109
Views
0
CrossRef citations to date
0
Altmetric
Research Article

Monophasic coamorphous sulpiride: a leap in physicochemical attributes and dual inhibition of GlyT1 and P-glycoprotein, supported by experimental and computational insights

, , , , , , & show all
Received 03 Oct 2023, Accepted 30 Dec 2023, Published online: 01 Feb 2024

References

  • Abdullahi, M., Uzairu, A., Shallangwa, G. A., Mamza, P. A., Ibrahim, M. T., Ahmad, I., & Patel, H. (2023). Structure-based drug design, molecular dynamics simulation, ADMET, and quantum chemical studies of some thiazolinones targeting influenza neuraminidase. Journal of Biomolecular Structure & Dynamics, 41(23), 13829–13843. https://doi.org/10.1080/07391102.2023.2208225
  • Al-Otaibi, J. S., Mary, Y. S., Armaković, S., & Thomas, R. (2020). Hybrid and bioactive cocrystals of pyrazinamide with hydroxybenzoic acids: Detailed study of structure, spectroscopic characteristics, other potential applications and noncovalent interactions using SAPT. Journal of Molecular Structure, 1202, 127316. https://doi.org/10.1016/j.molstruc.2019.127316
  • Andronis, V., Yoshioka, M., & Zografi, G. (1997). Effects of sorbed water on the crystallization of indomethacin from the amorphous state. Journal of Pharmaceutical Sciences, 86(3), 346–351. https://doi.org/10.1021/js9602711
  • Arulraj, R. A., Gajjela, R., Chauthe, S. K., Bagadi, M., & Mathur, A. J. C. (2022). Correction to: High-throughput GC-FID method for the determination of residual solvents in early-phase drug discovery samples. Chromatographia, 85(6), 497–506. https://doi.org/10.1007/s10337-022-04157-9
  • Ayoub, A. M., Ibrahim, M. M., Abdallah, M. H., & Mahdy, M. A. (2016). Sulpiride microemulsions as antipsychotic nasal drug delivery systems: In-vitro and pharmacodynamic study. Journal of Drug Delivery Science and Technology, 36, 10–22. https://doi.org/10.1016/j.jddst.2016.09.002
  • Baird, J. A., & Taylor, L. S. (2012). Evaluation of amorphous solid dispersion properties using thermal analysis techniques. Advanced Drug Delivery Reviews, 64(5), 396–421. https://doi.org/10.1016/j.addr.2011.07.009
  • Baluom, M. Friedman, M.,& Rubinstein, A (2001). Improved intestinal absorption of sulpiride in rats with synchronized oral delivery systems. Journal of Controlled Release, 70(1-2), 139–147. https://doi.org/10.1016/S0168-3659(00)00337-0.
  • Banerjee, P., Chandra, A., Mohammad, T., Singh, N., Hassan, M. I., & Qamar, I. (2023). Identification of high-affinity pyridoxal kinase inhibitors targeting cancer therapy: An integrated docking and molecular dynamics simulation approach. Journal of Biomolecular Structure & Dynamics, 42 (1), 1–18. https://doi.org/10.1080/07391102.2023.2246580
  • Bharti, S., Rani, N., Krishnamurthy, B., & Arya, D. S. (2014). Preclinical evidence for the pharmacological actions of naringin: A review. Planta Medica, 80(6), 437–451. https://doi.org/10.1055/s-0034-1368351
  • Bochevarov, A. D., Harder, E., Hughes, T. F., Greenwood, J. R., Braden, D. A., Philipp, D. M., Rinaldo, D., Halls, M. D., Zhang, J., & Friesner, R. A. (2013). Jaguar: A high-performance quantum chemistry software program with strengths in life and materials sciences. International Journal of Quantum Chemistry, 113(18), 2110–2142. https://doi.org/10.1002/qua.24481
  • Castner, S. A., Murthy, N. V., Ridler, K., Herdon, H., Roberts, B. M., Weinzimmer, D. P., Huang, Y., Zheng, M. Q., Rabiner, E. A., Gunn, R. N., Carson, R. E., Williams, G. V., & Laruelle, M. (2014). Relationship between glycine transporter 1 inhibition as measured with positron emission tomography and changes in cognitive performances in nonhuman primates. Neuropsychopharmacology, 39(12), 2742–2749. https://doi.org/10.1038/npp.2014.4
  • Castro-Alvarez, A., Costa, A., & Vilarrasa, J. (2017). The performance of several docking programs at reproducing protein–macrolide-like crystal structures. Molecules (Basel, Switzerland), 22(1), 136. https://doi.org/10.3390/molecules22010136
  • Cavia-Saiz, M., Busto, M. D., Pilar-Izquierdo, M. C., Ortega, N., Perez-Mateos, M., & Muñiz, P. (2010). Antioxidant properties, radical scavenging activity and biomolecule protection capacity of flavonoid naringenin and its glycoside naringin: A comparative study. Journal of the Science of Food and Agriculture, 90(7), 1238–1244. https://doi.org/10.1002/jsfa.3959
  • Celiz, G., Suarez, S. A., Arias, A., Molina, J., Brondino, C. D., & Doctorovich, F. (2019). Synthesis, structural elucidation and antiradical activity of a copper (II) naringenin complex. Biometals: An International Journal on the Role of Metal Ions in Biology, Biochemistry, and Medicine, 32(4), 595–610. https://doi.org/10.1007/s10534-019-00187-3
  • Chieng, N., Rehder, S., Saville, D., Rades, T., & Aaltonen, J. (2009). Quantitative solid-state analysis of three solid forms of ranitidine hydrochloride in ternary mixtures using Raman spectroscopy and X-ray powder diffraction. Journal of Pharmaceutical and Biomedical Analysis, 49(1), 18–25. https://doi.org/10.1016/j.jpba.2008.09.054
  • Chintha, N., Jupudi, S., Palathoti, N., Bharathi J, J., & Justin, A. (2023). In-silico docking and molecular dynamic introspective study of multiple targets of AChE with Rivastigmine and NMDA receptors with Riluzole for Alzheimer’s disease. Journal of Biomolecular Structure & Dynamics, 41(22), 12620–12631. https://doi.org/10.1080/07391102.2023.2167119
  • Chitneni, M., Peh, K. K., Darwis, D., Abdulkarim, M., Abdullah, G. Z., & Qureshi, M. J. (2011). Intestinal permeability studies of sulpiride incorporated into self-microemulsifying drug delivery system (SMEDDS). Pakistan Journal of Pharmaceutical Sciences, 24(2), 113–121. PMID:21454158
  • Clark, J. J., Benson, M. L., Smith, R. D., & Carlson, H. A. (2019). Inherent versus induced protein flexibility: Comparisons within and between apo and holo structures. PLoS Computational Biology, 15(1), e1006705. https://doi.org/10.1371/journal.pcbi.1006705
  • Dasmahapatra, U., Kumar, C. K., Das, S., Subramanian, P. T., Murali, P., Isaac, A. E., Ramanathan, K., Mm, B., & Chanda, K. (2022). In-silico molecular modelling, MM/GBSA binding free energy and molecular dynamics simulation study of novel pyrido fused imidazo[4,5-c]quinolines as potential anti-tumor agents. Frontiers in Chemistry, 10, 991369. https://doi.org/10.3389/fchem.2022.991369
  • De Oliveira, M. V. D., Bittencourt Fernandes, G. M., Da Costa, K. S., Vakal, S., & Lima, A. H. (2022). Virtual screening of natural products against 5-enolpyruvylshikimate-3-phosphate synthase using the Anagreen herbicide-like natural compound library. RSC Advances, 12(29), 18834–18847. https://doi.org/10.1039/d2ra02645g
  • De Vivo, M., Masetti, M., Bottegoni, G., & Cavalli, A. (2016). Role of molecular dynamics and related methods in drug discovery. Journal of Medicinal Chemistry, 59(9), 4035–4061. https://doi.org/10.1021/acs.jmedchem.5b01684
  • Dengale, S. J., Grohganz, H., Rades, T., & Löbmann, K. (2016). Recent advances in co-amorphous drug formulations. Advanced Drug Delivery Reviews, 100, 116–125. https://doi.org/10.1016/j.addr.2015.12.009
  • Dengale, S. J., Ranjan, O. P., Hussen, S. S., Krishna, B., Musmade, P. B., Shenoy, G. G., & Bhat, K. (2014). Preparation and characterization of co-amorphous Ritonavir–Indomethacin systems by solvent evaporation technique: Improved dissolution behavior and physical stability without evidence of intermolecular interactions. European Journal of Pharmaceutical Sciences, 62, 57–64. https://doi.org/10.1016/j.ejps.2014.05.015
  • DiLabio, G. A., Koleini, M., & Torres, E. (2013). Extension of the B3LYP–dispersion-correcting potential approach to the accurate treatment of both inter- and intra-molecular interactions. Theoretical Chemistry Accounts, 132(10), 1389. https://doi.org/10.1007/s00214-013-1389-x
  • El Fadili, M., Er-Rajy, M., Ali Eltayb, W., Kara, M., Assouguem, A., Saleh, A., Al Kamaly, O., Zarougui, S., & Elhallaoui, M. (2023). In-silico screening based on molecular simulations of 3,4-disubstituted pyrrolidine sulfonamides as selective and competitive GlyT1 inhibitors. Arabian Journal of Chemistry, 16(10), 105105. https://doi.org/10.1016/j.arabjc.2023.105105
  • El Fadili, M., Er-Rajy, M., Kara, M., Assouguem, A., Belhassan, A., Alotaibi, A., Mrabti, N. N., Fidan, H., Ullah, R., Ercisli, S., Zarougui, S., & Elhallaoui, M. (2022). QSAR, ADMET In silico pharmacokinetics, molecular docking and molecular dynamics studies of novel bicyclo (aryl methyl) benzamides as potent GlyT1 inhibitors for the treatment of schizophrenia. Pharmaceuticals (Basel, Switzerland), 15(6), 670. https://doi.org/10.3390/ph15060670
  • Elangovan, N. D., Dhanabalan, A. K., Gunasekaran, K., Kandimalla, R., & Sankarganesh, D. (2021). Screening of potential drug for Alzheimer’s disease: A computational study with GSK-3 β inhibition through virtual screening, docking, and molecular dynamics simulation. Journal of Biomolecular Structure & Dynamics, 39(18), 7065–7079. https://doi.org/10.1080/07391102.2020.1805362
  • El Fadili, M., Er-Rajy, M., Imtara, H., Noman, O. M., Mothana, R. A., Abdullah, S., Zerougui, S., & Elhallaoui, M. (2023). QSAR, ADME-Tox, molecular docking and molecular dynamics simulations of novel selective glycine transporter type 1 inhibitors with memory enhancing properties. Heliyon, 9(2), e13706. https://doi.org/10.1016/j.heliyon.2023.e13706
  • Fael, H. D., & Levent, A. (2021). Indomethacin co-amorphous drug-drug systems with improved solubility, supersaturation, dissolution rate and physical stability. International Journal of Pharmaceutics, 600, 120448. https://doi.org/10.1016/j.ijpharm.2021.120448
  • Fang, X., Hu, Y., Huang, Z., Han, L., Li, B., Lu, S., & Cao, Y. (2022). Exploring the formation mechanism of coamorphous andrographolide-oxymatrine based on molecular dynamics and spectroscopy. Journal of Pharmaceutical Sciences, 111(7), 2056–2071. https://doi.org/10.1016/j.xphs.2022.02.003
  • Fawcett, R. W. (1973). A radial distribution function analysis of an amorphous calcium phosphate with calcium to phosphorus molar ratio of 1.42. Calcified Tissue Research, 13(4), 319–325. https://doi.org/10.1007/bf02015422
  • Feng, J., Chen, X., Lu, S., Li, W., Yang, D., Su, W., Wang, X., & Shen, J. (2018). Naringin attenuates cerebral ischemia-reperfusion injury through inhibiting peroxynitrite-mediated mitophagy activation. Molecular Neurobiology, 55(12), 9029–9042. https://doi.org/10.1007/s12035-018-1027-7
  • Firdhouse, M. J., & Lalitha, P. (2015). Maestro 9.4 as a tool in the structure based screening of glycoalkaloids and related compounds, targeting aldose reductase. Trends in Bioinformatics, 8(1), 26–36. https://doi.org/10.3923/tb.2015.26.36
  • Garg, A. ,Bhalala, K., Tomar, D. S., & Wahajuddin. (2017). In-situ single pass intestinal permeability andpharmacokinetic study of developed Lumefantrine loaded solid lipid nanoparticles. International Journal of Pharmaceutics, 516(1-2), 120–130. https://doi.org/10.1016/j.ijpharm.2016.10.064.
  • Gollavilli, H., Hegde, A. R., Managuli, R. S., Bhaskar, K. V., Dengale, S. J., Reddy, M. S., Kalthur, G., & Mutalik, S. (2020). Naringin nano-ethosomal novel sunscreen creams: Development and performance evaluation. Colloids and Surfaces B, Biointerfaces, 193, 111122. https://doi.org/10.1016/j.colsurfb.2020.111122
  • Han, J., Wei, Y., Lu, Y., Wang, R., Zhang, J., Gao, Y., & Qian, S. (2020). Co-amorphous systems for the delivery of poorly water-soluble drugs: Recent advances and an update. Expert Opinion on Drug Delivery, 17(10), 1411–1435. https://doi.org/10.1080/17425247.2020.1796631
  • Han, Y., Pan, Y., Lv, J., Guo, W., & Wang, J. (2016). Powder grinding preparation of co-amorphous β-azelnidipine and maleic acid combination: Molecular interactions and physicochemical properties. Powder Technology, 291, 110–120. https://doi.org/10.1016/j.powtec.2015.11.068
  • Hao, H., Zhang, Y., Hu, X., Guo, W., Yang, C., & Wang, J. (2023). Cocrystallization of 5-fluorouracil with gallic acid: A novel 5-fluorouracil cocrystal displaying synergistic anti-tumor activity both in oral and intraperitoneal injection administration. European Journal of Pharmaceutics and Biopharmaceutics, 187, 12–23. https://doi.org/10.1016/j.ejpb.2023.04.001
  • Harvey, R. J., & Yee, B. K. (2013). Glycine transporters as novel therapeutic targets in schizophrenia, alcohol dependence and pain. Nature Reviews Drug Discovery, 12(11), 866–885. https://doi.org/10.1038/nrd3893
  • Hosen, S. Z., Dash, R., Khatun, M., Akter, R., Bhuiyan, M. H. R., Karim, M. R., Mouri, N. J., Ahamed, F., Islam, K. S., & Afrin, S. (2017). In silico ADME/T and 3D QSAR analysis of KDR inhibitors. Journal of Applied Pharmaceutical Science, 7(1), 120–128. https://doi.org/10.7324/JAPS.2017.70116
  • Hu, D., Chen, X., Li, D., Zhang, H., Duan, Y., & Huang, Y. (2023). Tranilast-matrine co-amorphous system: Strong intermolecular interactions, improved solubility, and physiochemical stability. International Journal of Pharmaceutics, 635, 122707. https://doi.org/10.1016/j.ijpharm.2023.122707
  • Ibrahim, W. M., AlOmrani, A. H., & Yassin, A. E. B. (2014). Novel sulpiride-loaded solid lipid nanoparticles with enhanced intestinal permeability. International Journal of Nanomedicine, 9, 129–144. https://doi.org/10.2147/IJN.S54413
  • Impurities: Guideline for Residual Solvents Q3C(R8). (2021). Ich harmonised guideline. https://www.ema.europa.eu/documents/regulatory-procedural-guideline/ich-guideline-q3c-r8-impurities-guideline-residual-solvents-step-5_en.pdf
  • Javitt, D. C. (2012). Glycine transport inhibitors in the treatment of schizophrenia. Novel antischizophrenia treatments (pp. 367–399). https://doi.org/10.1007/978-3-642-25758-2_12
  • Kagami, L. P., das Neves, G. M., Timmers, L. F. S. M., Caceres, R. A., & Eifler-Lima, V. L. (2020). Geo-Measures: A PyMOL plugin for protein structure ensembles analysis. Computational Biology and Chemistry, 87, 107322. https://doi.org/10.1016/j.compbiolchem.2020.107322
  • Kalam, M. A., Alshamsan, A., Alkholief, M., Alsarra, I. A., Ali, R., Haq, N., Anwer, M. K., & Shakeel, F. (2020). Solubility measurement and various solubility parameters of glipizide in different neat solvents. ACS Omega, 5(3), 1708–1716. https://doi.org/10.1021/acsomega.9b04004
  • Kasten, G., Löbmann, K., Grohganz, H., & Rades, T. (2019). Co-former selection for co-amorphous drug-amino acid formulations. International Journal of Pharmaceutics, 557, 366–373. https://doi.org/10.1016/j.ijpharm.2018.12.036
  • Kecel-Gunduz, S., Budama-Kilinc, Y., Cakir-Koc, R., Zorlu, T., Bicak, B., Kokcu, Y., Kaya, Z., Ozel, A. E., & Akyuz, S. (2020). In silico analysis of sulpiride, synthesis, characterization and in vitro studies of its nanoparticle for the treatment of schizophrenia. Current Computer-Aided Drug Design, 16(2), 104–121. https://doi.org/10.2174/1573409915666190627125643
  • Kim, D. S., Choi, J. S., Kim, D. W., Kim, K. S., Seo, Y. G., Cho, K. H., Kim, J. O., Yong, C. S., Youn, Y. S., Lim, S. J., Jin, S. G., & Choi, H. G. (2016). Comparison of solvent wetted and kneaded l-sulpiride loaded solid dispersions: Powder characterization and in vivo evaluation. International Journal of Pharmaceutics, 511(1), 351–358. https://doi.org/10.1016/j.ijpharm.2016.07.006
  • Kim, D. S., Kim, D. W., Kim, K. S., Choi, J. S., Seo, Y. G., Youn, Y. S., Oh, K. T., Yong, C. S., Kim, J. O., Jin, S. G., & Choi, H. G. (2016). Development of a novel l-sulpiride-loaded quaternary microcapsule: Effect of TPGS as an absorption enhancer on physicochemical characterization and oral bioavailability. Colloids and Surfaces B, Biointerfaces, 147, 250–257. https://doi.org/10.1016/j.colsurfb.2016.08.010
  • Kirubakaran, P., Karthikeyan, M., Singh, K. D., Nagamani, S., & Premkumar, K. (2013). In silico structural and functional analysis of the human TOPK protein by structure modeling and molecular dynamics studies. Journal of Molecular Modeling, 19(1), 407–419. https://doi.org/10.1007/s00894-012-1566-1
  • Kohri, N., Naasani, I., Iseki, K., & Miyazaki, K. (1996). Improving the oral bioavailability of sulpiride by a gastric-retained form in rabbits. The Journal of Pharmacy and Pharmacology, 48(4), 371–374. https://doi.org/10.1111/j.2042-7158.1996.tb05935.x
  • Ks, N. S., Dengale, S. J., Mutalik, S., & Bhat, K. (2022). Raloxifene HCl–quercetin co-amorphous system: Preparation, characterization, and investigation of its behavior in phosphate buffer. Drug development industrial pharmacy (just-accepted) (pp. 1–34). https://doi.org/10.1080/03639045.2022.2104308
  • Kumar, N., James, R., Sinha, S., Kinra, M., Anuranjana, P., & Nandakumar, K. (2021). Naringin exhibited Anti-Parkinsonian like effect against haloperidol-induced catalepsy in mice. Research Journal of Pharmacy and Technology, 14(2), 662–666. https://doi.org/10.5958/0974-360X.2021.00118.9
  • Kumari, P., Sharma, B., & Som, A. (2023). (2-Cyclohexyl-1-methylpropyl) cyclohexane isolated from garlic extract exhibits antidepressant-like activity: Extraction, docking, drug-like properties, molecular dynamics simulations and MM/GBSA studies. Journal of Biomolecular Structure & Dynamics, 41(7), 1–13. https://doi.org/10.1080/07391102.2023.2202250
  • Lai, E. C. C., Hsieh, C. Y., Kao Yang, Y. H., & Lin, S. J. (2014). Detecting potential adverse reactions of sulpiride in schizophrenic patients by prescription sequence symmetry analysis. PLoS One, 9(2), e89795. https://doi.org/10.1371/journal.pone.0089795
  • Lai, M., Zhang, X., & Fang, F. (2017). Crystal orientation effect on the subsurface deformation of monocrystalline germanium in nanometric cutting. Nanoscale Research Letters, 12(1), 296. https://doi.org/10.1186/s11671-017-2047-3
  • Laitinen, R., Löbmann, K., Strachan, C. J., Grohganz, H., & Rades, T. (2013). Emerging trends in the stabilization of amorphous drugs. International Journal of Pharmaceutics, 453(1), 65–79. https://doi.org/10.1016/j.ijpharm.2012.04.066
  • Łątka, K., & Bajda, M. (2022). Analysis of binding determinants for different classes of competitive and noncompetitive inhibitors of glycine transporters. International Journal of Molecular Sciences, 23(14), 8050. https://doi.org/10.3390/ijms23148050
  • Li, B., Hu, Y., Guo, Y., Xu, R., Fang, X., Xiao, X., Jiang, C., & Lu, S. (2021). Coamorphous system of Florfenicol-Oxymatrine for improving the solubility and dissolution rate of Florfenicol: Preparation, characterization and molecular dynamics simulation. Journal of Pharmaceutical Sciences, 110(6), 2544–2554. https://doi.org/10.1016/j.xphs.2021.02.005
  • Li, B., Wang, Y., Feng, Y., Yuan, D., Xu, R., Jiang, C., Xiao, X., & Lu, S. (2022). Design and molecular insights of drug-active metabolite based co-amorphous formulation: A case study of toltrazuril-ponazuril co-amorphous. International Journal of Pharmaceutics, 615, 121475. https://doi.org/10.1016/j.ijpharm.2022.121475
  • Liu, W., Pan, H., Zhang, C., Zhao, L., Zhao, R., Zhu, Y., & Pan, W. (2016). Developments in methods for measuring the intestinal absorption of nanoparticle-bound drugs. International Journal of Molecular Sciences, 17(7), 1171. https://doi.org/10.3390/ijms17071171
  • Löbmann, K., Grohganz, H., Laitinen, R., Strachan, C., & Rades, T. (2013). Amino acids as co-amorphous stabilizers for poorly water soluble drugs–Part 1: Preparation, stability and dissolution enhancement. European Journal of Pharmaceutics and Biopharmaceutics, 85(3 Pt B), 873–881. https://doi.org/10.1016/j.ejpb.2013.03.014
  • Lokhande, K. B., Doiphode, S., Vyas, R., & Swamy, K. V. (2021). Molecular docking and simulation studies on SARS-CoV-2 Mpro reveals Mitoxantrone, Leucovorin, Birinapant, and Dynasore as potent drugs against COVID-19. Journal of Biomolecular Structure & Dynamics, 39(18), 7294–7305. https://doi.org/10.1080/07391102.2020.1805019
  • Lu, L. (2015). Can B3LYP be improved by optimization of the proportions of exchange and correlation functionals? International Journal of Quantum Chemistry, 115(8), 502–509. https://doi.org/10.1002/qua.24876
  • M’bitsi-Ibouily, G., Marimuthu, T., Kumar, P., Choonara, Y., Toit, L., Pradeep, P., Modi, G., & Pillay, V. (2019). Synthesis, characterisation and in vitro permeation, dissolution and cytotoxic evaluation of ruthenium(II)-liganded sulpiride and amino alcohol. Scientific Reports, 9(1), 4146. https://doi.org/10.1038/s41598-019-40538-1
  • Malgarim Cordenonsi, L., Sponchiado, R., Campanharo, S. C., Garcia, C., Raffin, R., & Schapoval, E. (2017). Study of flavonoids present in pomelo (Citrus Maxima) BY DSC, UV-VIS, IR, 1H and 13C NMR and MS. Drug Analytical Research, 1(1), 31–37. https://doi.org/10.22456/2527-2616.74097
  • Mizoguchi, R., Waraya, H., & Hirakura, Y. (2019). Application of co-amorphous technology for improving the physicochemical properties of amorphous formulations. Molecular Pharmaceutics, 16(5), 2142–2152. https://doi.org/10.1021/acs.molpharmaceut.9b00105
  • Mohamed, G. G., & Soliman, M. H. (2010). Synthesis, spectroscopic and thermal characterization of sulpiride complexes of iron, manganese, copper, cobalt, nickel, and zinc salts. Antibacterial and antifungal activity. Spectrochimica Acta Part A, Molecular and Biomolecular Spectroscopy, 76(3–4), 341–347. https://doi.org/10.1016/j.saa.2010.03.016
  • Moinuddin, S. M., Ruan, S., Huang, Y., Gao, Q., Shi, Q., Cai, B., & Cai, T. (2017). Facile formation of co-amorphous atenolol and hydrochlorothiazide mixtures via cryogenic-milling: Enhanced physical stability, dissolution and pharmacokinetic profile. International Journal of Pharmaceutics, 532(1), 393–400. https://doi.org/10.1016/j.ijpharm.2017.09.020
  • Nagamani, S., Muthusamy, K., Kirubakaran, P., Singh, K. D., & Krishnasamy, G. (2012). Theoretical studies on benzimidazole derivatives as E. coli biotin carboxylase inhibitors. Medicinal Chemistry Research, 21(9), 2169–2180. https://doi.org/10.1007/s00044-011-9738-6
  • Naik, B., Mattaparthi, V. S. K., Gupta, N., Ojha, R., Das, P., Singh, S., Prajapati, V. K., & Prusty, D. (2022). Chemical system biology approach to identify multi-targeting FDA inhibitors for treating COVID-19 and associated health complications. Journal of Biomolecular Structure & Dynamics, 40(19), 9543–9567. https://doi.org/10.1080/07391102.2021.1931451
  • Otte, A., Zhang, Y., Carvajal, M. T., & Pinal, R. (2012). Milling induces disorder in crystalline griseofulvin and order in its amorphous counterpart. CrystEngComm, 14(7), 2560–2570. https://doi.org/10.1039/c2ce06365d
  • Panda, S. P., Roy, P., Soren, D., Ranjan Sahoo, D., Dehury, B., Rout, A. K., Behera, B. K., & Das, B. K. (2023). Structural insights of Labeo catla (catla) myxovirus resistance protein,GTP binding recognition and constitutive expression induced with Poly I:C. Journal of Biomolecular Structure & Dynamics, 42 (1), 1–15. https://doi.org/10.1080/07391102.2023.2213345
  • Pandey, R. K., Kumbhar, B. V., Srivastava, S., Malik, R., Sundar, S., Kunwar, A., & Prajapati, V. K. (2017). Febrifugine analogues as Leishmania donovani trypanothione reductase inhibitors: Binding energy analysis assisted by molecular docking, ADMET and molecular dynamics simulation. Journal of Biomolecular Structure & Dynamics, 35(1), 141–158. https://doi.org/10.1080/07391102.2015.1135298
  • Pandi, P., Bulusu, R., Kommineni, N., Khan, W., & Singh, M. (2020). Amorphous solid dispersions: An update for preparation, characterization, mechanism on bioavailability, stability, regulatory considerations and marketed products. International Journal of Pharmaceutics, 586, 119560. https://doi.org/10.1016/j.ijpharm.2020.119560
  • Pang, W., Lv, J., Du, S., Wang, J., Wang, J., & Zeng, Y. (2017). Preparation of curcumin-piperazine coamorphous phase and fluorescence spectroscopic and density functional theory simulation studies on the interaction with bovine serum albumin. Molecular Pharmaceutics, 14(9), 3013–3024. https://doi.org/10.1021/acs.molpharmaceut.7b00217
  • Panwar, U., & Singh, S. K. (2021). Atom-based 3D-QSAR, molecular docking, DFT, and simulation studies of acylhydrazone, hydrazine, and diazene derivatives as IN-LEDGF/p75 inhibitors. Structural Chemistry, 32(1), 337–352. https://doi.org/10.1007/s11224-020-01628-3
  • Pardhi, E., Tomar, D. S., Khemchandani, R., Samanthula, G., Singh, P. K., & Mehra, N. K. (2023). Design, development and characterization of the Apremilast and Indomethacin coamorphous system. Journal of Molecular Structure, 1299, 137045. https://doi.org/10.1016/j.molstruc.2023.137045
  • PerkinElmer Informatics 2020, ChemDraw 20.0. https://perkinelmerinformatics.com
  • Pešić, N., Dapčević, A., Ivković, B., Kachrimanis, K., Mitrić, M., Ibrić, S., & Medarević, D. (2021). Potential application of low molecular weight excipients for amorphization and dissolution enhancement of carvedilol. International Journal of Pharmaceutics, 608, 121033. https://doi.org/10.1016/j.ijpharm.2021.121033
  • Pinheiro Barros, M. C., Sousa Lima, M. A., Braz-Filho, R., & Rocha Silveira, E. (2003). 1H and 13C NMR assignments of abietane diterpenes from Aegiphila lhotzkyana. Magnetic Resonance in Chemistry, 41(9), 731–734. https://doi.org/10.1002/mrc.1236
  • R Sá, M., Sarraguça, J. M. G., de Sousa, F. F., Sarraguça, M. S. C., Lopes, J. A., Lima, A. D. D S. G., Lage, M. R., & Ribeiro, P. R. S. (2022). Structural, thermal, vibrational, solubility and DFT studies of a tolbutamide co-amorphous drug delivery system for treatment of diabetes. International Journal of Pharmaceutics, 615, 121500. https://doi.org/10.1016/j.ijpharm.2022.121500
  • Raghunathan, S., El Hage, K., Desmond, J. L., Zhang, L., & Meuwly, M. (2018). The role of water in the stability of wild-type and mutant insulin dimers. The Journal of Physical Chemistry B, 122(28), 7038–7048. https://doi.org/10.1021/acs.jpcb.8b04448
  • Rahmani, R., Boukabcha, N., Chouaih, A., Hamzaoui, F., & Goumri-Said, S. (2018). On the molecular structure, vibrational spectra, HOMO-LUMO, molecular electrostatic potential, UV–Vis, first order hyperpolarizability, and thermodynamic investigations of 3-(4-chlorophenyl)-1-(1yridine-3-yl) prop-2-en-1-one by quantum chemistry calculations. Journal of Molecular Structure, 1155, 484–495. https://doi.org/10.1016/j.molstruc.2017.11.033
  • Rosenbrock, H., Desch, M., & Wunderlich, G. (2023). Development of the novel GlyT1 inhibitor, iclepertin (BI 425809), for the treatment of cognitive impairment associated with schizophrenia. European Archives of Psychiatry and Clinical Neuroscience, 273(7), 1557–1566. https://doi.org/10.1007/s00406-023-01576-z
  • Russo, M. G., Baldoni, H. A., Dávila, Y. A., Brusau, E. V., Ellena, J. A., & Narda, G. E. (2018). Rational design of a famotidine–ibuprofen coamorphous system: An experimental and theoretical study. The Journal of Physical Chemistry. B, 122(37), 8772–8782. https://doi.org/10.1021/acs.jpcb.8b06105
  • Salam, N. K., Nuti, R., & Sherman, W. (2009). Novel method for generating structure-based pharmacophores using energetic analysis. Journal of Chemical Information and Modeling, 49(10), 2356–2368. https://doi.org/10.1021/ci900212v
  • Shahsavar, A., Stohler, P., Bourenkov, G., Zimmermann, I., Siegrist, M., Guba, W., Pinard, E., Sinning, S., Seeger, M. A., Schneider, T. R., Dawson, R. J. P., & Nissen, P. (2021). Structural insights into the inhibition of glycine reuptake. Nature, 591(7851), 677–681. https://doi.org/10.1038/s41586-021-03274-z
  • Shi, Q., Moinuddin, S. M., & Cai, T. (2019). Advances in coamorphous drug delivery systems. Acta Pharmaceutica Sinica B, 9(1), 19–35. https://doi.org/10.1016/j.apsb.2018.08.002
  • Shirasaka, Y., Li, Y., Shibue, Y., Kuraoka, E., Spahn-Langguth, H., Kato, Y., Langguth, P., & Tamai, I. (2009). Concentration-dependent effect of naringin on intestinal absorption of β1-adrenoceptor antagonist talinolol mediated by P-glycoprotein and organic anion transporting polypeptide (Oatp). Pharmaceutical Research, 26(3), 560–567. https://doi.org/10.1007/s11095-008-9771-4
  • Shukr, M. H., & Ahmed Farid, O. A. (2018). Amisulpride–CD-loaded liposomes: Optimization and in vivo evaluation. AAPS PharmSciTech, 19(6), 2658–2671. https://doi.org/10.1208/s12249-018-1079-z
  • Simoben, C., Robaa, D., Chakrabarti, A., Schmidtkunz, K., Marek, M., Lancelot, J., Kannan, S., Melesina, J., Shaik, T., Pierce, R., Romier, C., Jung, M., & Sippl, W. (2018). A novel class of Schistosoma mansoni histone deacetylase 8 (HDAC8) inhibitors identified by structure-based virtual screening and in vitro testing. Molecules (Basel, Switzerland), 23(3), 566. https://doi.org/10.3390/molecules23030566
  • Singh, K. D., Kirubakaran, P., Nagarajan, S., Sakkiah, S., Muthusamy, K., Velmurgan, D., & Jeyakanthan, J. (2012). Homology modeling, molecular dynamics, e-pharmacophore mapping and docking study of Chikungunya virus nsP2 protease. Journal of Molecular Modeling, 18(1), 39–51. https://doi.org/10.1007/s00894-011-1018-3
  • Skotnicki, M., Jadach, B., Skotnicka, A., Milanowski, B., Tajber, L., Pyda, M., & Kujawski, J. (2021). Physicochemical characterization of a co-amorphous atorvastatin-irbesartan system with a potential application in fixed-dose combination therapy. Pharmaceutics, 13(1), 118. https://doi.org/10.3390/pharmaceutics13010118
  • Srolovitz, D., Egami, T., & Vitek, V. (1981). Radial distribution function and structural relaxation in amorphous solids. Physical Review B, 24(12), 6936–6944. https://doi.org/10.1103/PhysRevB.24.6936
  • Surampalli, G. Nanjwade, B. K., & Patil, P.(2013). Drug Development and Industrial Pharmacy. Drug Development and Industrial Pharmacy, 1, 170–176. https://doi.org/10.3109/03639045.2013.850716
  • Teja, A., Musmade, P. B., Khade, A. B., & Dengale, S. J. (2015). Simultaneous improvement of solubility and permeability by fabricating binary glassy materials of Talinolol with Naringin: Solid state characterization, in-vivo in-situ evaluation. European Journal of Pharmaceutical Sciences, 78, 234–244. https://doi.org/10.1016/j.ejps.2015.08.002
  • Terban, M. W., & Billinge, S. J. (2021). Structural analysis of molecular materials using the pair distribution function. Chemical Reviews, 122(1), 1208–1272. https://doi.org/10.1021/acs.chemrev.1c00237
  • Tiwari, S., Arya, N., Mishra, S. K., & Suryaprakash, N. (2021). Competing HB acceptors: An extensive NMR investigations corroborated by single crystal XRD and DFT calculations. RSC Advances, 11(25), 15195–15202. https://doi.org/10.1039/d1ra02538d
  • Tomar, D., Singh, P. K., Hoque, S., Modani, S., Sriram, A., Kumar, R., Madan, J., Khatri, D., & Dua, K. (2022). Amorphous systems for delivery of nutraceuticals: Challenges opportunities. Critical Reviews in Food Science and Nutrition, 62(5), 1204–1221. https://doi.org/10.1080/10408398.2020.1836607
  • Uppala, S., Vullendula, S. K. A., Yarlagadda, D. L., & Dengale, S. J. (2022). Exploring the utility of co-amorphous materials to concurrently improve the solubility and permeability of Fexofenadine. Journal of Drug Delivery Science and Technology, 72, 103431. https://doi.org/10.1016/j.jddst.2022.103431
  • Vullendula, S. K. A., Nair, A. R., Yarlagadda, D. L., Navya Sree, K. S., Bhat, K., & Dengale, S. J. (2022). Polymeric solid dispersion Vs co-amorphous technology: A critical comparison. Journal of Drug Delivery Science and Technology, 78, 103980. https://doi.org/10.1016/j.jddst.2022.103980
  • Wang, E., Sun, H., Wang, J., Wang, Z., Liu, H., Zhang, J. Z. H., & Hou, T. (2019). End-point binding free energy calculation with MM/PBSA and MM/GBSA: Strategies and applications in drug design. Chemical Reviews, 119(16), 9478–9508. https://doi.org/10.1021/acs.chemrev.9b00055
  • Wang, W., Li, X., Wang, Q., Zhu, X., Zhang, Q., & Du, L. (2018). The acidic pH-induced structural changes in apo-CP43 by spectral methodologies and molecular dynamics simulations. Journal of Molecular Structure, 1152, 177–188. https://doi.org/10.1016/j.molstruc.2017.09.082
  • Warren, H. R., & Hejmadi, M. (2016). Effect of hypoxia on chemosensitivity to 5-fluorouracil in SH-SY5Y neuroblastoma cells. Bioscience Horizons, 9, hzw005. https://doi.org/10.1093/biohorizons/hzw005
  • Wei, Y., Zhou, S., Hao, T., Zhang, J., Gao, Y., & Qian, S. (2019). Further enhanced dissolution and oral bioavailability of docetaxel by coamorphization with a natural P-gp inhibitor myricetin. European Journal of Pharmaceutical Sciences, 129, 21–30. https://doi.org/10.1016/j.ejps.2018.12.016
  • Xu, Y., Shrestha, N., Préat, V., & Beloqui, A. (2021). An overview of in vitro, ex vivo and in vivo models for studying the transport of drugs across intestinal barriers. Advanced Drug Delivery Reviews, 175, 113795. https://doi.org/10.1016/j.addr.2021.05.005
  • Yang, C., Di, P., Fu, J., Xiong, H., Jing, Q., Ren, G., Tang, Y., Zheng, W., Liu, G., & Ren, F. (2017). Improving the physicochemical properties of bicalutamide by complex formation with bovine serum albumin. European Journal of Pharmaceutical Sciences, 106, 381–392. https://doi.org/10.1016/j.ejps.2017.05.059
  • Yang, Y., Tang, W., Liu, S., Han, D., Liu, Y., & Gong, J. (2017). Solubility of benzoin in three binary solvent mixtures and investigation of intermolecular interactions by molecular dynamic simulation. Journal of Molecular Liquids, 243, 472–483. https://doi.org/10.1016/j.molliq.2017.07.125
  • Yarlagadda, D. L., Sai Krishna Anand, V., Nair, A. R., Navya Sree, K. N., Dengale, S. J., & Bhat, K. (2021a). Considerations for the selection of co-formers in the preparation of co-amorphous formulations. International Journal of Pharmaceutics, 602, 120649. https://doi.org/10.1016/j.ijpharm.2021.120649
  • Yarlagadda, D. L., Sai Krishna Anand, V., Nair, A. R., Navya Sree, K. S., Dengale, S. J., & Bhat, K. (2021b). Considerations for the selection of co-formers in the preparation of co-amorphous formulations .  International Journal of Pharmaceutics, 602, 120649. https://doi.org/10.1016/j.ijpharm.2021.120649
  • Yoda, O., Kuriyama, I., & Odajima, A. (1978). RDF analysis for the degree of interchain ordering in amorphous solid polyethylene. Journal of Applied Physics, 49(11), 5468–5472. https://doi.org/10.1063/1.324516
  • Zhang, X., Perez-Sanchez, H., & Lightstone, F. C. (2017). A comprehensive docking and MM/GBSA rescoring study of ligand recognition upon binding antithrombin. Current Topics in Medicinal Chemistry, 17(14), 1631–1639. https://doi.org/10.2174/1568026616666161117112604
  • Zhang, Y., Huo, M., Zhou, J., Zou, A., Li, W., Yao, C., & Xie, S. (2010). DDSolver: An add-in program for modeling and comparison of drug dissolution profiles. The AAPS Journal, 12(3), 263–271. https://doi.org/10.1208/s12248-010-9185-1
  • Zhu, S., Gao, H., Babu, S., & Garad, S. (2018). Co-amorphous formation of high-dose zwitterionic compounds with amino acids to improve solubility and enable parenteral delivery. Molecular Pharmaceutics, 15(1), 97–107. https://doi.org/10.1021/acs.molpharmaceut.7b00738
  • Zochedh, A., Priya, M., Chakaravarthy, C., Sultan, A. B., & Kathiresan, T. (2023). Experimental and computational evaluation of syringic acid – structural, spectroscopic, biological activity and docking simulation. Polycyclic Aromatic Compounds, 43(7), 6516–6548. https://doi.org/10.1080/10406638.2022.2118332

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.