471 in patients with ER+/HER2- locally advanced or metastatic breast cancer (mBC). https://clinicaltrials.gov/ct2/show/NCT04072952 (2019).

  51. Flanagan, J. J. & Neklesa, T. K. Targeting nuclear receptors with PROTAC degraders. Mol. Cell. Endocrinol. 493, 110452 (2019).

    CAS  PubMed  Google Scholar 

  52. Salami, J. et al. Androgen receptor degradation by the proteolysis-targeting chimera ARCC-4 outperforms enzalutamide in cellular models of prostate cancer drug resistance. Commun. Biol. 1, 100 (2018).

    PubMed  PubMed Central  Google Scholar 

  53. Arvinas, Inc. A phase 1 clinical trial of ARV-110 in patients with metastatic castration-resistant prostate cancer (mCRPC). https://clinicaltrials.gov/ct2/show/NCT03888612 (2019).

  54. 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 316, 354–356 (1985).

    ADS  CAS  PubMed  Google Scholar 

  55. Staerz, U. D. & Bevan, M. J. Hybrid hybridoma producing a bispecific monoclonal antibody that can focus effector T-cell activity. Proc. Natl Acad. Sci. USA 83, 1453–1457 (1986).These two papers 54,55launched the field of bispecific CD3 engagers for use in cancer immunotherapy.

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  56. Riechelmann, H. et al. Adoptive therapy of head and neck squamous cell carcinoma with antibody coated immune cells: a pilot clinical trial. Cancer Immunol. Immunother. 56, 1397–1406 (2007).

    CAS  PubMed  Google Scholar 

  57. Löffler, A. et al. A recombinant bispecific single-chain antibody, CD19 x CD3, induces rapid and high lymphoma-directed cytotoxicity by unstimulated T lymphocytes. Blood 95, 2098–2103 (2000).

    PubMed  Google Scholar 

  58. Bargou, R. et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science 321, 974–977 (2008). More than 20 years after research on bispecific CD3 engagers began, this paper provided the first compelling demonstration of the tremendous clinical potential of this class of bispecific molecules.

    ADS  CAS  PubMed  Google Scholar 

  59. Curran, E. & Stock, W. Taking a “BiTE out of ALL”: blinatumomab approval for MRD-positive ALL. Blood 133, 1715–1719 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Przepiorka, D. et al. FDA approval: blinatumomab. Clin. Cancer Res. 21, 4035–4039 (2015).

    CAS  PubMed  Google Scholar 

  61. Viardot, A. & Bargou, R. Bispecific antibodies in haematological malignancies. Cancer Treat. Rev. 65, 87–95 (2018).

    CAS  PubMed  Google Scholar 

  62. Gauthier, L. et al. Multifunctional natural killer cell engagers targeting NKp46 trigger protective tumor immunity. Cell 177, 1701–1713 (2019).

    CAS  PubMed  Google Scholar 

  63. Nayyar, G., Chu, Y. & Cairo, M. S. Overcoming resistance to natural killer cell based immunotherapies for solid tumors. Front. Oncol. 9, 51 (2019).

    PubMed  PubMed Central  Google Scholar 

  64. Sampei, Z. et al. Identification and multidimensional optimization of an asymmetric bispecific IgG antibody mimicking the function of factor VIII cofactor activity. PLoS ONE 8, e57479 (2013). This paper reports on the long campaign that led to discovery of emicizumab, a landmark achievement in the history of bispecific biologic drugs.

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  65. Kitazawa, T. & Shima, M. Emicizumab, a humanized bispecific antibody to coagulation factors IXa and X with a factor VIIIa-cofactor activity. Int. J. Hematol. 111, 20–30 (2018).

    PubMed  Google Scholar 

  66. Foster, D. J. et al. Advanced siRNA designs further improve in vivo performance of GalNAc–siRNA conjugates. Mol. Ther. 26, 708–717 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Khvorova, A. & Watts, J. K. The chemical evolution of oligonucleotide therapies of clinical utility. Nat. Biotechnol. 35, 238–248 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Egli, M. & Manoharan, M. Re-engineering RNA molecules into therapeutic agents. Acc. Chem. Res. 52, 1036–1047 (2019).

    CAS  PubMed  Google Scholar 

  69. Matsuda, S. et al. siRNA conjugates carrying sequentially assembled trivalent N-acetylgalactosamine linked through nucleosides elicit robust gene silencing in vivo in hepatocytes. ACS Chem. Biol. 10, 1181–1187 (2015).

    CAS  PubMed  Google Scholar 

  70. Levin, A. A. Treating disease at the RNA level with oligonucleotides. N. Engl. J. Med. 380, 57–70 (2019).

    PubMed  Google Scholar 

  71. Winkler, J. Extrahepatic targeting of oligonucleotides with receptor-binding non-immunoglobulin scaffold proteins. Nucleic Acid Ther. 28, 137–145 (2018).

    CAS  PubMed  Google Scholar 

  72. Matthews, P. M. Chronic inflammation in multiple sclerosis – seeing what was always there. Nat. Rev. Neurol. 15, 582–593 (2019).

    CAS  PubMed  Google Scholar 

  73. Freskgård, P. O. & Urich, E. Antibody therapies in CNS diseases. Neuropharmacology 120, 38–55 (2017).

    PubMed  Google Scholar 

  74. Poduslo, J. F., Curran, G. L. & Berg, C. T. Macromolecular permeability across the blood–nerve and blood–brain barriers. Proc. Natl Acad. Sci. USA 91, 5705–5709 (1994).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  75. Sevigny, J. et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature 537, 50–56 (2016).

    ADS  CAS  PubMed  Google Scholar 

  76. Lajoie, J. M. & Shusta, E. V. Targeting receptor-mediated transport for delivery of biologics across the blood–brain barrier. Annu. Rev. Pharmacol. Toxicol. 55, 613–631 (2015).

    CAS  PubMed  Google Scholar 

  77. Mäger, I. et al. Targeting blood–brain-barrier transcytosis – perspectives for drug delivery. Neuropharmacology 120, 4–7 (2017).

    PubMed  Google Scholar 

  78. Pardridge, W. M. Delivery of biologics across the blood–brain barrier with molecular Trojan horse technology. BioDrugs 31, 503–519 (2017).

    CAS  PubMed  Google Scholar 

  79. Zuchero, Y. J. et al. Discovery of novel blood–brain barrier targets to enhance brain uptake of therapeutic antibodies. Neuron 89, 70–82 (2016).

    CAS  PubMed  Google Scholar 

  80. Stutz, C. C., Zhang, X. & Shusta, E. V. Combinatorial approaches for the identification of brain drug delivery targets. Curr. Pharm. Des. 20, 1564–1576 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Chan, K. Y. et al. Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat. Neurosci. 20, 1172–1179 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Le, R. Q. et al. FDA approval summary: tocilizumab for treatment of chimeric antigen receptor T cell-induced severe or life-threatening cytokine release syndrome. Oncologist 23, 943–947 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Li, J. et al. CD3 bispecific antibody-induced cytokine release is dispensable for cytotoxic T cell activity. Sci. Transl. Med. 11, eaax8861 (2019). These authors report that it is possible to uncouple cytokine release from the anti-tumour activity of BCEs, which could lead to an enhanced therapeutic index for BCEs.

    PubMed  Google Scholar 

  84. Topp, M. S. et al. Long-term follow-up of hematologic relapse-free survival in a phase 2 study of blinatumomab in patients with MRD in B-lineage ALL. Blood 120, 5185–5187 (2012).

    CAS  PubMed  Google Scholar 

  85. Balakrishnan, A. et al. Multispecific targeting with synthetic ankyrin repeat motif chimeric antigen receptors. Clin. Cancer Res. 25, 7506–7516 (2019).

    PubMed  PubMed Central  Google Scholar 

  86. Zhao, J., Song, Y. & Liu, D. Clinical trials of dual-target CAR T cells, donor-derived CAR T cells, and universal CAR T cells for acute lymphoid leukemia. J. Hematol. Oncol. 12, 17 (2019).

    PubMed  PubMed Central  Google Scholar 

  87. Kebenko, M. et al. A multicenter phase 1 study of solitomab (MT110, AMG 110), a bispecific EpCAM/CD3 T-cell engager (BiTE®) antibody construct, in patients with refractory solid tumors. OncoImmunology 7, e1450710 (2018).

    PubMed  PubMed Central  Google Scholar 

  88. Mau-Sørensen, M. et al. A phase I trial of intravenous catumaxomab: a bispecific monoclonal antibody targeting EpCAM and the T cell coreceptor CD3. Cancer Chemother. Pharmacol. 75, 1065–1073 (2015).

    PubMed  Google Scholar 

  89. Desnoyers, L. R. et al. Tumor-specific activation of an EGFR-targeting probody enhances therapeutic index. Sci. Transl. Med. 5, 207ra144 (2013).

    PubMed  Google Scholar 

  90. Kloss, C. C., Condomines, M., Cartellieri, M., Bachmann, M. & Sadelain, M. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat. Biotechnol. 31, 71–75 (2013).

    CAS  PubMed  Google Scholar 

  91. Roybal, K. T. et al. Precision tumor recognition by T cells with combinatorial antigen-sensing circuits. Cell 164, 770–779 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Srivastava, S. et al. Logic-gated ROR1 chimeric antigen receptor expression rescues T cell-mediated toxicity to normal tissues and enables selective tumor targeting. Cancer Cell 35, 489–503 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Langan, R. A. et al. De novo design of bioactive protein switches. Nature 572, 205–210 (2019) Computational protein design is used to construct a synthetic AND gate that may have applicability to future therapeutic agents.

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  94. de Sostoa, J. et al. Targeting the tumor stroma with an oncolytic adenovirus secreting a fibroblast activation protein-targeted bispecific T-cell engager. J. Immunother. Cancer 7, 19 (2019).

    PubMed  PubMed Central  Google Scholar 

  95. Brey, C. U. et al. A gB/CD3 bispecific BiTE antibody construct for targeting human cytomegalovirus-infected cells. Sci. Rep. 8, 17453 (2018).

    ADS  PubMed  PubMed Central  Google Scholar 

  96. Brozy, J. et al. Antiviral activity of HIV gp120-targeting bispecific T cell engager antibody constructs. J. Virol. 92, e00491-18 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Aghajanian, H. et al. Targeting cardiac fibrosis with engineered T cells. Nature 573, 430–433 (2019).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  98. Ellebrecht, C. T. et al. Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease. Science 353, 179–184 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  99. Baker, D. J. et al. Naturally occurring p16Ink4a-positive cells shorten healthy lifespan. Nature 530, 184–189 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  100. Igawa, T. et al. Engineered monoclonal antibody with novel antigen-sweeping activity in vivo. PLoS ONE 8, e63236 (2013).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  101. Banik, S., Pedram, K., Wisnovsky, S., Riley, N. & Bertozzi, C. Lysosome targeting chimeras (LYTACs) for the degradation of secreted and membrane proteins. Preprint at https://chemrxiv.org/articles/Lysosome_Targeting_Chimeras_LYTACs_for_the_Degradation_of_Secreted_and_Membrane_Proteins/7927061 (2019).

  102. Takahashi, D. et al. AUTACs: cargo-specific degraders using selective autophagy. Mol. Cell 76, 797–810.e10 (2019).

    CAS  PubMed  Google Scholar 

  103. Ito, C. et al. Endogenous nitrated nucleotide is a key mediator of autophagy and innate defense against bacteria. Mol. Cell 52, 794–804 (2013

    CAS  PubMed  Google Scholar 

  104. Yamazoe, S. et al. Heterobifunctional molecules induce dephosphorylation of kinases-a proof of concept study. J. Med. Chem. https://doi.org/10.1021/acs.jmedchem.9b01167 (2020).

  105. Zhang, Z. & Shokat, K. M. Bifunctional small-molecule ligands of K-Ras induce its association with immunophilin proteins. Angew. Chem. Int. Edn Engl. 58, 16314–16319 (2019).

    CAS  Google Scholar 

  106. Costales, M. G. et al. Small-molecule targeted recruitment of a nuclease to cleave an oncogenic RNA in a mouse model of metastatic cancer. Proc. Natl Acad. Sci. USA 117, 2406–2411 (2020). This study reports on ribonuclease-targeting chimaeras, which potentially represent an entire new class of therapeutic agent that targets the elimination of specific RNAs.

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Klemm, J. D., Beals, C. R. & Crabtree, G. R. Rapid targeting of nuclear proteins to the cytoplasm. Curr. Biol. 7, 638–644 (1997).

    CAS  PubMed  Google Scholar 

  108. Haruki, H., Nishikawa, J. & Laemmli, U. K. The anchor-away technique: rapid, conditional establishment of yeast mutant phenotypes. Mol. Cell 31, 925–932 (2008).

    CAS  PubMed  Google Scholar 

  109. Wang, Q. et al. Design and production of bispecific antibodies. Antibodies (Basel) 8, 43 (2019).

    CAS  Google Scholar 

  110. Bratt, J., Linderholm, A., Monroe, B. & Chamow, S. Therapeutic IgG-like bispecific antibodies. Bioprocess Int. 16, 40–49 (2018).

    CAS  Google Scholar 

  111. Ponce, R. et al. Immunogenicity of biologically-derived therapeutics: assessment and interpretation of nonclinical safety studies. Regul. Toxicol. Pharmacol. 54, 164–182 (2009).

    CAS  PubMed  Google Scholar 

  112. Datta-Mannan, A. et al. Aberrant bispecific antibody pharmacokinetics linked to liver sinusoidal endothelium clearance mechanism in cynomolgus monkeys. MAbs 8, 969–982 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Harper, J. et al. An approved in vitro approach to preclinical safety and efficacy evaluation of engineered T cell receptor anti-CD3 bispecific (ImmTAC) molecules. PLoS ONE 13, e0205491 (2018).

    PubMed  PubMed Central  Google Scholar 

  114. Saber, H., Del Valle, P., Ricks, T. K. & Leighton, J. K. An FDA oncology analysis of CD3 bispecific constructs and first-in-human dose selection. Regul. Toxicol. Pharmacol. 90, 144–152 (2017).

    CAS  PubMed  Google Scholar 

  115. US Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research & Center for Biologics Evaluation and Research. Bispecific antibody development programs Guidance for industry, https://www.fda.gov/media/123313/download (2019).

  116. Lipinski, C. A., Lombardo, F., Dominy, B. W. & Feeney, P. J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 23, 3–25 (1997).

    CAS  Google Scholar 

  117. Dauvois, S., Danielian, P. S., White, R. & Parker, M. G. Antiestrogen ICI 164,384 reduces cellular estrogen receptor content by increasing its turnover. Proc. Natl Acad. Sci. USA 89, 4037–4041 (1992).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  118. Bernstein, I. D. Monoclonal antibodies to the myeloid stem cells: therapeutic implications of CMA-676, a humanized anti-CD33 antibody calicheamicin conjugate. Leukemia 14, 474–475 (2000).

    CAS  PubMed  Google Scholar 

  119. Guan, J. et al. Therapeutic ligands antagonize estrogen receptor function by impairing its mobility. Cell 178, 949–963 (2019).

    CAS  PubMed  Google Scholar 

  120. Du, J. et al. Structural basis for the blockage of IL-2 signaling by therapeutic antibody basiliximab. J. Immunol. 184, 1361–1368 (2010).

    CAS  PubMed  Google Scholar 

  121. Gadd, M. S. et al. Structural basis of PROTAC cooperative recognition for selective protein degradation. Nat. Chem. Biol. 13, 514–521 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

I thank C. Kiefer and L. Jia for artwork, J. Bailis, R. Desphande, H. Hsu, G. Moffat, J. Murry, N. Ogbechie, and D. Rock for supplying content and comments, and D. Baker, M. Chu-Moyer, A. Gouliaev, S. Haldar, E. Levy, F. Martin, D. Reese, E. Sigal, K. Stefánsson, P. Tagari, and S. Wang for comments on the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Raymond J. Deshaies.

Ethics declarations

Competing interests

R.J.D. is an employee, officer and shareholder of Amgen.

Additional information

Peer review information Nature thanks Alessio Ciulli, Ira Mellman and Nurulain Zaveri for their contribution to the peer review of this work.

Supplementary information

Supplementary Tables

This file contains Supplementary Tables 1-3.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Deshaies, R.J. Multispecific drugs herald a new era of biopharmaceutical innovation. Nature 580, 329–338 (2020). https://doi.org/10.1038/s41586-020-2168-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2168-1

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research