Skip to main content

Advertisement

Log in

The Evolving Druggability and Developability Space: Chemically Modified New Modalities and Emerging Small Molecules

  • Review Article
  • Published:
The AAPS Journal Aims and scope Submit manuscript

Abstract

The druggability and developability space is rapidly evolving in the post-genomic era. In the past, Lipinski’s rule-of-five (Ro5) emerged and served as a guide for drug-like molecule design for oral delivery in the traditional druggable target space. In contrast, in this new era, a transition is occurring in drug discovery towards novel approaches to bind and modulate challenging biological targets that have led to transformative treatments for patients. Consequently, drugging novel targets using a variety of emerging molecular modalities, namely beyond the Ro5 (bRo5) small molecules (such as protein-protein interaction modulators, protein-targeted chimeras, or PROTACs), peptide/peptidomimetics, and nucleic acid-based modalities, have become a key focus in drug discovery. Herein, the emerging druggability and developability space is discussed side by side to build a general understanding of the potential development challenges of these novel modalities. An overview is provided on the evolving novel targets and molecular modalities, followed by a detailed analysis of the druggability aspects as well as the strategies used to progress drug candidate, and the trending chemistry and formulation strategies used to assess developability.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Hopkins AL, Groom CR. The druggable genome. Nat Rev Drug Discov. 2002;1(9):727–30.

    Article  CAS  PubMed  Google Scholar 

  2. Abi Hussein H, Geneix C, Petitjean M, Borrel A, Flatters D, Camproux AC. Global vision of druggability issues: applications and perspectives. Drug Discov Today. 2017;22(2):404–15.

    Article  PubMed  Google Scholar 

  3. Makley LN, Gestwicki JE. Expanding the number of 'druggable' targets: non-enzymes and protein-protein interactions. Chem Biol Drug Des. 2013;81(1):22–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Saxena V, Panicucci R, Joshi Y, Garad S. Developability assessment in pharmaceutical industry: an integrated group approach for selecting developable candidates. J Pharm Sci. 2009;98(6):1962–79.

    Article  CAS  PubMed  Google Scholar 

  5. Venkatesh S, Lipper RA. Role of the development scientist in compound lead selection and optimization. J Pharm Sci. 2000;89(2):145–54.

    Article  CAS  PubMed  Google Scholar 

  6. Bak A, Leung D, Barrett SE, Forster S, Minnihan EC, Leithead AW, et al. Physicochemical and formulation developability assessment for therapeutic peptide delivery--a primer. AAPS J. 2015;17(1):144–55.

    Article  CAS  PubMed  Google Scholar 

  7. Mathias NR, Hussain MA. Non-invasive systemic drug delivery: Developability considerations for alternate routes of administration. J Pharm Sci. 2009;99(1):1–20.

    Article  CAS  Google Scholar 

  8. Furman JL, Chiu M, Hunter MJ. Early engineering approaches to improve peptide Developability and manufacturability. AAPS J. 2015;17(1):111–20.

    Article  CAS  PubMed  Google Scholar 

  9. Kwong E. Advancing drug discovery: a pharmaceutics perspective. J Pharm Sci. 2015;104(3):865–71.

    Article  CAS  PubMed  Google Scholar 

  10. Ramachander RRN. Molecule and Manufacturability Assessment Leading to Robust Commercial Formulation for Therapeutic Proteins. In: Parag Kolhe MS, Rathore N, editors. Sterile product development AAPS Advances in the Pharmaceutical Sciences Series. 1st ed. New York: Springer-Verlag; 2013. p. 33–45.

    Google Scholar 

  11. Almagro J, Mascioni A. Best practices in assessing the developability of biopharmaceutical candidates. 2015;195–220.

  12. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev. 2001;46(1–3):3–26.

    Article  CAS  PubMed  Google Scholar 

  13. Bayliss MK, Butler J, Feldman PL, Green DV, Leeson PD, Palovich MR, et al. Quality guidelines for oral drug candidates: dose, solubility and lipophilicity. Drug Discov Today. 2016;21(10):1719–27.

    Article  CAS  PubMed  Google Scholar 

  14. Paul SM, Mytelka DS, Dunwiddie CT, Persinger CC, Munos BH, Lindborg SR, et al. How to improve R&D productivity: the pharmaceutical industry's grand challenge. Nat Rev Drug Discov. 2010;9(3):203–14.

    Article  CAS  PubMed  Google Scholar 

  15. Bunnage ME. Getting pharmaceutical R&D back on target. Nat Chem Biol. 2011;7(6):335–9.

    Article  CAS  PubMed  Google Scholar 

  16. Gulcher J, Stefansson K. Population genomics: laying the groundwork for genetic disease modeling and targeting. Clin Chem Lab Med. 1998;36(8):523–7.

    Article  CAS  PubMed  Google Scholar 

  17. Sakharkar MK, Sakharkar KR, Pervaiz S. Druggability of human disease genes. Int J Biochem Cell Biol. 2007;39(6):1156–64.

    Article  CAS  PubMed  Google Scholar 

  18. Lazo JS, Sharlow ER. Drugging Undruggable molecular Cancer targets. Annu Rev Pharmacol Toxicol. 2016;56:23–40.

    Article  CAS  PubMed  Google Scholar 

  19. Leeson PD. Molecular inflation, attrition and the rule of five. Adv Drug Deliv Rev. 2016;101:22–33.

    Article  CAS  PubMed  Google Scholar 

  20. DeGoey DA, Chen HJ, Cox PB, Wendt MD. Beyond the rule of 5: lessons Learned from AbbVie's drugs and compound collection. J Med Chem. 2018;61(7):2636–51.

    Article  CAS  PubMed  Google Scholar 

  21. Shultz MD. Two decades under the influence of the rule of five and the changing properties of approved Oral drugs. J Med Chem. 2019;62(4):1701–14.

    Article  CAS  PubMed  Google Scholar 

  22. Valeur E, Gueret SM, Adihou H, Gopalakrishnan R, Lemurell M, Waldmann H, et al. New modalities for challenging targets in drug discovery. Angew Chem Int Engl. 2017;56(35):10294–323.

    Article  CAS  Google Scholar 

  23. Churcher I. Protac-induced protein degradation in drug discovery: breaking the rules or just making new ones? J Med Chem. 2018;61(2):444–52.

    Article  CAS  PubMed  Google Scholar 

  24. Matsson P, Kihlberg J. How big is too big for cell permeability? J Med Chem. 2017;60(5):1662–4.

    Article  CAS  PubMed  Google Scholar 

  25. Matsson P, Doak BC, Over B, Kihlberg J. Cell permeability beyond the rule of 5. Adv Drug Deliv Rev. 2016;101:42–61.

    Article  CAS  PubMed  Google Scholar 

  26. Doak BC, Over B, Giordanetto F, Kihlberg J. Oral druggable space beyond the rule of 5: insights from drugs and clinical candidates. Chem Biol. 2014;21(9):1115–42.

    Article  CAS  PubMed  Google Scholar 

  27. Edmondson SD, Yang B, Fallan C. Proteolysis targeting chimeras (PROTACs) in ‘beyond rule-of-five’ chemical space: recent Progress and future challenges. Bioorg Med Chem Lett. 2019.

  28. Bergström CAS, Porter CJH. Understanding the challenge of beyond-rule-of-5 compounds. Adv Drug Deliv Rev. 2016;101:1–5.

    Article  PubMed  CAS  Google Scholar 

  29. Choo EF, Boggs J, Zhu C, Lubach JW, Catron ND, Jenkins G, et al. The role of lymphatic transport on the systemic bioavailability of the Bcl-2 protein family inhibitors navitoclax (ABT-263) and ABT-199. Drug Metab Dispos. 2014;42(2):207–12.

    Article  CAS  PubMed  Google Scholar 

  30. Neklesa T, Snyder LB, Willard RR, Vitale N, Pizzano J, Gordon DA, et al. ARV-110: An oral androgen receptor PROTAC degrader for prostate cancer. 2019;37(7_suppl):259.

  31. Flanagan J, Qian Y, Gough S, Andreoli M, Bookbinder M, Cadelina G, et al. Abstract P5–04-18: ARV-471, an oral estrogen receptor PROTAC degrader for breast cancer. 2019;79(4 Supplement):P5–04-18-P5–04-18.

  32. Lau JL, Dunn MK. Therapeutic peptides: historical perspectives, current development trends, and future directions. Bioorg Med Chem. 2018;26(10):2700–7.

    Article  CAS  PubMed  Google Scholar 

  33. Henninot A, Collins JC, Nuss JM. The current state of peptide drug discovery: Back to the future? J Med Chem. 2018;61(4):1382–414.

    Article  CAS  PubMed  Google Scholar 

  34. Zorzi A, Deyle K, Heinis C. Cyclic peptide therapeutics: past, present and future. Curr Opin Chem Biol. 2017;38:24–9.

    Article  CAS  PubMed  Google Scholar 

  35. Aguirre TA, Teijeiro-Osorio D, Rosa M, Coulter IS, Alonso MJ, Brayden DJ. Current status of selected oral peptide technologies in advanced preclinical development and in clinical trials. Adv Drug Deliv Rev. 2016;106(Pt B):223–41.

    Article  CAS  PubMed  Google Scholar 

  36. Linzess (linaclotide). Chemistry review. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2012/202811Orig1s000ChemR.pdf. FDA. 2011.

  37. Tyagi P, Pechenov S, Anand SJ. Oral peptide delivery: translational challenges due to physiological effects. J Control Release. 2018;287:167–76.

    Article  CAS  PubMed  Google Scholar 

  38. Semaglutide Label. FDA;2019. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/213051s000lbl.pdf. Accessed 27 Sept 2019

  39. Octreotide capsules. Chiasma, Inc.; 2019 [Available from: http://www.chiasmapharma.com/octreotide-capsules. Accessed 10 Sept 2019

  40. Stein CA, Castanotto D. FDA-approved oligonucleotide therapies in 2017. Mol Ther. 2017;25(5):1069–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Juliano RL. The delivery of therapeutic oligonucleotides. Nucleic Acids Res. 2016;44(14):6518–48.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Kulkarni JA, Witzigmann D, Chen S, Cullis PR, van der Meel R. Lipid Nanoparticle Technology for Clinical Translation of siRNA Therapeutics. Acc Chem Res. 2019.

  43. Tegsedi (inotersen). Clinical review. 2018. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2018/211172Orig1s000MedR.pdf. Accessed 6 May 2019

  44. Prakash TP, Graham MJ, Yu J, Carty R, Low A, Chappell A, et al. Targeted delivery of antisense oligonucleotides to hepatocytes using triantennary N-acetyl galactosamine improves potency 10-fold in mice. Nucleic Acids Res. 2014;42(13):8796–807.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Maier MA, Jayaraman M, Matsuda S, Liu J, Barros S, Querbes W, et al. Biodegradable lipids enabling rapidly eliminated lipid nanoparticles for systemic delivery of RNAi therapeutics. Mol Ther. 2013;21(8):1570–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. The medicines company presents results from ORION-11, first phase 3 trial of inclisiran, showing durable and potent lowering of LDL-C with twice-yearly dosing. : The Medicines Company. September 2, 2019 [Available from: https://www.themedicinescompany.com/investor/pr/4000045/. Accessed 1 Oct 2019

  47. Arkin MR, Tang Y, Wells JA. Small-molecule inhibitors of protein-protein interactions: progressing toward the reality. Chem Biol. 2014;21(9):1102–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Petta I, Lievens S, Libert C, Tavernier J, De Bosscher K. Modulation of protein-protein interactions for the development of novel therapeutics. Mol Ther. 2016;24(4):707–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Smith MC, Gestwicki JE. Features of protein-protein interactions that translate into potent inhibitors: topology, surface area and affinity. Expert Rev Mol Med. 2012;14:e16.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Doak BC, Zheng J, Dobritzsch D, Kihlberg J. How beyond rule of 5 drugs and clinical candidates bind to their targets. J Med Chem. 2016;59(6):2312–27.

    Article  CAS  PubMed  Google Scholar 

  51. Kramer SD, Aschmann HE, Hatibovic M, Hermann KF, Neuhaus CS, Brunner C, et al. When barriers ignore the "rule-of-five". Adv Drug Deliv Rev. 2016;101:62–74.

    Article  CAS  PubMed  Google Scholar 

  52. Fischer ES, Park E, Eck MJ, Thoma NH. SPLINTS: small-molecule protein ligand interface stabilizers. Curr Opin Struct Biol. 2016;37:115–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Mullard A. Pioneering apoptosis-targeted cancer drug poised for FDA approval. Nat Rev Drug Discov. 2016;15(3):147–9.

    Article  CAS  PubMed  Google Scholar 

  54. Venclexta (venetoclax). Clinical pharmacology and biopharmaceutics review. FDA; 2016. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2016/208573Orig1s000ClinPharmR.pdf. Accessed 6 May 2019

  55. Ottis P, Toure M, Cromm PM, Ko E, Gustafson JL, Crews CM. Assessing different E3 ligases for small molecule induced protein Ubiquitination and degradation. ACS Chem Biol. 2017;12(10):2570–8.

    Article  CAS  PubMed  Google Scholar 

  56. Gu S, Cui D, Chen X, Xiong X, Zhao Y. PROTACs: An Emerging Targeting Technique for Protein Degradation in Drug Discovery. BioEssays. 2018;40(4):e1700247.

    Article  PubMed  CAS  Google Scholar 

  57. Maple HJ, Clayden N, Baron A, Stacey C, Felix R. Developing degraders: principles and perspectives on design and chemical space. MedChemComm. 2019.

  58. Cromm PM, Crews CM. Targeted protein degradation: from chemical biology to drug discovery. Cell Chem Biol. 2017;24(9):1181–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Bondeson DP, Mares A, Smith IED, Ko E, Campos S, Miah AH, et al. Catalytic in vivo protein knockdown by small-molecule PROTACs. Nat Chem Biol. 2015;11(8):611–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Galdeano C. Drugging the undruggable: targeting challenging E3 ligases for personalized medicine. Future Med Chem. 2017;9(4):347–50.

    Article  CAS  PubMed  Google Scholar 

  61. Neklesa TK, Winkler JD, Crews CM. Targeted protein degradation by PROTACs. Pharmacol Ther. 2017;174:138–44.

    Article  CAS  PubMed  Google Scholar 

  62. Crews CM. Inducing protein degradation as a therapeutic strategy. J Med Chem. 2018;61(2):403–4.

    Article  CAS  PubMed  Google Scholar 

  63. Erak M, Bellmann-Sickert K, Els-Heindl S, Beck-Sickinger AG. Peptide chemistry toolbox – transforming natural peptides into peptide therapeutics. Bioorg Med Chem. 2018;26(10):2759–65.

    Article  CAS  PubMed  Google Scholar 

  64. Pelay-Gimeno M, Glas A, Koch O, Grossmann TN. Structure-based Design of Inhibitors of protein-protein interactions: mimicking peptide binding epitopes. Angew Chem Int Engl. 2015;54(31):8896–927.

    Article  CAS  Google Scholar 

  65. Gil-Martin M, Pardo PG, Lopez-Tarruella S, Manso L, Perez-Fidalgo JA, Ademuyiwa FO, et al. Phase I study of the combination of balixafortide (CXCR4 inhibitor) and eribulin in HER2-negative metastatic breast cancer (MBC) patients (pts). 2017;35(15_suppl):2555-.

  66. Lau J, Bloch P, Schäffer L, Pettersson I, Spetzler J, Kofoed J, et al. Discovery of the once-weekly glucagon-like Peptide-1 (GLP-1) analogue Semaglutide. J Med Chem. 2015;58(18):7370–80.

    Article  CAS  PubMed  Google Scholar 

  67. Yu M, Benjamin MM, Srinivasan S, Morin EE, Shishatskaya EI, Schwendeman SP, et al. Battle of GLP-1 delivery technologies. Adv Drug Deliv Rev. 2018;130:113–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Zarei-Ghanavati S, Alizadeh R, Deng S. Topical interferon alpha-2b for treatment of noninvasive ocular surface squamous neoplasia with 360° limbal involvement. J Ophthalmic Vis Res. 2014;9(4):423–6.

    Article  PubMed  PubMed Central  Google Scholar 

  69. McCormack PL. Linaclotide: a review of its use in the treatment of irritable bowel syndrome with constipation. Drugs. 2014;74(1):53–60.

    Article  CAS  PubMed  Google Scholar 

  70. Schwochert J, Turner R, Thang M, Berkeley RF, Ponkey AR, Rodriguez KM, et al. Peptide to Peptoid substitutions increase cell permeability in cyclic Hexapeptides. Org Lett. 2015;17(12):2928–31.

    Article  CAS  PubMed  Google Scholar 

  71. Whitty A, Zhong M, Viarengo L, Beglov D, Hall DR, Vajda S. Quantifying the chameleonic properties of macrocycles and other high-molecular-weight drugs. Drug Discov Today. 2016;21(5):712–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Over B, Matsson P, Tyrchan C, Artursson P, Doak BC, Foley MA, et al. Structural and conformational determinants of macrocycle cell permeability. Nat Chem Biol. 2016;12(12):1065–74.

    Article  CAS  PubMed  Google Scholar 

  73. Rezai T, Bock JE, Zhou MV, Kalyanaraman C, Lokey RS, Jacobson MP. Conformational flexibility, internal hydrogen bonding, and passive membrane permeability: successful in silico prediction of the relative permeabilities of cyclic peptides. J Am Chem Soc. 2006;128(43):14073–80.

    Article  CAS  PubMed  Google Scholar 

  74. Knudsen LB, Lau J. The Discovery and Development of Liraglutide and Semaglutide. 2019;10(155).

  75. Buckley ST, Bækdal TA, Vegge A, Maarbjerg SJ, Pyke C, Ahnfelt-Rønne J, et al. Transcellular stomach absorption of a derivatized glucagon-like peptide-1 receptor agonist. 2018;10(467):eaar7047.

  76. Tuvia S, Pelled D, Marom K, Salama P, Levin-Arama M, Karmeli I, et al. A novel suspension formulation enhances intestinal absorption of macromolecules via transient and reversible transport mechanisms. Pharm Res. 2014;31(8):2010–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Mullard A. FDA approves landmark RNAi drug. Nat Rev Drug Discov. 2018;17:613.

    PubMed  Google Scholar 

  78. Khorkova O, Hsiao J, Wahlestedt C. Basic biology and therapeutic implications of lncRNA. Adv Drug Deliv Rev. 2015;87:15–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Juliano RL, Ming X, Carver K, Laing B. Cellular uptake and intracellular trafficking of oligonucleotides: implications for oligonucleotide pharmacology. Nucleic Acid Ther. 2014;24(2):101–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Abe K, Fujiyoshi Y. Cryo-electron microscopy for structure analyses of membrane proteins in the lipid bilayer. Curr Opin Struct Biol. 2016;39:71–8.

    Article  CAS  PubMed  Google Scholar 

  81. Doherty GJ, McMahon HT. Mechanisms of endocytosis. Annu Rev Biochem. 2009;78:857–902.

    Article  CAS  PubMed  Google Scholar 

  82. Eckstein F. Phosphorothioates, essential components of therapeutic oligonucleotides. Nucleic Acid Ther. 2014;24(6):374–87.

    Article  CAS  PubMed  Google Scholar 

  83. Agrawal S, Goodchild J, Civeira MP, Thornton AH, Sarin PS, Zamecnik PC. Oligodeoxynucleoside phosphoramidates and phosphorothioates as inhibitors of human immunodeficiency virus. Proc Natl Acad Sci U S A. 1988;85(19):7079–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Dirin M, Winkler J. Influence of diverse chemical modifications on the ADME characteristics and toxicology of antisense oligonucleotides. Expert Opin Biol Ther. 2013;13(6):875–88.

    Article  CAS  PubMed  Google Scholar 

  85. Stanton MG, Colletti SL. Medicinal chemistry of siRNA delivery. J Med Chem. 2010;53(22):7887–901.

    Article  CAS  PubMed  Google Scholar 

  86. Kenski DM, Cooper AJ, Li JJ, Willingham AT, Haringsma HJ, Young TA, et al. Analysis of acyclic nucleoside modifications in siRNAs finds sensitivity at position 1 that is restored by 5′-terminal phosphorylation both in vitro and in vivo. Nucleic Acids Res. 2010;38(2):660–71.

    Article  CAS  PubMed  Google Scholar 

  87. Parmar R, Willoughby JL, Liu J, Foster DJ, Brigham B, Theile CS, et al. 5′-(E)-Vinylphosphonate: a stable phosphate mimic can improve the RNAi activity of siRNA-GalNAc conjugates. Chembiochem. 2016;17(11):985–9.

    Article  CAS  PubMed  Google Scholar 

  88. Lam JK, Chow MY, Zhang Y, Leung SW. siRNA Versus miRNA as Therapeutics for Gene Silencing. Mol Ther Nucleic Acids. 2015;4:e252.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Stephenson ML, Zamecnik PC. Inhibition of Rous sarcoma viral RNA translation by a specific oligodeoxyribonucleotide. Proc Natl Acad Sci U S A. 1978;75(1):285–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Weidner DA, Valdez BC, Henning D, Greenberg S, Busch H. Phosphorothioate oligonucleotides bind in a non sequence-specific manner to the nucleolar protein C23/nucleolin. FEBS Lett. 1995;366(2–3):146–50.

    Article  CAS  PubMed  Google Scholar 

  91. Aartsma-Rus A. New momentum for the field of oligonucleotide therapeutics. Mol Ther. 2016;24(2):193–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Verma A. Recent advances in antisense oligonucleotide therapy in genetic neuromuscular diseases. Ann Indian Acad Neurol. 2018;21(1):3–8.

    PubMed  PubMed Central  Google Scholar 

  93. Shen X, Corey DR. Chemistry, mechanism and clinical status of antisense oligonucleotides and duplex RNAs. Nucleic Acids Res. 2018;46(4):1584–600.

    Article  CAS  PubMed  Google Scholar 

  94. Yu RZ, Graham MJ, Post N, Riney S, Zanardi T, Hall S, et al. Disposition and pharmacology of a GalNAc3-conjugated ASO targeting human lipoprotein (a) in mice. Mol Ther Nucleic Acids. 2016;5:e317.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Willoughby JLS, Chan A, Sehgal A, Butler JS, Nair JK, Racie T, et al. Evaluation of GalNAc-siRNA conjugate activity in pre-clinical animal models with reduced Asialoglycoprotein receptor expression. Mol Ther. 2018;26(1):105–14.

    Article  CAS  PubMed  Google Scholar 

  96. Scharner J, Qi S, Rigo F, Bennett CF, Krainer AR. Delivery of GalNAc-conjugated splice-switching ASOs to non-hepatic cells through ectopic expression of Asialoglycoprotein receptor. Mol Ther Nucleic Acids. 2019;16:313–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Egli M, Manoharan M. Re-engineering RNA molecules into therapeutic agents. Acc Chem Res. 2019;52(4):1036–47.

    Article  CAS  PubMed  Google Scholar 

  98. Khvorova A, Watts JK. The chemical evolution of oligonucleotide therapies of clinical utility. Nat Biotechnol. 2017;35(3):238–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998;391(6669):806–11.

    Article  CAS  PubMed  Google Scholar 

  100. Zimmermann TS, Lee AC, Akinc A, Bramlage B, Bumcrot D, Fedoruk MN, et al. RNAi-mediated gene silencing in non-human primates. Nature. 2006;441(7089):111–4.

    Article  CAS  PubMed  Google Scholar 

  101. Alnylam Pharmaceuticals I. Methods of treating transthyretin (TTR) mediated amyloidosis. US10060921B2. 2018.

  102. Sabnis S, Kumarasinghe ES, Salerno T, Mihai C, Ketova T, Senn JJ, et al. A novel amino lipid series for mRNA delivery: improved Endosomal escape and sustained pharmacology and safety in non-human primates. Mol Ther. 2018;26(6):1509–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Givosiran Label. FDA; 2019. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/0212194s000lbl.pdf. Accessed 22 Nov 2019

  104. Gan LM, Lagerstrom-Fermer M, Carlsson LG, Arfvidsson C, Egnell AC, Rudvik A, et al. Intradermal delivery of modified mRNA encoding VEGF-A in patients with type 2 diabetes. Nat Commun. 2019;10(1):871.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Patra JK, Das G, Fraceto LF, Campos EVR, Rodriguez-Torres MDP, Acosta-Torres LS, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnol. 2018;16(1):71.

    Article  CAS  Google Scholar 

  106. Landis MS, Bhattachar S, Yazdanian M, Morrison J. Commentary: why pharmaceutical scientists in early drug discovery are critical for influencing the design and selection of optimal drug candidates. AAPS PharmSciTech. 2018;19(1):1–10.

    Article  CAS  PubMed  Google Scholar 

  107. Bhattachar SN, Bender DM, Sweetana SA, Wesley JA. Discovery formulations: approaches and practices in early preclinical development. In: Templeton AC, Byrn SR, Haskell RJ, Prisinzano TE, editors. Discovering and developing molecules with optimal drug-like properties. New York: Springer New York; 2015. p. 49–94.

    Chapter  Google Scholar 

  108. Yang NJ, Hinner MJ. Getting across the cell membrane: an overview for small molecules, peptides, and proteins. Methods Mol Biol. 2015;1266:29–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Transforming the market. Rani Therapeutics; 2019. Available from: https://www.ranitherapeutics.com/. Accessed 1 Oct 2019

  110. Abramson A, Caffarel-Salvador E, Khang M, Dellal D, Silverstein D, Gao Y, et al. An ingestible self-orienting system for oral delivery of macromolecules. Science. 2019;363(6427):611–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

The authors thank Carolyn A. Heusser at Novartis Institutes for BioMedical Research for carefully reviewing the manuscripts and providing helpful comments for revision. The authors also thank Sara Danforth at Novartis Institutes for BioMedical Research for her key assistance on creating the figures.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Wenzhan Yang.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

ESM 1

(DOCX 2244 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, W., Gadgil, P., Krishnamurthy, V.R. et al. The Evolving Druggability and Developability Space: Chemically Modified New Modalities and Emerging Small Molecules. AAPS J 22, 21 (2020). https://doi.org/10.1208/s12248-019-0402-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1208/s12248-019-0402-2

KEY WORDS

Navigation