Solid Lipid Nanoparticles for the Delivery of Plant-derived Bioactive Compounds in the Treatment of Cancer Disorders – A Review

Jump To References Section

Authors

  • Faculty of Pharmaceutical Science, Assam Down Town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati – 781026, Assam ,IN
  • Department of Pharmacognosy and Phytochemistry, Pratiksha Institute of Pharmaceutical Sciences, Panikhaiti – 781026, Guwahati, Assam ,IN
  • Department of Pharmacology, Vaagdevi Pharmacy College, Bollikunta, Warangal – 506005, Telangana ,IN
  • Department of Pharmaceutics, S. R. R. College of Pharmaceutical Sciences, Valbhapur, Warangal – 505476, Telangana ,IN

DOI:

https://doi.org/10.18311/jnr/2024/36537

Keywords:

Anticancer, Non-small-cell Lung Cancer, Paclitaxel, Phyto-bioactive Compounds, Solid Lipid Nanoparticles

Abstract

Background: The study explores phyto-bioactive compounds in Solid Lipid Nanoparticles (SLN) and their potential use in cancer therapy. Objective: Most phyto-bioactive compounds have properties such as anti-inflammatory agents, antioxidants, anti-microbe agents, anti-arthritic agents, hypoglycemic agents, cardioprotective agents, and anti-cancer agents. Phyto-bioactive compounds are abundant in fruits, vegetables, and whole grains, and may impact metabolic processes improve health, and prevent illness. Breast, colon, and prostate cancers, as well as other malignancies, are now being studied for their potential prevention and treatment using therapies based on phyto-bioactive compounds in in vitro, in vivo, and clinical studies. Methods: More than 10 million individuals have lost their lives to cancer this year alone. Cancer is one of the most malignant and deadly diseases afflicting mankind today, and its death toll continues to rise. SLNs have been extensively used by several research groups to efficiently transport phyto-bioactive substances with enhanced anticancer effects. Now is the time for really groundbreaking ideas and innovations in the field of medicinal nanocarriers. Conclusion: Lipid nanoparticles’ size-dependent properties might lead to new therapeutics. Small size, large surface area, high drug loading, control pattern release, targeted drug release, and interface phase interaction are their benefits. Nanoformulations are undeniably powerful resources for therapeutic delivery applications; the current difficulty is in ensuring their safety, efficacy, and scalability for industrial production and eventual clinical use. Our review offers a novel perspective by focusing on the phyto-bioactive chemicals carried by these SLN nano-carriers, describing recent advancements and their potential applications to cancer therapy.

Downloads

Download data is not yet available.

Metrics

Metrics Loading ...

Downloads

Published

2024-07-31

How to Cite

Samanthula, K. S., Obbalareddy, S., Thatipelli, R. C., & Bairi, A. G. (2024). Solid Lipid Nanoparticles for the Delivery of Plant-derived Bioactive Compounds in the Treatment of Cancer Disorders – A Review. Journal of Natural Remedies, 24(7), 1445–1465. https://doi.org/10.18311/jnr/2024/36537
Received 2024-02-12
Accepted 2024-06-24
Published 2024-07-31

 

References

Witkamp RF, van Norren K. Let thy food be thy medicine when possible. Eur J Pharmacol. 2018; 836:102-14. https://doi.org/10.1016/j.ejphar.2018.06.026

Hamzalıoglu A, Gokmen V. Chapter 18 - Interaction between bioactive carbonyl compounds and asparagine and impact on acrylamide. Acryl Food. 2016:355-76. http://dx.doi.org/10.1016/B978-0-12-802832-2.00018-8

Devkota HP, Paudel KR, Lall N, Tomczyk M, Atanasov AG. Pharmacology of plant polyphenols in human health and diseases. Front Pharmacol. 2022; 13:945033. https://doi.org/10.3389/fphar.2022.945033

Teodoro AJ. Bioactive compounds of food: Their role in the prevention and treatment of diseases. Oxid Med Cell Longev. 2019. https://doi.org/10.1155/2019/3765986

Mondal S, Soumya NP, Mini S, Sivan SK. Bioactive compounds in functional food and their role as therapeutics. Bioact Compd Health Dis. 2021; 4(3):24-39. https://doi.org/10.31989/bchd.v4i3.786

Andonova V, Peneva P. Characterization methods for Solid Lipid Nanoparticles (SLN) and Nanostructured Lipid Carriers (NLC). Curr Pharm Des. 2017; 23(43):6630-42. https://doi.org/10.2174/1381612823666171115105721

Ahmed MF, Swain K, Pattnaik S, Dey BK. Quality by design based development of electrospun Nanofibrous Solid Dispersion mats for oral delivery of Efavirenz. Acta Chim Slov. 2024; 71(1). https://doi.org/10.17344/acsi.2023.8538

Chakraborty S, Dhibar M, Das A, Swain K, Pattnaik S. A critical appraisal of lipid nanoparticles deployed in cancer pharmacotherapy. Recent Adv Drug Deliv Formul. 2023; 17(2):132-51. https://doi.org/10.2174/2667387817666230726140745

Yadav YC, Pattnaik S. Hesperetin-loaded polymeric nanofibers: Assessment of bioavailability and neuroprotective effect. Drug Dev Ind Pharm. 2023; 49(2):240-7. https://doi.org/10.1080/03639045.2023.2201625

Duan Y, Dhar A, Patel C, Khimani M, Neogi S, Sharma P, et al. A brief review on solid lipid nanoparticles: Part and parcel of contemporary drug delivery systems. RSC Adv. 2020; 10(45):26777-91. https://doi.org/10.1039/D0RA03491F

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: Cancer J Clin. 2018; 68(6):394-424. https://doi.org/10.3322/caac.21492

Nguyen TT, Nguyen TT, Tran NM, Van Vo G. Lipid-based nanocarriers via nose-to-brain pathway for central nervous system disorders. Neurochem Res. 2022:1-22. https://doi.org/10.1007/s11064-021-03488-7

Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA: Cancer J Clin. 2023; 73(1):17-48. https://doi.org/10.3322/caac.21763

Lee WL, Huang JY, Shyur LF. Phytoagents for cancer management: regulation of nucleic acid oxidation, ROS, and related mechanisms. Oxid Med Cell Longev. 2013; 2013. https://doi.org/10.1155/2013/925804

Tan RS. Glycosylated and non-glycosylated quantum dot-displayed peptides trafficked indiscriminately inside lung cancer cells but discriminately sorted in normal lung cells: An indispensable part in nanoparticle-based intracellular drug delivery. Asian J Pharm Sci. 2018; 13(3):197-211. https://doi.org/10.1016/j.ajps.2017.12.002

De Santis CE, Lin CC, Mariotto AB, Siegel RL, Stein KD, Kramer JL, et al. Cancer treatment and survivorship statistics, 2014. CA: Cancer J Clin. 2014; 64(4):252-71. https://doi.org/10.3322/caac.21235

Fridlender M, Kapulnik Y, Koltai H. Plant derived substances with anti-cancer activity: From folklore to practice. Front Plant Sci. 2015; 6:799. https://doi.org/10.3389/fpls.2015.00799

Choudhari AS, Mandave PC, Deshpande M, Ranjekar P, Prakash O. Phytochemicals in cancer treatment: From preclinical studies to clinical practice. Front Pharmacol. 2020; 10:1614. https://doi.org/10.3389/fphar.2019.01614

Maheshwari RA, Raghunathan NJ, Maheshwari A, Mehta SP, Balaraman R. Traditional medicine-a gold mine in the treatment of cancer. J Nat Med. 2022; 22(4):540-7. http://doi.org/10.18311/jnr/2022/30377

Wang TY, Li Q, Bi KS. Bioactive flavonoids in medicinal plants: Structure, activity and biological fate. Asian J Pharm Sci. 2018; 13(1):12-23. https://doi.org/10.1016/j.ajps.2017.08.004

Van Agtmael MA, Eggelte TA, Van Boxtel CJ. Artemisinin drugs in the treatment of malaria: from medicinal herb to registered medication. Trends Pharmacol Sci. 1999; 20(5):199-205. https://doi.org/10.1016/S0165-6147(99)01302-4

Lai HC, Singh NP, Sasaki T. Development of artemisinin compounds for cancer treatment. Invest New Drugs. 2013; 31(1):230-46. https://doi.org/10.1007/s10637-012-9873-z

Rogliani P, Calzetta L, Coppola A, Cavalli F, Ora J, Puxeddu E, et al. Optimizing drug delivery in COPD: The role of inhaler devices. Respir Med. 2017; 124:6-14. https://doi.org/10.1016/j.rmed.2017.01.006

Mundy C, Kirkpatrick P. Tiotropium bromide. Nat Rev Drug Discov. 2004; 3(8):643-4. doi: 10.1038/nrd1472 https://doi.org/10.1038/nrd1472

Tembe-Fokunang EA, Charles F, Kaba N, Donatien G, Michael A, Bonaventure N. The potential pharmacological and medicinal properties of neem (Azadirachta indica A. Juss) in the drug development of phytomedicine. J Complement Altern Med Res. 2019; 7(1):1-8. https://doi.org/10.9734/jocamr/2019/v7i130093

Singh S, Pathak N, Fatima E, Negi AS. Plant isoquinoline alkaloids: Advances in the chemistry and biology of berberine. Eur J Med Chem. 2021; 226:113839. https://doi.org/10.1016/j.ejmech.2021.113839

Das AM. Clinical utility of nitisinone for the treatment of hereditary tyrosinemia Type-1 (HT-1). CA: Cancer J Clin. 2017:43-8. http://dx.doi.org/10.2147/TACG.S113310

Pasierski M, Szulczyk B. Beneficial effects of capsaicin in disorders of the central nervous system. Molecules. 2022; 27(8):2484. https://doi.org/10.3390/molecules27082484

Chand J, Panda SR, Jain S, Murty US, Das AM, Kumar GJ, et al. Phytochemistry and polypharmacology of cleome species: A comprehensive ethnopharmacological review of the medicinal plants. J Ethnopharmacol. 2022; 282:114600. https://doi.org/10.1016/j.jep.2021.114600

Hewlings SJ, Kalman DS. Curcumin: A review of its effects on human health. Foods. 2017; 6(10):92. https://doi.org/10.3390/foods6100092

Heinrich M, Teoh HL. Galanthamine from snowdrop — The development of a modern drug against Alzheimer’s disease from local Caucasian knowledge. J Ethnopharmacol. 2004; 92(2-3):147-62. https://doi.org/10.1016/j.jep.2004.02.012

Tuli HS, Tuorkey MJ, Thakral F, Sak K, Kumar M, Sharma AK, et al. Molecular mechanisms of action of genistein in cancer: Recent advances. Front Pharmacol. 2019; 10:1336. https://doi.org/10.3389/fphar.2019.01336

Tsai MJ, Huang YB, Wu PC, Fu YS, Kao YR, Fang JY, et al. Oral apomorphine delivery from solid lipid nanoparticles with different monostearate emulsifiers: Pharmacokinetic and behavioral evaluations. J Pharm Sci. 2011; 100(2):547-57. https://doi.org/10.1002/jps.22285

Deleu D, Hanssens Y, Northway MG. Subcutaneous apomorphine: An evidence-based review of its use in Parkinson’s disease. Drugs Aging. 2004; 21:687-709. https://doi.org/10.2165/00002512-200421110-00001

Chelora J, Zhang J, Wan Y, Cui X, Zhao J, Meng XM, et al. Plant-derived single-molecule-based nanotheranostics for photoenhanced chemotherapy and ferroptotic like cancer cell death. ACS Appl Bio Mater. 2019; 2(6):2643-9. https://doi.org/10.1021/acsabm.9b00311

Ubaidulla U, Sinha P, Sangavi T, Rathnam G. Development of Silymarin entrapped chitosan phthalate nanoparticles for targeting colon cancer. J Nat Remedies; 2022; 22 (4):559-671. http://doi.org/10.18311/jnr/2022/29816

Mamadalieva NZ, Mamedov NA. Taxus brevifolia a highvalue medicinal plant, as a source of taxol. In: Máthé Á, editor. Medicinal and Aromatic Plants of North America. Cham: Springer; 2020. p. 201-18. https://doi.org/10.1007/978-3-030-44930-8_9

Paliwal R, Paliwal SR, Kenwat R, Kurmi BD, Sahu MK. Solid lipid nanoparticles: A review on recent perspectives and patents. Expert Opin Ther Pat. 2020; 30(3):179-94. https://doi.org/10.1080/13543776.2020.1720649

Samanthula KS, Alli R, Gorre T. Preliminary studies on optimization of Anti-Parkinson drug loaded lipid nanoparticles enriched hydrogel formulations for management of Parkinson’s Disease. Curr Nanomed. 2021; 11(2):112-26. https://doi.org/10.2174/2468187311666210311114908

Peng Y, Chen L, Ye S, Kang Y, Liu J, Zeng S, et al. Research and development of drug delivery systems based on drug transporter and nano-formulation. Asian J Pharm Sci. 2020; 15(2):220-36. https://doi.org/10.1016/j.ajps.2020.02.004

Montoto SS, Muraca G, Ruiz ME. Solid lipid nanoparticles for drug delivery: Pharmacological and biopharmaceutical aspects. Front Mol Biosci. 2020; 7:319. https://doi.org/10.3389/fmolb.2020.587997

Akbari-Alavijeh S, Shaddel R, Jafari SM. Encapsulation of food bioactives and nutraceuticals by various chitosan-based nanocarriers. Food Hydrocoll. 2020; 105:105774. https://doi.org/10.1016/j.foodhyd.2020.105774

Awasthi R, Roseblade A, Hansbro PM, Rathbone MJ, Dua K, Bebawy M. Nanoparticles in cancer treatment: Opportunities and obstacles. Curr Drug Targets. 2018; 19(14):1696-709. https://doi.org/10.2174/1389450119666180326122831

Montoto SS, Muraca G, Ruiz ME. Solid lipid nanoparticles for drug delivery: Pharmacological and biopharmaceutical aspects. Front Mol Biosci. 2020; 7:319. https://doi.org/10.3389/fmolb.2020.587997

Tubtimsri S, Limmatvapirat C, Limsirichaikul S, Akkaramongkolporn P, Inoue Y, Limmatvapirat S. Fabrication and characterization of spearmint oil loaded nanoemulsions as cytotoxic agents against oral cancer cell. Asian J Pharm Sci. 2018; 13(5):425-37. https://doi.org/10.1016/j.ajps.2018.02.003

Das A, Edwin AR, Krushnakeerthana R, Bindhu J. Comparative analysis of bioactive compounds and anticancerous activities in leaf and stem extract of Physalis minima. J Nat Remedies. 2020:42-52. https://doi.org/10.18311/jnr/2020/24166

Gordaliza M. Natural products as leads to anticancer drugs. Clin Transl Oncol. 2007; 9:767-76. https://doi.org/10.1007/s12094-007-0138-9

Deng QP, Wang MJ, Zeng X, Chen GG, Huang RY. Effects of glycyrrhizin in a mouse model of lung adenocarcinoma. Cell Physiol Biochem. 2017; 41(4):1383-92. https://doi.org/10.1159/000467897

Wani MC, Taylor HL, Wall ME, Coggon P, McPhail AT. Plant antitumor agents. VI. Isolation and structure of taxol, a novel antileukemic and antitumor agent from Taxus brevifolia. J Am Chem Soc. 1971; 93(9):2325-7. https://doi.org/10.1021/ja00738a045

Gallego-Jara J, Lozano-Terol G, Sola-Martínez RA, Cánovas-Díaz M, de Diego Puente T. A compressive review about Taxol®: History and future challenges. Molecules. 2020; 25(24):5986. https://doi.org/10.3390/molecules25245986

Kumaran RS, Muthumary J, Hur BK. Taxol from Phyllosticta citricarpa, a leaf spot fungus of the angiosperm Citrus medica. J Biosci Bioeng. 2008; 106(1):103-6. https://doi.org/10.1263/jbb.106.103

Kingston DG. Taxol, a molecule for all seasons. Chem Comm. 2001(10):867-80. https://doi.org/10.1039/B100070P

Isah T. Anticancer alkaloids from trees: Development into drugs. Phcog Rev. 2016; 10(20):90. https://doi.org/10.4103/0973-7847.194047

Gowda JI, Nandibewoor ST. Electrochemical behavior of paclitaxel and its determination at glassy carbon electrode. Asian J Pharm Sci. 2014; 9(1):42-9. https://doi.org/10.1016/j.ajps.2013.11.007

Kotsakis A, Matikas A, Koinis F, Kentepozidis N, Varthalitis II, Karavassilis V, et al. A multicentre Phase II trial of cabazitaxel in patients with advanced non-small-cell lung cancer progressing after docetaxel-based chemotherapy. Br J Cancer. 2016; 115(7):784-8. https://doi.org/10.1038/bjc.2016.281

Barbuti AM, Chen ZS. Paclitaxel through the ages of anticancer therapy: Exploring its role in chemoresistance and radiation therapy. Cancers. 2015; 7(4):2360-71. https://doi.org/10.3390/cancers7040897

Oudard S, Fizazi K, Sengeløv L, Daugaard G, Saad F, Hansen S, et al. Cabazitaxel versus docetaxel as first-line therapy for patients with metastatic castration-resistant prostate cancer: A randomized Phase III trial — FIRSTANA. J Clin Oncol. 2017; 35(28):3189-97. https://doi.org/10.1200/JCO.2016.72.1068

Stierle A, Strobel G, Stierle D. Taxol and taxane production by Taxomyces andreanae, an endophytic fungus of Pacific yew. Science. 1993; 260(5105):214-6. https://doi.org/10.1126/science.8097061

Yuan JI, Jian-Nan BI, Bing YA, Xu-Dong Z. Taxol-producing fungi: A new approach to industrial production of taxol. Chin J Biotechnol. 2006; 22(1):1-6. https://doi.org/10.1016/S1872-2075(06)60001-0

Shakya AK, Naik RR. The chemotherapeutic potentials of compounds isolated from the plant, marine, fungus, and microorganism: Their mechanism of action and prospects. J Trop Med. 2022; 2022. https://doi.org/10.1155/2022/5919453

Guenard D, Gueritte-Voegelein F, Potier P. Taxol and taxotere: Discovery, chemistry, and structure-activity relationships. Acc Chem Res. 1993; 26(4):160-7. https://doi.org/10.1021/ar00028a005

Kingston DG. Taxol, a molecule for all seasons. Chem Comm. 2001; (10):867-80. https://doi.org/10.1039/B100070P

Malik S, Cusidó RM, Mirjalili MH, Moyano E, Palazón J, Bonfill M. Production of the anticancer drug taxol in Taxus baccata suspension cultures: A review. Process Biochem. 2011; 46(1):23-34. https://doi.org/10.1016/j.procbio.2010.09.004

Moudi M, Go R, Yien CY, Nazre M. Vinca alkaloids. Int J Prev Med. 2013; 4(11):1231. PMID: 24404355; PMCID: PMC3883245.

Chandraprasad MS, Dey A, Swamy MK. Introduction to cancer and treatment approaches. In Paclitaxel 2022 Jan 1 (pp. 1-27). Academic Press. https://doi.org/10.1016/B978-0-323-90951-8.00010-2

Haque A, Rahman MA, Faizi MS, Khan MS. Next generation antineoplastic agents: A review on structurally modified Vinblastine (VBL) analogues. Curr Med Chem. 2018; 25(14):1650-62. https://doi.org/10.2174/0929867324666170502123639

Stephens DM, Boucher K, Kander E, Parikh SA, Parry EM, Shadman M, et al. Hodgkin lymphoma arising in patients with chronic lymphocytic leukemia: Outcomes from a large multi-center collaboration. Haematologica. 2021; 106(11):2845. https://doi.org/10.3324/haematol.2020.256388

Martino E, Casamassima G, Castiglione S, Cellupica E, Pantalone S, Papagni F, et al. Vinca alkaloids and analogues as anti-cancer agents: Looking back, peering ahead. Bioorg Med Chem Lett. 2018; 28(17):2816-26. https://doi.org/10.1016/j.bmcl.2018.06.044

Uzma F, Mohan CD, Hashem A, Konappa NM, Rangappa S, Kamath PV, et al. Endophytic fungi — Alternative sources of cytotoxic compounds: A review. Front Pharmacol. 2018; 9:309. https://doi.org/10.3389/fphar.2018.00309

Ardalani H, Avan A, Ghayour-Mobarhan M. Podophyllotoxin: A novel potential natural anticancer agent. Avicenna J Phytomed. 2017; 7(4):285. PMID: 28884079; PMCID: PMC5580867.

Cao B, Chen H, Gao Y, Niu C, Zhang Y, Li L. CIP-36, a novel Topoisomerase II-targeting agent, induces the apoptosis of multidrug-resistant cancer cells in vitro. Int J Mol Med. 2015; 35(3):771-6. https://doi.org/10.3892/ijmm.2015.2068

Kumar P, Pal T, Sharma N, Kumar V, Sood H, Chauhan RS. Expression analysis of biosynthetic pathway genes vis-à-vis podophyllotoxin content in Podophyllum hexandrum Royle. Protoplasma. 2015; 252:1253-62. https://doi.org/10.1007/s00709-015-0757-x

You Y. Podophyllotoxin derivatives: Current synthetic approaches for new anticancer agents. Curr Pharm Des. 2005; 11(13):1695-717. https://doi.org/10.2174/1381612053764724

Kour A, Shawl AS, Rehman S, Sultan P, Qazi PH, Suden P, et al. Isolation and identification of an endophytic strain of Fusarium oxysporum producing podophyllotoxin from Juniperus recurva. World J Microbiol Biotechnol. 2008; 24:1115-21. https://doi.org/10.1007/s11274-007-9582-5

Nadeem M, Ram M, Alam P, Ahmad MM, Mohammad A, Al-Qurainy F, et al. Fusarium solani, P1, a new endophytic podophyllotoxin-producing fungus from roots of Podophyllum hexandrum. Afr J Microbiol Res. 2012; 6(10):2493-9. https://doi.org/10.5897/AJMR11.1596

Puri SC, Nazir A, Chawla R, Arora R, Riyaz-ul-Hasan S, Amna T, et al. The endophytic fungus Trametes hirsuta as a novel alternative source of podophyllotoxin and related aryl tetralin lignans. J Biotech. 2006; 122(4):494-510. https://doi.org/10.1016/j.jbiotec.2005.10.015

Huang JX, Zhang J, Zhang XR, Zhang K, Zhang X, He XR. Mucor fragilis as a novel source of the key pharmaceutical agents podophyllotoxin and kaempferol. Pharm Biol. 2014; 52(10):1237-43. https://doi.org/10.3109/13880209.2014.885061

Kusari S, Zühlke S, Spiteller M. Chemometric evaluation of the anti‐cancer pro‐drug podophyllotoxin and potential therapeutic analogues in Juniperus and Podophyllum species. Phytochem Anal. 2011; 22(2):128-43. https://doi.org/10.1002/pca.1258

Hertzberg RP, Caranfa MJ, Hecht SM. On the mechanism of Topoisomerase I inhibition by camptothecin: Evidence for binding to an enzyme-DNA complex. Biochemist. 1989; 28(11):4629-38. https://doi.org/10.1021/bi00437a018

Hsiang YH, Hertzberg R, Hecht S, Liu LF. Camptothecin induces protein-linked DNA breaks via mammalian DNA topoisomerase I. J Biol Chem. 1985; 260(27):14873-8. https://doi.org/10.1016/S0021-9258(17)38654-4

Blagosklonny MV. Analysis of FDA approved anticancer drugs reveals the future of cancer therapy. Cell Cycle. 2004; 3(8):1033-40. https://doi.org/10.4161/cc.3.8.1023

Oberlies NH, Kroll DJ. Camptothecin and taxol: Historic achievements in natural products research. J Nat Prod. 2004; 67(2):129-35. https://doi.org/10.1021/np030498t

Amna T, Puri SC, Verma V, Sharma JP, Khajuria RK, Musarrat J, et al. Bioreactor studies on the endophytic fungus Entrophospora infrequens for the production of an anticancer alkaloid camptothecin. Can J Microbiol. 2006; 52(3):189-96. https://doi.org/10.1139/w05-122

Puri SC, Nazir A, Chawla R, Arora R, Riyaz-ul-Hasan S, Amna T, et al. The endophytic fungus Trametes hirsuta as a novel alternative source of podophyllotoxin and related aryl tetralin lignans. J Biotech. 2006; 122(4):494-510. https://doi.org/10.1016/j.jbiotec.2005.10.015

Xu XY, Meng X, Li S, Gan RY, Li Y, Li HB. Bioactivity, health benefits, and related molecular mechanisms of curcumin: Current progress, challenges, and perspectives. Nutrients. 2018; 10(10):1553. https://doi.org/10.3390/nu10101553

Tomeh MA, Hadianamrei R, Zhao X. A review of curcumin and its derivatives as anticancer agents. Int J Mol Sci. 2019; 20(5):1033. https://doi.org/10.3390/ijms20051033

Agarwal A, Kasinathan A, Ganesan R, Balasubramanian A, Bhaskaran J, Suresh S, et al. Curcumin induces apoptosis and cell cycle arrest via the activation of reactive oxygen species–independent mitochondrial apoptotic pathway in Smad4 and p53 mutated colon adenocarcinoma HT29 cells. Nutr Res. 2018; 51:67-81. https://doi.org/10.1016/j.nutres.2017.12.011

Doello K, Ortiz R, Alvarez PJ, Melguizo C, Cabeza L, Prados J. Latest in vitro and in vivo assay, clinical trials and patents in cancer treatment using curcumin: A literature review. Nutr Cancer. 2018; 70(4):569-78. https://doi.org/10.1080/01635581.2018.1464347

Pastorelli D, Fabricio AS, Giovanis P, D’Ippolito S, Fiduccia P, Soldà C, et al. Phytosome complex of curcumin as complementary therapy of advanced pancreatic cancer improves safety and efficacy of gemcitabine: Results of a prospective Phase II trial. Pharmacol Res. 2018; 132:72-9. https://doi.org/10.1016/j.phrs.2018.03.013

Ismail NI, Othman I, Abas F, Lajis NH, Naidu R. Mechanism of apoptosis induced by curcumin in colorectal cancer. Int J Mol Sci. 2019; 20(10):2454. https://doi.org/10.3390/ijms20102454

Ahmed K, Zaidi SF, Cui ZG, Zhou D, Saeed SA, Inadera H. Potential proapoptotic phytochemical agents for the treatment and prevention of colorectal cancer. Oncology letters. 2019 Jul 1;18(1):487-98. https://doi.org/10.3892/ol.2019.10349

Kunnumakkara AB, Bordoloi D, Harsha C, Banik K, Gupta SC, Aggarwal BB. Curcumin mediates anticancer effects by modulating multiple cell signaling pathways. Clin Sci. 2017 Jul 5;131(15):1781-99. https://doi.org/10.1042/CS20160935

Klippstein R, Bansal SS, Al-Jamal KT. Doxorubicin enhances curcumin’s cytotoxicity in human prostate cancer cells in vitro by enhancing its cellular uptake. Int J Pharm. 2016; 514(1):169-75. https://doi.org/10.1016/j.ijpharm.2016.08.003

Mock CD, Jordan BC, Selvam C. Recent advances of curcumin and its analogues in breast cancer prevention and treatment. RSC Adv. 2015; 5(92):75575-88. https://doi.org/10.1039/C5RA14925H

Kumar G, Mittal S, Sak K, Tuli HS. Molecular mechanisms underlying chemopreventive potential of curcumin: Current challenges and future perspectives. Life Sci. 2016; 148:313-28. https://doi.org/10.1016/j.lfs.2016.02.022

Anand P, Sundaram C, Jhurani S, Kunnumakkara AB, Aggarwal BB. Curcumin and cancer: An "old-age" disease with an "age-old" solution. Cancer Lett. 2008 Aug 18; 267(1):133-64. https://doi.org/10.1016/j.canlet.2008.03.025

Zhou DC, Zittoun R, Marie JP. Homoharringtonine: An effective new natural product in cancer chemotherapy. Bull Cancer. 1995; 82(12):987-95. PMID: 8745664.

Meng H, Yang C, Jin J, Zhou Y, Qian W. Homoharringtonine inhibits the AKT pathway and induces in vitro and in vivo cytotoxicity in human multiple myeloma cells. Leuk Lymphoma. 2008; 49(10):1954-62. https://doi.org/10.1080/10428190802320368

Yakhni M, Briat A, El Guerrab A, Furtado L, Kwiatkowski F, Miot-Noirault E, et al. Homoharringtonine, an approved anti-leukemia drug, suppresses triple negative breast cancer growth through a rapid reduction of anti-apoptotic protein abundance. Am J Cancer Res. 2019; 9(5):1043. PMID: 31218111; PMCID: PMC6556597.

Weng TY, Wu HF, Li CY, Hung YH, Chang YW, Chen YL, et al. Homoharringtonine induced immune alteration for an efficient anti-tumor response in mouse models of non-small cell lung adenocarcinoma expressing Kras mutation. Scient Rep. 2018; 8(1):8216. https://doi.org/10.1038/s41598-018-26454-w

Skroza N, Bernardini N, Proietti I, Potenza C. Clinical utility of ingenol mebutate in the management of actinic keratosis: Perspectives from clinical practice. Ther Clin Risk Manag. 2018:1879-85. https://doi.org/10.2147/TCRM.S145779

Ali FR, Wlodek C, Lear JT. The role of ingenol mebutate in the treatment of actinic keratoses. Dermatol Ther. 2012; 2:1-8. https://doi.org/10.1007/s13555-012-0008-4

Bobyr I, Campanati A, Consales V, Giuliodori K, Scalise A, Offidani A. Efficacy, safety and tolerability of field treatment of actinic keratosis with ingenol mebutate 0.015% gel: A single center case series. SpringerPlus. 2016; 5:1-6. https://doi.org/10.1186/s40064-016-2290-6

Rosen RH, Gupta AK, Tyring SK. Dual mechanism of action of ingenol mebutate gel for topical treatment of actinic keratoses: Rapid lesion necrosis followed by lesion-specific immune response. J Am Acad Dermatol. 2012; 66(3):486-93. https://doi.org/10.1016/j.jaad.2010.12.038

Bernabeu E, Cagel M, Lagomarsino E, Moretton M, Chiappetta DA. Paclitaxel: What has been done and the challenges remain ahead. Int J Pharm. 2017; 526(1-2):474-95. https://doi.org/10.1016/j.ijpharm.2017.05.016

Casado P, Zuazua-Villar P, del Valle E, Martínez-Campa C, Lazo PS, Ramos S. Vincristine regulates the phosphorylation of the antiapoptotic protein HSP27 in breast cancer cells. Cancer Lett. 2007; 247(2):273-82. https://doi.org/10.1016/j.canlet.2006.05.005

Liu J, Geng G, Liang G, Wang L, Luo K, Yuan J, et al. A novel topoisomerase I inhibitor DIA-001 induces DNA damage mediated cell cycle arrest and apoptosis in cancer cell. Ann Transl Med. 2020; 8(4). https://doi.org/10.21037/atm.2019.12.138

Jonsson E, Dhar S, Jonsson B, Nygren P, Graf W, Larsson R. Differential activity of topotecan, irinotecan and SN-38 in fresh human tumour cells but not in cell lines. Eur J Cancer. 2000; 36(16):2120-7. https://doi.org/10.1016/S0959-8049(00)00289-6

Goto K, Ohe Y, Shibata T, Seto T, Takahashi T, Nakagawa K, et al. Combined chemotherapy with cisplatin, etoposide, and irinotecan versus topotecan alone as second-line treatment for patients with sensitive relapsed small-cell lung cancer (JCOG0605): A multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2016; 17(8):1147-57. https://doi.org/10.1016/S1470-2045(16)30104-8

Seca AM, Pinto DC. Plant secondary metabolites as anticancer agents: Successes in clinical trials and therapeutic application. Int J Mol Sci. 2018; 19(1):263. https://doi.org/10.3390/ijms19010263

Wen G, Qu XX, Wang D, Chen XX, Tian XC, Gao F, et al. Recent advances in design, synthesis and bioactivity of paclitaxel-mimics. Fitoterapia. 2016; 110:26-37. https://doi.org/10.1016/j.fitote.2016.02.010

Wang X, Song Y, Su Y, Tian Q, Li B, Quan J, et al. Are PEGylated liposomes better than conventional liposomes? A special case for vincristine. Drug Deliv. 2016; 23(4):1092-100. https://doi.org/10.3109/10717544.2015.1027015

Hertzberg RP, Caranfa MJ, Hecht SM. On the mechanism of topoisomerase I inhibition by camptothecin: Evidence for binding to an enzyme-DNA complex. Biochem. 1989; 28(11):4629-38. https://doi.org/10.1021/bi00437a018

Stohs SJ, Chen O, Ray SD, Ji J, Bucci LR, Preuss HG. Highly bioavailable forms of curcumin and promising avenues for curcumin-based research and application: A review. Molecules. 2020; 25(6):1397. https://doi.org/10.3390/molecules25061397

Chen Y, Lu Y, Lee RJ, Xiang G. Nano encapsulated curcumin: and its potential for biomedical applications. Int J Nanomedicine. 2020:3099-120. https://doi.org/10.2147/IJN.S210320

Kanai M, Otsuka Y, Otsuka K, Sato M, Nishimura T, Mori Y, et al. A phase I study investigating the safety and pharmacokinetics of highly bioavailable curcumin (Theracurmin®) in cancer patients. Cancer Chemother Pharmacol. 2013; 71:1521-30. https://doi.org/10.1007/s00280-013-2151-8

Belcaro G, Hosoi M, Pellegrini L, Appendino G, Ippolito E, Ricci A, et al. A controlled study of a lecithinized delivery system of curcumin (Meriva®) to alleviate the adverse effects of cancer treatment. Phytother Res. 2014; 28(3):444-50. https://doi.org/10.1002/ptr.5014

Lam SS, Ho ES, He BL, Wong WW, Cher CY, Ng NK, et al. Homoharringtonine (omacetaxine mepesuccinate) as an adjunct for FLT3-ITD acute myeloid leukemia. Sci Transl Med. 2016; 8(359):359ra129. https://doi.org/10.1126/scitranslmed.aaf3735

Tzogani K, Nagercoil N, Hemmings RJ, Samir B, Gardette J, Demolis P, et al. The European medicines agency approval of ingenol mebutate (Picato) for the cutaneous treatment of non-hyperkeratotic, non-hypertrophic actinic keratosis in adults: Summary of the scientific assessment of the Committee for Medicinal Products for Human Use (CHMP). Eur J Dermatol. 2014; 24:457-63. https://doi.org/10.1684/ejd.2014.2368

Eng QY, Thanikachalam PV, Ramamurthy S. Molecular understanding of Epigallocatechin Gallate (EGCG) in cardiovascular and metabolic diseases. J Ethnopharmacol. 2018; 210:296-310. https://doi.org/10.1016/j.jep.2017.08.035

Musial C, Kuban-Jankowska A, Gorska-Ponikowska M. Beneficial properties of green tea catechins. Int J Mol Sci. 2020; 21(5):1744. https://doi.org/10.3390/ijms21051744b

Schulze J, Melzer L, Smith L, Teschke R. Green tea and its extracts in cancer prevention and treatment. Beverages. 2017; 3(1):17. https://doi.org/10.3390/beverages3010017

Ho HC, Huang CC, Lu YT, Yeh CM, Ho YT, Yang SF, et al. Epigallocatechin‐3‐gallate inhibits migration of human nasopharyngeal carcinoma cells by repressing MMP‐2 expression. J Cell Physiol. 2019; 234(11):20915-24. https://doi.org/10.1002/jcp.28696

Safwat MA, Kandil BA, Elblbesy MA, Soliman GM, Eleraky NE. Epigallocatechin-3-gallate-loaded gold nanoparticles: Preparation and evaluation of anticancer efficacy in ehrlich tumor-bearing mice. Pharmaceuticals. 2020; 13(9):254. https://doi.org/10.3390/ph13090254

Zhang L, Chen W, Tu G, Chen X, Lu Y, Wu L, et al. Enhanced chemotherapeutic efficacy of PLGA-encapsulated Epigallocatechin Gallate (EGCG) against human lung cancer. Int J Nanomedicine. 2020:4417-29. https://doi.org/10.2147/IJN.S243657

Mukund V, Mukund D, Sharma V, Mannarapu M, Alam A. Genistein: Its role in metabolic diseases and cancer. Crit Rev Oncol Hematol. 2017; 119:13-22. https://doi.org/10.1016/j.critrevonc.2017.09.004

Hsiao YC, Peng SF, Lai KC, Liao CL, Huang YP, Lin CC, et al. Genistein induces apoptosis in vitro and has antitumor activity against human leukemia HL‐60 cancer cell xenograft growth in vivo. Environ Toxicol. 2019; 34(4):443-56. https://doi.org/10.1002/tox.22698

Gu Y, Zhu CF, Iwamoto H, Chen JS. Genistein inhibits invasive potential of human hepatocellular carcinoma by altering cell cycle, apoptosis, and angiogenesis. World J Gastroenterol. 2005; 11(41):6512. https://doi.org/10.3748/wjg.v11.i41.6512

Eckelbarger JD, Wilmot JT, Epperson MT, Thakur CS, Shum D, Antczak C, et al. Synthesis of antiproliferative Cephalotaxus esters and their evaluation against several human hematopoietic and solid tumor cell lines: uncovering differential susceptibilities to multidrug resistance. Chem. 2008; 14(14):4293-306. https://doi.org/10.1002/chem.200701998

Kantarjian HM, Talpaz M, Santini V, Murgo A, Cheson B, O’Brien SM. Homoharringtonine: History, current research, and future directions. Cancer. 2001; 92(6):1591-605. https://doi.org/10.1002/1097-0142(20010915)92:6<1591::AID-CNCR1485>3.0.CO;2-U

Li J, Gao J, Liu A, Liu W, Xiong H, Liang C, et al. Homoharringtonine - Based Induction regimen improved the remission rate and survival rate in chinese childhood AML: A report from the CCLG-AML 2015 protocol study. J Clin Oncol. 2023:JCO-22. https://creativecommons.org/licenses/by-nc-nd/4.0/

Wei W, Huang S, Ling Q, Mao S, Qian Y, Ye W, et al. Homoharringtonine is synergistically lethal with BCL-2 inhibitor APG-2575 in acute myeloid leukemia. J Transl Med. 2022; 20(1):299. https://doi.org/10.1186/s12967-022-03497-2

Weng TY, Wu HF, Li CY, Hung YH, Chang YW, Chen YL, et al. Homoharringtonine induced immune alteration for an efficient anti-tumor response in mouse models of non-small cell lung adenocarcinoma expressing Kras mutation. Sci Rep. 2018; 8(1):8216. https://doi.org/10.1038/s41598-018-26454-w

Varshney S, Shankar K, Beg M, Balaramnavar VM, Mishra SK, Jagdale P, et al. Rohitukine inhibits in vitro adipogenesis arresting mitotic clonal expansion and improves dyslipidemia in vivo [S]. J Lipid Res. 2014; 55(6):1019-32. https://doi.org/10.1194/jlr.M039925

Kumara PM, Soujanya KN, Ravikanth G, Vasudeva R, Ganeshaiah KN, Shaanker RU. Rohitukine, a chromone alkaloid and a precursor of flavopiridol, is produced by endophytic fungi isolated from Dysoxylum binectariferum Hook. f and Amoora rohituka (Roxb). Wight and Arn. Phytomedicine. 2014; 21(4):541-6. https://doi.org/10.1016/j.phymed.2013.09.019

Karatoprak GŞ, Akkol EK, Genç Y, Bardakcı H, Yücel Ç, Sobarzo-Sánchez E. Combretastatins: An overview of structure, probable mechanisms of action and potential applications. Molecules. 2020; 25(11):2560. https://doi.org/10.3390/molecules25112560

Tozer GM, Kanthou C, Parkins CS, Hill SA. The biology of the combretastatins as tumour vascular targeting agents. Int J Exp Pathol. 2002; 83(1):21-38. https://doi.org/10.1046/j.1365-2613.2002.00211.x

Marrelli M, Conforti F, Statti GA, Cachet X, Michel S, Tillequin F, et al. Biological potential and structure-activity relationships of most recently developed vascular disrupting agents: An overview of new derivatives of natural combretastatin A-4. Current medicinal chemistry. 2011; 18(20):3035-81. https://doi.org/10.2174/092986711796391642

Kupchan SM, Yokoyama N. The structure, configuration and synthesis of thalicarpine, a novel dimeric aporphine-benzylisoquinoline alkaloid. J Am Chem Soc. 1963; 85(9):1361-2. https://doi.org/10.1021/ja00892a041

Xu W, Huang Z, Ji X, Lumb JP. Catalytic aerobic cross-dehydrogenative coupling of phenols and catechols. ACS Catalysis. 2019; 9(5):3800-10. https://doi.org/10.1021/acscatal.8b04443

Allen LM, Creaven PJ. Binding of a new antitumor agent, thalicarpine, to DNA. J Pharm Sci. 1974; 63(3):474-5. https://doi.org/10.1002/jps.2600630343

Frédérich M, Bentires-Alj M, Tits M, Angenot L, Greimers R, Gielen J, et al. Isostrychnopentamine, an indolomonoterpenic alkaloid from Strychnos usambarensis, induces cell cycle arrest and apoptosis in human colon cancer cells. J Pharmacol Exp Ther. 2003; 304(3):1103-10. https://doi.org/10.1124/jpet.102.044867

Nistor I, Cao M, Debrus B, Lebrun P, Lecomte F, Rozet E, et al. Application of a new optimization strategy for the separation of tertiary alkaloids extracted from Strychnos usambarensis leaves. J Pharm Biomed Anal. 2011; 56(1):30-7. https://doi.org/10.1016/j.jpba.2011.04.027

Liu W, Wang M, Guo Z, He Y, Jia H, He J, et al. Inspired by bis-β-carboline alkaloids: Construction and antitumor evaluation of a novel bis-β-carboline scaffold as potent antitumor agents. Bioorg Chem. 2023; 133:106401. https://doi.org/10.1016/j.bioorg.2023.106401

Belmont P, Bosson J, Godet T, Tiano M. Acridine and acridone derivatives, anticancer properties and synthetic methods: Where are we now?. Anticancer Agents Med Chem. 2007; 7(2):139-69. https://doi.org/10.2174/187152007780058669

Fujiwara M, Okamoto M, Okamoto M, Watanabe M, Machida H, Shigeta S, et al. Acridone derivatives are selective inhibitors of HIV-1 replication in chronically infected cells. Antivir Res. 1999; 43(3):189-99. https://doi.org/10.1016/S0166-3542(99)00045-5

Sepúlveda CS, Fascio ML, García CC, D’Accorso NB, Damonte EB. Acridones as antiviral agents: Synthesis, chemical and biological properties. Curr Med Chem. 2013; 20(19):2402-14. https://doi.org/10.1002/chin.201343237

Dalton LK, Demerac S, Elmes BC, Loder JW, Swan JM, Teitei T. Synthesis of the tumour-inhibitory alkaloids, ellipticine, 9-methoxyellipticine, and related pyrido [4, 3-b] carbazoles. Aust J Chem. 1967; 20(12):2715-27. https://doi.org/10.1071/CH9672715

Andrews WJ, Panova T, Normand C, Gadal O, Tikhonova IG, Panov KI. Old drug, new target: ellipticines selectively inhibit RNA polymerase I transcription. J Biol Chem. 2013; 288(7):4567-82. https://doi.org/10.1074/jbc.M112.411611

Krishna PM, Knv R, Banji D. A review on phytochemical, ethnomedical and pharmacological studies on genus Sophora, Fabaceae. Rev Bras Farmacogn. 2012; 22:1145-54. https://doi.org/10.1590/S0102-695X2012005000043

Luo C, Zhong HJ, Zhu LM, Wu XG, Ying JE, Wang XH, et al. Inhibition of matrine against gastric cancer cell line MNK45 growth and its anti-tumor mechanism. Mol Biol Rep. 2012; 39:5459-64. https://doi.org/10.1007/s11033-011-1346-5

Li D, Wang G, Jin G, Yao K, Zhao Z, Bie L, et al. Resveratrol suppresses colon cancer growth by targeting the AKT/STAT3 signaling pathway. Int J Mol Med. 2019; 43(1):630-40. https://doi.org/10.3892/ijmm.2018.3969

Paller CJ, Zhou XC, Heath EI, Taplin ME, Mayer T, Stein MN, et al. Muscadine grape skin extract (MPX) in men with biochemically recurrent prostate cancer: A randomized, multicenter, placebo-controlled clinical trial. Clin Cancer Res. 2018; 24(2):306-15. https://doi.org/10.1158/1078-0432.CCR-17-1100

Ganesan P, Ramalingam P, Karthivashan G, Ko YT, Choi DK. Recent developments in solid lipid nanoparticle and surface-modified solid lipid nanoparticle delivery systems for oral delivery of phyto-bioactive compounds in various chronic diseases. Int J Nanomedicine. 2018:1569-83. http://dx.doi.org/10.2147/IJN.S155593

Aditya NP, Ko S. Solid Lipid Nanoparticles (SLNs): Delivery vehicles for food bioactives. RSC Adv. 2015; 5(39):30902-11. https://doi.org/10.1039/C4RA17127F

Weber S, Zimmer A, Pardeike J. Solid Lipid Nanoparticles (SLN) and Nanostructured Lipid Carriers (NLC) for pulmonary application: A review of the state of the art. Eur J Pharm Biopharm. 2014; 86(1):7-22. https://doi.org/10.1016/j.ejpb.2013.08.013

Khan I, Saeed K, Khan I. Nanoparticles: Properties, applications and toxicities. Arab J Chem. 2019; 12(7):908-31. https://doi.org/10.1016/j.arabjc.2017.05.011

Baig N, Kammakakam I, Falath W. Nanomaterials: A review of synthesis methods, properties, recent progress, and challenges. Mater Adv. 2021; 2(6):1821-71. https://doi.org/10.1039/D0MA00807A

Duan H, Wang T, Su Z, Pang H, Chen C. Recent progress and challenges in plasmonic nanomaterials. Nanotechnol Rev. 2022; 11(1):846-73. https://doi.org/10.1515/ntrev-2022-0039

Ruiz ME, Montoto SS. Routes of drug administration. ADME Processes Pharm Sci. Routes of drug administration; 2018. p. 97-133. https://doi.org/10.1007/978-3-319-99593-9_6

Tan ME, He CH, Jiang W, Zeng C, Yu N, Huang W, et al. Development of solid lipid nanoparticles containing total flavonoid extract from Dracocephalum moldavica L. and their therapeutic effect against myocardial ischemia–reperfusion injury in rats. Int J Nanomedicine. 2017:3253-65. https://doi.org/10.2147/IJN.S131893

Nunes S, Madureira AR, Campos D, Sarmento B, Gomes AM, Pintado M, et al. Solid lipid nanoparticles as oral delivery systems of phenolic compounds: Overcoming pharmacokinetic limitations for nutraceutical applications. Crit Rev Food Sci Nutr. 2017; 57(9):1863-73. https://doi.org/10.1080/10408398.2015.1031337