1932

Abstract

Innovative formulation technologies can play a crucial role in transforming a novel molecule to a medicine that significantly enhances patients’ lives. Improved mechanistic understanding of diseases has inspired researchers to expand the druggable space using new therapeutic modalities such as interfering RNA, protein degraders, and novel formats of monoclonal antibodies. Sophisticated formulation strategies are needed to deliver the drugs to their sites of action and to achieve patient centricity, exemplified by messenger RNA vaccines and oral peptides. Moreover, access to medical information via digital platforms has resulted in better-informed patient groups that are requesting consideration of their needs during drug development. This request is consistent with health authority efforts to upgrade their regulations to advance age-appropriate product development for patients. This review describes formulation innovations contributingto improvements in patient care: convenience of administration, preferred route of administration, reducing dosing burden, and achieving targeted delivery of new modalities.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pharmtox-052120-093517
2022-01-06
2024-05-11
Loading full text...

Full text loading...

/deliver/fulltext/pharmtox/62/1/annurev-pharmtox-052120-093517.html?itemId=/content/journals/10.1146/annurev-pharmtox-052120-093517&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Seyhan AA, Carini C. 2019. Are innovation and new technologies in precision medicine paving a new era in patients centric care?. J. Transl. Med. 17:1114
    [Google Scholar]
  2. 2. 
    Yeoman G, Furlong P, Seres M, Binder H, Chung H et al. 2017. Defining patient centricity with patients for patients and caregivers: a collaborative endeavour. BMJ Innov 3:276–83
    [Google Scholar]
  3. 3. 
    Burke MD, Keeney M, Kleinberg R, Burlage R 2019. Challenges and opportunities for patient centric drug product design: industry perspectives. Pharm. Res. 36:685
    [Google Scholar]
  4. 4. 
    Hoos A, Anderson J, Boutin M, Dewulf L, Geissler J et al. 2015. Partnering with patients in the development and lifecycle of medicines. Ther. Innov. Regul. Sci. 49:6929–39
    [Google Scholar]
  5. 5. 
    Menditto E, Orlando V, Rosa GD, Minghetti P, Musazzi UM et al. 2020. Patient centric pharmaceutical drug product design—the impact on medication adherence. Pharmaceutics 12:144
    [Google Scholar]
  6. 6. 
    Khan MS, Roberts MS. 2018. Challenges and innovations of drug delivery in older age. Adv. Drug Deliv. Rev. 135:3–38
    [Google Scholar]
  7. 7. 
    Gutierrez L, Cauchon NS, Christian TR, Giffin M, Abernathy MJ 2020. The confluence of innovation in therapeutics and regulation: recent CMC considerations. J. Pharm. Sci. 109:123524–34
    [Google Scholar]
  8. 8. 
    Blanco M-J, Gardinier KM. 2020. New chemical modalities and strategic thinking in early drug discovery. ACS Med. Chem. Lett. 11:3228–31
    [Google Scholar]
  9. 9. 
    Valeur E, Guéret SM, Adihou H, Gopalakrishnan R, Lemurell M et al. 2017. New modalities for challenging targets in drug discovery. Angew. Chem. Int. Ed. 56:3510294–323
    [Google Scholar]
  10. 10. 
    Yang W, Gadgil P, Krishnamurthy VR, Landis M, Mallick P et al. 2020. The evolving druggability and developability space: chemically modified new modalities and emerging small molecules. AAPS J 22:221
    [Google Scholar]
  11. 11. 
    Mullard A. 2020. 2019. FDA drug approvals. Nat. Rev. Drug Discov. 19:279–84
    [Google Scholar]
  12. 12. 
    Shaer DA, Musaimi OA, Albericio F, de la Torre BG. 2020. 2019. FDA TIDES (peptides and oligonucleotides) harvest. Pharmaceuticals 13:340
    [Google Scholar]
  13. 13. 
    Anderson SL, Beutel TR, Trujillo JM. 2020. Oral semaglutide in type 2 diabetes. J. Diabetes Complicat. 34:4107520
    [Google Scholar]
  14. 14. 
    Isaacs DM, Kruger DF, Spollett GR. 2020. Optimizing therapeutic outcomes with oral semaglutide: a patient-centered approach. Diabetes Spectr 34:17–19
    [Google Scholar]
  15. 15. 
    FDA (US Food Drug Admin.) 2020. Advancing health through innovation: new drug therapy approvals 2019 Rep., Cent. Drug Eval. Res., FDA Silver Spring, MD: https://www.fda.gov/media/134493/download
  16. 16. 
    FDA (US Food Drug Admin.) 2021. Advancing health through innovation: new drug therapy approvals 2020 Rep., Cent. Drug Eval. Res., FDA Silver Spring, MD: https://mcusercontent.com/7a354eb20ba2e6447b22e9a28/files/ac585a6b-5cf0-498d-9558-97770306587b/FINAL_NewDrugsApprovalReport_Final2020_210108_0948_FINAL.pdf
  17. 17. 
    FDA (US Food Drug Admin.) 2016. Best Pharmaceuticals for Children Act and Pediatric Research Equity Act: July 2016 status report to Congress Rep., Dept. Health Hum. Serv FDA, Silver Spring, MD: https://www.fda.gov/media/99184/download
  18. 18. 
    Comm. Eur. Parliam. Counc 2017. State of paediatric medicines in the EU: 10 years of the EU Paediatric Regulation Rep., Eur. Med. Agency Amsterdam, Neth: https://ec.europa.eu/health/sites/default/files/files/paediatrics/docs/2017_childrensmedicines_report_en.pdf
  19. 19. 
    Walsh J, Ranmal SR, Ernest TB, Liu F 2018. Patient acceptability, safety and access: a balancing act for selecting age-appropriate oral dosage forms for paediatric and geriatric populations. Int. J. Pharm. 536:2547–62
    [Google Scholar]
  20. 20. 
    Comm. Med. Prod. Hum. Use 2020. Reflection paper on the pharmaceutical development of medicines for use in the older population Rep., Eur. Med. Agency, Amsterdam, Neth https://www.ema.europa.eu/en/documents/scientific-guideline/reflection-paper-pharmaceutical-development-medicines-use-older-population-first-version_en.pdf
  21. 21. 
    NIH (Natl. Inst. Health) 2020. DailyMed database Database, Natl. Libr. Med NIH, Bethesda, MD: https://dailymed.nlm.nih.gov
  22. 22. 
    Datapharm 2020. Electronic medicines compendium Database, Datapharm Ltd Leatherhead, UK: https://www.medicines.org.uk/emc
  23. 23. 
    Walsh J, Cram A, Woertz K, Breitkreutz J, Winzenburg G et al. 2014. Playing hide and seek with poorly tasting paediatric medicines: Do not forget the excipients. Adv. Drug Deliv. Rev. 73:14–33
    [Google Scholar]
  24. 24. 
    Berkland C, Dormer N. 2017. Controlled release—leveraging Precision Particle Fabrication® technology to create patient-friendly dosage forms. Drug Development & Delivery May. https://drug-dev.com/controlled-release-leveraging-precision-particle-fabrication-technology-to-create-patient-friendly-dosage-forms/
    [Google Scholar]
  25. 25. 
    Waybrant B. 2017. Enabling taste-masking with lipid-based multiparticulate dosage forms Poster presented at the AAPS Annual Meeting and Exposition San Diego, CA:
  26. 26. 
    Spitz B, Nonoyama A, Miller C, Berkland C, Dormer N. 2019. Taste masking and immediate release: You can have both. Paper presented at the 11th Conference of the European Paediatric Formulation Initiative Malmö, Sweden:
  27. 27. 
    Strickley RG. 2018. Pediatric oral formulations: an updated review of commercially available pediatric oral formulations since 2007. J. Pharm. Sci. 108:41335–65
    [Google Scholar]
  28. 28. 
    Lee HS, Lee J-J, Kim M-G, Kim K-T, Cho C-W et al. 2019. Sprinkle formulations—a review of commercially available products. Asian J. Pharm. Sci. 15:3292–310
    [Google Scholar]
  29. 29. 
    Deleted in proof
  30. 30. 
    FDA (US Food Drug Admin.) 2018. Use of liquids and/or soft foods as vehicles for drug administration: general considerations for selection and in vitro methods for product quality assessments Draft Guid., Cent. Drug Eval. Res., FDA Silver Spring, MD: https://www.fda.gov/media/114872/download
  31. 31. 
    Cilurzo F, Musazzi UM, Franzé S, Selmin F, Minghetti P. 2017. Orodispersible dosage forms: biopharmaceutical improvements and regulatory requirements. Drug Discov. Today 23:2251–59
    [Google Scholar]
  32. 32. 
    Slavkova M, Breitkreutz J. 2015. Orodispersible drug formulations for children and elderly. Eur. J. Pharm. Sci. 75:2–9
    [Google Scholar]
  33. 33. 
    Seager H. 1998. Drug-delivery products and the Zydis fast-dissolving dosage form. J. Pharm. Pharmacol. 50:375–82
    [Google Scholar]
  34. 34. 
    Cui M, Pan H, Fang D, Qiao S, Wang S, Pan W 2020. Fabrication of high drug loading levetiracetam tablets using semi-solid extrusion 3D printing. J. Drug Deliv. Sci. Technol. 57:101683
    [Google Scholar]
  35. 35. 
    Zhu X, Li H, Huang L, Zhang M, Fan W, Cui L 2020. 3D printing promotes the development of drugs. Biomed. Pharmacother. 131:110644
    [Google Scholar]
  36. 36. 
    Elkasabgy NA, Mahmoud AA, Maged A 2020. 3D printing: an appealing route for customized drug delivery systems. Int. J. Pharm. 588:119732
    [Google Scholar]
  37. 37. 
    Atkinson MJ, Sinha A, Hass SL, Colman SS, Kumar RN et al. 2004. Validation of a general measure of treatment satisfaction, the Treatment Satisfaction Questionnaire for Medication (TSQM), using a national panel study of chronic disease. Health Qual. Life Outcomes 2:12
    [Google Scholar]
  38. 38. 
    Malori A. 2018. Oral drug reconstitution: making it easy and accurate via packaging innovation. ONdrugDelivery 88:18–21
    [Google Scholar]
  39. 39. 
    Rosemont Pharm 2020. For the first licensed liquid omeprazole. 2020. Rosemont Pharmaceuticals https://www.rosemontpharma.com/omeprazole
    [Google Scholar]
  40. 40. 
    Shah N, Iyer RM, Mair H, Choi DS, Tian H et al. 2013. Improved human bioavailability of vemurafenib, a practically insoluble drug, using an amorphous polymer-stabilized solid dispersion prepared by a solvent-controlled coprecipitation process. J. Pharm. Sci. 102:3967–81
    [Google Scholar]
  41. 41. 
    Jain S, Patel N, Lin S 2014. Solubility and dissolution enhancement strategies: current understanding and recent trends. Drug Dev. Ind. Pharm. 41:6875–87
    [Google Scholar]
  42. 42. 
    Göke K, Lorenz T, Repanas A, Schneider F, Steiner D et al. 2018. Novel strategies for the formulation and processing of poorly water-soluble drugs. Eur. J. Pharm. Biopharm. 126:40–56
    [Google Scholar]
  43. 43. 
    Tan DK, Davis DA, Miller DA, Williams RO, Nokhodchi A 2020. Innovations in thermal processing: hot-melt extrusion and KinetiSol® dispersing. AAPS PharmSciTech 21:8312
    [Google Scholar]
  44. 44. 
    Glover ZWK, Gennaro L, Yadav S, Demeule B, Wong PY, Sreedhara A. 2013. Compatibility and stability of pertuzumab and trastuzumab admixtures in i.v. infusion bags for coadministration. J. Pharm. Sci. 102:3794–812
    [Google Scholar]
  45. 45. 
    Kirschbrown WP, Wynne C, Kågedal M, Wada R, Li H et al. 2019. Development of a subcutaneous fixed-dose combination of pertuzumab and trastuzumab: results from the phase Ib dose-finding study. J. Clin. Pharmacol. 59:5702–16
    [Google Scholar]
  46. 46. 
    Gao JJ, Osgood CL, Gong Y, Zhang H, Bloomquist EW et al. 2020. FDA approval summary: pertuzumab, trastuzumab, and hyaluronidase-zzxf injection for subcutaneous use in patients with HER2-positive breast cancer. Clin. Cancer Res 27:82126–129
    [Google Scholar]
  47. 47. 
    Bittner B, Richter W, Schmidt J. 2018. Subcutaneous administration of biotherapeutics: an overview of current challenges and opportunities. BioDrugs 32:5425–40
    [Google Scholar]
  48. 48. 
    Pivot X, Semiglazov V, Chen S, Moodley SD, Manihkas A et al. 2012. Subcutaneous injection of trastuzumab—analysis of administration time and injection site reactions. Ann. Oncol. 23:Suppl. 9ix103
    [Google Scholar]
  49. 49. 
    Gatlin L, Gatlin C 1999. Minimizing injection pain and damage. Injectable Drug Development: Techniques to Reduce Pain and Irritation PK Gupta, GA Brazeau 401–21 Boca Raton, FL: CRC Press
    [Google Scholar]
  50. 50. 
    Locke KW, Maneval DC, LaBarre MJ. 2019. ENHANZE® drug delivery technology: a novel approach to subcutaneous administration using recombinant human hyaluronidase 20. Drug Deliv 26:198–106
    [Google Scholar]
  51. 51. 
    Garidel P, Kuhn AB, Schäfer LV, Karow-Zwick AR, Blech M. 2017. High-concentration protein formulations: How high is high?. Eur. J. Pharm. Biopharm. 119:353–60
    [Google Scholar]
  52. 52. 
    Shire SJ. 2015. Strategies to deal with challenges of developing high-concentration subcutaneous (SC) formulations for monoclonal antibodies (mAbs). Monoclonal Antibodies: Meeting the Challenges in Manufacturing, Formulation, Delivery and Stability of Final Drug Product139–52 Cambridge, UK: Woodhead Publ.
    [Google Scholar]
  53. 53. 
    Brayden DJ, Hill TA, Fairlie DP, Maher S, Mrsny RJ. 2020. Systemic delivery of peptides by the oral route: formulation and medicinal chemistry approaches. Adv. Drug Deliv. Rev. 157:2–36
    [Google Scholar]
  54. 54. 
    Buckley ST, Bækdal TA, Vegge A, Maarbjerg SJ, Pyke C et al. 2018. Transcellular stomach absorption of a derivatized glucagon-like peptide-1 receptor agonist. Sci. Transl. Med. 10:467eaar7047
    [Google Scholar]
  55. 55. 
    Zizzari AT, Pliatsika D, Gall FM, Fischer T, Riedl R. 2021. New perspectives in oral peptide delivery. Drug Discov. Today 26:1097–105
    [Google Scholar]
  56. 56. 
    Balson MF-M, Leeuw BD. 2020. The rise of complex oral delivery systems. ONdrugDelivery 109:35–39
    [Google Scholar]
  57. 57. 
    Abbasi J. 2020. Biotech innovations—oral injections tested in proof-of-concept trial. JAMA 323:10916
    [Google Scholar]
  58. 58. 
    Progenity 2021. Progenity initiates safety and tolerability study of its smart capsule-based oral drug delivery system for GI diseases Press Release, Febr 17 https://investors.progenity.com/news-releases/news-release-details/progenity-initiates-safety-and-tolerability-study-its-smart
  59. 59. 
    Han L, Tang C, Yin C. 2014. Oral delivery of shRNA and siRNA via multifunctional polymeric nanoparticles for synergistic cancer therapy. Biomaterials 35:154589–600
    [Google Scholar]
  60. 60. 
    Coffey JW, Gaiha GD, Traverso G. 2021. Oral biologic delivery: advances toward oral subunit, DNA, and mRNA vaccines and the potential for mass vaccination during pandemics. Annu. Rev. Pharmacol. Toxicol. 61:517–40
    [Google Scholar]
  61. 61. 
    Crawford L, Rosch J, Putnam D. 2016. Concepts, technologies, and practices for drug delivery past the blood-brain barrier to the central nervous system. J. Control. Release 240:251–66
    [Google Scholar]
  62. 62. 
    Arora J, Saville S, Waybrant B 2018. Multiparticulate system—advances in lipid multiparticulate technologies for controlled release. Drug Development & Delivery June. https://drug-dev.com/multiparticulate-system-advances-in-lipid-multiparticulate-technologies-for-controlled-release/
    [Google Scholar]
  63. 63. 
    Davis SS 2005. Formulation strategies for absorption windows. Drug Discov. Today 10:249–57
    [Google Scholar]
  64. 64. 
    Laffleur F, Keckeis V. 2020. Advances in drug delivery systems: work in progress still needed?. Int. J. Pharm. 590:119912
    [Google Scholar]
  65. 65. 
    Lopes CM, Bettencourt C, Rossi A, Buttini F, Barata P. 2016. Overview on gastroretentive drug delivery systems for improving drug bioavailability. Int. J. Pharm. 510:1144–58
    [Google Scholar]
  66. 66. 
    Navon N. 2019. The Accordion Pill®: unique oral delivery to enhance pharmacokinetics and therapeutic benefit of challenging drugs. Ther. Deliv. 10:7433–42
    [Google Scholar]
  67. 67. 
    Deleted in proof
  68. 68. 
    Kanasty R, Low S, Bhise N, Yang J, Peeke E et al. 2019. A pharmaceutical answer to nonadherence: once weekly oral memantine for Alzheimer's disease. J. Control. Release 303:34–41
    [Google Scholar]
  69. 69. 
    LeWitt PA, Giladi N, Navon N 2019. Pharmacokinetics and efficacy of a novel formulation of carbidopa-levodopa (Accordion Pill®) in Parkinson's disease. Parkinsonism Relat. Disord 65:131–38
    [Google Scholar]
  70. 70. 
    Lewis AL, Richard J. 2015. Challenges in the delivery of peptide drugs: an industry perspective. Ther. Deliv. 6:2149–63
    [Google Scholar]
  71. 71. 
    Maji SK, Schubert D, Rivier C, Lee S, Rivier JE, Riek R. 2008. Amyloid as a depot for the formulation of long-acting drugs. PLOS Biol 6:2e17
    [Google Scholar]
  72. 72. 
    Roberge C, Cros J-M, Serindoux J, Cagnon M-E, Samuel R et al. 2020. BEPO®: bioresorbable diblock mPEG-PDLLA and triblock PDLLA-PEG-PDLLA based in situ forming depots with flexible drug delivery kinetics modulation. J. Control. Release 319:416–27
    [Google Scholar]
  73. 73. 
    Wright J, Matriano J 2016. Long acting injections and implants. Drug Delivery: Fundamentals & Applications A Hillery, K Park 137–70 Boca Raton, FL: CRC Press
    [Google Scholar]
  74. 74. 
    Pavel M, Borson-Chazot F, Cailleux A, Hörsch D, Lahner H et al. 2019. Octreotide SC depot in patients with acromegaly and functioning neuroendocrine tumors: a phase 2, multicenter study. Cancer Chemother. Pharm. 83:2375–85
    [Google Scholar]
  75. 75. 
    Adamis AP, Brittain CJ, Dandekar A, Hopkins JJ 2020. Building on the success of anti-vascular endothelial growth factor therapy: a vision for the next decade. Eye 34:111966–72
    [Google Scholar]
  76. 76. 
    Famili A, Crowell SR, Loyet KM, Mandikian D, Boswell CA et al. 2019. Hyaluronic acid-antibody fragment bioconjugates for extended ocular pharmacokinetics. Bioconjug. Chem 30:112782–89
    [Google Scholar]
  77. 77. 
    Campochiaro PA, Marcus DM, Awh CC, Regillo C, Adamis AP et al. 2019. The port delivery system with ranibizumab for neovascular age-related macular degeneration results from the randomized phase 2 Ladder clinical trial. Ophthalmology 126:81141–54
    [Google Scholar]
  78. 78. 
    Thackaberry EA, Farman C, Zhong F, Lorget F, Staflin K et al. 2017. Evaluation of the toxicity of intravitreally injected PLGA microspheres and rods in monkeys and rabbits: effects of depot size on inflammatory response. Investig. Ophth. Vis. Sci. 58:104274–85
    [Google Scholar]
  79. 79. 
    Fernández-García R, Prada M, Bolás-Fernández F, Ballesteros MP, Serrano DR. 2020. Oral fixed-dose combination pharmaceutical products: industrial manufacturing versus personalized 3D printing. Pharm. Res. 37:7132
    [Google Scholar]
  80. 80. 
    Sadia M, Isreb A, Abbadi I, Isreb M, Aziz D et al. 2018. From ‘fixed dose combinations’ to ‘a dynamic dose combiner’: 3D printed bi-layer antihypertensive tablets. Eur. J. Pharm. Sci. 123:484–94
    [Google Scholar]
  81. 81. 
    Fang D, Yang Y, Cui M, Pan H, Wang L et al. 2021. Three-dimensional (3D)-printed zero-order released platform: a novel method of personalized dosage form design and manufacturing. AAPS PharmSciTech 22:137
    [Google Scholar]
  82. 82. 
    Varum F, Freire AC, Fadda HM, Bravo R, Basit AW. 2020. A dual pH and microbiota-triggered coating (Phloral™) for fail-safe colonic drug release. Int. J. Pharm. 583:119379
    [Google Scholar]
  83. 83. 
    Packiaraj JM, Venkateswaran CS, Janakiraman K 2013. Formulation and evaluation of modified-release tablets of corticosteroid. Indo Am. J. Pharm. Res 3:18
    [Google Scholar]
  84. 84. 
    Deleted in proof
  85. 85. 
    Palmer PP, Hamel LG, Dasu B. 2013. Sufentanil NanoTab technology: revolutionizing patient-controlled analgesia. Drug Development & Delivery April. https://drug-dev.com/therapeutic-focus-sufentanil-nanotab-technology-revolutionizing-patient-controlled-analgesia/
    [Google Scholar]
  86. 86. 
    Deleted in proof
  87. 87. 
    Borkar N, Mu H, Holm R. 2018. Challenges and trends in apomorphine drug delivery systems for the treatment of Parkinson's disease. Asian J. Pharm. Sci. 13:6507–17
    [Google Scholar]
  88. 88. 
    Deleted in proof
  89. 89. 
    Rathbone MJ, Pather I, Şenel S 2015. Overview of oral mucosal delivery. Oral Mucosal Drug Delivery and Therapy MJ Rathbone, S Şenel, I Pather 17–29 New York: Springer
    [Google Scholar]
  90. 90. 
    Deleted in proof
  91. 91. 
    Quinn M, Duval V 2020. Röchling Medical's Sympfiny®. ONdrugDelivery 109:20–21
    [Google Scholar]
  92. 92. 
    Tumuluri V 2020. Pharmaceutical mini-tablets: a revived trend. Drug Delivery Trends, Vol. 3: Expectations and Realities of Multifunctional Drug Delivery Systems R Shegokar 123–39 Amsterdam: Elsevier
    [Google Scholar]
  93. 93. 
    Sun X, Gao H, Yang Y, He M, Wu Y et al. 2019. PROTACs: great opportunities for academia and industry. Signal Transduct. Target Ther. 4:164
    [Google Scholar]
  94. 94. 
    Berlin M. 2020. Targeted protein degradation as a clinical stage modality: insights from ARV-110 and other PROTAC protein degraders Rep., Arvinas New Haven, CT:
  95. 95. 
    Cantrill C, Chaturvedi P, Rynn C, Schaffland JP, Walter I, Wittwer MB 2020. Fundamental aspects of DMPK optimization of targeted protein degraders. Drug Discov. Today 25:6969–82
    [Google Scholar]
  96. 96. 
    Arvinas 2020. Arvinas releases interim clinical data further demonstrating the powerful potential of PROTAC protein degraders ARV-471 and ARV-110 Press Release Dec. 14. https://ir.arvinas.com/node/8056/pdf
  97. 97. 
    Rathod D, Fu Y, Patel K. 2019. BRD4 PROTAC as a novel therapeutic approach for the treatment of vemurafenib resistant melanoma: preformulation studies, formulation development and in vitro evaluation. Eur. J. Pharm. Sci. 138:105039
    [Google Scholar]
  98. 98. 
    Saraswat A, Patki M, Fu Y, Barot S, Dukhande VV, Patel K. 2020. Nanoformulation of PROteolysis TArgeting Chimera targeting ‘undruggable’ c-Myc for the treatment of pancreatic cancer. Nanomedicine 15:181761–77
    [Google Scholar]
  99. 99. 
    Hu B, Zhong L, Weng Y, Peng L, Huang Y et al. 2020. Therapeutic siRNA: state of the art. Signal Transduct. Target. Ther. 5:1101
    [Google Scholar]
  100. 100. 
    Springer AD, Dowdy SF. 2018. GalNAc-siRNA conjugates: leading the way for delivery of RNAi therapeutics. Nucleic Acid Ther 28:3109–18
    [Google Scholar]
  101. 101. 
    Gosse C, Boutorine A, Aujard I, Chami M, Kononov A et al. 2004. Micelles of lipid−oligonucleotide conjugates: implications for membrane anchoring and base pairing. J. Phys. Chem. B 108:206485–97
    [Google Scholar]
  102. 102. 
    Nair JK, Willoughby JLS, Chan A, Charisse K, Alam MR et al. 2014. Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J. Am. Chem. Soc. 136:4916958–61
    [Google Scholar]
  103. 103. 
    Poecheim J, Graeser KA, Hoernschemeyer J, Becker G, Storch K, Printz M. 2018. Development of stable liquid formulations for oligonucleotides. Eur. J. Pharm. Biopharm. 129:80–87
    [Google Scholar]
  104. 104. 
    de Fougerolles AR. 2008. Delivery vehicles for small interfering RNA in vivo. Hum. Gene Ther. 19:2125–32
    [Google Scholar]
  105. 105. 
    Morrissey DV, Lockridge JA, Shaw L, Blanchard K, Jensen K et al. 2005. Potent and persistent in vivo anti-HBV activity of chemically modified siRNAs. Nat. Biotechnol. 23:81002–7
    [Google Scholar]
  106. 106. 
    Tam YYC, Chen S, Cullis PR 2013. Advances in lipid nanoparticles for siRNA delivery. Pharmaceutical 5:3498–507
    [Google Scholar]
  107. 107. 
    Juliano RL, Akhtar S. 1992. Liposomes as a drug delivery system for antisense oligonucleotides. Antisense Res. Dev. 2:2165–76
    [Google Scholar]
  108. 108. 
    Rungta RL, Choi HB, Lin PJ, Ko RW, Ashby D et al. 2013. Lipid nanoparticle delivery of siRNA to silence neuronal gene expression in the brain. Mol. Ther. Nucleic Acids 2:11e136
    [Google Scholar]
  109. 109. 
    Hoy SM. 2018. Patisiran: first global approval. Drugs 78:151625–31
    [Google Scholar]
  110. 110. 
    Deleted in proof
  111. 111. 
    Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. 2016. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv. Drug Deliv. Rev. 99:Pt. A28–51
    [Google Scholar]
  112. 112. 
    Sun D, Sun W, Gao S-Q, Wei C, Naderi A et al. 2021. Formulation and efficacy of ECO/pRHO-ABCA4-SV40 nanoparticles for nonviral gene therapy of Stargardt disease in a mouse model. J. Control. Release 330:329–40
    [Google Scholar]
  113. 113. 
    Zhou Z, Liu X, Zhu D, Wang Y, Zhang Z et al. 2017. Nonviral cancer gene therapy: delivery cascade and vector nanoproperty integration. Adv. Drug Deliv. Rev. 115:115–54
    [Google Scholar]
  114. 114. 
    Coelho T, Adams D, Silva A, Lozeron P, Hawkins PN et al. 2013. Safety and efficacy of RNAi therapy for transthyretin amyloidosis. N. Engl. J. Med. 369:9819–29
    [Google Scholar]
  115. 115. 
    Verbeke R, Lentacker I, Smedt SCD, Dewitte H. 2019. Three decades of messenger RNA vaccine development. Nano Today 28:100766
    [Google Scholar]
  116. 116. 
    ModernaTX 2020. Moderna COVID-19 vaccine. FDA Brief. Doc., FDA Silver Spring, MD: https://www.fda.gov/media/144434/download
  117. 117. 
    Pfizer, BioNTech 2020. Pfizer-BioNTech COVID-19 vaccine FDA Brief. Doc: FDA, Silver Spring, MD https://www.fda.gov/media/144245/download
  118. 118. 
    Carter PJ, Lazar GA. 2018. Next generation antibody drugs: pursuit of the “high-hanging fruit. .” Nat. Rev. Drug Discov. 17:3197–223
    [Google Scholar]
  119. 119. 
    Sonzini S, Greco ML, Cailleau T, Adams L, Masterson L et al. 2019. Improved physical stability of an antibody-drug conjugate using host-guest chemistry. Bioconjug. Chem. 31:1123–29
    [Google Scholar]
  120. 120. 
    Klein C, Schaefer W, Regula JT. 2016. The use of CrossMAb technology for the generation of bi- and multispecific antibodies. mAbs 8:61010–20
    [Google Scholar]
  121. 121. 
    Labrijn AF, Janmaat ML, Reichert JM, Parren PWHI. 2019. Bispecific antibodies: a mechanistic review of the pipeline. Nat. Rev. Drug Discov. 18:8585–608
    [Google Scholar]
  122. 122. 
    Beckmann R, Jensen K, Fenn S, Speck J, Krause K et al. 2021. DutaFabs are engineered therapeutic Fab fragments that can bind two targets simultaneously. Nat. Commun. 12:1708
    [Google Scholar]
  123. 123. 
    Gentiluomo L, Svilenov HL, Augustijn D, Bialy IE, Greco ML et al. 2020. Advancing therapeutic protein discovery and development through comprehensive computational and biophysical characterization. Mol. Pharm. 17:426–40
    [Google Scholar]
  124. 124. 
    Jain T, Sun T, Durand S, Hall A, Houston NR et al. 2017. Biophysical properties of the clinical-stage antibody landscape. PNAS 114:5944–49
    [Google Scholar]
  125. 125. 
    Lai P-K, Fernando A, Cloutier TK, Gokarn Y, Zhang J et al. 2021. Machine learning applied to determine the molecular descriptors responsible for the viscosity behavior of concentrated therapeutic antibodies. Mol. Pharm. 18:1167–75
    [Google Scholar]
  126. 126. 
    Narayanan H, Dingfelder F, Butté A, Lorenzen N, Sokolov M, Arosio P. 2021. Machine learning for biologics: opportunities for protein engineering, developability, and formulation. Trends Pharmacol. Sci 42:151–65
    [Google Scholar]
  127. 127. 
    Bak A, Friis KP, Wu Y, Ho RJY. 2019. Translating cell and gene biopharmaceutical products for health and market impact product scaling from clinical to marketplace: lessons learned and future outlook. J. Pharm. Sci. 108:103169–75
    [Google Scholar]
  128. 128. 
    Jere D, Sediq AS, Huwyler J, Vollrath I, Kardorff M, Mahler H-C. 2020. Challenges for cell-based medicinal products from a pharmaceutical product perspective. J. Pharm. Sci. 110:51900–8
    [Google Scholar]
  129. 129. 
    Hoogendoorn KH, Crommelin DJA, Jiskoot W. 2020. Formulation of cell-based medicinal products: a question of life or death?. J. Pharm. Sci. 110:1885–94
    [Google Scholar]
  130. 130. 
    Buchanan SS, Pyatt DW, Carpenter JF. 2010. Preservation of differentiation and clonogenic potential of human hematopoietic stem and progenitor cells during lyophilization and ambient storage. PLOS ONE 5:9e12518
    [Google Scholar]
/content/journals/10.1146/annurev-pharmtox-052120-093517
Loading
/content/journals/10.1146/annurev-pharmtox-052120-093517
Loading

Data & Media loading...

Supplemental Material

Supplementary Data

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error