Drug Res (Stuttg) 2021; 71(08): 417-428
DOI: 10.1055/a-1518-6606
Review

Exploring Nanoemulsions for Prostate Cancer Therapy

Sushmita Srivastava
1   Faculty of Pharmacy, Integral University, Lucknow, India
,
Md. Faheem Haider
1   Faculty of Pharmacy, Integral University, Lucknow, India
,
Afroz Ahmad
1   Faculty of Pharmacy, Integral University, Lucknow, India
,
Usama Ahmad
1   Faculty of Pharmacy, Integral University, Lucknow, India
,
Muhammad Arif
1   Faculty of Pharmacy, Integral University, Lucknow, India
,
Asad Ali
1   Faculty of Pharmacy, Integral University, Lucknow, India
› Institutsangaben

Abstract

Prostate carcinoma is typical cancer. It is the second most common cancer globally. The estimated new cases in 2020 was 191 930 and estimated deaths was 33 330. Age, family history, & genetic factors are major factors that drive prostate cancer. Although, for treating metastatic disease, the major therapies available are radiation,bisphosphonate, and palliative chemotherapy. But the major drawback is therapy is disease-driven and later becomes metastatic and requires treatment. The ability to revolutionize cancer treatment by major targeting vehicles via the exploration of nanoemulsion suggests a potential for cancer treatment. The unique property of a biphasic liquid dosage form called nanoemulsion to reach leaky tumor vasculature is due to its nano-meter oil-droplet size of 20–200 nm. Recent reporting on nanoemulsions disclose their embracing and lay alternative for re-purposing herbal and synthetic drugs and their combination especially for targeting prostate cancer formulating an obtainable nanomedicine. So, this article emphasizes the use of nanoemulsions incorporating therapeutic agents for successful and targeted delivery for prostate cancer.



Publikationsverlauf

Eingereicht: 23. Februar 2021

Angenommen: 24. Mai 2021

Artikel online veröffentlicht:
22. Juni 2021

© 2021. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Golban R. Immunology and immunopathology. Lecture course, Chisinau, uasm. Moodle. md, UASM 2015; 4: 155 p
  • 2 Ferlay J, Colombet M, Soerjomataram I. et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer 2018; 144: 1941-1953
  • 3 Kattumuri V, Katti K, Bhaskaran S. et al. Gum arabic as a phytochemical construct for the stabilization of gold nanoparticles: in vivo pharmacokinetics and X-ray-contrast-imaging studies. Small. 2007; 3: 333-341
  • 4 Kapoor DK, Bansal A, Sharma R. et al. Advanced nanomedicine: Present contributions and future expectations. American J Phytomed Clin Therapeutics 2013; 1: 124-139
  • 5 Shukla R, Nune SK, Chanda N. et al. Soybeans as a phytochemical reservoir for the production and stabilization of biocompatible gold nanoparticles. Small. 2008; 4: 1425-1436
  • 6 Sikora A, Bartczak D, Geißler D. et al. A systematic comparison of different techniques to determine the zeta potential of silica nanoparticles in biological medium. Analytical Methods 2015; 7: 9835-9843
  • 7 Golubnitschaja O, Sridhar KC. Liver metastatic disease: new concepts and biomarker panels to improve individual outcomes. Clinical & Experimental Metastasis 2016; 33: 743-755
  • 8 Golubnitschaja O. Feeling cold and other underestimated symptoms in breast cancer: Anecdotes or individual profiles for advanced patient stratification?. EPMA Journal 2017; 8: 17-22
  • 9 Zubor P, Gondova A, Polivka J. et al. Breast cancer and Flammer syndrome: Any symptoms in common for prediction, prevention and personalised medical approach?. EPMA Journal 2017; 8: 129-140
  • 10 Fröhlich H, Patjoshi S, Yeghiazaryan K. et al. Premenopausal breast cancer: potential clinical utility of a multi-omics based machine learning approach for patient stratification. EPMA Journal 2018; 9: 175-186
  • 11 Zubor P, Kubatka P, Dankova Z. et al. miRNA in a multiomic context for diagnosis, treatment monitoring and personalized management of metastatic breast cancer. Future Oncology 2018; 14: 1847-1867
  • 12 "Prostate Cancer". National Cancer Institute. January 1980. Archived from the original on 12 October 2014. Retrieved 12 October 2014.
  • 13 "Chapter 5.11". World Cancer Report. World Health Organization. 2014. ISBN 978-9283204299
  • 14 "Chapter 1.1". World Cancer Report. World Health Organization. 2014. ISBN 978-9283204299
  • 15 Bray F, Ferlay J, Soerjomataram I. 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: 394-424 DOI: 10.3322/caac.21492.
  • 16 Baade PD, Youlden DR, Krnjacki LJ. International epidemiology of prostate cancer: Geographical distribution and secular trends. Molecular Nutrition & Food Research 2009; 53: 171-184 DOI: 10.1002/mnfr.200700511. PMID 19101947
  • 17 Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin 63: 11-30 2013;
  • 18 "Prostate Cancer Statistics". Laparoscopic Urology. Archived from the originalon 24 June 2016. Retrieved 19 June 2016
  • 19 Overview: Prostate Cancer – What Causes Prostate Cancer? Archived2006-04-04 at the Wayback Machine American Cancer Society (2 May 2006). Retrieved on 5 April 2007
  • 20 Prostate Cancer FAQs. Archived 2006-05-29 at the Wayback MachineState University of New York School of Medicine Department of Urology (31 August 2006). Retrieved on 5 April 200
  • 21 Rendon RA, Mason RJ, Marzouk K. et al. Canadian Urological Association recommendations on prostate cancer screening and early diagnosis. Canadian Urological Association Journal 2017; 11: 298-310 DOI: 10.5489/cuaj.4888. PMC 5659858. PMID 29381452
  • 22 "Prostate cancer statistics". Cancer Research UK. Archived from the original on 6 October 2014. Retrieved 3 October 2014
  • 23 Ferlay JEM, Lam F, Colombet M. et al. Global cancer observatory: cancer tomorrow. Lyon, France: International Agency for Research on Cancer; Available from: https://gco.iarc.fr/tomorrow, Accessed 02 February 2019
  • 24 Huggins C, Hodges CV. Studies on prostatic cancer. I. The effect of castration, of estrogen and androgen injection on serum phosphatases in metastatic carcinoma of the prostate. CA Cancer J Clin 1972; 22: 232-240
  • 25 Kirby M, Hirst C, Crawford ED. Characterising the castration, resistant prostate cancer population: a systematic review. International Journal of Clinical Practice 2011; 65: 1180-1192
  • 26 Siegel R, Ma J, Zou Z. et al. Cancer statistics, 2014. CA Cancer J Clin 2014; 64: 9-29
  • 27 Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J. et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer 2013; 49: 1374-1403
  • 28 Bostwick DG, Burke HB, Djakiew D. et al. Human prostate cancer risk factors. Cancer 2004; 101: 2371-2490
  • 29 Dagnelie PC, Schuurman AG, Goldbohm RA. et al. Diet, anthropometric measures and prostate cancer risk: a review of prospective cohort and intervention studies. BJU Int 2004; 93: 1139-1150
  • 30 Pienta KJ, Esper PS. Risk factors for prostate cancer. Ann Intern Med 1993; 118: 793-803
  • 31 Rybak AP, Bristow RG, Kapoor A. 2015; Prostate cancer stem cells: deciphering the origins and pathways involved in prostate tumorigenesis and aggression. Oncotarget 6 p 1900
  • 32 "Prostate Cancer Treatment (PDQ) – Patient Version". National Cancer Institute. 2014-04-08. Archived from the original on 5 July 2014. Retrieved 1 July 2014.
  • 33 Anand P, Kunnumakkara AB, Newman RA. et al. Bioavailability of curcumin: problems and promises. Molecular Pharmaceutics 03.12.2007; 4: 807-818
  • 34 Sharma RA, Steward WP, Gescher AJ. Pharmacokinetics and pharmacodynamics of curcumin. In The molecular targets and therapeutic uses of curcumin in health and disease. 2007: pp. 453-pp. 470 Springer; Boston, MA:
  • 35 Shaikh J, Ankola DD, Beniwal V. et al. Nanoparticle encapsulation improves oral bioavailability of curcumin by at least 9-fold when compared to curcumin administered with piperine as absorption enhancer. European Journal of Pharmaceutical Sciences 2009; 37: 223-230
  • 36 Moorthi C, Krishnan K, Manavalan R. et al. Preparation and characterization of curcumin–piperine dual drug loaded nanoparticles. Asian Pacific Journal of Tropical Biomedicine 2012; 2: 841-848
  • 37 Yu H, Li J, Shi K. et al. Structure of modified ε-polylysine micelles and their application in improving cellular antioxidant activity of curcuminoids. Food & Function 2011; 2: 373-380
  • 38 Yu H, Huang Q. Improving the oral bioavailability of curcumin using novel organogel-based nanoemulsions. Journal of Agricultural and Food Chemistry 2012; 60: 5373-5379
  • 39 Méndez FB, Medrano FJ, Pariona N. et al. Recent Progress in Antitumoral Nanotechnology. Int J Nanoparticles Nanotech 2015; 1: 2-10
  • 40 Nishiyama N, Kataoka K. Current state, achievements, and future prospects of polymeric micelles as nanocarriers for drug and gene delivery. Pharmacology & Therapeutics 2006; 112: 630-648
  • 41 Torchilin VP. Micellar nanocarriers: pharmaceutical perspectives. Pharmaceutical Research 2007; 24: 1
  • 42 Mulik RS, Mönkkönen J, Juvonen RO. et al. Transferrin mediated solid lipid nanoparticles containing curcumin: enhanced in vitro anticancer activity by induction of apoptosis. International Journal of Pharmaceutics 2010; 398: 190-203
  • 43 Anat EB, Keren M, Joaquin S. et al. Biomater 32 2011; 3862-3874
  • 44 Danhier F, Vroman B, Lecouturier N. et al. Targeting of tumor endothelium by RGD-grafted PLGA-nanoparticles loaded with paclitaxel. Journal of Controlled Release 2009; 140: 166-173
  • 45 Ganta S, Paxton JW, Baguley BC. et al. Pharmacokinetics and pharmacodynamics of chlorambucil delivered in parenteral emulsion. Int J Pharm 2008; 360: 115-121
  • 46 Ganta S, Sharma P, Paxton JW. et al. Pharmacokinetics and pharmacodynamics of chlorambucil delivered in long-circulating nanoemulsion. Journal of Drug Targeting 2010; 18: 125-133
  • 47 Talekar M, Ganta S, Singh A. et al. Phosphatidylinositol 3-kinase inhibitor (PIK75) containing surface functionalized nanoemulsion for enhanced drug delivery, cytotoxicity and pro-apoptotic activity in ovarian cancer cells. Pharm Res 2012; 29: 2874-2886
  • 48 Patlolla RR, Vobalaboina V. Pharmacokinetics and tissue distribution of etoposide delivered in parenteral emulsion. J Pharm Sci 2005; 94: 437-445
  • 49 Tiwari S, Tan YM, Amiji M. Preparation and in vitro characterization of multifunctional nanoemulsions for simultaneous MR imaging and targeted drug delivery. Journal of Biomedical Nanotechnology 2006; 2: 217-224
  • 50 Gianella A, Jarzyna PA, Mani V. et al. Multifunctional nanoemulsion platform for imaging guided therapy evaluated in experimental cancer. ACS Nano 2011; 5: 4422-4433
  • 51 Gupta S, Kesarla R, Omri A. Formulation strategies to improve the bioavailability of poorly absorbed drugs with special emphasis on self-emulsifying systems. ISRN Pharm 2013; 2013: 848043
  • 52 Rahman A, Harwansh R, Mirza A. et al. Oral lipid based drug delivery system: formulation, characterization and application: a review. Curr Drug Deliv 2011; 8: 330-345
  • 53 Jemal A, Bray F, Center MM. et al. Global cancer statistics. CA Cancer J. Clin 2011; 61: 69-90
  • 54 Reya T, Morrison SJ, Clarke MF. et al. Stem cells, cancer, and cancer stem cells. Nat. Cell Boil 2001; 414: 105-111
  • 55 Murali R, Varghese BA, Nair R. et al. The role of cancer stem cells in tumor heterogeneity and resistance to therapy. Can. J. Physiol. Pharmacol. 2017; 95: 1-15
  • 56 Ahmad G, Sadda El, Botchkina R. et al. Formulation of a Novel Taxoid DHA-SBT-1214 Inhibits Prostate Cancer Stem Cell-Induced Tumor Growth. Cancer Lett 2017; 406: 71-80
  • 57 Gillet J-P, Calcagno AM, Varma S. et al. Redefining the relevance of established cancer cell lines to the study of mechanisms of clinical anti-cancer drug resistance. Proc Natl Acad Sci USA 2011; 108: 18708-18713
  • 58 McClements DJ, Rao J. Food-grade nanoemulsions: formulation, fabrication, properties, performance, biological fate, and potential toxicity. Critical Reviews in Food Science and Nutrition 2011; 51: 285-330
  • 59 Salvia-Trujillo L, Rojas-Graü MA, Soliva-Fortuny R. et al. Use of antimicrobial nanoemulsions as edible coatings: Impact on safety and quality attributes of fresh-cut Fuji apples. Postharvest Biology and Technology 2015; 105: 8-16
  • 60 Tadros T, Izquierdo R, Esquena J. et al. 2004; Formation and stability of nano-emulsions. Advances in Colloid and Interface Science 108–109: 303-318
  • 61 Wooster T, Golding M, Sanguansri P. 2008; Impact of oil type on nanoemulsion formation and Ostwald ripening stability. Langmuir 24: 12758-12765
  • 62 McClements DJ. Nanoemulsions versus microemulsions: terminology, differences and similarities. Soft Matter 2012; 8: 1719-1729
  • 63 Ganta S, Talekar M, Singh A. et al. Nanoemulsions in Translational Research—Opportunities and Challenges in Targeted Cancer Therapy. AAPS PharmSciTech 2014; 15: 694-708
  • 64 Gi HJ, Chen SN, Hwang JS. et al. Studies of Formation and Interface of Oil-Water Microemulsion. Chin. J. Phys 1992; 30: 665-678
  • 65 Devarajan V, Ravichandran V. Nanoemulsions: As modified drug delivery tool. Pharm. Glob 2011; 2: 1-6
  • 66 Kale SN, Deore SL. Emulsion micro emulsion and nano emulsion: A review. Syst Rev Pharm 2016; 8: 39-47
  • 67 Danhier F, Feron O, Préat V. To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J Control Release 2010; 148: 135-146
  • 68 Qadir A, Faiyazuddin M, Hussain MT. et al. Critical steps and energetics involved in a successful development of a stable nanoemulsion. J Mol Liq 2016; 214: 7-18
  • 69 Amin N, Das B. A Review on Formulation and Characterization of Nanoemulsion. Int J Curr Pharm Res 2019; 11: 42-46
  • 70 Yuliani S, Noveriza R. Effect of carrier oil and co-solvent on the formation of clove oil nanoemulsion by phase inversion technique. IOP Conf. Ser. Earth Environ Sci. 2019; 309
  • 71 Ganta S, Talekar M, Singh A. et al. Nanoemulsions in Translational Research—Opportunities and Challenges in Targeted Cancer Therapy. AAPS PharmSciTech 2014; 15: 694-708
  • 72 Gupta A, Burak HE, Hatton TA. et al. Nanoemulsions: formation, properties and applications. Soft Matter 2016; 12: 2826e41
  • 73 Bae YH. Drug targeting and tumor heterogeneity. J Control Release 2009; 133: 2-3
  • 74 Peer D, Karp JM, Hong S. et al. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol 2007; 2: 751-760
  • 75 Hedeman M, Brøndsted H, Müllertz A. et al. Fat emulsions based on structured lipids (1, 3-specific triglycerides): an investigation of the in vivo fate. Pharmaceutical Research 01.05.1996; 13: 725-728
  • 76 Haider MF, Khan S, Gaba B. et al. Optimization of rivastigmine nanoemulsion for enhanced brain delivery: in-vivo and toxicity evaluation. J Molecular Liquids 2018; 255: 384-396
  • 77 Tiwari S, Tan Y-M, Amiji M. Preparation and in vitro characterization of multifunctional nanoemulsions for simultaneous MR imaging and targeted drug delivery. J Biomed Nanotechnol 2006; 2: 3-4
  • 78 Gallarate M, Chirio D, Bussano R. et al. Development of O/W nanoemulsions for ophthalmic administration of timolol. Int J Pharm 2013; 440: 126-134 15
  • 79 Nesamony J, Shah IS, Kalra A. et al. Nebulized oil-in-water nanoemulsion mists for pulmonary delivery: development, physico-chemical characterization and in vitro evaluation. Drug Dev Ind Pharm 2013; 40: 1253-1263
  • 80 Khandavilli S, Panchagnula R. Nanoemulsions as versatile formulations for paclitaxel delivery: peroral and dermal delivery studies in rats. J Investig Dermatol 2007; 127: 154-162
  • 81 Jaiswal M, Dudhe R, Sharma PK. Nanoemulsion: An advanced mode of drug delivery system. 3 Biotech 2015; 5: 123-127
  • 82 Ahmad U, Akhtar J, Singh SP. et al. Silmayrinnanoemulsion against human hepatocellular carcinoma: development and optimization. Artificial Cells, Nanomedicine, and Biotechnology 2018; 46: 231-241
  • 83 Chaudhry Q, Scotter M, Blackburn J. et al. Applications and implications of nanotechnologies for the food sector. Food additives and contaminants 2008; 25: 241-258
  • 84 Jiang JG. et al. 2009; Characterization of size, surface ¨ charge, and agglomeration state of nanoparticle dispersions for toxicological studies. Journal of Nanoparticle Research 11: 77-89
  • 85 Dube A, Nicolazzo JA, Larson I. Chitosan nanoparticles enhance the intestinal absorption of the green tea catechins (+)-catechin and (-)-epigallocatechin gallate. Eur J Pharm Sci 2010; 41: 219-225
  • 86 Kale A, Gawande S, Kotwal S. et al. Studies on the effects of oral administration of nutrient mixture, quercetin and red onions on the bioavailability of epigallocatechin gallate from green tea extract. Phytother Res 2010; 24: 48-55
  • 87 Lante A, Friso D. Oxidative stability and rheological properties of nanoemulsion with ultrasonic extracted green tea infusion. Food Res Int 2013; 54: 269-276
  • 88 Kim YJ, Houng SJ, Kim JH. et al. Nanoemulsified green tea extract shows improved hypocholesterolemic effects in C57BL/6 mice. J NutrBiochem 2012; 23: 186-191
  • 89 Tsai YJ, Chen BH. Preparation of catechin extracts and nanoemulsions from green tea leaf waste and their inhibition effect on prostate cancer cell PC-3. International Journal of Nanomedicine 2016; 11: 1907
  • 90 Khan N, Afaq F, Saleem M. et al. Targeting multiple signaling pathways by green tea polyphenol (−)-epigallocatechin-3-gallate. Cancer Res 2006; 66: 2500-2505
  • 91 Sun J, Bi C, Chan HM. et al. Curcumin-loaded solid lipid nanoparticles have prolonged in vitro antitumour activity, cellular uptake and improved in vivo bioavailability. Colloids Surf B Biointerfaces 2013; 111: 367-375
  • 92 Bisht S, Feldmann G, Soni S. et al. Polymeric nanoparticle-encapsulated curcumin (‘‘nanocurcumin’’): a novel strategy for human cancer therapy. J Nanobiotechnol 2007; 5: 3-10
  • 93 Ucisik MH, Küpcü S, Schuster B. et al. Characterization of CurcuEmulsomes: nanoformulation for enhanced solubility and delivery of curcumin. J Nanobiotechnol 2013; 11: 11-37
  • 94 Guan YB, Zhou SY, Zhang YQ. et al. Therapeutic effects of curcumin nanoemulsions on prostate cancer. Journal of Huazhong University of Science and Technology [Medical Sciences] 2017; 37: 371-378
  • 95 Teiten MH, Gaascht F, Eifes S. et al. Chemopreventive potential of curcumin in prostate cancer. Genes & Nutrition 2010; 5: 61-74
  • 96 Chrastina A, Baron VT, Abedinpour P. et al. Plumbagin-loaded nanoemulsion drug delivery formulation and evaluation of antiproliferative effect on prostate cancer cells. BioMed Research International 2018; 11: 2018
  • 97 Panichayupakaranant P, Ahmad MI. Plumbagin and its role in chronic diseases. In Drug Discovery from Mother Nature. 2016: pp. 229-pp. 246 Springer; Cham:
  • 98 Ahmad M, Sahabjada JA, Hussain A. et al. Development of a new rutinnanoemulsion and its application on prostate carcinoma PC3 cell line. Excli Journal 2017; 16: 810
  • 99 Goldstein D, Gofrit O, Nyska A. et al. Anti-HER2 cationic immunoemulsion as a potential targeted drug delivery system for the treatment of prostate cancer. Cancer Research 2007; 67: 269-275
  • 100 Castilla C, Flores ML, Medina R. et al. Prostate cancer cell response to paclitaxel is affected by abnormally expressed securin PTTG1. Molecular Cancer Therapeutics 2014; 13: 2372-2383
  • 101 Cockshott ID, Oliver SD, Young JJ. et al. The effect of food on the pharmacokinetics of the bicalutamide (‘Casodex’) enantiomers. Biopharm. Drug Dispos. 1997; 18: 499-507
  • 102 Fradet Y. Bicalutamide (Casodex) in the treatment of prostate cancer. Expert Rev. Anticancer Ther 2004; 4: 37-48
  • 103 Schellhammer PF, Davis JW. An evaluation of bicalutamide in the treatment of prostate cancer. Clin. Prostate Cancer 2004; 2: 213-219
  • 104 Takegami S, Uchida S, Aoi E. et al. Effect of bovine serum albumin on the stability of bicalutamide-encapsulated lipid nano-emulsion in bovine serum. Current Nanoscience 2012; 8: 187-192
  • 105 Floyd MS, Teahan SJ, Fitzpatrick JM. et al. Differential mechanisms of bicalutamide-induced apoptosis in prostate cell lines. Prostate Cancer and Prostatic Diseases 03/2009; 12: 25-33
  • 106 Ali MS, Alam MS, Alam N. et al. Preparation, characterization and stability study of dutasteride loaded nanoemulsion for treatment of benign prostatic hypertrophy. Iranian Journal of Pharmaceutical Research: IJPR 2014; 13: 1125
  • 107 Schmidt LJ, Murillo H, Tindall DJ. Gene expression in prostate cancer cells treated with the dual 5 alpha-reductase inhibitor dutasteride. Journal of Andrology 2004; 25: 944-953
  • 108 Chourasia Mk, Khan Fa. Docetaxel Nanoemulsion For Effective Chemotherapy of Prostate Cancer (Doctoral dissertation).
  • 109 Yang C, Zhang W, Wang J. et al. Effect of docetaxel on the regulation of proliferation and apoptosis of human prostate cancer cells. Molecular Medicine Reports 2019; 19: 3864-3870
  • 110 Komura K, Jeong SH, Hinohara K. et al. Resistance to docetaxel in prostate cancer is associated with androgen receptor activation and loss of KDM5D expression. Proceedings of the National Academy of Sciences 2016; 113: 6259-6264
  • 111 Alhakamy NA, Fahmy UA, Ahmed OA. Attenuation of Benign Prostatic Hyperplasia by Optimized Tadalafil Loaded Pumpkin Seed Oil-Based Self Nanoemulsion: In Vitro and In Vivo Evaluation. Pharmaceutics. 2019; 11: 640
  • 112 Shukla P, Mathur V, Kumar A. et al. Nanoemulsion based concomitant delivery of curcumin and etoposide: Impact on cross talk between prostate cancer cells and osteoblast during metastasis. Journal of Biomedical Nanotechnology 2014; 10: 3381-3391
  • 113 Hilchie AL, Furlong SJ, Sutton K. et al. Curcumin-induced apoptosis in PC3 prostate carcinoma cells is caspase-independent and involves cellular ceramide accumulation and damage to mitochondria. Nutrition and cancer 2010; 62: 379-389
  • 114 Mukhopadhyay A, Bueso-Ramos C, Chatterjee D. et al. Curcumin downregulates cell survival mechanisms in human prostate cancer cell lines. Oncogene 2001; 20: 7597-7609
  • 115 de Abreu LC, de Souza Furtado P, da Silva Honorio T. et al. A synergistic nanoformulation of babassu and copaiba oils as natural alternative for prevention of benign prostatic hyperplasia. Journal of Drug Delivery Science and Technology 2018; 47: 167-175
  • 116 Guo Y, Liu X, Sun X. et al. Mannosylated lipid nano-emulsions loaded with lycorine-oleic acid ionic complex for tumor cell-specific delivery. Theranostics. 2012; 2: 1104
  • 117 Mohamed FT, Abdelghfour AY, Morsy BA. et al. Promising inhibitor against mitogen-activated protein kinase-dependent inflammation and ameliorates nicotine induced-lung toxicity in rats. Free Radicals and Antioxidants 2020; 10: 35-41
  • 118 Pouton CW. Lipid formulations for oral administration of drugs: non-emulsifying, self-emulsifying and ‘self-microemulsifying’drug delivery systems. European Journal of Pharmaceutical Sciences 2000; 11: S93-S98
  • 119 Kushwaha RS, Chasta P, Ahmed A. et al. A Novel Approach on Thalidomide and their Analogues with their Therapeutic uses and Clinical Application. IJTSRD 2019; 3: 1022-1036
  • 120 Khuroo T, Verma D, Talegaonkar S. et al. Topotecan tamoxifen duple PLGA polymeric nanoparticles: Investigation of in vitro, in vivo and cellular uptake potential. Int J Pharmaceutics 2014; 473: 384-394