191
Views
0
CrossRef citations to date
0
Altmetric
Review

Role of nanotechnology in the prolonged release of drugs by the subcutaneous route

& ORCID Icon
Pages 559-577 | Received 13 Nov 2022, Accepted 11 May 2023, Published online: 12 Jun 2023

References

  • Le J. Drug administration - drugs - MSD Manual Consumer Version. 2016. [cited Apr, 2022]. Available from: https://www.msdmanuals.com/home/drugs/administration-and-kinetics-of-drugs/drug-administration
  • Zhou Q, Jin J, Zhu L, et al. The optimal choice of medication administration route regarding intravenous, intramuscular, and subcutaneous injection. Patient Prefer Adherence. 2015;9:923.
  • Elsamany SA, Jafal M, Hassanin F. Pharmaco-economic assessment of subcutaneous compared to intravenous trastuzumab in HER2-positive breast cancer patients: a single institution experience. J Clin Oncol. 2020;38:e19409.
  • Viola M, Sequeira J, Seiça R, et al. Subcutaneous delivery of monoclonal antibodies: how do we get there? J Control Release. 2018;286:301–314.
  • Patra JK, Das G, Fraceto LF, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnology. 2018;16:1–33.
  • Halwani AA. Development of pharmaceutical nanomedicines: from the bench to the market. Pharmaceutics. 2022;14:106.
  • Kalaydina RV, Bajwa K, Qorri B, et al. Recent advances in “smart” delivery systems for extended drug release in cancer therapy. Int J Nanomedicine. 2018;13:4727–4745.
  • Farjadian F, Ghasemi A, Gohari O, et al. Nanopharmaceuticals and nanomedicines currently on the market: challenges and opportunities. Nanomedicine. 2019;14:93–126.
  • FDA approves first COVID-19 vaccine.[cited Jun 21, 2022]. Available from: https://www.fda.gov/news-events/press-announcements/fda-approves-first-covid-19-vaccine
  • Coronavirus (COVID-19) update: fDA takes key action by approving second COVID-19vaccine. [cited Jun 21, 2022]. Available from: https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-takes-key-action-approving-second-covid-19-vaccine
  • Richter WF, Bhansali SG, Morris ME. Mechanistic determinants of biotherapeutics absorption following SC administration. Aaps J. 2012;14:559–570.
  • Luo L, Liu M. Adipose tissue in control of metabolism. J Endocrinol. 2016;231:R77–99.
  • Kendall RT, Feghali-Bostwick CA. Fibroblasts in fibrosis: novel roles and mediators. Front Pharmacol. 2014;5:77–99.
  • Frost GI. Recombinant human hyaluronidase (rHuph20): an enabling platform for subcutaneous drug and fluid administration. Expert Opin Drug Deliv. 2007;4:427–440.
  • Varkhede N, Bommana R, Schöneich C, et al. Proteolysis and oxidation of therapeutic proteins after intradermal or subcutaneous administration. J Pharm Sci. 2020;109:191–205.
  • Sequeira JAD, Santos AC, Serra J, et al. Subcutaneous delivery of biotherapeutics: challenges at the injection site. Expert Opin Drug Deliv. 2019;16:143–151.
  • Rasmussen CH, Røge RM, Ma Z, et al. Insulin aspart pharmacokinetics: an assessment of its variability and underlying mechanisms. Eur J Pharm Sci. 2014;62:65–75.
  • Breslin JW, Yang Y, Scallan JP, et al. Lymphatic vessel network structure and physiology. Compr Physiol Wiley. 2018;9:207–299.
  • Moore JE, Bertram CD. Lymphatic system flows. 2017; [cited Mar 21, 2023]. Available from: http://doi.org/10.1146/annurev-fluid-122316
  • Charman WN, Stella VJ. Lymphatic transport of drugs . New York: CRC Press; 1992.
  • McCright J, Naiknavare R, Yarmovsky J, et al. Targeting lymphatics for nanoparticle drug delivery. Front Pharmacol. 2022;13:887402.
  • Stack T, Liu Y, Frey M, et al. Enhancing subcutaneous injection and target tissue accumulation of nanoparticles: via co-administration with macropinocytosis inhibitory nanoparticles (MiNP). Nanoscale Horiz. 2021;6:393–400.
  • Liu J, Liu Z, Pang Y, et al. The interaction between nanoparticles and immune system: application in the treatment of inflammatory diseases. J Nanobiotechnology. 2022;20:1–25.
  • Mitchell MJ, Billingsley MM, Haley RM, et al. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov. 2021;20:101–124.
  • Li Z, Zhu Y, Zeng H, et al. Mechano-boosting nanomedicine antitumour efficacy by blocking the reticuloendothelial system with stiff nanogels. Nat Commun. 2023;14:1437.
  • Burke JA, Zhang X, Bobbala S, et al. Subcutaneous nanotherapy repurposes the immunosuppressive mechanism of rapamycin to enhance allogeneic islet graft viability. Nat Nanotechnol. 2022;17:319–330.
  • Maisel K, Hrusch CL, Medellin JEG, et al. Pro-lymphangiogenic VEGFR-3 signaling modulates memory T cell responses in allergic airway inflammation. Mucosal Immunol. 2021;14:144–151.
  • McCright J, Skeen C, Yarmovsky J, et al. Nanoparticles with dense poly(ethylene glycol) coatings with near neutral charge are maximally transported across lymphatics and to the lymph nodes. Acta Biomater. 2022;145:146–158.
  • Nakamura T, Kawai M, Sato Y, et al. The effect of size and charge of lipid nanoparticles prepared by microfluidic mixing on their lymph node transitivity and distribution. Mol Pharm. 2020;17:944–953.
  • Rao DA, Forrest ML, Alani AWG, et al. Biodegradable PLGA based nanoparticles for sustained regional lymphatic drug delivery. J Pharm Sci. 2010;99:2018–2031.
  • Helle M, Rampazzo E, Monchanin M, et al. Surface chemistry architecture of silica nanoparticles determine the efficiency of in vivo fluorescence lymph node mapping. ACS Nano. 2013;7:8645–8657.
  • Rahimi E, Aramideh S, Han D, et al. Transport and lymphatic uptake of monoclonal antibodies after subcutaneous injection. Microvasc Res. 2022;139:104228.
  • Ke X, Howard GP, Tang H, et al. Physical and chemical profiles of nanoparticles for lymphatic targeting. Adv Drug Deliv Rev. 2019;151–152:72–93.
  • Bittner B, Richter W, Schmidt J. Subcutaneous administration of biotherapeutics: an overview of current challenges and opportunities. BioDrugs. 2018;32:425–440.
  • Reddy ST, Berk DA, Jain RK, et al. A sensitive in vivo model for quantifying interstitial convective transport of injected macromolecules and nanoparticles. J Appl Physiol. 2006;101:1162–1169.
  • Collins DS, Kourtis LC, Thyagarajapuram NR, et al. Optimizing the bioavailability of subcutaneously administered biotherapeutics through mechanochemical drivers. Pharm Res. 2017;34:2000–2011.
  • Vijayakumar A, Sharon E, Teena J, et al. A clinical study on drug-related problems associated with intravenous drug administration. J Basic Clin Pharm. 2014;5:49–53.
  • Haeri A, Osouli M, Bayat F, et al. Nanomedicine approaches for sirolimus delivery: a review of pharmaceutical properties and preclinical studies. Artif Cells Nanomed Biotechnol. 2018;46:1–14.
  • Davies N, Hovdal D, Edmunds N, et al. Functionalized lipid nanoparticles for subcutaneous administration of mRNA to achieve systemic exposures of a therapeutic protein. Mol Ther Nucleic Acids. 2021;24:369–384.
  • Wang L, Liu G, Hu Y, et al. Doxorubicin-loaded polypyrrole nanovesicles for suppressing tumor metastasis through combining photothermotherapy and lymphatic system-targeted chemotherapy. Nanoscale. 2022;14:3097–3111.
  • Clair-Jones A S, Prignano F, Goncalves J, et al. Understanding and minimising injection-site pain following subcutaneous administration of biologics: a narrative review. Rheumatol Ther. 2020;7:741–757.
  • Woodley WD, Morel DR, Sutter DE, et al. Clinical evaluation of large volume subcutaneous injection tissue effects, pain, and acceptability in healthy adults. Clin Transl Sci. 2022;15:92–104.
  • Jiskoot W, Hawe A, Menzen T, et al. Ongoing challenges to develop high concentration monoclonal antibody-based formulations for subcutaneous administration: quo vadis? J Pharm Sci. 2022;111:861–867.
  • Heo YA, Syed YY. Subcutaneous trastuzumab: a review in HER2-Positive Breast Cancer. Target Oncol. 2019;14:749–758.
  • Wasserman RL. Clinical practice experience with HyQvia in adults using alternative dosing regimens and pediatric patients: a retrospective study. Adv Ther. 2020;37:1536–1549.
  • Locke KW, Maneval DC, LaBarre MJ. ENHANZE® drug delivery technology: a novel approach to subcutaneous administration using recombinant human hyaluronidase PH20. Drug Deliv. 2019;26:98–106.
  • Chen W, Yung BC, Qian Z, et al. Improving long-term subcutaneous drug delivery by regulating material-bioenvironment interaction. Adv Drug Deliv Rev. 2018;127:20–34.
  • Kastellorizios M, Tipnis N, Burgess DJ. Foreign body reaction to subcutaneous implants. Adv Exp Med Biol Springer New York. 2015;LLC:93–108.
  • Karabin NB, Allen S, Kwon H-K, et al. Sustained micellar delivery via inducible transitions in nanostructure morphology. Nat Commun. 2018;9:624.
  • Kharbikar BN, Chendke GS, Desai TA. Modulating the foreign body response of implants for diabetes treatment. Adv Drug Deliv Rev. 2021;174:87–113.
  • Jogpal V, Sanduja M, Dutt R, et al. Advancement of nanomedicines in chronic inflammatory disorders. Inflammopharmacology. 2022;30:355–368.
  • Sercombe L, Veerati T, Moheimani F, et al. Advances and challenges of liposome assisted drug delivery. Front Pharmacol. 2015;6:286.
  • Wang P, Perche F, Midoux P, et al. In Vivo bone tissue induction by freeze-dried collagen-nanohydroxyapatite matrix loaded with BMP2/NS1 mRnas lipopolyplexes. J Control Release. 2021;334:188–200.
  • Korupalli C, Pan WY, Yeh CY, et al. Single-injecting, bioinspired nanocomposite hydrogel that can recruit host immune cells in situ to elicit potent and long-lasting humoral immune responses. Biomaterials. 2019;216:119268.
  • Zhang ZQ, Kim YM, Song SC. Injectable and quadruple-functional hydrogel as an alternative to intravenous delivery for enhanced tumor targeting. ACS Appl Mater Interfaces. 2019;11:34634–34644.
  • Suk JS, Xu Q, Kim N, et al. Pegylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev. 2016;99:28–51.
  • Leung AKK, Tam YYC, Chen S, et al. Microfluidic mixing: a general method for encapsulating macromolecules in lipid nanoparticle systems. J Phys Chem B. 2015;119:8698–8706.
  • Berraondo P, Martini PGV, Avila MA, et al. Messenger RNA therapy for rare genetic metabolic diseases. Gut. 2019;68:1323–1330.
  • Kulkarni JA, Witzigmann D, Leung J, et al. On the role of helper lipids in lipid nanoparticle formulations of siRNA. Nanoscale. 2019;11:21733–21739.
  • Volpatti LR, Matranga MA, Cortinas AB, et al. Glucose-responsive nanoparticles for rapid and extended self-regulated insulin delivery. ACS Nano. 2020;14:488–497.
  • Yang W, Liang H, Ma S, et al. Gold nanoparticle based photothermal therapy: development and application for effective cancer treatment. Sustainable Mater Technol. 2019;22:e00109.
  • Tanaka M, Hayashi M, Roach L, et al. Synthesis of near-infrared absorbing triangular Au nanoplates using biomineralisation peptides. Acta Biomater. 2021;131:519–531.
  • Arias L, Pessan J, Vieira A, et al. Iron oxide nanoparticles for biomedical applications: a perspective on synthesis, drugs, antimicrobial activity, and toxicity. Antibiotics. 2018;7:46.
  • Kim SH, Thambi T, Giang Phan VH, et al. Modularly engineered alginate bioconjugate hydrogel as biocompatible injectable scaffold for in situ biomineralization. Carbohydr Polym. 2020;233:233.
  • Bahari Javan N, Montazeri H, Rezaie Shirmard L, et al. Preparation, characterization and in vivo evaluation of a combination delivery system based on hyaluronic acid/jeffamine hydrogel loaded with PHBV/PLGA blend nanoparticles for prolonged delivery of Teriparatide. Eur J Pharm Sci. 2017;101:167–181.
  • Cao S, Liu Y, Shang H, et al. Supramolecular nanoparticles of calcitonin and dipeptide for long-term controlled release. J Control Release. 2017;256:182–192.
  • Bordat A, Boissenot T, Ibrahim N, et al. A Polymer prodrug strategy to switch from intravenous to subcutaneous cancer therapy for irritant/vesicant drugs. J Am Chem Soc. 2022;144:18844–18860.
  • Thambi T, Giang Phan VH, Kim SH, et al. Smart injectable biogels based on hyaluronic acid bioconjugates finely substituted with poly(β-amino ester urethane) for cancer therapy. Biomater Sci. 2019;7:5424–5437.
  • Vogus DR, Evans MA, Pusuluri A, et al. A hyaluronic acid conjugate engineered to synergistically and sequentially deliver gemcitabine and doxorubicin to treat triple negative breast cancer. J Control Release. 2017;267:191–202.
  • Lee ALZ, Yang C, Gao S, et al. Subcutaneous vaccination using injectable biodegradable hydrogels for long-term immune response. Nanomedicine. 2019;21:102056.
  • Kristó K, Szekeres M, Makai Z, et al. Preparation and investigation of core-shell nanoparticles containing human interferon-α. Int J Pharm. 2020;573:118825.
  • Rezaie Shirmard L, Ghofrani M, Bahari Javan N, et al. Improving the in-vivo biological activity of fingolimod loaded PHBV nanoparticles by using hydrophobically modified alginate. Drug Dev Ind Pharm. 2020;46:318–328.
  • Jeong DI, Kim S, Kim M-H, et al. Donepezil hydrochloride-reinforced cellulose nanocrystal-aggregated gel structure for long-acting drug delivery. Carbohydr Polym. 2022;296:119887.
  • Chendke GS, Faleo G, Juang C, et al. Supporting survival of transplanted stem‐cell‐derived insulin‐producing cells in an encapsulation device augmented with controlled release of amino acids. Adv Biosyst. 2019;3:1900086.
  • Trinh TA, Duy Le TM, HGV H, et al. A novel injectable Ph-temperature sensitive hydrogel containing chitosan-insulin electrosprayed nanosphere composite for an insulin delivery system in type i diabetes treatment. Biomater Sci. 2020;8:3830–3843.
  • Tong F. Preparation of exenatide-loaded linear poly(ethylene glycol)-brush poly(l-lysine) block copolymer: potential implications on diabetic nephropathy. Int J Nanomedicine. 2017;12:4663–4678.
  • Dong N, Zhu C, Jiang J, et al. Development of composite PLGA microspheres containing exenatide-encapsulated lecithin nanoparticles for sustained drug release. Asian J Pharm Sci. 2020;15:347–355.
  • Seo BB, Park MR, Song SC. Sustained release of exendin 4 using injectable and ionic-nano-complex forming polymer hydrogel system for long-term treatment of type 2 diabetes mellitus. ACS Appl Mater Interfaces. 2019;11:15201–15211.
  • Filipović N, Veselinović L, Ražić S, et al. Poly (ε-caprolactone) microspheres for prolonged release of selenium nanoparticles. Mater Sci Eng C. 2019;96:776–789.
  • Sigfridsson K, Xue A, Goodwin K, et al. Sustained release and improved bioavailability in mice after subcutaneous administration of griseofulvin as nano- and microcrystals. Int J Pharm. 2019;566:565–572.
  • Tanaudommongkon I, Tanaudommongkon A, Dong X. Development of in situ self-assembly nanoparticles to encapsulate lopinavir and ritonavir for long-acting subcutaneous injection. Pharmaceutics. 2021;13:904.
  • Duong V-A, Nguyen T-T-L, Maeng H-J, et al. Preparation of ondansetron hydrochloride-loaded nanostructured lipid carriers using solvent injection method for enhancement of pharmacokinetic properties. Pharm Res. 2019;36:138.
  • Elbrink K, van Hees S, Chamanza R, et al. Application of solid lipid nanoparticles as a long-term drug delivery platform for intramuscular and subcutaneous administration: in vitro and in vivo evaluation. Eur J Pharm Biopharm. 2021;163:158–170.
  • Prakapenka AV, Quihuis AM, Carson CG, et al. Poly(lactic-co-glycolic acid) nanoparticle encapsulated 17β-estradiol improves spatial memory and increases uterine stimulation in middle-aged ovariectomized rats. Front Behav Neurosci. 2020;14:597690.
  • Park J-S, Lee D, Yang S, et al. Methotrexate-loaded nanoparticles ameliorate experimental model of autoimmune arthritis by regulating the balance of interleukin-17-producing T cells and regulatory T cells. J Transl Med. 2022;20:1–11.
  • Kovalainen M, Mönkäre J, Kaasalainen M, et al. Development of porous silicon nanocarriers for parenteral peptide delivery. Mol Pharm. 2013;10:353–359.
  • Sozen T, Ozisik L, Calik Basaran N. An overview and management of osteoporosis. Eur J Rheumatol. 2017;4:46–56.
  • Chesnut CH, Azria M, Silverman S, et al. Salmon calcitonin: a review of current and future therapeutic indications. Osteoporos Int. 2008;19:479–491.
  • Canalis E. Management of endocrine disease: novel anabolic treatments for osteoporosis. Eur J Endocrinol. 2018;178:33–44.
  • Reid IR, Billington EO. Drug therapy for osteoporosis in older adults. Lancet. 2022;399:1080–1092.
  • Sun B, Luo C, Yu H, et al. Disulfide bond-driven oxidation- and reduction-responsive prodrug nanoassemblies for cancer therapy. Nano Lett. 2018;18:3643–3650.
  • Li N, Guo W, Li Y, et al. Construction and anti-tumor activities of disulfide-linked docetaxel-dihydroartemisinin nanoconjugates. Colloids Surf B Biointerfaces. 2020;191:111018.
  • Wang Z, Ling L, Du Y, et al. Reduction responsive liposomes based on paclitaxel-ss-lysophospholipid with high drug loading for intracellular delivery. Int J Pharm. 2019;564:244–255.
  • Lu J, Huang Y, Zhao W, et al. PEG-derivatized embelin as a nanomicellar carrier for delivery of paclitaxel to breast and prostate cancers. Biomaterials. 2013;34:1591–1600.
  • Barbee MS, Owonikoko TK, Harvey RD. Taxanes: vesicants, irritants, or just irritating? Ther Adv Med Oncol. 2014;6:16–20.
  • Zhao N, Woodle CM, Mixson AJ. Advances in delivery systems for doxorubicin. J Nanomed Nanotechnol. 2018;09:1000519.
  • Kanwal U, Irfan Bukhari N, Ovais M, et al. Advances in nano-delivery systems for doxorubicin: an updated insight. J Drug Target. 2018;26:296–310.
  • Wang Y, Yang ST, Wang Y, et al. Adsorption and desorption of doxorubicin on oxidized carbon nanotubes. Colloids Surf B Biointerfaces. 2012;97:62–69.
  • Zha Z, Yue X, Ren Q, et al. Uniform polypyrrole nanoparticles with high photothermal conversion efficiency for photothermal ablation of cancer cells. Adv Mater. 2013;25:777–782.
  • Wang T, Yang L, Xie Y, et al. An injectable hydrogel/staple fiber composite for sustained release of CA4P and doxorubicin for combined chemotherapy of xenografted breast tumor in mice. Nan Fang Yi Ke Da Xue Xue Bao. 2022;42:625–632.
  • Rosenblum D, Joshi N, Tao W, et al. Progress and challenges towards targeted delivery of cancer therapeutics. Nat Commun. 2018;9:1410.
  • Vincent MP, Bobbala S, Karabin NB, et al. Surface chemistry-mediated modulation of adsorbed albumin folding state specifies nanocarrier clearance by distinct macrophage subsets. Nat Commun. 2021;12:648.
  • Vincent MP, Navidzadeh JO, Bobbala S, et al. Leveraging self-assembled nanobiomaterials for improved cancer immunotherapy. Cancer Cell. 2022;40:255–276.
  • Cohen JA, Tenenbaum N, Bhatt A, et al. Extended treatment with fingolimod for relapsing multiple sclerosis: the 14-year LONGTERMS study results. Ther Adv Neurol Disord. 2019;12:175628641987832.
  • Singer BA. Initiating oral fingolimod treatment in patients with multiple sclerosis. Ther Adv Neurol Disord. 2013;6:269–275.
  • Vargas WS, Perumal JS. Fingolimod and cardiac risk: latest findings and clinical implications. Ther Adv Drug Saf. 2013;4:119–124.
  • Genuth SM, Palmer JP, Nathan DM. Classification and diagnosis of diabetes. Diabetes in America. Bethesda (MD): National Institute of Diabetes and Digestive and Kidney Diseases (US); 2018. p. 1–39.
  • Hansen KB, Vilsbøll T, Knop FK. Incretin mimetics: a novel therapeutic option for patients with type 2 diabetes - a review. Diabetes Metab Syndr Obes. 2010;3:155–163.
  • Seo BB, Park MR, Chun CJ, et al. The biological efficiency and bioavailability of human growth hormone delivered using injectable, ionic, thermosensitive poly(organophosphazene)-polyethylenimine conjugate hydrogels. Biomaterials. 2011;32:8271–8280.
  • Plotkin SA. Correlates of protection induced by vaccination. Clin Vaccin Immunol. 2010;17:1055–1065.
  • Sallusto F, Lanzavecchia A, Araki K, et al. From Vaccines to Memory and Back. Immunity. 2010;33:451–463.
  • Roth GA, Gale EC, Alcantara-Hernandez M, et al. Injectable hydrogels for sustained codelivery of subunit vaccines enhance humoral immunity. ACS Cent Sci. 2020;6:1800–1812.
  • Bailon L, Mothe B, Berman L, et al. Novel Approaches Towards a Functional Cure of HIV/AIDS. Drugs. 2020;80:859–868.
  • Adefolalu A, Nkosi Z. The complex nature of adherence in the management of HIV/AIDS as a chronic medical condition. Diseases. 2013;1:18–35.
  • Zingg R, Fischer M. The consolidation of nanomedicine. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2019;11:1–6.
  • Foulkes R, Man E, Thind J, et al. The regulation of nanomaterials and nanomedicines for clinical application: current and future perspectives. Biomater Sci. 2020;8:4653–4664.
  • Committee for Medicinal Products for Human Use (CHMP) EMA. Reflection paper on non-clinical studies for generic nanoparticle iron medicinal product applications, EMA/CHMP/SWP/100094/2011. [cited May 24, 2022]. Available from: https://www.ema.europa.eu/en/documents/scientific-guideline/reflection-paper-non-clinical-studies-generic-nanoparticle-iron-medicinal-product-applications_en.pdf
  • Soares S, Sousa J, Pais A, et al. Nanomedicine: principles, properties, and regulatory issues. Front Chem. 2018;6:1–15.
  • Allen RP, Bolandparvaz A, Ma JA, et al. Latent, immunosuppressive nature of poly(lactic- co -glycolic acid) microparticles. ACS Biomater Sci Eng. 2018;4:900–918.
  • Epstein RS. Payer perspectives on intravenous versus subcutaneous administration of drugs. Clinicoecon Outcomes Res. 2021;13:801–807.
  • Danaei M, Dehghankhold M, Ataei S, et al. Impact of particle size and polydispersity index on the clinical applications of lipidic nanocarrier systems. Pharmaceutics. 2018;10:57.
  • Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol. 2015;33:941–951.
  • Bastogne T. Quality-by-design of nanopharmaceuticals – a state of the art. Nanomedicine. 2017;13:2151–2157.
  • Rawal M, Singh A, Amiji MM. Quality-by-design concepts to improve nanotechnology-based drug development. Pharm Res. 2019;36:1–20.
  • Pallagi E, Ismail R, Paál TL, et al. Initial risk assessment as part of the quality by design in peptide drug containing formulation development. Eur J Pharm Sci. 2018;122:160–169.
  • Thanki K, Zeng X, Justesen S, et al. Engineering of small interfering RNA-loaded lipidoid-poly(DL-lactic-co-glycolic acid) hybrid nanoparticles for highly efficient and safe gene silencing: a quality by design-based approach. Eur J Pharm Biopharm. 2017;120:22–33.
  • Gupta A, Costa AP, Xu X, et al. Formulation and characterization of curcumin loaded polymeric micelles produced via continuous processing. Int J Pharm. 2020;583:119340.
  • Alexis F, Pridgen E, Molnar LK, et al. Factors affecting the clearance and biodistribution of polymeric nanoparticles. Mol Pharm. 2008;5:505–515.
  • Zagalo DM, Silva BMA, Silva C, et al. A quality by design (QbD) approach in pharmaceutical development of lipid-based nanosystems: a systematic review. J Drug Deliv Sci Technol. 2022;70:103207.
  • Omar Zaki SS, Ibrahim MN, Katas H. Particle size affects concentration-dependent cytotoxicity of chitosan nanoparticles towards mouse hematopoietic stem cells. J Nanotechnol. 2015;2015:1–5.
  • Egorov E, Pieters C, Korach-Rechtman H, et al. Robotics, microfluidics, nanotechnology and AI in the synthesis and evaluation of liposomes and polymeric drug delivery systems. Drug Deliv Transl Res. 2021;11:345–352.
  • Villa Nova M, Lin TP, Shanehsazzadeh S, et al. Nanomedicine ex machina: between model-informed development and artificial intelligence. Front Digit Health. 2022;4:1–12.
  • Sarker IH. Machine learning: algorithms, real-world applications and research directions. SN Comput Sci. 2021;2:1–21.
  • Shabanzadeh P, Senu N, Shameli K, et al. Artificial intelligence in numerical modeling of silver nanoparticles prepared in montmorillonite interlayer space. J Chem. 2013;2013:1–8.
  • Li Y, Abbaspour MR, Grootendorst PV, et al. Optimization of controlled release nanoparticle formulation of verapamil hydrochloride using artificial neural networks with genetic algorithm and response surface methodology. Eur J Pharm Biopharm. 2015;94:170–179.
  • Zaki MR, Varshosaz J, Fathi M. Preparation of agar nanospheres: comparison of response surface and artificial neural network modeling by a genetic algorithm approach. Carbohydr Polym. 2015;122:314–320.
  • Hashad RA, Ishak RAH, Fahmy S, et al. Chitosan-tripolyphosphate nanoparticles: optimization of formulation parameters for improving process yield at a novel pH using artificial neural networks. Int j biol macromol. 2016;86:50–58.
  • Baharifar H, Amani A. Size, loading efficiency, and cytotoxicity of albumin-loaded chitosan nanoparticles: an artificial neural networks study. J Pharm Sci. 2017;106:411–417.
  • Youshia J, Ali ME, Lamprecht A. Artificial neural network based particle size prediction of polymeric nanoparticles. Eur J Pharm Biopharm. 2017;119:333–342.
  • Kanwal U, Bukhari NI, Rana NF, et al. Doxorubicin-loaded quaternary ammonium palmitoyl glycol chitosan polymeric nanoformulation: uptake by cells and organs. Int J Nanomedicine. 2019;14:1–15.
  • Öztürk AA, Gündüz AB, Ozisik O. Supervised machine learning algorithms for evaluation of solid lipid nanoparticles and particle size. Comb Chem High Throughput Screen. 2018;21:693–699.
  • He Y, Ye Z, Liu X, et al. Can machine learning predict drug nanocrystals? J Control Release. 2020;322:274–285.
  • Lazarovits J, Sindhwani S, Tavares AJ, et al. Supervised learning and mass spectrometry predicts the in vivo fate of nanomaterials. ACS Nano. 2019;13:8023–8034.
  • Li F, Han J, Cao T, et al. Design of self-assembly dipeptide hydrogels and machine learning via their chemical features. PNAS. 2019;116:11259–11264.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.