|
1.Fradet, Y., et al., The burden of prostate cancer in Canada. Canadian Urological Association Journal, 2009. 3(3 Suppl 2): p. S92. 2.Bray, F., et al., Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a cancer journal for clinicians, 2018. 68(6): p. 394-424. 3.Ferlay, J., et al., Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer, 2019. 144(8): p. 1941-1953. 4.Barbieri, C.E., et al., The mutational landscape of prostate cancer. Eur Urol, 2013. 64(4): p. 567-76. 5.Bergengren, O., et al., 2022 Update on Prostate Cancer Epidemiology and Risk Factors-A Systematic Review. Eur Urol, 2023. 84(2): p. 191-206. 6.Wang, G., et al., Genetics and biology of prostate cancer. Genes Dev, 2018. 32(17-18): p. 1105-1140. 7.Chen, J., et al., Translational bioinformatics for diagnostic and prognostic prediction of prostate cancer in the next-generation sequencing era. Biomed Res Int, 2013. 2013: p. 901578. 8.Hjelmborg, J.B., et al., The heritability of prostate cancer in the Nordic Twin Study of Cancer. Cancer Epidemiol Biomarkers Prev, 2014. 23(11): p. 2303-2310. 9.Chopra, S., et al., Comparing oxygen-sensitive MRI (BOLD R2*) with oxygen electrode measurements: A pilot study in men with prostate cancer. International Journal of Radiation Biology, 2009. 85(9): p. 805-813. 10.De Marzo, A.M., et al., Inflammation in prostate carcinogenesis. Nat Rev Cancer, 2007. 7(4): p. 256-69. 11.Hickling, D.R., T.T. Sun, and X.R. Wu, Anatomy and physiology of the urinary tract: relation to host defense and microbial infection. Urinary tract infections: Molecular pathogenesis and clinical management, 2017: p. 1-25. 12.Yamada, Y. and H. Beltran, Clinical and Biological Features of Neuroendocrine Prostate Cancer. Curr Oncol Rep, 2021. 23(2): p. 15. 13.Shen, M.M. and C. Abate-Shen, Molecular genetics of prostate cancer: new prospects for old challenges. Genes Dev, 2010. 24(18): p. 1967-2000. 14.Takayama, K.I., Splicing Factors Have an Essential Role in Prostate Cancer Progression and Androgen Receptor Signaling. Biomolecules, 2019. 9(4). 15.Tannock, I.F., et al., Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med, 2004. 351(15): p. 1502-12. 16.Fizazi, K., et al., Novel and bone-targeted agents for CRPC. Annals of Oncology, 2012. 23: p. x264-x267. 17.Tannock, I.F., et al., Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. New England Journal of Medicine, 2004. 351(15): p. 1502-1512. 18.Frieling, J.S., D. Basanta, and C.C. Lynch, Current and emerging therapies for bone metastatic castration-resistant prostate cancer. Cancer Control, 2015. 22(1): p. 109-20. 19.Ryan, C.J., et al., Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med, 2013. 368(2): p. 138-48. 20.Barrie, S., et al., Pharmacology of novel steroidal inhibitors of cytochrome P45017α (17α-hydroxylase/C17–20 lyase). The Journal of steroid biochemistry and molecular biology, 1994. 50(5-6): p. 267-273. 21.Potter, G.A., et al., Novel steroidal inhibitors of human cytochrome P45017. alpha.-Hydroxylase-C17, 20-lyase): potential agents for the treatment of prostatic cancer. Journal of medicinal chemistry, 1995. 38(13): p. 2463-2471. 22.Vasaitis, T.S., R.D. Bruno, and V.C. Njar, CYP17 inhibitors for prostate cancer therapy. The Journal of steroid biochemistry and molecular biology, 2011. 125(1-2): p. 23-31. 23.Sonpavde, G., et al., The role of abiraterone acetate in the management of prostate cancer: a critical analysis of the literature. European urology, 2011. 60(2): p. 270-278. 24.Thakur, A., et al., Abiraterone acetate in the treatment of prostate cancer. Biomed Pharmacother, 2018. 101: p. 211-218. 25.Akula, S., A.K. Gurram, and S.R. Devireddy, Self-Microemulsifying Drug Delivery Systems: An Attractive Strategy for Enhanced Therapeutic Profile. Int Sch Res Notices, 2014. 2014: p. 964051. 26.Dokania, S. and A.K. Joshi, Self-microemulsifying drug delivery system (SMEDDS)--challenges and road ahead. Drug Deliv, 2015. 22(6): p. 675-90. 27.Dixit, A.R., S.J. Rajput, and S.G. Patel, Preparation and bioavailability assessment of SMEDDS containing valsartan. AAPS PharmSciTech, 2010. 11(1): p. 314-21. 28.Shah, N.H., et al., Self-emulsifying drug delivery systems (SEDDS) with polyglycolyzed glycerides for improving in vitro dissolution and oral absorption of lipophilic drugs. International Journal of Pharmaceutics, 1994. 106(1): p. 15-23. 29.Prince, L., Microemulsions theory and practice. 2012: Elsevier. 30.Hsieh, C.M., et al., Application of Design of Experiments in the Development of Self-Microemulsifying Drug Delivery Systems. Pharmaceuticals (Basel), 2023. 16(2). 31.Pawar, S.D., et al. SELF-MICRO EMULSIFYING DRUG DELIVERY SYSTEM ( SMEDDS ) : A PROMISING DRUG DELIVERY SYSTEM FOR ENHANCEMENT OF BIOAVAILABILITY. 2016. 32.Singh, A.K., et al., Oral bioavailability enhancement of exemestane from self-microemulsifying drug delivery system (SMEDDS). AAPS PharmSciTech, 2009. 10(3): p. 906-16. 33.Wang, Y., et al., Enhanced oral bioavailability of tacrolimus in rats by self-microemulsifying drug delivery systems. Drug development and industrial Pharmacy, 2011. 37(10): p. 1225-1230. 34.Gursoy, R.N. and S. Benita, Self-emulsifying drug delivery systems (SEDDS) for improved oral delivery of lipophilic drugs. Biomedicine & pharmacotherapy, 2004. 58(3): p. 173-182. 35.Prajapati, H.N., et al., Effect of difference in fatty acid chain lengths of medium-chain lipids on lipid-surfactant-water phase diagrams and drug solubility. Journal of Excipients and Food Chemicals, 2011. 2(3): p. 73-88. 36.Stegemann, S., et al., When poor solubility becomes an issue: from early stage to proof of concept. European journal of pharmaceutical sciences, 2007. 31(5): p. 249-261. 37.Gurram, A.K., et al., Role of Components in the Formation of Self-microemulsifying Drug Delivery Systems. Indian J Pharm Sci, 2015. 77(3): p. 249-57. 38.Rosen, M.J. and J.T. Kunjappu, Surfactants and interfacial phenomena. 2012: John Wiley & Sons. 39.Cortés, H., et al., Non-Ionic Surfactants for Stabilization of Polymeric Nanoparticles for Biomedical Uses. Materials, 2021. 14(12): p. 3197. 40.Tapia-Guerrero, Y., et al., Effect of UV and gamma irradiation sterilization processes in the properties of different polymeric nanoparticles for biomedical applications. Materials, 2020. 13(5): p. 1090. 41.Sahu, R.K. and J. Khan, Chapter 7 - Formulation strategies to improve the bioavailability of poorly absorbed drugs, in Advances and Challenges in Pharmaceutical Technology, A.K. Nayak, et al., Editors. 2021, Academic Press. p. 229-242. 42.Čerpnjak, K., et al., Lipid-based systems as promising approach for enhancing the bioavailability of poorly water-soluble drugs. Acta pharmaceutica, 2013. 63(4): p. 427-445. 43.Al-Akayleh, F., et al., Antimicrobial potential of natural deep eutectic solvents. Lett Appl Microbiol, 2022. 75(3): p. 607-615. 44.Buya, A.B., et al., Design and evaluation of self-nanoemulsifying drug delivery systems (SNEDDSs) for senicapoc. International Journal of Pharmaceutics, 2020. 580: p. 119180. 45.Sun, D., et al., In vitro testing of drug absorption for drug 'developability' assessment: Forming an interface between in vitro preclinical data and clinical outcome. Current Opinion in Drug Discovery and Development, 2004. 7(1): p. 75-85. 46.Amidon, G.L., et al., A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharmaceutical research, 1995. 12: p. 413-420. 47.Wu, C.Y. and L.Z. Benet, Predicting drug disposition via application of BCS: transport/absorption/ elimination interplay and development of a biopharmaceutics drug disposition classification system. Pharm Res, 2005. 22(1): p. 11-23. 48.Rane, S.S. and B.D. Anderson, What determines drug solubility in lipid vehicles: is it predictable? Advanced drug delivery reviews, 2008. 60(6): p. 638-656. 49.Colbourn, E. and R. Rowe, Encyclopaedia of pharmaceutical technology. 2005, Marcel Dekker: New York, NY, USA. 50.Wasylaschuk, W.R., et al., Evaluation of hydroperoxides in common pharmaceutical excipients. J Pharm Sci, 2007. 96(1): p. 106-16. 51.Porter, C.J.H. and W.N. Charman, Uptake of drugs into the intestinal lymphatics after oral administration. Advanced Drug Delivery Reviews, 1997. 25(1): p. 71-89. 52.O’Driscoll, C.M., Lipid-based formulations for intestinal lymphatic delivery. European Journal of Pharmaceutical Sciences, 2002. 15(5): p. 405-415. 53.Humberstone, A.J. and W.N. Charman, Lipid-based vehicles for the oral delivery of poorly water soluble drugs. Advanced Drug Delivery Reviews, 1997. 25(1): p. 103-128. 54.Rahman, M.A., et al., Oral lipid based drug delivery system (LBDDS): formulation, characterization and application: a review. Curr Drug Deliv, 2011. 8(4): p. 330-45. 55.Kim, D.W., et al., Comparison of a solid SMEDDS and solid dispersion for enhanced stability and bioavailability of clopidogrel napadisilate. Carbohydr Polym, 2014. 114: p. 365-374. 56.Ziaee, A., et al., Spray drying of pharmaceuticals and biopharmaceuticals: Critical parameters and experimental process optimization approaches. European Journal of Pharmaceutical Sciences, 2019. 127: p. 300-318. 57.Schuck, P., Spray drying of dairy products: state of the art. Le Lait, 2002. 82(4): p. 375-382. 58.Truong, V., B.R. Bhandari, and T. Howes, Optimization of cocurrent spray drying process for sugar-rich foods. Part II—Optimization of spray drying process based on glass transition concept. Journal of Food Engineering, 2005. 71(1): p. 66-72. 59.Couto, R.O., et al., Spray drying of Eugenia dysenterica extract: effects of in-process parameters on product quality. Revista brasileira de Farmacognosia, 2013. 23(1): p. 115-123. 60.Dufour, G., et al., Interest of cyclodextrins in spray-dried microparticles formulation for sustained pulmonary delivery of budesonide. International journal of pharmaceutics, 2015. 495(2): p. 869-878. 61.Liu, Y., et al., The optimization of spray drying process of Lactobacillus reuteri. LWT-Food Science and Technology, 2016. 68: p. 615-618. 62.Lechanteur, A. and B. Evrard, Influence of Composition and Spray-Drying Process Parameters on Carrier-Free DPI Properties and Behaviors in the Lung: A review. Pharmaceutics, 2020. 12(1). 63.Yu, L.X., et al., Understanding pharmaceutical quality by design. Aaps j, 2014. 16(4): p. 771-83. 64.Astakhov, V.P., Design of experiment methods in manufacturing: basics and practical applications, in Statistical and computational techniques in manufacturing. 2012, Springer. p. 1-54. 65.Fisher, R.A., Design of Experiments. British Medical Journal, 1936. 1(3923): p. 554-554. 66.Bezerra, M.A., et al., Response surface methodology (RSM) as a tool for optimization in analytical chemistry. Talanta, 2008. 76(5): p. 965-977. 67.Lee, R., Statistical design of experiments for screening and optimization. Chemie ingenieur technik, 2019. 91(3): p. 191-200. 68.Hanrahan, G. and K. Lu, Application of Factorial and Response Surface Methodology in Modern Experimental Design and Optimization. Critical Reviews in Analytical Chemistry, 2006. 36(3-4): p. 141-151. 69.Jebali, S., et al., Application of Factorial and Doehlert Designs for the Optimization of the Simultaneous Separation and Determination of Antimigraine Drugs in Pharmaceutical Formulations by RP‐HPLC‐UV. International journal of analytical chemistry, 2019. 2019(1): p. 9685750. 70.Bastogne, T., Quality-by-design of nanopharmaceuticals–a state of the art. Nanomedicine: Nanotechnology, Biology and Medicine, 2017. 13(7): p. 2151-2157. 71.Viegas, I.M.A., et al., Multivariate optimization of optical properties of CdSe quantum dots obtained by a facile one-pot aqueous synthesis. Inorganic Chemistry Frontiers, 2019. 6(6): p. 1350-1360. 72.Karimifard, S. and M.R. Alavi Moghaddam, Application of response surface methodology in physicochemical removal of dyes from wastewater: A critical review. Science of The Total Environment, 2018. 640-641: p. 772-797. 73.Wu, C.J. and M.S. Hamada, Experiments: planning, analysis, and optimization. 2011: John Wiley & Sons. 74.Ferreira, S.C., et al., Box-Behnken design: an alternative for the optimization of analytical methods. Analytica chimica acta, 2007. 597(2): p. 179-186. 75.Mousavi, L., Z. Tamiji, and M.R. Khoshayand, Applications and opportunities of experimental design for the dispersive liquid–liquid microextraction method–A review. Talanta, 2018. 190: p. 335-356. 76.Shimojo, A.A., et al., Evaluation of the influence of process parameters on the properties of resveratrol-loaded NLC using 22 full factorial design. Antioxidants, 2019. 8(8): p. 272. 77.Myers, R.H., D.C. Montgomery, and C.M. Anderson-Cook, Response surface methodology: process and product optimization using designed experiments. 2016: John Wiley & Sons. 78.Sy Mohamad, S.F., et al., Application of experimental designs and response surface methods in screening and optimization of reverse micellar extraction. Critical reviews in biotechnology, 2020. 40(3): p. 341-356. 79.Box, G.E. and N.R. Draper, Response surfaces. Mixtures, and Ridge Analyses, 2007. 649. 80.Arepally, D., R.S. Reddy, and T.K. Goswami, Encapsulation of Lactobacillus acidophilus NCDC 016 cells by spray drying: characterization, survival after in vitro digestion, and storage stability. Food Funct, 2020. 11(10): p. 8694-8706. 81.Fernandes, K.A., et al., Availability of Guanitoxin in Water Samples Containing Sphaerospermopsis torques-reginae Cells Submitted to Dissolution Tests. Pharmaceuticals (Basel), 2020. 13(11). 82.Stippler, E., S. Kopp, and J. Dressman, Comparison of US Pharmacopeia Simulated Intestinal Fluid TS (without pancreatin) and Phosphate Standard Buffer pH 6.8, TS of the International Pharmacopoeia with Respect to Their Use in In Vitro Dissolution Testing. Dissolution Technologies, 2004. 11: p. 6-10. 83.Potter, G.A., et al., Novel steroidal inhibitors of human cytochrome P45017 alpha (17 alpha-hydroxylase-C17,20-lyase): potential agents for the treatment of prostatic cancer. J Med Chem, 1995. 38(13): p. 2463-71. 84.Liu, Y., et al., Development of Abiraterone Acetate Nanocrystal Tablets to Enhance Oral Bioavailability: Formulation Optimization, Characterization, In Vitro Dissolution and Pharmacokinetic Evaluation. Pharmaceutics, 2022. 14(6). 85.Lee, J.Y., et al., Soluplus®/TPGS-based solid dispersions prepared by hot-melt extrusion equipped with twin-screw systems for enhancing oral bioavailability of valsartan. Drug Des Devel Ther, 2015. 9: p. 2745-56.
|