1,042
Views
48
CrossRef citations to date
0
Altmetric
Review

Co-amorphous systems for the delivery of poorly water-soluble drugs: recent advances and an update

, , , , , & ORCID Icon show all
Pages 1411-1435 | Received 25 Apr 2020, Accepted 14 Jul 2020, Published online: 17 Sep 2020

References

  • Di L, Fish PV, Mano T. Bridging solubility between drug discovery and development. Drug Discov Today. 2012;17(9–10):486–495.
  • Shi Q, Moinuddin SM, Cai T. Advances in coamorphous drug delivery systems. Acta pharm Sin B. 2019;9(1):19–35.
  • Qian S, Heng W, Wei Y, et al. Coamorphous lurasidone hydrochloride–saccharin with charge-assisted hydrogen bonding interaction shows improved physical stability and enhanced dissolution with pH-independent solubility behavior. Cryst Growth Des. 2015;15(6):2920–2928.
  • Hsu CM, Yu SC, Tsai FJ, et al. Characterization of in vitro and in vivo bioactivity of a ferulic acid-2-Hydroxypropyl-beta-cyclodextrin inclusion complex. Colloids Surf B: Biointerfaces. 2019;180:68–74.
  • Ahmed S, Corvis Y, Gahoual R, et al. Conception of nanosized hybrid liposome/poloxamer particles to thicken the interior core of liposomes and delay hydrophilic drug delivery. Int J Pharm. 2019;567:118488.
  • Vasconcelos T, Marques S, Das Neves J, et al. Amorphous solid dispersions: rational selection of a manufacturing process. Adv Drug Deliv Rev. 2016;100:85–101.
  • 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.
  • Hou G, Wang Z, Ma H, et al. High-temperature stable plasmonic and cavity resonances in metal nanoparticle-decorated silicon nanopillars for strong broadband absorption in photothermal applications. Nanoscale. 2019;11:14777–14784.
  • Trubitsyn G, Nga Nguyen V, Di Tommaso C, et al. Impact of covalently nile red and covalently rhodamine labeled fluorescent polymer micelles for the improved imaging of the respective drug delivery system. Eur J Pharm Biopharm. 2019;142:480–487.
  • Sverdlov Arzi R, Sosnik A. Electrohydrodynamic atomization and spray-drying for the production of pure drug nanocrystals and co-crystals. Adv Drug Deliv Rev. 2018;131:79–100.
  • Wang R, Han J, Jiang A, et al. Involvement of metabolism-permeability in enhancing the oral bioavailability of curcumin in excipient-free solid dispersions co-formed with piperine. Int J Pharm. 2019;561:9–18.
  • Kasten G, Lobmann K, Grohganz H, et al. Co-former selection for co-amorphous drug-amino acid formulations. Int J Pharm. 2019;557:366–373.
  • Shi X, Song S, Ding Z, et al. Improving the solubility, dissolution, and bioavailability of ibrutinib by preparing it in a coamorphous state with saccharin. J Pharm Sci. 2019;108:3020–3028.
  • Wu W, Lobmann K, Rades T, et al. On the role of salt formation and structural similarity of co-formers in co-amorphous drug delivery systems. Int J Pharm. 2018;535(1–2):86–94.
  • Fung MH, DeVault M, Kuwata KT, et al. Drug-excipient interactions: effect on molecular mobility and physical stability of ketoconazole-organic acid coamorphous systems. Mol Pharm. 2018;15(3):1052–1061.
  • Chavan RB, Thipparaboina R, Kumar D, et al., Co amorphous systems: a product development perspective. Int J Pharm. 2016;515(1–2): 403–415.
  • Lu W, Rades T, Rantanen J, et al. Inhalable co-amorphous budesonide-arginine dry powders prepared by spray drying. Int J Pharm. 2019;565:1–8.
  • Moinuddin SM, Ruan S, Huang Y, et al. Facile formation of co-amorphous atenolol and hydrochlorothiazide mixtures via cryogenic-milling: enhanced physical stability, dissolution and pharmacokinetic profile. Int J Pharm. 2017;532(1):393–400.
  • Li B, Konecke S, Wegiel LA, et al. Both solubility and chemical stability of curcumin are enhanced by solid dispersion in cellulose derivative matrices. Carbohyd Polym. 2013;98(1):1108–1116.
  • Korhonen O, Pajula K, Laitinen R. Rational excipient selection for co-amorphous formulations. Expert Opin Drug Deliv. 2017;14(4):551–569.
  • Knapik-Kowalczuk J, Chmiel K, Jurkiewicz K, et al. Physical stability and viscoelastic properties of co-amorphous ezetimibe/simvastatin system. Pharmaceuticals. 2019;12(1):40.
  • Bohr A, Nascimento TL, Harmankaya N, et al. Efflux inhibitor bicalutamide increases oral bioavailability of the poorly soluble efflux substrate docetaxel in co-amorphous anti-cancer combination therapy. Molecules. 2019;24(2):266.
  • Russo MG, Baldoni HA, Davila YA, et al. Rational design of a famotidine-ibuprofen coamorphous system: an experimental and theoretical study. J Phys Chem B. 2018;122(37):8772–8782.
  • Meng-Lund H, Kasten G, Jensen KT, et al. The use of molecular descriptors in the development of co-amorphous formulations. Eur J Pharm Sci. 2018;119:31–38.
  • Tilborg A, Norberg B, Wouters J. Pharmaceutical salts and cocrystals involving amino acids: A brief structural overview of the state-of-art. Eur J Med Chem. 2014;74:411–426.
  • Kasten G, Lobo L, Dengale S, et al. In vitro and in vivo comparison between crystalline and co-amorphous salts of naproxen-arginine. Eur J Pharm Biopharm. 2018;132:192–199.
  • Ojarinta R, Saarinen J, Strachan CJ, et al. Preparation and characterization of multi-component tablets containing co-amorphous salts: combining multimodal non-linear optical imaging with established analytical methods. Eur J Pharm Biopharm. 2018;132:112–126.
  • Mishra J, Rades T, Lobmann K, et al. Influence of solvent composition on the performance of spray-dried co-amorphous formulations. Pharmaceutics. 2018;10(2):47.
  • Kasten G, Nouri K, Grohganz H, et al. Performance comparison between crystalline and co-amorphous salts of indomethacin-lysine. Int J Pharm. 2017;533(1):138–144.
  • Ojarinta R, Lerminiaux L, Laitinen R. Spray drying of poorly soluble drugs from aqueous arginine solution. Int J Pharm. 2017;532(1):289–298.
  • Kasten G, Grohganz H, Rades T, et al. Development of a screening method for co-amorphous formulations of drugs and amino acids. Eur J Pharm Sci. 2016;95:28–35.
  • Wu W, Ueda H, Lobmann K, et al. Organic acids as co-formers for co-amorphous systems - influence of variation in molar ratio on the physicochemical properties of the co-amorphous systems. Eur J Pharm Biopharm. 2018;131:25–32.
  • Wu W, Wang Y, Lobmann K, et al. Transformations between co-amorphous and co-crystal systems and their influence on the formation and physical stability of co-amorphous systems. Mol Pharm. 2019;16(3):1294–1304.
  • An JH, Lim C, Kiyonga AN, et al. Co-amorphous screening for the solubility enhancement of poorly water-soluble mirabegron and investigation of their intermolecular interactions and dissolution behaviors. Pharmaceutics. 2018;10(3):149.
  • Gniado K, MacFhionnghaile P, McArdle P, et al. The natural bile acid surfactant sodium taurocholate (NaTC) as a coformer in coamorphous systems: enhanced physical stability and dissolution behavior of coamorphous drug-NaTc systems. Int J Pharm. 2018;535(1–2):132–139.
  • Fung M, Be Rzins KR, Suryanarayanan R. Physical stability and dissolution behavior of ketoconazole-organic acid coamorphous systems. Mol Pharm. 2018;15(5):1862–1869.
  • Lobmann K, Laitinen R, Strachan C, et al. Amino acids as co-amorphous stabilizers for poorly water-soluble drugs–Part 2: molecular interactions. Eur J Pharm Biopharm. 2013;85:882–888.
  • Kissi EO, Kasten G, Lobmann K, et al. The role of glass transition temperatures in coamorphous drug-amino acid formulations. Mol Pharm. 2018;15(9):4247–4256.
  • Petry I, Lobmann K, Grohganz H, et al. In situ co-amorphisation in coated tablets - The combination of carvedilol with aspartic acid during immersion in an acidic medium. Int J Pharm. 2019;558:357–366.
  • Laitinen R, Lobmann K, Grohganz H, et al. Amino acids as co-amorphous excipients for simvastatin and glibenclamide: physical properties and stability. Mol Pharm. 2014;11(7):2381–2389.
  • Ruponen M, Visti M, Ojarinta R, et al. Permeability of glibenclamide through a PAMPA membrane: the effect of co-amorphization. Eur J Pharm Biopharm. 2018;129:247–256.
  • Franca MT, Marcos TM, Pereira RN, et al. Could the small molecules such as amino acids improve aqueous solubility and stabilize amorphous systems containing Griseofulvin? Eur J Pharm Sci. 2019;143:105178.
  • Petry I, Lobmann K, Grohganz H, et al. Undesired co-amorphisation of indomethacin and arginine during combined storage at high humidity conditions. Int J Pharm. 2018;544(1):172–180.
  • Wu W, Lobmann K, Schnitzkewitz J, et al. Dipeptides as co-formers in co-amorphous systems. Eur J Pharm Biopharm. 2019;134:68–76.
  • Craye G, Lobmann K, Grohganz H, et al. Characterization of amorphous and co-amorphous simvastatin formulations prepared by spray drying. Molecules. 2015;20(12):21532–21548.
  • Lu W, Rades T, Rantanen J, et al. Amino acids as stabilizers for spray-dried simvastatin powder for inhalation. Int J Pharm. 2019;572:118724.
  • Huang Y, Zhang Q, Wang JR, et al. Amino acids as co-amorphous excipients for tackling the poor aqueous solubility of valsartan. Pharm Dev Technol. 2017;22(1):69–76.
  • Pan Y, Pang W, Lv J, et al. Solid state characterization of azelnidipine-oxalic acid co-crystal and co-amorphous complexes: the effect of different azelnidipine polymorphs. J Pharm Biomed Anal. 2017;138:302–315.
  • Ali AM, Ali AA, Maghrabi IA. Clozapine-carboxylic acid plasticized co-amorphous dispersions: preparation, characterization and solution stability evaluation. Acta Pharm. 2015;65(2):133–146.
  • Al-Hamidi H, Edwards AA, Mohammad MA, et al. To enhance dissolution rate of poorly water-soluble drugs: glucosamine hydrochloride as a potential carrier in solid dispersion formulations. Colloids Surf B: Biointerfaces. 2010;76(1):170–178.
  • Madgulkar A, Bandivadekar M, Shid T, et al. Sugars as solid dispersion carrier to improve solubility and dissolution of the BCS class II drug: clotrimazole. Drug Dev Ind Pharm. 2016;42(1):28–38.
  • Takeda K, Sekitoh T, Fujioka A, et al. Physical stability of an amorphous sugar matrix dried from methanol as an amorphous solid dispersion carrier and the influence of heat treatment. J Pharm Sci. 2019;108(6):2056–2062.
  • Kaminska E, Adrjanowicz K, Tarnacka M, et al. Impact of inter- and intramolecular interactions on the physical stability of indomethacin dispersed in acetylated saccharides. Mol Pharm. 2014;11(8):2935–2947.
  • Kaminska E, Adrjanowicz K, Zakowiecki D, et al. Enhancement of the physical stability of amorphous indomethacin by mixing it with octaacetylmaltose. inter and intra molecular studies. Pharm Res. 2014;31(10):2887–2903.
  • Kaminska E, Madejczyk O, Tarnacka M, et al. Studying of crystal growth and overall crystallization of naproxen from binary mixtures. Eur J Pharm Biopharm. 2017;113:75–87.
  • Zajc N, Obreza A, Bele M, et al. Physical properties and dissolution behaviour of nifedipine/mannitol solid dispersions prepared by hot melt method. Int J Pharm. 2005;291(1–2):51–58.
  • Kaminska E, Tarnacka M, Wlodarczyk P, et al. Studying the impact of modified saccharides on the molecular dynamics and crystallization tendencies of model API nifedipine. Mol Pharm. 2015;12(8):3007–3019.
  • Shayanfar A, Ghavimi H, Hamishehkar H, et al. Coamorphous atorvastatin calcium to improve its physicochemical and pharmacokinetic properties. J Pharm Pharm Sci. 2013;16(4):577–587.
  • Khan MZ, Rausl D, Zanoski R, et al. Classification of loratadine based on the biopharmaceutics drug classification concept and possible in vitro-in vivo correlation. Biol Pharm Bull. 2004;27(10):1630–1635.
  • Sora DI, Udrescu S, David V, et al. Validated ion pair liquid chromatography/fluorescence detection method for assessing the variability of the loratadine metabolism occurring in bioequivalence studies. Biomed Chromatogr. 2007;21(10):1023–1029.
  • Wang J, Chang R, Zhao Y, et al. Coamorphous loratadine-citric acid system with enhanced physical stability and bioavailability. AAPS PharmSciTech. 2017;18(7):2541–2550.
  • Ainurofiq A, Mauludin R, Mudhakir D, et al. A novel desloratadine-benzoic acid co-amorphous solid: preparation, characterization, and stability evaluation. Pharmaceutics. 2018;10(3):85.
  • Martinez LM, Videa M, Lopez Silva T, et al. Two-phase amorphous-amorphous solid drug dispersion with enhanced stability, solubility and bioavailability resulting from ultrasonic dispersion of an immiscible system. Eur J Pharm Biopharm. 2017;119:243–252.
  • Takeda K, Gotoda Y, Hirota D, et al. Surfactant-free solid dispersions of hydrophobic drugs in an amorphous sugar matrix dried from an organic solvent. Mol Pharm. 2017;14(3):791–798.
  • Satoh T, Hidaka F, Miyake K, et al. Surfactant-free solid dispersion of fat-soluble flavour in an amorphous sugar matrix. Food Chem. 2016;197:1136–1142.
  • Das A, Nayak AK, Mohanty B, et al. Solubility and dissolution enhancement of etoricoxib by solid dispersion technique using sugar carriers. ISRN Pharm. 2011;2011:819765.
  • Huang R, Han J, Wang R, et al. Surfactant-free solid dispersion of BCS class IV drug in an amorphous chitosan oligosaccharide matrix for concomitant dissolution in vitro - permeability increase. Eur J Pharm Sci. 2019;130:147–155.
  • Kulthe VV, Chaudhari PD, Aboul-Enein HY. Freeze-dried amorphous dispersions for solubility enhancement of thermosensitive API having low molecular lipophilicity. Drug Res. 2014;64(9):493–498.
  • Kaminska E, Tarnacka M, Kolodziejczyk K, et al. Impact of low molecular weight excipient octaacetylmaltose on the liquid crystalline ordering and molecular dynamics in the supercooled liquid and glassy state of itraconazole. Eur J Pharm Biopharm. 2014;88(3):1094–1104.
  • Bi Y, Xiao D, Ren S, et al. The binary system of ibuprofen-nicotinamide under nanoscale confinement: from cocrystal to coamorphous state. J Pharm Sci. 2017;106(10):3150–3155.
  • Gniado K, Lobmann K, Rades T, et al. The influence of co-formers on the dissolution rates of co-amorphous sulfamerazine/excipient systems. Int J Pharm. 2016;504(1–2):20–26.
  • Wu W, Lobmann K, Schnitzkewitz J, et al. Aspartame as a co-former in co-amorphous systems. Int J Pharm. 2018;549(1–2):380–387.
  • Gao Y, Liao J, Qi X, et al. Coamorphous repaglinide-saccharin with enhanced dissolution. Int J Pharm. 2013;450(1–2):290–295.
  • Ueda H, Kadota K, Imono M, et al. Co-amorphous formation induced by combination of tranilast and diphenhydramine hydrochloride. J Pharm Sci. 2017;106(1):123–128.
  • Dengale SJ, Grohganz H, Rades T, et al. Recent advances in co-amorphous drug formulations. Adv Drug Deliv Rev. 2016;100:116–125.
  • Haneef J, Chadha R. Drug-drug multicomponent solid forms: cocrystal, coamorphous and eutectic of three poorly soluble antihypertensive drugs using mechanochemical approach. AAPS PharmSciTech. 2017;18(6):2279–2290.
  • Hirakawa Y, Ueda H, Wakabayashi R, et al. A novel binary supercooled liquid formulation for transdermal drug delivery. Biol Pharm Bull. 2020;43(3):393–398.
  • Wairkar S, Gaud R. Co-amorphous combination of nateglinide-metformin hydrochloride for dissolution enhancement. AAPS PharmSciTech. 2016;17(3):673–681.
  • Lodagekar A, Chavan RB, Mannava MKC, et al. Co amorphous valsartan nifedipine system: preparation, characterization, in vitro and in vivo evaluation. Eur J Pharm Sci. 2019;139:105048.
  • Mannava MKC, Suresh K, Kumar Bommaka M, et al. Curcumin-artemisinin coamorphous solid: xenograft model preclinical study. Pharmaceutics. 2018;10(1):7.
  • Lababidi N, Ofosu Kissi E, Elgaher WAM, et al. Spray-drying of inhalable, multifunctional formulations for the treatment of biofilms formed in cystic fibrosis. J Control Release. 2019;314:62–71.
  • Dengale SJ, Hussen SS, Krishna BSM, et al. Fabrication, solid state characterization and bioavailability assessment of stable binary amorphous phases of ritonavir with quercetin. Eur J Pharm Biopharm. 2015;89:329–338.
  • Sanphui P, Devi VK, Clara D, et al. Cocrystals of hydrochlorothiazide: solubility and diffusion/permeability enhancements through drug-coformer interactions. Mol Pharm. 2015;12(5):1615–1622.
  • Lobmann K, Laitinen R, Grohganz H, et al. Coamorphous drug systems: enhanced physical stability and dissolution rate of indomethacin and naproxen. Mol Pharm. 2011;8(5):1919–1928.
  • Li H, Pan T, Cui Y, et al. Improved oral bioavailability of poorly water-soluble glimepiride by utilizing microemulsion technique. Int J Nanomedicine. 2016;11:3777–3788.
  • Zhang L, Zhu W, Lin Q, et al. Hydroxypropyl-beta-cyclodextrin functionalized calcium carbonate microparticles as a potential carrier for enhancing oral delivery of water-insoluble drugs. Int J Nanomedicine. 2015;10:3291–3302.
  • Cruz-Angeles J, Videa M, Martinez LM. Highly soluble glimepiride and irbesartan co-amorphous formulation with potential application in combination therapy. AAPS PharmSciTech. 2019;20(4):144.
  • Lobmann K, Strachan C, Grohganz H, et al. Co-amorphous simvastatin and glipizide combinations show improved physical stability without evidence of intermolecular interactions. Eur J Pharm Biopharm. 2012;81(1):159–169.
  • Leng D, Kissi EO, Lobmann K, et al. Design of inhalable solid dosage forms of budesonide and theophylline for pulmonary combination therapy. AAPS PharmSciTech. 2019;20(3):137.
  • Arnfast L, Kamruzzaman M, Lobmann K, et al. Melt extrusion of high-dose co-amorphous drug-drug combinations. Pharma Res. 2017;34(12):2689–2697.
  • Martinez-Jimenez C, Cruz-Angeles J, Videa M, et al. Co-amorphous simvastatin-nifedipine with enhanced solubility for possible use in combination therapy of hypertension and hypercholesterolemia. Molecules. 2018;23(9):2161.
  • Newman A, Reutzel-Edens SM, Zografi G. Coamorphous active pharmaceutical ingredient-small molecule mixtures: considerations in the choice of coformers for enhancing dissolution and oral bioavailability. J Pharm Sci. 2018;107(1):5–17.
  • Lobmann K, Grohganz H, Laitinen R, et al. Amino acids as co-amorphous stabilizers for poorly water soluble drugs–Part 1: preparation, stability and dissolution enhancement. Eur J Pharm Biopharm. 2013;85:873–881.
  • Mohammad MA, Alhalaweh A, Velaga SP. Hansen solubility parameter as a tool to predict cocrystal formation. Int J Pharm. 2011;407(1–2):63–71.
  • Salem A, Nagy S, Pal S, et al. Reliability of the Hansen solubility parameters as co-crystal formation prediction tool. Int J Pharm. 2019;558:319–327.
  • Metre S, Mukesh S, Samal SK, et al., Enhanced biopharmaceutical performance of rivaroxaban through polymeric amorphous solid dispersion. Mol Pharm. 2018;15(2): 652–668.
  • Pajula K, Wittoek L, Lehto VP, et al. Phase separation in coamorphous systems: in silico prediction and the experimental challenge of detection. Mol Pharm. 2014;11(7):2271–2279.
  • Baird JA, Taylor LS. Evaluation of amorphous solid dispersion properties using thermal analysis techniques. Adv Drug Deliv Rev. 2012;64(5):396–421.
  • Lopez FL, Shearman GC, Gaisford S, et al. Amorphous formulations of indomethacin and griseofulvin prepared by electrospinning. Mol Pharm. 2014;11(12):4327–4338.
  • Safna Hussan KP, Thayyil MS, Deshpande SK, et al. Molecular dynamics, physical and thermal stability of neat amorphous amlodipine besylate and in binary mixture. Eur J Pharm Sci. 2018;119:268–278.
  • Cordeiro T, Castineira C, Mendes D, et al. Stabilizing unstable amorphous menthol through inclusion in mesoporous silica hosts. Mol Pharm. 2017;14(9):3164–3177.
  • Zhu S, Gao H, Babu S, et al. Co-amorphous formation of high-dose zwitterionic compounds with amino acids to improve solubility and enable parenteral delivery. Mol Pharm. 2018;15(1):97–107.
  • Russo MG, Sancho MI, Silva LMA, et al. Looking for the interactions between omeprazole and amoxicillin in a disordered phase. An experimental and theoretical study. Spectrochim Acta A. 2016;156:70–77.
  • Sormunen H, Ruponen M, Laitinen R. The effect of co-amorphization of glibenclamide on its dissolution properties and permeability through an MDCKII-MDR1 cell layer. Int J Pharm. 2019;570:118653.
  • Murtaza G. Solubility enhancement of simvastatin: A review. Acta Pol Pharm. 2012;69(4):581–590.
  • Keratichewanun S, Yoshihashi Y, Sutanthavibul N, et al. An investigation of nifedipine miscibility in solid dispersions using raman spectroscopy. Pharm Res. 2015;32(7):2458–2473.
  • Bates S, Zografi G, Engers D, et al. Analysis of amorphous and nanocrystalline solids from their X-ray diffraction patterns. Pharm Res. 2006;23(10):2333–2349.
  • Healy AM, Worku ZA, Kumar D, et al. Pharmaceutical solvates, hydrates and amorphous forms: a special emphasis on cocrystals. Adv Drug Deliv Rev. 2017;117:25–46.
  • Maher EM, Ali AM, Salem HF, et al. In vitro/in vivo evaluation of an optimized fast dissolving oral film containing olanzapine co-amorphous dispersion with selected carboxylic acids. Drug Deliv. 2016;23(8):3088–3100.
  • Karagianni A, Kachrimanis K, Nikolakakis I. Co-amorphous solid dispersions for solubility and absorption improvement of drugs: composition, preparation, characterization and formulations for oral delivery. Pharmaceutics. 2018;10(3):98.
  • Singh A. Van den Mooter G. Spray drying formulation of amorphous solid dispersions. Adv Drug Deliv Rev. 2016;100:27–50.
  • Sai Krishna Anand V, Sakhare SD, Navya SKS, et al. The relevance of co-amorphous formulations to develop supersaturated dosage forms: in-vitro, and ex-vivo investigation of ritonavir-lopinavir co-amorphous materials. Eur J Pharm Sci. 2018;123:124–134.
  • Lenz E, Jensen KT, Blaabjerg LI, et al. Solid-state properties and dissolution behaviour of tablets containing co-amorphous indomethacin-arginine. Eur J Pharm Biopharm. 2015;96:44–52.
  • Jensen KT, Blaabjerg LI, Lenz E, et al. Preparation and characterization of spray-dried co-amorphous drug-amino acid salts. J Pharm Pharmacol. 2016;68(5):615–624.
  • Beyer A, Radi L, Grohganz H, et al. Preparation and recrystallization behavior of spray-dried co-amorphous naproxen-indomethacin. Eur J Pharm Biopharm. 2016;104:72–81.
  • Ali AM, Al-Remawi MM. Freeze dried quetiapine-nicotinamide binary solid dispersions: a new strategy for improving physicochemical properties and ex vivo diffusion. J Pharm. 2016;2016:2126056.
  • Ohori R, Akita T, Yamashita C. Mechanism of collapse of amorphous-based lyophilized cake induced by slow ramp during the shelf ramp process. Int J Pharm. 2019;564:461–471.
  • Lenz E, Lobmann K, Rades T, et al. Hot melt extrusion and spray drying of co-amorphous indomethacin-arginine with polymers. J Pharm Sci. 2017;106(1):302–312.
  • Hitzer P, Bauerle T, Drieschner T, et al. Process analytical techniques for hot-melt extrusion and their application to amorphous solid dispersions. Anal Bioanal Chem. 2017;409(18):4321–4333.
  • Netchacovitch L, Thiry J, De Bleye C, et al. Vibrational spectroscopy and microspectroscopy analyzing qualitatively and quantitatively pharmaceutical hot melt extrudates. J Pharm Biomed Anal. 2015;113:21–33.
  • Descamps M, Willart JF. Perspectives on the amorphisation/milling relationship in pharmaceutical materials. Adv Drug Deliv Rev. 2016;100:51–66.
  • Krupa A, Descamps M, Willart JF, et al. High-energy ball milling as green process to vitrify tadalafil and improve bioavailability. Mol Pharm. 2016;13(11):3891–3902.
  • Jensen KT, Larsen FH, Cornett C, et al. Formation mechanism of coamorphous drug-amino acid mixtures. Mol Pharm. 2015;12(7):2484–2492.
  • Willart JF, Descamps M. Solid state amorphization of pharmaceuticals. Mol Pharm. 2008;5(6):905–920.
  • Ojarinta R, Heikkinen AT, Sievanen E, et al. Dissolution behavior of co-amorphous amino acid-indomethacin mixtures: the ability of amino acids to stabilize the supersaturated state of indomethacin. Eur J Pharm Biopharm. 2017;112:85–95.
  • Pang W, Lv J, Du S, et al. Preparation of curcumin-piperazine coamorphous phase and fluorescence spectroscopic and density functional theory simulation studies on the interaction with bovine serum albumin. Mol Pharm. 2017;14(9):3013–3024.
  • Sou T, Bergstrom CAS. Automated assays for thermodynamic (equilibrium) solubility determination. Drug Discov Today Technol. 2018;27:11–19.
  • Konczol A, Dargo G. Brief overview of solubility methods: recent trends in equilibrium solubility measurement and predictive models. Drug Discov Today Technol. 2018;27:3–10.
  • Prachi BS, Varsha BP. Understanding peroral absorption: regulatory aspects and contemporary approaches to tackling solubility and permeability hurdles. Acta Pharm Sin B. 2017;7(3):260–280.
  • Maleki A, Kettiger H, Schoubben A, et al. Mesoporous silica materials: from physico-chemical properties to enhanced dissolution of poorly water-soluble drugs. J Control Release. 2017;262:329–347.
  • Priemel PA, Grohganz H, Rades T. Unintended and in situ amorphisation of pharmaceuticals. Adv Drug Deliv Rev. 2016;100:126–132.
  • Dahan A, Beig A, Lindley D, et al. The solubility-permeability interplay and oral drug formulation design: two heads are better than one. Adv Drug Deliv Rev. 2016;101:99–107.
  • Newman A, Nagapudi K, Wenslow R. Amorphous solid dispersions: A robust platform to address bioavailability challenges. Ther Deliv. 2015;6(2):247–261.
  • Yamamoto K, Kojima T, Karashima M, et al. Physicochemical evaluation and developability assessment of co-amorphouses of low soluble drugs and comparison to the co-crystals. Chem Pharm Bull. 2016;64(12):1739–1746.
  • Laitinen R, Lobmann K, Grohganz H, et al., Supersaturating drug delivery systems: the potential of co-amorphous drug formulations. Int J Pharm. 2017;532(1): 1–12.
  • Sun DD, Wen H, Taylor LS. Non-sink dissolution conditions for predicting product quality and in vivo performance of supersaturating drug delivery systems. J Pharm Sci. 2016;105(9):2477–2488.
  • Taylor LS, Zhang GGZ. Physical chemistry of supersaturated solutions and implications for oral absorption. Adv Drug Deliv Rev. 2016;101:122–142.
  • Wei Y, Zhou S, Hao T, et al. Further enhanced dissolution and oral bioavailability of docetaxel by coamorphization with a natural P-gp inhibitor myricetin. Eur J Pharm Sci. 2019;129:21–30.
  • Alhalaweh A, Bergstrom CAS, Taylor LS. Compromised in vitro dissolution and membrane transport of multidrug amorphous formulations. J Control Release. 2016;229:172–182.
  • Trasi NS, Taylor LS. Dissolution performance of binary amorphous drug combinations–Impact of a second drug on the maximum achievable supersaturation. Int J Pharm. 2015;496(2):282–290.
  • Trasi NS, Taylor LS. Thermodynamics of highly supersaturated aqueous solutions of poorly water-soluble drugs-impact of a second drug on the solution phase behavior and implications for combination products. J Pharm Sci. 2015;104(8):2583–2593.
  • Arca HC, Mosquera-Giraldo LI, Dahal D, et al. Multidrug, anti-HIV amorphous solid dispersions: nature and mechanisms of impacts of drugs on each other’s solution concentrations. Mol Pharm. 2017;14(11):3617–3627.
  • Nuxoll E. Added release time in diffusion/dissolution coupled release. Int J Pharm. 2015;494(1):195–204.
  • Mori K, Hori S, Kawata T, et al. The in vitro release of indomethacin from suppositories: effects of bases and comparison of different dissolution methods. Chem Pharm Bull. 2017;65(7):674–677.
  • Wairkar S, Gaud R. Development and characterization of microstructured, spray-dried co-amorphous mixture of antidiabetic agents stabilized by silicate. AAPS PharmSciTech. 2019;20(3):141.
  • Alleso M, Chieng N, Rehder S, et al., Enhanced dissolution rate and synchronized release of drugs in binary systems through formulation: amorphous naproxen-cimetidine mixtures prepared by mechanical activation. J Control Release. 2009;136(1): 45–53.
  • Aljohani M, MacFhionnghaile P, McArdle P, et al. Investigation of the formation of drug-drug cocrystals and coamorphous systems of the antidiabetic drug gliclazide. Int J Pharm. 2019;561:35–42.
  • Heng W, Wei Y, Xue Y, et al. Gel formation induced slow dissolution of amorphous indomethacin. Pharm Res. 2019;36(11):159.
  • Pacult J, Rams-Baron M, Chmiel K, et al. How can we improve the physical stability of co-amorphous system containing flutamide and bicalutamide? The case of ternary amorphous solid dispersions. Eur J Pharm Sci. 2019;136:104947.
  • Phan AD, Knapik-Kowalczuk J, Paluch M, et al. Theoretical model for the structural relaxation time in coamorphous drugs. Mol Pharm. 2019;16(7):2992–2998.
  • Riekes MK, Engelen A, Appeltans B, et al. New perspectives for fixed dose combinations of poorly water-soluble compounds: A case study with ezetimibe and lovastatin. Pharm Res. 2016;33(5):1259–1275.
  • Qian S, Li Z, Heng WL, et al. Charge-assisted intermolecular hydrogen bond formed in coamorphous system is important to relieve the pH-dependent solubility behavior of lurasidone hydrochloride. RSC Adv. 2016;6(108):106396–106412.
  • DeBoyace K, Wildfong PLD. The application of modeling and prediction to the formation and stability of amorphous solid dispersions. J Pharm Sci. 2018;107(1):57–74.
  • Yuan X, Sperger D, Munson EJ. Investigating miscibility and molecular mobility of nifedipine-PVP amorphous solid dispersions using solid-state NMR spectroscopy. Mol Pharm. 2014;11(1):329–337.
  • Tian Y, Jones DS, Andrews GP. An investigation into the role of polymeric carriers on crystal growth within amorphous solid dispersion systems. Mol Pharm. 2015;12(4):1180–1192.
  • Tian B, Tang X, Taylor LS. Investigating the correlation between miscibility and physical stability of amorphous solid dispersions using fluorescence-based techniques. Mol Pharm. 2016;13(11):3988–4000.
  • Beyer A, Grohganz H, Lobmann K, et al. Influence of the cooling rate and the blend ratio on the physical stability of co-amorphous naproxen/indomethacin. Eur J Pharm Biopharm. 2016;109:140–148.
  • Grzybowska K, Capaccioli S, Paluch M. Recent developments in the experimental investigations of relaxations in pharmaceuticals by dielectric techniques at ambient and elevated pressure. Adv Drug Deliv Rev. 2016;100:158–182.
  • Heng W, Su M, Cheng H, et al. Incorporation of complexation into a coamorphous system dramatically enhances dissolution and eliminates gelation of amorphous lurasidone hydrochloride. Mol Pharm. 2020;17:84–97.
  • Hirakawa Y, Ueda H, Miyano T, et al. New insight into transdermal drug delivery with supersaturated formulation based on co-amorphous system. Int J Pharm. 2019;569:118582.
  • Liu W, Liu Y, Huang J, et al. Identification and investigation of the vibrational properties of crystalline and co-amorphous drugs with Raman and terahertz spectroscopy. Biomed Opt Express. 2019;10(8):4290–4304.
  • Wang C, Paul S, Wang K, et al. Relationships among crystal structures, mechanical properties, and tableting performance probed using four salts of diphenhydramine. Cryst Growth Des. 2017;17(11):6030–6040.
  • Petry I, Lobmann K, Grohganz H, et al. Solid state properties and drug release behavior of co-amorphous indomethacin-arginine tablets coated with kollicoat (R) protect. Eur J Pharm Biopharm. 2017;119:150–160.
  • Suresh K, Mannava MKC, Nangia A. A novel curcumin-artemisinin coamorphous solid: physical properties and pharmacokinetic profile. RSC Adv. 2014;4(102):58357–58361.
  • Khajuria A, Thusu N, Zutshi U. Piperine modulates permeability characteristics of intestine by inducing alterations in membrane dynamics: influence on brush border membrane fluidity, ultrastructure and enzyme kinetics. Phytomedicine. 2002;9(3):224–231.
  • Di X, Wang X, Di X, et al. Effect of piperine on the bioavailability and pharmacokinetics of emodin in rats. J Pharm Biomed Anal. 2015;115:144–149.

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.