Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 The results of biological activity studies have shown that SDs, as a tool of drug release, do not constitute a limiting factor for the execution of in vitro and in vivo tests; indeed, they stand out as a promising system in which the active principle and + 2325 word(s) 2325 2020-10-07 05:04:15 |
2 format correct Meta information modification 2325 2020-10-22 05:09:57 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Oliveira, V.D.S.; De Almeida, A.S.; Albuquerque, I.D.S.; Duarte, F.�.C.; Queiroz, B.C.S.H.; Converti, A.; Lima, �.A.N.D. Therapeutic Applications of Solid Dispersions. Encyclopedia. Available online: https://encyclopedia.pub/entry/2583 (accessed on 29 March 2024).
Oliveira VDS, De Almeida AS, Albuquerque IDS, Duarte F�C, Queiroz BCSH, Converti A, et al. Therapeutic Applications of Solid Dispersions. Encyclopedia. Available at: https://encyclopedia.pub/entry/2583. Accessed March 29, 2024.
Oliveira, Verônica Da Silva, Amanda Silva De Almeida, Ingrid Da Silva Albuquerque, Fernanda Ílary Costa Duarte, Bárbara Cristina Silva Holanda Queiroz, Attilio Converti, Ádley Antonini Neves De Lima. "Therapeutic Applications of Solid Dispersions" Encyclopedia, https://encyclopedia.pub/entry/2583 (accessed March 29, 2024).
Oliveira, V.D.S., De Almeida, A.S., Albuquerque, I.D.S., Duarte, F.�.C., Queiroz, B.C.S.H., Converti, A., & Lima, �.A.N.D. (2020, October 14). Therapeutic Applications of Solid Dispersions. In Encyclopedia. https://encyclopedia.pub/entry/2583
Oliveira, Verônica Da Silva, et al. "Therapeutic Applications of Solid Dispersions." Encyclopedia. Web. 14 October, 2020.
Therapeutic Applications of Solid Dispersions
Edit

Solid dispersions (SDs) are a technological strategy to improve the pharmacological potential of natural or synthetic bioactive molecules, due to the increase in its solubility and bioavailability, leading to a possible improvement of its biological activities. In this sense, the review sought to synthesize and critically examine the studies that address SDs with therapeutic applications, evaluated through in vitro and/or in vivo tests. This bibliographic survey shows the significant therapeutic potential of SDs in the context of the most diverse biological activities. Among these, including in vitro and/or in vivo antitumor, antiparasitic, antimicrobial, antioxidant, anti-inflammatory or cytoprotective activities, while additional activities, such as gastroprotective, hepatoprotective, antidiabetic or antinociceptive, were highlighted by in vivo studies.

Although SDs have already been studied and cited in the literature, the number of studies published with a focus on in vitro and in vivo trials is still relatively small, considering the great potential of these formulations in pharmaceutical technology and with the most diverse applications. The results of biological activity studies showed that SDs, as a drug release tool, is not a limiting factor for the execution of in vitro and in vivo tests. Additionally, it stands out as a promising system in which the active principle and the carrier interact, allowing, in most cases, an increase in the pharmacological potential due to changes in the physicochemical properties of the constituents. Thus, SDs can represent a safe and effective alternative for the development and improvement of drugs directed to a wide range of pharmacological treatments.

solid dispersions drug delivery biological applications

1. Introduction

Many drug candidates have low aqueous solubility, which can make their oral absorption inadequate. According to the literature, approximately 40% of marketed drugs are poorly soluble in water, as are, according to the Biopharmaceutical Classification System, about 40–90% of new drug candidates [1][2][3][4].

The slow dissolution rate and low solubility of some drugs lead to unpredictable bioavailability, non-reproducible clinical response or treatment inefficiency due to low therapeutic plasma levels; therefore, their use must be optimized using formulation strategies capable of improving their administration [5].

Several technological strategies have been developed and employed to circumvent this situation such as complexation with cyclodextrins, particle size reduction by micronization, crystal development, nanotechnology [5], salt formation and solid dispersions (SDs) [6][7]. However, some of these strategies have shown disadvantages including the development of active forms in vivo, high execution cost and considerable levels of toxicity. Some of these limits can be overcome by employing solid dispersions, a viable, well-established and widely-used strategy to increase the dissolution rate and solubility of poorly water-soluble drugs [4][6][7].

SDs can be defined as molecular mixtures of drugs that are not soluble in hydrophilic carriers, which exhibit a drug release profile driven by the polymer properties [8]. The so-called first-generation crystalline SDs, which use crystalline carriers such as urea and sugars including sucrose, dextrose and galactose, have the disadvantage of high thermodynamic stability that prevents a quick release of drugs [9]. In second-generation SDs drugs are dispersed in usually polymeric amorphous carriers [8], while the carriers used in third-generation SDs are surfactants or a mixture of amorphous polymers and surfactants [10].

The use of hydrophilic polymeric matrices to develop SDs for the release of commercial drugs or new drugs candidates has been shown to be a promising alternative able to improve their pharmacokinetic properties [7].

This review deals with SDs, mainly those belonging to the second-generation class that use polymers as carriers. It describes the main in vitro and in vivo activities (Figure 1) of SDs having different compositions and various preparation methods, intending to innovate and enhance the release of poorly water-soluble drugs, as well as improving the biological activities of the loaded bioactive compounds.

Figure 1. Representation of the application of solid dispersions in biological assays.

2. In Vitro Study of Solid Dispersions in Polymeric Matrices 

This section deals with solid dispersions (SDs) prepared to improve the properties and release of poorly soluble drugs and drug candidates of natural or synthetic origin. The preparation and use of SDs have been reported in several in vitro studies, which have been numerically quantified and classified in Figure 2, based on the biological activities of their active compounds, while the main information on these studies is summarized in Table 1.

Figure 2. Quantification and classification of in vitro studies on solid dispersions published in the period from 2009 to 2020.

Table 1. In vitro studies on solid dispersions.

Carrier Type  Substance Cell Type  Activity Improved Characteristics  Reference
OHPP Niclosamide PC-3, HeLa, A549 Anticancer SDs showed higher cytotoxicity to target cells (lower IC50) than the niclosamide solution. [11]
OHPP Paclitaxel PC-3, HeLa, A549 Anticancer SDs showed significantly higher cytotoxicity to target
cells (lower IC50) than the paclitaxel solution.
[12]
PVP/VA
TPGS
Paclitaxel BT-474, MCF-7, SK-BR-3 Anticancer SDs showed higher cytotoxicity against cancer cells compared to the pure drug. [13]
Brij®L4 Chrysin HT29 Anticancer The higher solubility of chrysin in SDs compared to
water solution increased cytotoxicity.
[14]
Poloxamer 407 Curcumin (CM) NCIH460, HeLa, HepG2,
MCF-7 and PLP2
Anticancer Anticancer SDs showed cytotoxicity against all tumor cell lines tested, but no toxic effects on non-tumor cells. [15]
AChE, BChE, GST,
MAO A-B
Enzyme inhibitory
/Antioxidant
SD was able to inhibit the activities of AChE, BChE and
GST in aqueous medium.
LPS-stimulated murine
macrophages (RAW 264.7)
Anti-inflammatory IC50 (inhibitory concentration of 50% NO production by macrophages) > 400 µg/mL.
PVP K30 Zn(II)-curcumin
complex
HepG2, SK-HEP1 Anticancer SD of Zn(II)-curcumin complex had a potent anticancer effect. [16]
HPMC,
PVP K30,
PEG 6000
Telaprevir HepG2 Anticancer The antitumor activity was dose dependent and even with the addition of the polymer the drug maintained its efficacy. [17]
Soluplus ® Angelica gigas Nakai HeLa, HEK 293 Anticancer SD at the concentration of 200 µg/mL showed a significant decrease (to only 17.37%) in cell viability. There was no toxicity to normal cells. [18]
Eudragit S-100 Berberine
hydrochloride (HB)
SW480, HCT116, Caco-2 Anticancer The release of HB from SDs was effective and cell viability was reduced in a dose and time dependent manner. [19]
PVP K30 IIIM-290
Ehrlich ascites
carcinoma cells
Cytotoxic Despite the reduced amount of IIIM-290 in SD, the IC50
value of SD was lower than that of IIIM-290 alone.
[20]
Poloxamer 407  Benznidazole Epimastigotes of
Trypanosoma cruzi
Antichagasic SDs enhanced drug solubility, release kinetics and parasitic activity. [21]
Low-substituted HPC  Benznidazole Epimastigotes and
intracellular amastigotes
of T. cruzi (CL-B5)
Antichagasic SDs had higher antiparasitic activity against amastigotes than epimastigotes. [22]
Gelucire 50/13 Ursolic acid Trypomastigotes of
T. cruzi Y
Antichagasic  Increased antiparasitic activity. [23]
PVP K30,PVP/VA,
Kollidon-CL-M,
sodium starch glycolate
Praziquantel Adult schistosomes of
Schistosoma mansoni
Antischistosomal Increased solubility, better bioavailability and stronger
antiparasitic activity.
[24]
PVP K30 Praziquantel Newly transformed
schistosomula of
S. mansoni and adults
Antischistosomal Increased solubility, reduced dosage especially for
children and increased antiparasitic activity
[25]
Soluplus, PEG 400,
Lutrol F127 and
Lutrol F68
Artemether Schizonts of
Plasmodium falciparum 3D7
Antimalarial Increased dissolution rate, amorphous form, increased
solubility and, mainly, increased antimalarial activity.
[26]
Soluplus, Kollidon
VA64, Plasdone S630
Lumefantrine  ITG cells Antimalarial  Increased antiparasitic activity. [27]
Chitosan  Abietic acid Staphylococcus epidermidis Antimicrobial SD exhibited better MIC values against S. epidermidis
than chitosan and abietic acid alone.
[28]
DPPH radical scavenging Antioxidant SD had higher antioxidant power (IC50 of 0.61 mg/mL)
than abietic acid alone (IC50 of 11 mg/mL).
PVP K30 and HPMCAS Griseofulvin Dermatophytes of
Trichophyton rubrum
NCPF 935
Antimicrobial SDs significantly reduced biofilm formation when
compared to the control.
[29]
Pluronic F127  Gatifloxacin Staphylococcus aureus Antimicrobial The gatifloxacin/Pluronic F127 system exhibited
antimicrobial efficacy when compared to commercialized eye drops.
[30]
PVP K30 Curcumin Salmonella enteritidis Antimicrobial SD had a strong antimicrobial effect on S. enteritidis, while CM alone did not show antimicrobial activity in vitro [31]
HPMC Curcumin Escherichia coli Antimicrobial SD used to prepare phototoxic supersaturated solutions showed significant bactericidal activity against E. coli. [32]
Polaxamer 407 Curcumin E. coli, Pseudomonas
aeruginosa and S. aureus
Antimicrobial The association between SD and silver nanoparticles
increased CM antimicrobial and antioxidant activities.
[33]
DPPH radical scavenging Antioxidant
PVP K25  Quercetin DPPH radical scavenging Antioxidant Increased quercetin antioxidant activity in SD (0.61 ± 0.03 ≤ IC50 ≤ 1.00 ± 0.02 µg/mL). [34]
Mannitol  Coenzyme Q10 Intracellular ROS level Antioxidant The SD with the smallest particle size showed the
greatest absorption of UVB radiation as well as the highest antioxidant activity in vitro.
[35]
PVP K30 Usnic acid DPPH radical scavenging Antioxidant Increased usnic acid solubility and antioxidant activity. [36]
PEG 4000 Luteolin DPPH Antioxidant  Polymers increased luteolin solubility and antioxidant activity. [37]
PVP K30, PEG 6000 and
HPMC 
α,β-Amyrin LPS-stimulated
macrophages J774 
Anti-inflammatory SDs enhanced the anti-inflammatory activity of α,β-amyrin. [7]
HPMC Curcumin HepG2  Cytoprotective SDs showed better cytoprotective activity than pure CM and inhibited cell death induced by t-BHP. [38]

OHPP, Octenylsuccinate hydroxypropyl phytoglycogen; IC50, Half inhibitory concentration; PVP/VA, Polyvinylpyrrolidone/vinyl acetate; TPGS, D-α-toco-pheryl polyethylene glycol-1000-succinate; PVP, Polyvinylpyrrolidone; SD, Solid dispersion; AChE, Acetylcholinesterase; BChE, Butyrylcholinesterase; CM, Curcumin; GST, Glutathione S-transferase; MAO, Monoamine oxidase; LPS, Lipopolysaccharide; NO, Nitric oxide; PEG, Polyethylene glycol; HPMC, Hydroxypropyl methylcellulose; ITG (chloroquine-resistant cell line); DPPH, 2,2-diphenyl-1-picryl-hydrazyl-hydrate; HPMCAS, Hydroxypropyl methylcellulose acetate succinate; MIC, Minimum inhibitory concentration; ROS, Reactive oxygen species; t-BHP, tert-Butylhydroperoxide.

3. In Vivo Studies of Solid Dispersions in Polymeric Matrices

As already mentioned, solid dispersions (SDs) have been used as a strategy in pharmaceutical technology to circumvent some limitations presented by drugs and new bioactive compounds such as low solubility and bioavailability. In this sense, this section addresses in vivo studies on SDs with different biological activities, as shown in Figura 3 and quantitatively expressed in Figure 4 . The main information about these studies is summarized in Table 2.

Figure 3. In vivo activities of solid dispersions.

Figure 4. Quantification and classification of in vivo studies on solid dispersions published in the period from 2009 to 2020.

Table 2. In vivo studies of solid dispersions.

Carrier Type Substance Animal Dose Activity Improved Characteristics  References
PVP K30 IIIM-290 Swiss male mice
(18–23 g)
25, 50 and 75 mg/kg Anticancer SD was able to reduce the IIIM-290 dose by at least 1.5-fold thanks to its efficiency in Ehrlich solid tumor model. [20]
Soluplus® 9-Nitrocamptothecin Male
Sprague-Dawley rats
(20 ± 2 g)
4 mg/kg Anticancer SD showed higher tumor growth inhibitory rate than the pure compound due to improved oral bioavailability. [39]
(+)-Xylitol  (−)-Oleocanthal Athymic nude mice  10 mg/kg Anticancer Treatment with SD showed prevention, lower growth rate and less recurrence of tumor. [40]
PVP K30 Zinc(II)-curcumin
complex
B-NDG, BALB/c mice  100 mg/kg Anticancer SD reduced tumor size and weight in animals. [17]
PVP K30 Selaginella doederleinii
Hieron
BALB/c mice  200 mg/kg Anticancer SD reduced tumor size as well as the level of tumor angiogenesis. [41]
Low substituted HPC Benznidazole Female NMRI mice
(25 ± 2 g) 
25 mg/kg/day  Antichagasic The best SD showed a 96.65% trypanocidal activity, expressed as percentage reduction in the area under the parasitic curve. [22]
PVP K30 Curcumin (CM)  Male Cobb-Vantress
broiler chickens
1 g/kg of feed Antimicrobial The synergistic effect of 0.05% CM/PVP SD with 0.05% boric acid reduced colonization of Salmonella enteritidis in crop and ceca-cecal tonsils. [31]
PVP K30 Taurine-zinc complex Female
Sprague-Dawley rats
(240–260 g)
100 and 200
mg/kg/day
Antioxidant SDs protected rat gastric mucosa from
ethanol-induced injury and increased SOD activity and glutathione level.
[42]]
Gastroprotective
Kollidon (VA64) Triacetylated
andrographolide (TA)
Male
Kunming mice
50, 100 and 200
mg/kg/day
Anti-inflammatory TA-SD prepared with VA64 significantly improved the drug activity against ulcerative colitis. [43]
PVP K30, Poloxamer 188  Curcumin  Female CD-1 mice 100 mg/kg oral doses Anti-inflammatory CM-SD prepared with PVP decreased matrix metallo-peptidase 9 expression and levels of IL-1β and IL-6 cytokines. [44]
Gelucire®50/13-Aerosil®  Curcumin  Rat  10 to 100 mg/kg Anti-inflammatory A CM-SD dose of 100 mg/kg was more effective than 5 mg/kg indomethacin in reducing edema [45]
HPMC, lecithin
and isomalt
 Curcumin Male
Sprague-Dawley rats
5 mg/kg Anti-inflammatory A CM-SD dose of 5 mg/kg had greater anti-inflammatory activity than 50 mg/kg curcumin alone. [46]
Crospovidone  Aceclofenac Male
Sprague-Dawley rats
1 g/cm2
(topical)
Anti-inflammatory The enhanced drug permeation increased the intensity of the anti-inflammatory response. [47]
PEG 8000  Ibuprofen Wistar rats 20 mg/kg Anti-inflammatory All SDs showed better anti-inflammatory activity than the pure drug, allowing up to 90% edema inhibition after 6 h. [48]
Urea and mannitol Flurbiprofen Rat  11.69 mg/kg Anti-inflammatory SD showed better inhibition of rat paw edema up to 16 h. [49]
Paracetamol Meloxicam Rat - Anti-inflammatory SDs reduced by more than 50% the volume of carrageenan-induced tail edema compared to the physical mixture. [50]
PVP K30 Chelerythrine (CHE) Mice 10 mg/kg Anti-inflammatory SD enhanced CHE anti-inflammatory effect by reducing the levels of TNF-α, IL-6 and NO in mice serum. [51]
HPMC Curcumin Male BABL/c mice 200 and 400 mg/kg Hepatoprotective The best SD increased the hepatoprotective
efficacy of CM. 
[38]
HPC Nobiletin Male Sprague-Dawley rats (220 g) 2 mg of drug/kg Hepatoprotective SD was more effective than the crystalline drug in rats with acute liver injury [52]
PVP K30  Silymarin Adult male albino rats (150–200 g) 25 mg/kg Hepatoprotective The best SD improved biomarker rates and had a significantly better hepatoprotective effect than the commercial extract [53]
PVP K30  Silymarin Male Sprague-Dawley rats (190–210 g) 50 mg/kg Hepatoprotective SD improved drug solubility and hepatoprotective activity, reducing the AST levels.  [54]
Poloxamer 188 Repaglinide Wistar rats
(150–250 g) )
(1 mg of drug) Antihyperglycemic SD obtained by the microwave method improved the drug anti-hyperglycemic activity.  [55]
Soluplus1 and PEG 4000 Glimepiride Albino Wistar rats
(200–250 g) 
0.0285 mg of drug/kg Anti-diabetic SD reduced the glucose level in rats more than the pure drug and a commercial product [5]
PVP K17 Pioglitazone Male Swiss albino mice (25–30 g) 30 mg/kg SD  Antihyperglycemic SD reduced the mean glucose level in mice more than the pure drug and a commercial product. [56]
HPMC  Hecogenin acetate Male Swiss mice
(28–35 g)
40 mg/kg  Antinociceptive Both the drug alone and its SD with HPMC-reduced mechanical and thermal hyperalgesia induced by crushing of the sciatic nerve in mice. [57]

PVP, Polyvinylpyrrolidone; SD, Solid dispersion; HPC, Hydroxypropyl cellulose; CM, Curcumin; CM/PVP SD, curcumin/polyvinylpyrrolidone solid dispersion; SOD, Superoxide dismutase; PEG, Polyethylene glycol; TNF-α, tumor necrosis factor alpha; IL, interleukin; NO, Nitric oxide; HPMC, Hydroxypropyl methylcellulose; AST, Aspartate aminotransferase.

4. Conclusions

Solid dispersions are a technological strategy to improve the pharmacological potential of natural or synthetic bioactive molecules, thanks to an increase in their solubility and bioavailability, thus leading to possible enhancement of their biological activities. In this context, this review sought to summarize and critically examine studies conducted with this approach both in vitro and in vivo, which have provided evidence of a significant therapeutic potential that solid dispersions could offer in the context of the most diverse biological activities.

As poorly water-soluble active ingredients incorporated in solid dispersions exhibited improved physicochemical and pharmacological properties, these systems can be targeted in the pharmaceutical industry as possible therapeutic alternatives for certain diseases, with greater selectivity, safety and efficacy than the existing treatments.

References

  1. Kambayashi, A.; Kiyota, T.; Fujiwara, M.; Dressman, J.B. PBPK modeling coupled with biorelevant dissolution to forecast the oral performance of amorphous solid dispersion formulations. Eur. J. Pharm. Sci. 2019, 135, 83–90.
  2. Jermain, S.V.; Brough, C.; Williams, R.O. Amorphous solid dispersions and nanocrystal technologies for poorly water-soluble drug delivery—An update. Int. J. Pharm. 2018, 535, 379–392.
  3. Loftsson, T.; Brewster, M.E. Pharmaceutical applications of cyclodextrins: Basic science and product development. J. Pharm. Pharmacol. 2010, 62, 1607–1621.
  4. Worku, Z.A.; Aarts, J.; Singh, A.; Mooter, G.V.D. Drug–Polymer Miscibility across a Spray Dryer: A Case Study of Naproxen and Miconazole Solid Dispersions. Mol. Pharm. 2014, 11, 1094–1101.
  5. Reginald-Opara, J.N.; Attama, A.; Ofokansi, K.; Umeyor, C.; Kenechukwu, F. Molecular interaction between glimepiride and Soluplus®-PEG 4000 hybrid based solid dispersions: Characterisation and anti-diabetic studies. Int. J. Pharm. 2015, 496, 741–750.
  6. Danda, L.J.D.A.; Batista, L.D.M.; Melo, V.C.S.; Sobrinho, J.L.S.; Soares, M.F.D.L.R. Combining amorphous solid dispersions for improved kinetic solubility of posaconazole simultaneously released from soluble PVP/VA64 and an insoluble ammonio methacrylate copolymer. Eur. J. Pharm. Sci. 2019, 133, 79–85.
  7. Júnior, W.F.D.S.; Pinheiro, J.G.D.O.; De Menezes, D.L.B.; Silva, N.E.D.S.E.; De Almeida, P.D.O.; Lima, E.S.; Veiga-Junior, V.F.; De Azevedo, E.P.; Lima, E.S. Development, Physicochemical Characterization and In Vitro Anti-Inflammatory Activity of Solid Dispersions of α,β Amyrin Isolated from Protium Oilresin. Molecules 2017, 22, 1512.
  8. Vasconcelos, T.; Sarmento, B.; Costa, P.C. Solid dispersions as strategy to improve oral bioavailability of poor water soluble drugs. Drug Discov. Today 2007, 12, 1068–1075.
  9. Leuner, C. Improving drug solubility for oral delivery using solid dispersions. Eur. J. Pharm. Biopharm. 2000, 50, 47–60.
  10. Zhang, Z.; Chen, Y.; Deng, J.; Jia, X.-B.; Zhou, J.; Lv, H. Solid dispersion of berberine–phospholipid complex/TPGS 1000/SiO2: Preparation, characterization and in vivo studies. Int. J. Pharm. 2014, 465, 306–316.
  11. Xie, Y.; Yao, Y. Octenylsuccinate hydroxypropyl phytoglycogen enhances the solubility and in-vitro antitumor efficacy of niclosamide. Int. J. Pharm. 2018, 535, 157–163.
  12. Xie, Y.; Yao, Y. Incorporation with Dendrimer-Like Biopolymer Leads to Improved Soluble Amount and In Vitro Anticancer Efficacy of Paclitaxel. J. Pharm. Sci. 2019, 108, 1984–1990.
  13. Choi, J.-S.; Cho, N.H.; Kim, D.-H.; Park, J.-S. Comparison of paclitaxel solid dispersion and polymeric micelles for improved oral bioavailability and in vitro anti-cancer effects. Mater. Sci. Eng. C 2019, 100, 247–259.
  14. Lee, S.H.; Lee, Y.-S.; Song, J.G.; Han, H.-K. Improved In vivo Effect of Chrysin as an Absorption Enhancer Via the Preparation of Ternary Solid Dispersion with Brij®L4 and Aminoclay. Curr. Drug Deliv. 2018, 16, 86–92.
  15. De Sá, I.S.; Peron, A.P.; Leimann, F.V.; Bressan, G.N.; Krum, B.N.; Fachinetto, R.; Pinela, J.; Calhelha, R.C.; Barreiro, M.F.; Ferreira, I.C.; et al. In vitro and in vivo evaluation of enzymatic and antioxidant activity, cytotoxicity and genotoxicity of curcumin-loaded solid dispersions. Food Chem. Toxicol. 2019, 125, 29–37.
  16. Wu, R.; Mei, X.; Ye, Y.; Xue, T.; Wang, J.; Sun, W.; Lin, C.; Xue, R.; Zhang, J.; Xu, D. Zn(II)-curcumin solid dispersion impairs hepatocellular carcinoma growth and enhances chemotherapy by modulating gut microbiota-mediated zinc homeostasis. Pharmacol. Res. 2019, 150, 104454.
  17. Xiong, X.; Zhang, M.; Hou, Q.; Tang, P.; Suo, Z.; Zhu, Y.; Li, H. Solid dispersions of telaprevir with improved solubility prepared by co-milling: Formulation, physicochemical characterization and cytotoxicity evaluation. Mater. Sci. Eng. C 2019, 105, 110012.
  18. Jiang, Y.; Piao, J.; Cho, H.-J.; Kang, W.-S.; Kim, H. Improvement in antiproliferative activity of Angelica gigas Nakai by solid dispersion formation via hot-melt extrusion and induction of cell cycle arrest and apoptosis in HeLa cells. Biosci. Biotechnol. Biochem. 2015, 79, 1635–1643.
  19. Guo, S.; Wang, G.; Wu, T.; Bai, F.; Xu, J.; Zhang, X. Solid dispersion of berberine hydrochloride and Eudragit® S100: Formulation, physicochemical characterization and cytotoxicity evaluation. J. Drug Deliv. Sci. Technol. 2017, 40, 21–27.
  20. Kumar, V.; Mintoo, M.J.; Mondhe, D.M.; Bharate, S.B.; Vishwakarma, R.A.; Bharate, S.S. Binary and ternary solid dispersions of an anticancer preclinical lead, IIIM-290: In vitro and in vivo studies. Int. J. Pharm. 2019, 570, 118683.
  21. Simonazzi, A.; Davies, C.; Cid, A.G.; Gonzo, E.; Parada, L.; Bermúdez, J.M. Preparation and Characterization of Poloxamer 407 Solid Dispersions as an Alternative Strategy to Improve Benznidazole Bioperformance. J. Pharm. Sci. 2018, 107, 2829–2836.
  22. Fonseca-Berzal, C.; Palmeiro-Roldán, R.; Escario, J.A.; Torrado, S.; Arán, V.J.; Torrado-Santiago, S.; Gómez-Barrio, A. Novel solid dispersions of benznidazole: Preparation, dissolution profile and biological evaluation as alternative antichagasic drug delivery system. Exp. Parasitol. 2015, 149, 84–91.
  23. Eloy, J.O.; Saraiva, J.; De Albuquerque, S.; Marchetti, J.M. Solid Dispersion of Ursolic Acid in Gelucire 50/13: A Strategy to Enhance Drug Release and Trypanocidal Activity. AAPS PharmSciTech 2012, 13, 1436–1445.
  24. Perissutti, B.; Passerini, N.; Trastullo, R.; Keiser, J.; Zanolla, D.; Zingone, G.; Voinovich, D.; Albertini, B. An explorative analysis of process and formulation variables affecting comilling in a vibrational mill: The case of praziquantel. Int. J. Pharm. 2017, 533, 402–412.
  25. Albertini, B.; Perissutti, B.; Bertoni, S.; Zanolla, D.; Franceschinis, E.; Voinovich, D.; Lombardo, F.; Keiser, J.; Passerini, N. Combining Mechanochemistry and Spray Congealing for New Praziquantel Pediatric Formulations in Schistosomiasis Treatment. Int. J. Mol. Sci. 2019, 20, 1233.
  26. Fule, R.A.; Meer, T.A.; Sav, A.R.; Amin, P. Artemether-Soluplus Hot-Melt Extrudate Solid Dispersion Systems for Solubility and Dissolution Rate Enhancement with Amorphous State Characteristics. J. Pharm. 2013, 2013, 1–15.
  27. Fule, R.; Meer, T.; Sav, A.; Amin, P. Solubility and dissolution rate enhancement of lumefantrine using hot melt extrusion technology with physicochemical characterisation. J. Pharm. Investig. 2013, 43, 305–321.
  28. Crucitti, V.C.; Migneco, L.M.; Piozzi, A.; Taresco, V.; Garnett, M.; Argent, R.H.; Francolini, I. Intermolecular interaction and solid state characterization of abietic acid/chitosan solid dispersions possessing antimicrobial and antioxidant properties. Eur. J. Pharm. Biopharm. 2018, 125, 114–123.
  29. Al-Obaidi, H.; Kowalczyk, R.M.; Kalgudi, R.; Zariwala, M.G. Griseofulvin solvate solid dispersions with synergistic effect against fungal biofilms. Colloids Surfaces B Biointerfaces 2019, 184, 110540.
  30. Kanoujia, J.; Kushwaha, P.S.; Saraf, S.A. Evaluation of gatifloxacin pluronic micelles and development of its formulation for ocular delivery. Drug Deliv. Transl. Res. 2014, 4, 334–343.
  31. Hernandez-Patlan, D.; Solis-Cruz, B.; Pontin, K.P.; Latorre, J.D.; Baxter, M.F.A.; Hernandez-Velasco, X.; Merino-Guzman, R.; Méndez-Albores, A.; Hargis, B.M.; Lopez-Arellano, R.; et al. Evaluation of a Solid Dispersion of Curcumin With Polyvinylpyrrolidone and Boric Acid Against Salmonella Enteritidis Infection and Intestinal Permeability in Broiler Chickens: A Pilot Study. Front. Microbiol. 2018, 9, 1289.
  32. Hegge, A.B.; Vukićević, M.; Bruzell, E.; Kristensen, S.; Tønnesen, H. Solid dispersions for preparation of phototoxic supersaturated solutions for antimicrobial photodynamic therapy (aPDT). Eur. J. Pharm. Biopharm. 2013, 83, 95–105.
  33. Alves, T.; Chaud, M.; Grotto, D.; Jozala, A.F.; Pandit, R.; Rai, M.; Dos Santos, C.A. Association of Silver Nanoparticles and Curcumin Solid Dispersion: Antimicrobial and Antioxidant Properties. AAPS PharmSciTech 2017, 19, 225–231.
  34. Costa, A.R.D.M.; Marquiafável, F.S.; Vaz, M.M.D.O.L.L.; Rocha, B.A.; Bueno, P.C.P.; Amaral, P.L.M.; Barud, H.D.S.; Berreta-Silva, A.A. Quercetin-PVP K25 solid dispersions. J. Therm. Anal. Calorim. 2010, 104, 273–278.
  35. Ge, Y.; Zhao, X.; Wang, S.; Zu, Y.; Zhao, T.; Sang, M.; Sui, X.; Wang, K. 10 (CoQ10) Prepared by Heat Melt and High-pressure Homogenization Method. Curr. Nanosci. 2014, 10, 292–296.
  36. Fitriani, L.; Rismawati, E.; Umar, S.; Zaini, E. Solid dispersion of usnic acid-PVP K30 and evaluation of antioxidant activity. Rasayan J. Chem. 2018, 11, 1643–1648.
  37. Alshehri, S.; Imam, S.S.; Altamimi, M.A.; Hussain, A.; Shakeel, F.; Elzayat, E.M.; Mohsin, K.; Ibrahim, M.; Alanazi, F. Enhanced Dissolution of Luteolin by Solid Dispersion Prepared by Different Methods: Physicochemical Characterization and Antioxidant Activity. ACS Omega 2020, 5, 6461–6471.
  38. Shin, M.-S.; Yu, J.S.; Lee, J.; Ji, Y.S.; Joung, H.J.; Han, Y.-M.; Yoo, H.H.; Kang, K.; Shin, Y. A Hydroxypropyl Methylcellulose-Based Solid Dispersion of Curcumin with Enhanced Bioavailability and its Hepatoprotective Activity. Biomolecules 2019, 9, 281.
  39. Lian, X.; Dong, J.; Zhang, J.; Teng, Y.; Lin, Q.; Fu, Y.; Gong, T. Soluplus® based 9-nitrocamptothecin solid dispersion for peroral administration: Preparation, characterization, in vitro and in vivo evaluation. Int. J. Pharm. 2014, 477, 399–407.
  40. Qusa, M.H.; Siddique, A.B.; Nazzal, S.; El Sayed, K. Novel olive oil phenolic (−)-oleocanthal (+)-xylitol-based solid dispersion formulations with potent oral anti-breast cancer activities. Int. J. Pharm. 2019, 569, 118596.
  41. Chen, B.; Wang, X.; Zhang, Y.; Huang, K.; Liu, H.; Xu, D.; Li, S.; Liu, Q.; Huang, J.; Yao, H.; et al. Improved solubility, dissolution rate and oral bioavailability of main biflavonoids from Selaginella doederleinii extract by amorphous solid dispersion. Drug Deliv. 2020, 27, 309–322.
  42. Yu, C.; Mei, X.-T.; Zheng, Y.-P.; Xu, D. Gastroprotective effect of taurine zinc solid dispersions against absolute ethanol-induced gastric lesions is mediated by enhancement of antioxidant activity and endogenous PGE2 production and attenuation of NO production. Eur. J. Pharmacol. 2014, 740, 329–336.
  43. Gou, J.; Fei, S.; Xue, B.; Zhang, J.; Zhang, Y.; Wang, X.; Zhang, Y.; Yin, T.; He, H.; Tang, X. Triacetylated andrographolide solid dispersions: Preparation, stability study and in vivo anti-inflammation in mice ulcerative colitis model. J. Drug Deliv. Sci. Technol. 2019, 51, 91–100.
  44. He, Y.; Liu, H.; Bian, W.; Liu, Y.; Liu, X.; Ma, S.; Zheng, X.; Du, Z.; Zhang, K.; Ouyang, D. Molecular Interactions for the Curcumin-Polymer Complex with Enhanced Anti-Inflammatory Effects. Pharmaceutics 2019, 11, 442.
  45. Teixeira, C.C.C.; Mendonça, L.M.; Bergamaschi, M.M.; Queiroz, R.H.C.; Souza, G.E.P.; Antunes, L.M.G.; Freitas, L. Microparticles Containing Curcumin Solid Dispersion: Stability, Bioavailability and Anti-Inflammatory Activity. AAPS PharmSciTech 2015, 17, 252–261.
  46. Chuah, A.M.; Jacob, B.; Jie, Z.; Ramesh, S.; Mandal, S.; Puthan, J.K.; Deshpande, P.; Vaidyanathan, V.V.; Gelling, R.W.; Patel, G.; et al. Enhanced bioavailability and bioefficacy of an amorphous solid dispersion of curcumin. Food Chem. 2014, 156, 227–233.
  47. Jana, S.; Ali, S.A.; Nayak, A.K.; Sen, K.K.; Basu, S.K. Development of topical gel containing aceclofenac-crospovidone solid dispersion by “Quality by Design (QbD)” approach. Chem. Eng. Res. Des. 2014, 92, 2095–2105.
  48. Kenechukwu, F.C.; Ofokansi, K.C.; Ezugwu, R.O.; Attama, A.A. Improved dissolution and anti-inflammatory activity of ibuprofen-polyethylene glycol 8000 solid dispersion systems. Int. J. Pharm. Investig. 2016, 6, 139–147.
  49. Malipeddi, V.R.; Dua, K.; Awasthi, R. Development and characterization of solid dispersion-microsphere controlled release system for poorly water-soluble drug. Drug Deliv. Transl. Res. 2016, 6, 540–550.
  50. Al-Remawi, M.; Ali, A.M.A.; Khames, A.; Hamaidi, M. Meloxicam-Paracetamol Binary Solid Dispersion Systems with Enhanced Solubility and Dissolution Rate:Preparation, Characterization and In Vivo Evaluation. J. Pharm. Innov. 2017, 12, 206–215.
  51. Li, W.; Qing, S.; Zhi, W.; Yao, H.; Fu, C.; Niu, X. The pharmacokinetics and anti-inflammatory effects of chelerythrine solid dispersions in vivo. J. Drug Deliv. Sci. Technol. 2017, 40, 51–58.
  52. Onoue, S.; Nakamura, T.; Uchida, A.; Ogawa, K.; Yuminoki, K.; Hashimoto, N.; Hiza, A.; Tsukaguchi, Y.; Asakawa, T.; Kan, T.; et al. Physicochemical and biopharmaceutical characterization of amorphous solid dispersion of nobiletin, a citrus polymethoxylated flavone, with improved hepatoprotective effects. Eur. J. Pharm. Sci. 2013, 49, 453–460.
  53. Balata, G.; Shamrool, H. Spherical agglomeration versus solid dispersion as different trials to optimize dissolution and bioactivity of silymarin. J. Drug Deliv. Sci. Technol. 2014, 24, 478–485.
  54. Hwang, D.H.; Kim, Y.-I.; Cho, K.H.; Poudel, B.K.; Choi, J.Y.; Kim, D.-W.; Shin, Y.-J.; Bae, O.-N.; Yousaf, A.M.; Yong, C.S.; et al. A novel solid dispersion system for natural product-loaded medicine: Silymarin-loaded solid dispersion with enhanced oral bioavailability and hepatoprotective activity. J. Microencapsul. 2014, 31, 619–626.
  55. Zawar, L.R.; Bari, S.B. Preparation, characterization and in vivo evaluation of antihyperglycemic activity of microwave generated repaglinide solid dispersion. Chem. Pharm. Bull. 2012, 60, 482–487.
  56. Pokharkar, V.; Kutwal, M.; Mandpe, L. Pioglitazone Solid Dispersion System Prepared by Spray Drying Method: In Vitro and In Vivo Evaluation. PDA J. Pharm. Sci. Technol. 2013, 67, 23–34.
  57. Moreira, C.D.L.D.F.A.; Pinheiro, J.G.D.O.; Da Silva-Júnior, W.F.; Barbosa, E.G.; Lavra, Z.M.M.; Pereira, E.W.M.; Resende, M.M.; De Azevedo, E.P.; Quintans-Júnior, L.J.; Araújo, A.A.D.S.; et al. Amorphous solid dispersions of hecogenin acetate using different polymers for enhancement of solubility and improvement of anti-hyperalgesic effect in neuropathic pain model in mice. Biomed. Pharmacother. 2018, 97, 870–879.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , ,
View Times: 622
Revisions: 2 times (View History)
Update Date: 22 Oct 2020
1000/1000