312
Views
1
CrossRef citations to date
0
Altmetric
Review

Lipid-based solubilization technology via hot melt extrusion: promises and challenges

, & ORCID Icon
Pages 1013-1032 | Received 01 Mar 2022, Accepted 08 Aug 2022, Published online: 18 Aug 2022

References

  • Amidon GL, Lennernäs H, Shah VP, et al. A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm Res. 1995;12(3):413–420.
  • Ghadi R, Dand N. BCS class IV drugs: highly notorious candidates for formulation development. J Control Release. 2017;248:71–95.
  • H. Park E-SH, Kim M-S, Kim M-S. Current status of supersaturable self-emulsifying Drug Deliverysystems. Pharmaceutics. 2020;12(365):365.
  • Singh D, Bedi N, Tiwary, et al. Tiwary, Enhancing solubility of poorly aqueous soluble drugs: critical appraisal of techniques. J Pharm Invest. 2018;48(5):509–526.
  • Dhaval M, Vaghela P, Patel K, et al. Lipid-based emulsion Drug Delivery systems—a comprehensive review. Drug Deliv Transl Res. 2022;12:1616–1639.
  • Holmstock N, Bruyn T, Bevernage J, et al. Exploring food effects on indinavir absorption with human intestinal fluids in the mouse intestine. Eur J Pharm Sci. 2013;49(1):27–32.
  • Gupta R, Jain V, Nagar JC, et al. Bioavailability enhancement techniques for poorly soluble drugs: a review. Asian J Pharm Res Dev. 2020;8(2):75–78.
  • Dumont C, Bourgeois S, Fessi H, et al. Lipid-based nanosuspensions for oral delivery of peptides, a critical review. Int J Pharm. 2018;541(1–2):117–135.
  • Nakach M, Shegokar R, Authelin J-R, et al. Nano-crystalline suspensions by top down processes: review of manufacturing considerations. Drug Deliv Lett. 2018;8:184–199.
  • Joseph E, Singhvi G. Chapter 4 - Multifunctional nanocrystals for cancer therapy: a potential nanocarrier. In: Grumezescu, AM, editor. Nanomater Drug Deliv Ther. William Andrew Publishing. 2019;91–116.
  • Gupta D, Bhatia D, Dave V, et al. Salts of therapeutic agents: chemical, physicochemical, and biological considerations. Molecules. 2018;23(7):1719.
  • Alsalhi A, Ayon NJ, Coulibaly F, et al. Enhancing etoposide aqueous solubility and anticancer activity with L-Arginine. Assay Drug Dev Technol. 2021;19(8):508–525.
  • Emami S, Siahi-Shadbad M, Adibkia K, et al. Recent advances in improving oral drug bioavailability by cocrystals. Bioimpacts. 2018;8(4):305.
  • Pandi P, Bulusu R, Kommineni N, et al. Amorphous solid dispersions: an update for preparation, characterization, mechanism on bioavailability, stability, regulatory considerations and marketed products. Int J Pharm. 2020;586:119560.
  • Schittny A, Huwyler J, Puchkov M. Mechanisms of increased bioavailability through amorphous solid dispersions: a review. Drug Deliv. 2020;27(1):110–127.
  • Ricarte RG, van Zee NJ, Li Z, et al. Recent advances in understanding the micro-and nanoscale phenomena of amorphous solid dispersions. Mol Pharm. 2019;16(10):4089–4103.
  • Koch N, Jennotte O, Ziemons E, et al. Influence of API physico-chemical properties on amorphization capacity of several mesoporous silica loading methods. Int J Pharm. 2022;613:121372.
  • Kuentz M. Lipid-based formulations for oral delivery of lipophilic drugs, Drug Discovery today. Technologies. 2012;9:e97–e104.
  • Porter CJH, Trevaskis NL, Charman WN. Lipids and lipid-based formulations: optimizing the oral delivery of lipophilic drugs. Nat Rev Drug Discov. 2007;6(3):231–248.
  • Cannon JB, Long MA. Chapter 11 - Emulsions, Microemulsions, and Lipid-Based Drug Delivery Systems for Drug Solubilization and Delivery—Part II: Oral Applications. In: Liu R, editor. Water-Insoluble Drug Formulation. CRC Press. 2018;247–282.
  • Parikh KJ, Sawant KK. Solubilization of vardenafil HCl in lipid-based formulations enhances its oral bioavailability in vivo: a comparative study using Tween-20 and Cremophor-EL. J Mol Liq. 2019;277:189–199.
  • Zhang J, Lv Y, Wang B, et al. Influence of microemulsion–mucin interaction on the fate of microemulsions diffusing through pig gastric mucin solutions. Mol Pharm. 2015;12(3):695–705.
  • Sigurdsson HH, Kirch J, Lehr C-M. Mucus as a barrier to lipophilic drugs. Int J Pharm. 2013;453(1):56–64.
  • Menzel C, Bernkop-Schnürch A. Enzyme decorated drug carriers: targeted swords to cleave and overcome the mucus barrier. Adv Drug Deliv Rev. 2018;124:164–174.
  • Laffleur F, Bernkop-Schnürch A. Strategies for improving mucosal Drug Deliveryery. Nanomedicine. 2013;8(12):2061–2075.
  • Leichner C, Menzel C, Laffleur F, et al. Development and in vitro characterization of a papain loaded mucolytic self-emulsifying Drug Delivery system (SEDDS. Int J Pharm. 2017;530(1–2):346–353.
  • Abdulkarim M, Sharma PK, Gumbleton M. Self-emulsifying Drug Delivery system: mucus permeation and innovative quantification technologies. Adv Drug Deliv Rev. 2019;142:62–74.
  • Griesser J, Hetényi G, Kadas H, et al. Self-emulsifying peptide Drug Delivery systems: how to make them highly mucus permeating. Int J Pharm. 2018;538(1–2):159–166.
  • Li AP. In vitro human cell–based experimental models for the evaluation of enteric metabolism and drug interaction potential of drugs and natural products. Drug Metab Dispos. 2020;48(10):980–992.
  • Elmeliegy M, Vourvahis M, Guo C, et al. Effect of P-glycoprotein (P-gp) inducers on exposure of P-gp substrates: review of clinical drug–drug interaction studies. Clin Pharmacokinet. 2020;59(6):699–714.
  • Hammann F, Gutmann H, Jecklin U, et al. Development of decision tree models for substrates, inhibitors, and inducers of p-glycoprotein. Curr Drug Metab. 2009;10(4):339–346.
  • Dong J, Qin Z, Zhang W-D, et al. Medicinal chemistry strategies to discover P-glycoprotein inhibitors: an update. Drug Resist Updat. 2020;49:100681.
  • Srivalli KMR, Lakshmi PK. Overview of P-glycoprotein inhibitors: a rational outlook. Braz J Pharm Sci. 2012;48(3):353–367.
  • Chang C, Bahadduri PM, Polli JE, et al. Rapid identification of P-glycoprotein substrates and inhibitors. Drug Metab Dispos. 2006;34(12):1976–1984.
  • Rathod S, Desai H, Patil R, et al. nonionic surfactants as a P-Glycoprotein(P-gp) efflux inhibitor for optimal Drug Delivery—A Concise outlook. AAPS PharmSciTech. 2022;23(1):1–10.
  • Sohi H, Ahuja A, Ahmad F, et al. Critical evaluation of permeation enhancers for oral mucosal Drug Deliveryery. Drug Dev Ind Pharm. 2010;36(3):254–282.
  • Maher S, Mrsny R, D.b.-a. Intestinal permeation enhancers for oral peptide delivery. Adv Drug Deliv Rev. 2016;106:277–319.
  • Whitehead K, Karr N, Mitragotri S. Safe and effective permeation enhancers for oral Drug Deliveryery. Pharm Res. 2008;25(8):1782–1788.
  • Zhang Z, Gao F, Jiang S, et al. Nano-based Drug Delivery system enhances the oral absorption of lipophilic drugs with extensive presystemic metabolism. Curr Drug Metab. 2012;13:1110–1118.
  • Sousa IP, Bernkop-Schnürch A. Pre-systemic metabolism of orally administered drugs and strategies to overcome it. J Control Release. 2014;192:301–309.
  • Thummel KE, Kunze KL, Shen DD. Enzyme-catalyzed processes of first-pass hepatic and intestinal drug extraction. Adv Drug Deliv Rev. 1997;27(2–3):99–127.
  • Galetin A, Gertz M, Houston JB. Contribution of intestinal cytochrome P450-mediated metabolism to Drug-Drug inhibition and induction interactions. Drug Metab Pharmacokinet. 2010;25(1):28–47.
  • Kharasch ED, Walker A, Hoffer C, et al. Intravenous and oral alfentanil as in vivo probes for hepatic and first-pass cytochrome P450 3A activity: noninvasive assessment by use of pupillary miosis. Clin Pharmacol Ther. 2004;76(5):452–466.
  • Bylund J, Bueters T. Presystemic metabolism of AZ’0908, A novel mPGES-1 inhibitor: an in vitro and in vivo cross-species comparison. J Pharm Sci. 2013;102(3):1106–1115.
  • Bernkop-Schnürch A, Krajicek ME. Mucoadhesive polymers as platforms for peroral peptide delivery and absorption: synthesis and evaluation of different chitosan–EDTA conjugates. J Control Release. 1998;50(1–3):215–223.
  • Borchard G, Lueflen HL, Coos Verhoef J, et al. Mucoadhesive polymers in peroral peptide Drug Delivery. II. carbomer and polycarbophil are potent inhibitors of the intestinal proteolytic enzyme trypsin. Springer. 1995;12:1293–1298.
  • Eichenbaum G, Hsu CP, Subrahmanyam V, et al. Oral coadministration of β-glucuronidase to increase exposure of extensively glucuronidated drugs that undergo enterohepatic recirculation. J Pharm Sci. 2012;101(7):2545–2556.
  • Gonçalves A, Nikmaram N, Roohinejad S, et al. Production, properties, and applications of solid self-emulsifying delivery systems (S-SEDS) in the food and pharmaceutical industries. Colloids Surf A Physicochem Eng Asp. 2018;538:108–126.
  • Porter CJH, Kaukonen AM, Boyd BJ, et al. Susceptibility to lipase-mediated digestion reduces the oral bioavailability of danazol after administration as a medium-chain lipid-based microemulsion formulation. Pharm Res. 2004;21(8):1405–1412.
  • Brocks DR, Davies NM. Lymphatic drug absorption via the enterocytes: pharmacokinetic simulation, modeling, and considerations for optimal drug development. J Pharm Pharm Sci. 2018;21(1s):254s–270s.
  • Menzel C, Holzeisen T, Laffleur F, et al. In vivo evaluation of an oral self-emulsifying Drug Delivery system (SEDDS) for exenatide. J Control Release. 2018;277:165–172.
  • Schneider F, Koziolek M, Weitschies W. In vitro and in vivo test methods for the evaluation of gastroretentive dosage forms. Pharmaceutics. 2019;11(416):416. 11 (2019).
  • Feeney OM, Crum MF, McEvoy CL, et al. 50 years of oral lipid-based formulations: provenance, progress and future perspectives. Adv Drug Deliv Rev. 2016;101:167–194.
  • Gao P, Morozowich W. Development of supersaturatable self-emulsifying Drug Delivery system formulations for improving the oral absorption of poorly soluble drugs. Expert Opin Drug Deliv. 2006;3(1):97–110.
  • Holm R, Tonsberg H, Jorgensen EB, et al. Influence of bile on the absorption of halofantrine from lipid-based formulations. Eur J Pharm Biopharm. 2012;81(2):281–287.
  • Zupančič O, Partenhauser A, Lam HT, et al. Development and in vitro characterization of an oral self-emulsifying delivery system for daptomycin. Eur J Pharm Sci. 2016;81:129–136.
  • Friedl H, Dünnhaupt S, Hintzen F, et al. Development and evaluation of a novel mucus diffusion test system approved by self-nanoemulsifying Drug DeliverySystems. J Pharm Sci. 2013;102(12):4406–4413.
  • Dünnhaupt S, Kammona O, Waldner C, et al. Nano-carrier systems: strategies to overcome the mucus gel barrier. Eur J Pharm Biopharm. 2015;96:447–453.
  • Goo YT, Song SH, Yeom DW, et al. Enhanced oral bioavailability of valsartan in rats using a supersaturable self-microemulsifying. Drug Delivery system with P-glycoprotein Inhibitors. 2019;25:178–186.
  • Netsomboon K, Laffleur F, Suchaoin W, et al. Novel in vitro transport method for screening the reversibility of P-glycoprotein inhibitors. Eur J Pharm Biopharm. 2016;100:9–14.
  • Bogman K, Erne-Brand F, Alsenz J, et al. The role of surfactants in the reversal of active transport mediated by multidrug resistance proteins. J Pharm Sci. 2003;92(6):1250–1261.
  • Twarog C, Fattah S, Heade J, et al. Intestinal permeation enhancers for oral delivery of macromolecules: a comparison between salcaprozate sodium (SNAC) and sodium caprate (C 10. Pharmaceutics. 2019;11(2):78.
  • Gleeson JP, Ryan SM, Brayden DJ. Oral delivery strategies for nutraceuticals: delivery vehicles and absorption enhancers. Trends Food SciTechnol. 2016;53:90–101.
  • Bu P, Ji Y, Narayanan S, et al. Assessment of cell viability and permeation enhancement in presence of lipid-based self-emulsifying Drug Delivery systems using Caco-2 cell model: polysorbate 80 as the surfactant. Eur J Pharm Sci. 2017;99:350–360.
  • Kossena GA, Charman WN, Wilson CG, et al. Low dose lipid formulations: effects on gastric emptying and biliary secretion. Pharm Res. 2007;24(11):2084–2096.
  • Brown NJ, Read NW, Richardson A, et al. Characteristics of lipid substances activating the ileal brake in the rat. Gut. 1990;31:1126–1129.
  • Spiller RC, Trotman IF, Adrian TE, et al. Further characterization of the ’ileal brake’ reflex in man--effect of ileal infusion of partial digests of fat, protein, and starch on jejunal motility and release of neurotensin, enteroglucagon, and peptide YY. Gut. 1988;29(8):1042–1051.
  • Christiansen ML, Holm R, Abrahamsson B, et al. Effect of food intake and co-administration of placebo self-nanoemulsifying Drug Delivery systems on the absorption of cinnarizine in healthy human volunteers. Eur J Pharm Sci. 2016;84:77–82.
  • Siepmann J, Faham A, Clas SD, et al. Lipids and polymers in pharmaceutical technology: lifelong companions. Int J Pharm. 2019;558:128–142.
  • Chauhan G, Shaik AA, Kulkarni NS, et al. The preparation of lipid-based Drug Delivery system using melt extrusion. Drug Discov Today. 2020;25:1930–1943.
  • Mesut B, Güngör S, Husuzade G. Role of self-emulsifying Drug DeliverySystems in enhancing the oral bioavailability of BCS class II drugs. Proceedings. 2021;78:32.
  • Jianxian C, Saleem K, Ijaz M, et al. Development and in vitro evaluation of gastro-protective aceclofenac-loaded self-emulsifying Drug DeliverySystem. Int J Nanomedicine. 2020;15:5217.
  • Zakkula A, Gabani B, Jairam R, et al. Preparation and optimization of nilotinib self-micro-emulsifying Drug Delivery systems to enhance oral bioavailability. Drug Dev Ind Pharm. 2020;46(3):498–504.
  • Husuzade G, Mesut B, Güngor S. Development of a solid-type IV self-emulsifying Drug DeliverySystem of BCS class II drug. 1st International Electronic Conference on Pharmaceutics, Switzerland. 2020;78:32.
  • Leonaviciute G, Zupančič O, Prüfert F, et al. Impact of lipases on the protective effect of SEDDS for incorporated peptide drugs toward intestinal peptidases. Int J Pharm. 2016;508(1–2):102–108.
  • Zupančič O, Leonaviciute G, Lam HT, et al. Development and in vitro evaluation of an oral SEDDS for desmopressin. Drug Deliv. 2016;23(6):2074–2083.
  • Ijaz M, Bonengel S, Zupančič O, et al. Development of oral self nano-emulsifying delivery system (s) of lanreotide with improved stability against presystemic thiol-disulfide exchange reactions. Expert Opin Drug Deliv. 2016;137:923–929. 137
  • Zupančič O, Rohrer J, Thanh Lam H, et al. Development and in vitro characterization of self-emulsifying drug delivery system (SEDDS) for oral opioid peptide delivery. Drug Dev Ind Pharm. 2017;43(10):1694–1702.
  • Nazir I, Asim MH, Dizdarević A, et al. Self-emulsifying Drug Delivery systems: impact of stability of hydrophobic ion pairs on drug release. Int J Pharm. 2019;561:197–205.
  • Friedl JD, Jörgensen AM, Le‐Vinh B, et al. Solidification of self-emulsifying Drug Delivery systems (SEDDS): impact on storage stability of a therapeutic protein. J Colloid Interface Sci. 2021;584:684–697.
  • Zupančič O, Grieβinger JA, Rohrer J, et al. Development, in vitro and in vivo evaluation of a self-emulsifying Drug Delivery system (SEDDS) for oral enoxaparin administration. Eur J Pharm Biopharm. 2016;109:113–121.
  • Tan A, Rao S, Prestidge CA. Transforming lipid-based oral Drug Delivery systems into solid dosage forms: an overview of solid carriers, physicochemical properties, and biopharmaceutical performance. Pharm Res. 2013;30(12):2993–3017.
  • P. Joyce TJD, Meola TR, Schultz HB, et al. Solidification to improve the biopharmaceutical performance of SEDDS: opportunities and challenges. Adv Drug Deliv Rev. 2019;142:102–117. DOI:10.1016/j.addr.2018.11.006.
  • Wagh MP, Singh PK, Chaudhari CS, et al. Solid self-emulsifying Drug Delivery system: preparation techniques and dosage forms. Int J Biopharm. 2014;5:101–108.
  • Tang B, Cheng G, Gu J-C, et al. Development of solid self-emulsifying Drug Delivery systems: preparation techniques and dosage forms. Drug Discov Today. 2008;13(13–14):606–612.
  • Jannin V, Musakhanian J, Marchaud D. Approaches for the development of solid and semi-solid lipid-based formulations. Adv Drug Deliv Rev. 2008;60(6):734–746.
  • Maji I, Mahajan S, Sriram A, et al. Solid self emulsifying Drug Delivery system: superior mode for oral delivery of hydrophobic cargos. J Control Release. 2021;337:646–660. DOI:10.1016/j.jconrel.2021.08.013.
  • Sacher S, Poms J, Rehrl J, et al. PAT implementation for advanced process control in solid dosage manufacturing – a practical guide. Int J Pharm. 2022;613:121408.
  • Passerini N, Perissutti B, Albertini B, et al. A new approach to enhance oral bioavailability of silybum marianum dry extract: association of mechanochemical activation and spray congealing. Phytomedicine. 2012;19(2):160–168.
  • Albertini B, Di Sabatino M, Melegari C, et al. Formulation of spray congealed microparticles with self-emulsifying ability for enhanced glibenclamide dissolution performance. J Microencapsul. 2015;32(2):181–192.
  • van Speybroeck M, Williams HD, Nguyen TH, et al. Incomplete Desorption of Liquid Excipients Reduces the in Vitro and in Vivo Performance of Self-Emulsifying Drug Delivery Systems Solidified by Adsorption onto an Inorganic Mesoporous Carrier. Mol Pharm. 2012;9(9):2750–2760.
  • Maniruzzaman M, Boateng JS, Snowden MJ, et al. A review of hot-melt extrusion: process technology to pharmaceutical products. Int Sch Res Notices. 2012;2012. Article id: 436763.
  • Patil H, Kulkarni V, Majumdar S, et al. Continuous manufacturing of solid lipid nanoparticles by hot melt extrusion. Int J Pharm. 2014;471(1–2):153–156.
  • H. Patil XF, Repka MA, Repka MA, et al. Continuous production of fenofibrate solid lipid nanoparticles by hot-melt extrusion technology: a systematic study based on a quality by design approach. AAPS J. 2015;17(1):194–205.
  • Shadambikar G, Marathe S, Ji N, et al. Formulation development of itraconazole PEGylated nano-lipid carriers for pulmonary aspergillosis using hot-melt extrusion technology. Int J Pharm. 2021;X(3):100074.
  • Bhagurkar AM, Repka MA, Murthy SN. A novel approach for the development of a nanostructured lipid carrier formulation by hot-melt extrusion technology. J Pharm Sci. 2017;106(4):1085–1091.
  • Witschnigg A, Koscher G, Treffer D, et al. Micro-pelletizing of pharmaceutical HME formulations using a die face pelletizer. AIP Conf Proc. 2016;1779:130001.
  • Spoerk M, Kottlan A, Makert C, et al. Continuously processed micropellets by hot-melt extrusion. AAPS PharmSciTec. 2022.
  • Sacher S, Celikovic S, Rehrl J, et al. Toward a novel continuous HME-`ting line: process development and control concept. Eur J Pharm Sci. 2020;142:105097.
  • Eggenreich K, Windhab S, Schrank S, et al. Injection molding as a one-step process for the direct production of pharmaceutical dosage forms from primary powders. Int J Pharm. 2016;505(1–2):341–351.
  • Tan DK, Maniruzzaman M, Nokhodchi A. Advanced pharmaceutical applications of hot-melt extrusion coupled with Fused Deposition Modeling (FDM) 3D printing for personalized Drug Deliveryery. Pharmaceutics. 2018;10(4):203.
  • Zhang J, Feng X, Patil H, et al. Coupling 3D printing with hot-melt extrusion to produce controlled-release tablets. Int J Pharm. 2017;519(1–2):186–197.
  • Ren Y, Mei L, Zhou L, et al. Recent perspectives in hot melt extrusion-based polymeric formulations for Drug Delivery: applications and Innovations. AAPS PharmSciTech. 2019;20(3):1–12.
  • Fan R, Chuan D, Hou H, et al. Development and evaluation of a novel biodegradable implants with excellent inflammatory response suppression effect by hot-melt extrusion. Eur J Pharm Sci. 2021;166:105981.
  • Sacher S, Heindl N, Afonso Urich JA, et al. A solution for low-dose feeding in continuous pharmaceutical processes. Int J Pharm. 2020;591:119969.
  • Repka MA, Bandari S, Kallakunta VR, et al. Melt extrusion with poorly soluble drugs–An integrated review. Int J Pharm. 2018;535(1–2):68–85.
  • Khinast J, Bresciani M. Chapter 1 - Continuous Manufacturing: Definitions and Engineering Principles. In: Kleinebudde P, Khinast J, Rantanen J, editors. Continuous manufacturing of pharmaceuticals. John Wiley & Sons; 2017. pp. 1–31.
  • Kallakunta VR, Sarabu S, Bandari S, et al. An update on the contribution of hot-melt extrusion technology to novel Drug Delivery in the twenty-first century: part I. Expert Opin Drug Delivery. 2019;16(5):539–550.
  • Sacher S, Poms J, Rehrl J, et al. PAT implementation for advanced process control in solid dosage manufacturing – a practical guide. Int J Pharm. 2022;613:121408.
  • Rehrl J, Karttunen AP, Nicolaï N, et al. Control of three different continuous pharmaceutical manufacturing processes: use of soft sensors. Int J Pharm. 2018;543(1–2):60–72.
  • Rehrl J, Kirchengast M, Celikovic S, et al. Improving pellet quality in a pharmaceutical hot melt extrusion process via PID control and LOLIMOT-based MPC. J Pharm Innov. 2020;15(4):678–689.
  • Beretta M, Pinto JT, Laggner P, et al. Insights into the impact of nanostructural properties on powder tribocharging: the case of milled salbutamol sulfate. Mol Pharm. 2022;19(2):547–557.
  • Koutsamanis I, Spoerk M, Arbeiter F, et al. Development of porous polyurethane implants manufactured via hot-melt extrusion. Polymers (Basel). 2020;12(12):1–22.
  • Kruisz J, Rehrl J, Hörmann-Kincses TR, et al. Effects of signal processing on the relative standard deviation in powder feeding characterization for continuous manufacturing. Powder Technol. 2021;389:536–548.
  • Fathollahi S, Sacher S, Escotet-Espinoza MS, et al. Performance evaluation of a high-precision low-dose powder feeder. AAPS PharmSciTech. 2020;21(8). DOI:10.1208/s12249-020-01835-5
  • Silva LAD, Almeida SL, Alonso ECP, et al. Preparation of a solid self-microemulsifying drug delivery system by hot-melt extrusion. Int J Pharm. 2018;541(1–2):1–10.
  • Aldosari B. Development and evaluation of self-nanoemulsifying drug delivery systems for oral delivery of indomethacin. UCL (University College London); 2018.
  • Gumaste SG, Gupta SS, Serajuddin ATM. Investigation of polymer-surfactant and polymer-drug-surfactant miscibility for solid dispersion. AAPS J. 2016;18(5):1131–1143.
  • Bordos E, Islam MT, Florence AJ, et al. Use of terahertz-raman spectroscopy to determine solubility of the crystalline active pharmaceutical ingredient in polymeric matrices during hot melt extrusion. Mol Pharm. 2019;16(10):4361–4371.
  • Krumbholz N, Hochrein T, Vieweg N, et al. Monitoring polymeric compounding processes inline with THz time-domain spectroscopy. Polym Test. 2009;28(1):30–35.
  • Wietzke S, Rutz F, Jördens C, et al. Applications of terahertz spectroscopy in the plastics industry. Terahertz Photonics. 2007;6840:68400 V.
  • Koutsamanis I, Paudel A, Alva Zúñiga CP, et al. Novel polyester-based thermoplastic elastomers for 3D-printed long-acting Drug Deliveryapplications. J Control Release. 2021;335:290–305.
  • Kushwah V, Saraf I, Yeoh T, et al. Interplay of aging and lot-to-lot variability on the physical and chemical properties of excipients: a case study of mono- and diglycerides. Mol Pharm. 2021;18(3):862–877.
  • Kushwah V, Lopes DG, Koutsamanis I, et al. Evolution of the microstructure and the drug release upon annealing the drug loaded lipid-surfactant microspheres. Eur J Pharm Sci. 2020;147:105278.
  • Saraf I, Kushwah V, Weber H, et al. Quantitative chemical profiling of commercial glyceride excipients via 1 H NMR spectroscopy. AAPS PharmSciTech. 2021;22(1):1–9.
  • Sarabu S, Bandari S, Kallakunta VR, et al. An update on the contribution of hot-melt extrusion technology to novel Drug Delivery in the twenty-first century: part II. Expert Opin Drug Deliv. 2019;16(6):567–582.
  • Thakkar R, Thakkar R, Pillai A, et al. Systematic screening of pharmaceutical polymers for hot melt extrusion processing: a comprehensive review. Int J Pharm. 2020;576:118989.
  • Li L, Yi T, Lam CWK. Effects of spray-drying and choice of solid carriers on concentrations of labrasol® and transcutol® in solid Self-Microemulsifying Drug DeliverySystems (SMEDDS. Molecules. 2013;18(2013):545–560.
  • Abbasi S, Higashino H, Sato Y, et al. Maximizing the oral bioavailability of poorly water-soluble drugs using novel oil-like materials in lipid-based formulations. Mol Pharm. 2021;18(9):3281–3289.
  • Wu X, Zhu Q, Chen Z, et al. Ionic liquids as a useful tool for tailoring active pharmaceutical ingredients. J Control Release. 2021;338:268–283.
  • accessed on 17.May.2022 https://www.peristalticpumps.it/en/tag/densi-en/
  • Nardin I, Köllner S. Successful development of oral SEDDS: screening of excipients from the industrial point of view. Adv Drug Deliv Rev. 2019;142:128–140.
  • Hsiao CYY, Ottaway CA, Wetlaufer DB. Preparation of fully deuterated fatty acids by simple method. Lipids. 1974;9(11):913–915. 9 (1974).
  • Shchepinov MS. Polyunsaturated fatty acid deuteration against neurodegeneration. Trends Pharmacol Sci. 2020;41(4):236–248.
  • Beaudoin-Chabot C, Wang L, Smarun AV, et al. Deuterated polyunsaturated fatty acids reduce oxidative stress and extend the lifespan of C. Elegans,Front Physiol. 2019;10:641.
  • Firsov AM, Fomich MA, V. Bekish A, et al. Threshold protective effect of deuterated polyunsaturated fatty acids on peroxidation of lipid bilayers. FEBS J. 2019;286(11):2099–2117.
  • Russak E. E.B.-A. of Pharmacotherapy, undefined 2019, Impact of deuterium substitution on the pharmacokinetics of pharmaceuticals. Journals.Sagepub.Com. 2019;53:211–216.
  • Crowley MM, Zhang F, Repka MA, et al. Pharmaceutical applications of hot-melt extrusion: part I. Drug Dev Ind Pharm. 2007;33(9):909–926.
  • Becker K, Salar-Behzadi S, Zimmer A. Solvent-free melting techniques for the preparation of lipid-based solid oral formulations. Pharm Res. 2015;32(5):1519–1545.
  • Schulze S, Winter G. Lipid extrudates as novel sustained release systems for pharmaceutical proteins. J Control Release. 2009;134(3):177–185.
  • Silva L, Teixeira F, Serpa R. Evaluation of carvedilol compatibility with lipid excipients for the development of lipid-based Drug Deliverysystems. J Therm Anal Calorim. 2016;123(3):2337–2344.
  • Kumar N, Goindi S, Saini B, et al. Thermal characterization and compatibility studies of itraconazole and excipients for development of solid lipid nanoparticles. J Therm Anal Calorim. 2013;115(3):2375–2383.
  • Kaur A, Goindi S, Katare OP. Thermal analysis and quantitative characterization of compatibility between diflunisal and lipid excipients as raw materials for development of solid lipid nanoparticles. Thermochim acta. 2016;643:23–32.
  • Zhang N, Zhang W, Jin Y, et al. Studies on preparation of carbamazepine (CBZ) supersaturatable self-microemulsifying (S-SMEDDS) formulation and relative bioavailability in beagle dogs. Pharm Dev Technol. 2011;16(4):415–421.
  • Suys EJA, Chalmers DK, Pouton CW, et al. Polymeric precipitation inhibitors promote fenofibrate supersaturation and enhance drug absorption from a type IV lipid-based formulation. Mol Pharm. 2018;15(6):2355–2371.
  • Tung NT, Tran CS, Nguyen HA, et al. Formulation and biopharmaceutical evaluation of supersaturatable self-nanoemulsifying Drug Delivery systems containing silymarin. Int J Pharm. 2019;555:63–76.
  • Dash RN, Mohammed H, Humaira T, et al. Solid supersaturatable self-nanoemulsifying Drug Delivery systems for improved dissolution, absorption and pharmacodynamic effects of glipizide. J Drug DeliverySci Technol. 2015;28:28–36.
  • Dash RN, Mohammed H, Humaira T. Design, optimization, and evaluation of ezetimibe solid supersaturatable self-nanoemulsifying Drug Delivery for enhanced solubility and dissolution. J Pharm Invest. 2015;46(2):153–168. 46 (2015).
  • Anby MU, Williams HD, McIntosh M, et al. Lipid digestion as a trigger for supersaturation: evaluation of the impact of supersaturation stabilization on the in vitro and in vivo performance of self-emulsifying Drug DeliverySystems. Mol Pharm. 2012;9(7):2063–2079.
  • Gao P, Rush BD, Pfund WP, et al. Development of a supersaturable SEDDS (S‐SEDDS) formulation of paclitaxel with improved oral bioavailability. J Pharm Sci. 2003;92(12):2386–2398.
  • Shi Y, Gao P, Gong Y, et al. Application of a biphasic test for characterization of in vitro drug release of immediate release formulations of celecoxib and its relevance to in vivo absorption. Mol Pharm. 2010;7(5):1458–1465.
  • Jaisamut P, Wiwattanawongsa K, Graidist P, et al. Enhanced oral bioavailability of curcumin using a supersaturatable self-microemulsifying system incorporating a hydrophilic polymer; in vitro and in vivo investigations. AAPS PharmSciTech. 2017;19(2):730–740. 19 (2017).
  • Gosangari S, Dyakonov T. Enhanced dissolution performance of curcumin with the use of supersaturatable formulations. Pharm Dev Technol. 2013;18(2):475–480.
  • Chavan RB, Modi SR, Bansal AK. Role of solid carriers in pharmaceutical performance of solid supersaturable SEDDS of celecoxib. Int J Pharm. 2015;495(1):374–384.
  • Song WH, Yeom DW, Lee DH, et al. In situ intestinal permeability and in vivo oral bioavailability of celecoxib in supersaturating self-emulsifying Drug Deliverysystem. Arch Pharm Res. 2014;37(5):626–635.
  • Song WH, Park JH, Yeom DW, et al. Enhanced dissolution of celecoxib by supersaturating self-emulsifying Drug Delivery system (S-SEDDS) formulation. Arch Pharm Res. 2013;36(1):69–78.
  • Lee DR, Ho MJ, Jung HJ, et al. Enhanced dissolution and oral absorption of tacrolimus by supersaturable self-emulsifying Drug Deliverysystem. Int J Nanomedicine. 2016;11:1109–1117.
  • Lee JH, Kim HH, Cho YH, et al. Development and evaluation of raloxifene-hydrochloride-loaded supersaturatable SMEDDS containing an acidifier. Pharmaceutics. 2018;10(78):78.
  • Chapman D. The polymorphism of glycerides. Chem Rev. 1962;62(5):433–456.
  • Sato K. Crystallization behavior of fats and lipids—a review. Chem Eng Sci. 2001;56(7):2255–2265.
  • Lutton ES. Review of the polymorphism of saturated even glycerides. J Am Oil Chem’ Soc. 1950;27(7):276–281.
  • Himawan C, Starov V, Stapley AGF. Thermodynamic and kinetic aspects of fat crystallization. Adv Colloid Interface Sci. 2006;122(1–3):1–3. 3–33.
  • Ghotra BS, Dyal SD, Narine SS. Lipid shortenings: a review. Food Res Int. 2002;35(10):1015–1048.
  • Bertoni S, Passerini N, Albertini B. Liquid lipids act as polymorphic modifiers of tristearin-based formulations produced by melting technologies. Pharmaceutics. 2021;13(7):1089.
  • Brunsteiner M, Khinast J, Paudel A. Relative contributions of solubility and mobility to the stability of amorphous solid dispersions of poorly soluble drugs: a molecular dynamics simulation study. Pharmaceutics. 2018;10(3):101.
  • Iosio T, Voinovich D, Grassi M, et al. Bi-layered self-emulsifying pellets prepared by co-extrusion and spheronization: influence of formulation variables and preliminary study on the in vivo absorption. Eur J Pharm Biopharm. 2008;69(2):686–697.
  • Abdalla A, Klein S, Mäder K. A new self-emulsifying Drug Delivery system (SEDDS) for poorly soluble drugs: characterization, dissolution, in vitro digestion and incorporation into solid pellets. Eur J Pharm Sci. 2008;35(5):457–464.
  • Hülsmann S, Backensfeld T, Keitel S, et al. Melt extrusion–an alternative method for enhancing the dissolution rate of 17β-estradiol hemihydrate. Eur J Pharm Biopharm. 2000;49(3):237–242.
  • Serratoni M, Newton M, Booth S, et al. Controlled drug release from pellets containing water-insoluble drugs dissolved in a self-emulsifying system. Eur J Pharm Biopharm. 2007;65(1):94–98.
  • Iosio T, Voinovich D, Perissutti B, et al. Pinto Oral bioavailability of silymarin phytocomplex formulated as self-emulsifying pellets. Phytomedicine. 2011;18(6):505–512.
  • Nikolakakis I, Malamataris S. Self-Emulsifying pellets: relations between kinetic parameters of drug release and emulsion reconstitution—influence of formulation variables. J Pharm Sci. 2014;103(5):1453–1465.
  • Kate L, Gokarna V, Borhade V, et al. Bioavailability enhancement of atovaquone using hot melt extrusion technology. Eur J Pharm Sci. 2016;86:103–114.
  • Diril M, Karasulu Y, Toskas M, et al. Development and permeability testing of self-emulsifying atorvastatin calcium pellets and tablets of compressed pellets. Processes. 2019;7(6):365.
  • Newton M, Petersson J, Podczeck F, et al. The influence of formulation variables on the properties of pellets containing a self-emulsifying mixture. J Pharm Sci. 2001;90(8):987–995.
  • Abdalla A, Mäder K. Preparation and characterization of a self-emulsifying pellet formulation. Eur J Pharm Biopharm. 2007;66(2):220–226.
  • Miao Y, Chen G, Ren L, et al. Characterization and evaluation of self-nanoemulsifying sustained-release pellet formulation of ziprasidone with enhanced bioavailability and no food effect. Drug Deliv. 2016;23(7):2163–2172.
  • Hanada M, Jermain SV, Thompson SA, et al. Ternary amorphous solid dispersions containing a high-viscosity polymer and mesoporous silica enhance dissolution performance. Mol Pharm. 2021;18(1):198–213.
  • Solomon S, Iqbal J, Albadarin AB. Insights into the ameliorating ability of mesoporous silica in modulating drug release in ternary amorphous solid dispersion prepared by hot melt extrusion. Eur J Pharm Biopharm. 2021;165:244–258.
  • Albertini B, Perissutti B, Bertoni S, et al. Combining mechanochemistry and spray congealing for new praziquantel pediatric formulations in schistosomiasis treatment. Int J Mol Sci. 2019;20(5):1233.
  • Khinast J, Baumgartner R, Roblegg E. Nano-extrusion: a one-step process for manufacturing of solid nanoparticle formulations directly from the liquid phase. AAPS PharmSciTech. 2013;14(2):601–604. 14 (2013).
  • Baumgartner R, Matić J, Schrank S, et al. NANEX: process design and optimization. Int J Pharm. 2016;506(1–2):35–45.
  • Brinkmann J, Luebbert C, Zaitsau DH, et al. Thermodynamic modeling of triglycerides using PC-SAFT. J Chem Eng Data. 2019;64(4):1446–1453.
  • Luebbert C, Brinkmann J, Exner L, et al. PC-SAFT Modeling of Phase Equilibria Relevant for Lipid-Based Drug DeliverySystems. J Chem Eng Data. 2021;66(3):1280–1289.
  • Matić J, Alva C, Eder S, et al. Toward predicting the product quality in hot-melt extrusion: pilot plant scale extrusion. Int J Pharm. 2021;X(3):100084.
  • Matić J, Alva C, Witschnigg A, et al. Toward predicting the product quality in hot-melt extrusion: small scale extrusion. Int J Pharm. 2020;X(2):100062.
  • Salentinig S, Amenitsch H, Yaghmur A. In situ monitoring of nanostructure formation during the digestion of mayonnaise. ACS Omega. 2017;2(4):1441–1446.
  • Boyd BJ, Clulow AJ. The influence of lipid digestion on the fate of orally administered Drug Deliveryvehicles. Biochem Soc Trans. 2021;49(4):1749–1761.
  • Ben Messaoud G, le Griel P, Hermida-Merino D, et al. Effects of pH, temperature and shear on the structure–property relationship of lamellar hydrogels from microbial glucolipids probed by in sit rheo-SAXS. Soft Matter. 2020;16(10):2540–2551.
  • Wang R, Jin W, Huang X. Construction of zwitterionic surfactant-stabilized hydrophobic ionic liquid-based bicontinuous microemulsion and microstructure-dependent activity of solubilized lipase. J Mol Liq. 2020;317:114011.
  • Goswami D. Lipase Catalysis in Presence of Nonionic Surfactants. Appl Biochem Biotechnol. 2019;191(2):744–762.

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.