Curcumin-Based Nanoformulations: A Promising Adjuvant towards Cancer Treatment
Abstract
:1. Introduction
2. Curcumin as Hydrophobic Phytochemical in Medicine
2.1. Curcumin Structure
2.2. Molecular Targets of Curcumin
3. Advances in Curcumin Formulations
3.1. Synthesis of Curcumin Nanomaterials
3.2. Nano-Based Formulation Strategies of Curcumin
3.2.1. Micelle Structures
3.2.2. Liposome Structures
3.2.3. Cyclodextrin Structures
3.2.4. Conjugate Structures
3.2.5. Nano- and Nanosphere Structures
3.3. Anti-Cancer Activity of Curcumin
4. Curcumin and Nanocurcumins as Drugs: Comparable Properties and Effectiveness
5. Cancer Immunotherapy Using Nanocurcumin Formulations
6. Anti-Angiogenic Activity of (Nano)Curcumin for Cancer Treatment
7. Effects of Curcumin and Nanocurcumin toward Bacteria and Viruses Associated with Cancer
8. Clinical Trials of Curcumin Nanoformulations
9. Various Patents on Using Curcumin Nanoformulations against Cancer
10. Conclusions and Future Considerations
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
References
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Arruebo, M.; Vilaboa, N.; Sáez-Gutierrez, B.; Lambea, J.; Tres, A.; Valladares, M.; González-Fernández, Á. Assessment of the evolution of cancer treatment therapies. Cancers 2011, 3, 3279–3330. [Google Scholar] [CrossRef] [PubMed]
- D’Alterio, C.; Scala, S.; Sozzi, G.; Roz, L.; Bertolini, G. Paradoxical effects of chemotherapy on tumor relapse and metastasis promotion. Semin. Cancer Biol. 2020, 60, 351–361. [Google Scholar] [CrossRef] [PubMed]
- Greenwell, M.; Rahman, P.K.S.M. Medicinal Plants: Their Use in Anticancer Treatment. Int. J. Pharm. Sci. Res. 2015, 6, 4103–4112. [Google Scholar] [CrossRef] [PubMed]
- Khalifa, S.A.; Elias, N.; Farag, M.A.; Chen, L.; Saeed, A.; Hegazy, M.E.F.; Moustafa, M.S.; Abd El-Wahed, A.; Al-Mousawi, S.M.; Musharraf, S.G.; et al. Marine natural products: A source of novel anticancer drugs. Mar. Drugs 2019, 17, 491. [Google Scholar] [CrossRef]
- Sarker, S.D.; Nahar, L.; Miron, A.; Guo, M. Chapter 2—Anticancer natural products. In Medicinal Natural Products: A Disease-Focused Approach; Sarker, S.D., Nahar, L., Eds.; Academic Press: Cambridge, MA, USA, 2020; Volume 55, pp. 45–75. [Google Scholar] [CrossRef]
- Cristiano, M.C.; Froiio, F.; Spaccapelo, R.; Mancuso, A.; Nisticò, S.P.; Udongo, B.P.; Fresta, M.; Paolino, D. Sulforaphane-loaded ultradeformable vesicles as a potential natural nanomedicine for the treatment of skin cancer diseases. Pharmaceutics 2019, 12, 6. [Google Scholar] [CrossRef]
- Wolf, C.P.; Rachow, T.; Ernst, T.; Hochhaus, A.; Zomorodbakhsch, B.; Foller, S.; Rengsberger, M.; Hartmann, M.; Hübner, J. Interactions in cancer treatment considering cancer therapy, concomitant medications, food, herbal medicine and other supplements. J. Cancer Res. Clin. Oncol. 2022, 148, 461–473. [Google Scholar] [CrossRef]
- Khodavirdipour, A.; Zarean, R.; Safaralizadeh, R. Evaluation of the anti-cancer effect of Syzygium cumini ethanolic extract on HT-29 colorectal cell line. J. Gastrointest. Cancer 2020, 52, 575–581. [Google Scholar] [CrossRef]
- Ahmad, A.; Sakr, W.A.; Rahman, K.M. Novel targets for detection of cancer and their modulation by chemopreventive natural compounds. Front. Biosci. 2012, 4, 410–425. [Google Scholar] [CrossRef]
- Tagde, P.; Tagde, P.; Islam, F.; Tagde, S.; Shah, M.; Hussain, Z.D.; Rahman, M.H.; Najda, A.; Alanazi, I.S.; Germoush, M.O.; et al. The multifaceted role of curcumin in advanced nanocurcumin form in the treatment and management of chronic disorders. Molecules 2021, 26, 7109. [Google Scholar] [CrossRef]
- Mansouri, K.; Rasoulpoor, S.; Daneshkhah, A.; Abolfathi, S.; Salari, N.; Mohammadi, M.; Rasoulpoor, S.; Shabani, S. Clinical effects of curcumin in enhancing cancer therapy: A systematic review. BMC Cancer 2020, 20, 791. [Google Scholar] [CrossRef] [PubMed]
- Bachmeier, B.E.; Killian, P.H.; Melchart, D. The role of curcumin in prevention and management of metastatic disease. Int. J. Mol. Sci. 2018, 19, 1716. [Google Scholar] [CrossRef] [PubMed]
- Islam, M.R.; Islam, F.; Nafady, M.H.; Akter, M.; Mitra, S.; Das, R.; Urmee, H.; Shohag, S.; Akter, A.; Chidambaram, K.; et al. Natural small molecules in breast cancer treatment: Understandings from a therapeutic viewpoint. Molecules 2022, 27, 2165. [Google Scholar] [CrossRef] [PubMed]
- Chenthamara, D.; Subramaniam, S.; Ramakrishnan, S.G.; Krishnaswamy, S.; Essa, M.M.; Lin, F.-H.; Qoronfleh, M.W. Therapeutic efficacy of nanoparticles and routes of administration. Biomater. Res. 2019, 23, 20. [Google Scholar] [CrossRef]
- Chauhan, I.; Yasir, M.; Verma, M.; Singh, A.P. Nanostructured lipid carriers: A Groundbreaking approach for transdermal drug delivery. Adv. Pharm. Bull. 2020, 10, 150–165. [Google Scholar] [CrossRef]
- Yallapu, M.M.; Jaggi, M.; Chauhan, S.C. β-Cyclodextrin-curcumin self-assembly enhances curcumin delivery in prostate cancer cells. Colloids Surf. B Biointerfaces 2010, 79, 113–125. [Google Scholar] [CrossRef]
- Deljoo, S.; Rabiee, N.; Rabiee, M. Curcumin-hybrid nanoparticles in drug delivery system. Asian J. Nanosci. Mater. 2019, 2, 66–91. [Google Scholar] [CrossRef]
- Bagheri, M.; Fens, M.H.; Kleijn, T.G.; Capomaccio, R.B.; Mehn, D.; Krawczyk, P.M.; Scutigliani, E.M.; Gurinov, A.; Baldus, M.; van Kronenburg, N.C. In vitro and in vivo studies on HPMA-based polymeric micelles loaded with curcumin. Mol. Pharm. 2021, 18, 1247–1263. [Google Scholar] [CrossRef]
- Available online: http://www.ncbi.nlm.nih.gov/sites/entrez (accessed on 6 March 2020).
- Shaikh, J.; Ankola, D.; Beniwal, V.; Singh, D.; Kumar, M.R. Nanoparticle encapsulation improves oral bioavailability of curcumin by at least 9-fold when compared to curcumin administered with piperine as absorption enhancer. Eur. J. Pharm. Sci. 2009, 37, 223–230. [Google Scholar] [CrossRef]
- Jazayeri-Tehrani, S.A.; Rezayat, S.M.; Mansouri, S.; Qorbani, M.; Alavian, S.M.; Daneshi-Maskooni, M.; Hosseinzadeh-Attar, M.-J. Nano-curcumin improves glucose indices, lipids, inflammation, and Nesfatin in overweight and obese patients with non-alcoholic fatty liver disease (NAFLD): A double-blind randomized placebo-controlled clinical trial. Nutr. Metab. 2019, 16, 8. [Google Scholar] [CrossRef]
- Wang, Y.; Luo, J.; Li, S.-Y. Nano-curcumin simultaneously protects the blood–brain barrier and reduces M1 microglial activation during cerebral ischemia–reperfusion injury. ACS Appl. Mater. Interfaces 2019, 11, 3763–3770. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Liu, Q.; Liu, Y.; Ju, F.; Ma, Q.; He, Q. In vivo evaluation of enhanced drug carrier efficiency and cardiac anti-hypertrophy therapeutic potential of nano-curcumin encapsulated photo-plasmonic nanoparticles combined polymerized nano-vesicles: A novel strategy. J. Photochem. Photobiol. B Biol. 2019, 199, 111619. [Google Scholar] [CrossRef] [PubMed]
- Wu, B.; Yao, H.; Wang, S.; Xu, R. DAPK1 modulates a curcumin-induced G2/M arrest and apoptosis by regulating STAT3, NF-κB, and caspase-3 activation. Biochem. Biophys. Res. Commun. 2013, 434, 75–80. [Google Scholar] [CrossRef] [PubMed]
- Nelson, K.M.; Dahlin, J.L.; Bisson, J.; Graham, J.; Pauli, G.F.; Walters, M.A. The essential medicinal chemistry of curcumin: Miniperspective. J. Med. Chem. 2017, 60, 1620–1637. [Google Scholar] [CrossRef] [PubMed]
- Abd Wahab, N.A.; Lajis, N.H.; Abas, F.; Othman, I.; Naidu, R. Mechanism of anti-cancer activity of curcumin on androgen-dependent and androgen-independent prostate cancer. Nutrients 2020, 12, 679. [Google Scholar] [CrossRef]
- Sharifi-Rad, J.; Rayess, Y.E.; Rizk, A.A.; Sadaka, C.; Zgheib, R.; Zam, W.; Sestito, S.; Rapposelli, S.; Neffe-Skocińska, K.; Zielińska, D.; et al. Turmeric and its major compound curcumin on health: Bioactive effects and safety profiles for food, pharmaceutical, biotechnological and medicinal applications. Front. Pharmacol. 2020, 11, 01021. [Google Scholar] [CrossRef]
- Manolova, Y.; Deneva, V.; Antonov, L.; Drakalska, E.; Momekova, D.; Lambov, N. The effect of the water on the curcumin tautomerism: A quantitative approach. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2014, 132, 815–820. [Google Scholar] [CrossRef]
- Tønnesen, H.H.; Másson, M.; Loftsson, T. Studies of curcumin and curcuminoids. XXVII. Cyclodextrin complexation: Solubility, chemical and photochemical stability. Int. J. Pharm. 2002, 244, 127–135. [Google Scholar] [CrossRef]
- Priyadarsini, K.I. The chemistry of curcumin: From extraction to therapeutic agent. Molecules 2014, 19, 20091–20112. [Google Scholar] [CrossRef]
- Amalraj, A.; Pius, A.; Gopi, S.; Gopi, S. Biological activities of curcuminoids, other biomolecules from turmeric and their derivatives—A review. J. Tradit. Complement. Med. 2017, 7, 205–233. [Google Scholar] [CrossRef]
- Somparn, P.; Phisalaphong, C.; Nakornchai, S.; Unchern, S.; Morales, N.P. Comparative antioxidant activities of curcumin and its demethoxy and hydrogenated derivatives. Biol. Pharm. Bull. 2007, 30, 74–78. [Google Scholar] [CrossRef] [PubMed]
- Rajasekaran, S.A. Therapeutic potential of curcumin in gastrointestinal diseases. World J. Gastrointest. Pathophysiol. 2011, 2, 1–14. [Google Scholar] [CrossRef]
- Jeong, S.-O.; Oh, G.-S.; Ha, H.-Y.; Koo, B.S.; Kim, H.S.; Kim, Y.-C.; Kim, E.-C.; Lee, K.-M.; Chung, H.-T.; Pae, H.-O. Dimethoxycurcumin, a synthetic curcumin analogue, induces heme oxygenase-1 expression through Nrf2 activation in RAW264. 7 macrophages. J. Clin. Biochem. Nutr. 2009, 44, 79–84. [Google Scholar] [CrossRef] [PubMed]
- Gupta, S.C.; Patchva, S.; Aggarwal, B.B. Therapeutic roles of curcumin: Lessons learned from clinical trials. AAPS J. 2013, 15, 195–218. [Google Scholar] [CrossRef] [PubMed]
- Anand, P.; Kunnumakkara, A.B.; Newman, R.A.; Aggarwal, B.B. Bioavailability of curcumin: Problems and promises. Mol. Pharm. 2007, 4, 807–818. [Google Scholar] [CrossRef] [PubMed]
- Huang, Q.; Yu, H.; Ru, Q. Bioavailability and delivery of nutraceuticals using nanotechnology. J. Food Sci. 2010, 75, R50–R57. [Google Scholar] [CrossRef]
- Liu, W.; Zhai, Y.; Heng, X.; Che, F.Y.; Chen, W.; Sun, D.; Zhai, G. Oral bioavailability of curcumin: Problems and advancements. J. Drug Target. 2016, 24, 694–702. [Google Scholar] [CrossRef]
- Metzler, M.; Pfeiffer, E.; Schulz, S.I.; Dempe, J.S. Curcumin uptake and metabolism. Biofactors 2013, 39, 14–20. [Google Scholar] [CrossRef]
- Vareed, S.K.; Kakarala, M.; Ruffin, M.T.; Crowell, J.A.; Normolle, D.P.; Djuric, Z.; Brenner, D.E. Pharmacokinetics of curcumin conjugate metabolites in healthy human subjects. Cancer Epidemiol. Biomark. Prev. 2008, 17, 1411–1417. [Google Scholar] [CrossRef]
- El-Magboub, A.; Rojsitthisak, P.; Muangnoi, C.; Wichitnithad, W.; Romero, R.M.; Haworth, I.S. Biological targets and pharmacology of curcumin. In Curcumin: Synthesis, Emerging Role in Pain Management and Health Implications; Nova Science: New York, NY, USA, 2014; pp. 103–134. [Google Scholar]
- Aggarwal, B.B.; Sung, B. Pharmacological basis for the role of curcumin in chronic diseases: An age-old spice with modern targets. Trends Pharmacol. Sci. 2009, 30, 85–94. [Google Scholar] [CrossRef]
- Gupta, S.C.; Prasad, S.; Aggarwal, B.B. Anti-Inflammatory Nutraceuticals and Chronic Diseases; Springer: New York, NY, USA, 2016; Volume 928. [Google Scholar]
- Fu, H.; Wang, C.; Yang, D.; Wei, Z.; Xu, J.; Hu, Z.; Zhang, Y.; Wang, W.; Yan, R.; Cai, Q. Curcumin regulates proliferation, autophagy, and apoptosis in gastric cancer cells by affecting PI3K and P53 signaling. J. Cell. Physiol. 2018, 233, 4634–4642. [Google Scholar] [CrossRef] [PubMed]
- Rafiq, S.; Raza, M.H.; Younas, M.; Naeem, F.; Adeeb, R.; Iqbal, J.; Anwar, P.; Sajid, U.; Manzoor, H.M. Molecular targets of curcumin and future therapeutic role in leukemia. J. Biosci. Med. 2018, 6, 33. [Google Scholar] [CrossRef]
- Shanmugam, M.K.; Rane, G.; Kanchi, M.M.; Arfuso, F.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; Tan, B.K.; Kumar, A.P.; Sethi, G. The multifaceted role of curcumin in cancer prevention and treatment. Molecules 2015, 20, 2728–2769. [Google Scholar] [CrossRef] [PubMed]
- Patel, S.S.; Acharya, A.; Ray, R.S.; Agrawal, R.; Raghuwanshi, R.; Jain, P. Cellular and molecular mechanisms of curcumin in prevention and treatment of disease. Crit. Rev. Food Sci. Nutr. 2020, 60, 887–939. [Google Scholar] [CrossRef]
- Zendehdel, E.; Abdollahi, E.; Momtazi-Borojeni, A.A.; Korani, M.; Alavizadeh, S.H.; Sahebkar, A. The molecular mechanisms of curcumin’s inhibitory effects on cancer stem cells. J. Cell. Biochem. 2019, 120, 4739–4747. [Google Scholar] [CrossRef]
- Shehzad, A.; Wahid, F.; Lee, Y.S. Curcumin in cancer chemoprevention: Molecular targets, pharmacokinetics, bioavailability, and clinical trials. Arch. Pharm. 2010, 343, 489–499. [Google Scholar] [CrossRef] [PubMed]
- Bimonte, S.; Barbieri, A.; Palma, G.; Rea, D.; Luciano, A.; D’Aiuto, M.; Arra, C.; Izzo, F. Dissecting the role of curcumin in tumour growth and angiogenesis in mouse model of human breast cancer. BioMed Res. Int. 2015, 2015, 878134. [Google Scholar] [CrossRef]
- Wan Mohd Tajuddin, W.N.B.; Lajis, N.H.; Abas, F.; Othman, I.; Naidu, R. Mechanistic understanding of curcumin’s therapeutic effects in lung cancer. Nutrients 2019, 11, 2989. [Google Scholar] [CrossRef]
- Yang, C.L.; Liu, Y.Y.; Ma, Y.G.; Xue, Y.X.; Liu, D.G.; Ren, Y.; Liu, X.B.; Li, Y.; Li, Z. Curcumin blocks small cell lung cancer cells migration, invasion, angiogenesis, cell cycle and neoplasia through Janus kinase-STAT3 signalling pathway. PLoS ONE 2012, 7, e37960. [Google Scholar] [CrossRef]
- Liu, H.; Zhou, B.H.; Qiu, X.; Wang, H.S.; Zhang, F.; Fang, R.; Wang, X.F.; Cai, S.H.; Du, J.; Bu, X.Z. T63, a new 4-arylidene curcumin analogue, induces cell cycle arrest and apoptosis through activation of the reactive oxygen species–FOXO3a pathway in lung cancer cells. Free Radic. Biol. Med. 2012, 53, 2204–2217. [Google Scholar] [CrossRef]
- Wu, L.; Guo, L.; Liang, Y.; Liu, X.; Jiang, L.; Wang, L. Curcumin suppresses stem-like traits of lung cancer cells via inhibiting the JAK2/STAT3 signaling pathway. Oncol. Rep. 2015, 34, 3311–3317. [Google Scholar] [CrossRef] [PubMed]
- Yao, Q.; Lin, M.; Wang, Y.; Lai, Y.; Hu, J.; Fu, T.; Wang, L.; Lin, S.; Chen, L.; Guo, Y. Curcumin induces the apoptosis of A549 cells via oxidative stress and MAPK signaling pathways. Int. J. Mol. Med. 2015, 36, 1118–1126. [Google Scholar] [CrossRef]
- Qu, F.; Cao, P. Long noncoding RNA SOX2OT contributes to gastric cancer progression by sponging miR-194-5p from AKT2. Exp. Cell Res. 2018, 369, 187–196. [Google Scholar] [CrossRef]
- Li, S.; Zhang, L.; Li, S.; Zhao, H.; Chen, Y. Curcumin suppresses the progression of gastric cancer by regulating circ_0056618/miR-194-5p axis. Open Life Sci. 2021, 16, 937–949. [Google Scholar] [CrossRef] [PubMed]
- Rajalakshmi, N.; Dhivya, S. A Review on the preparation methods of curcumin nanoparticles. PharmaTutor 2018, 6, 6–10. [Google Scholar] [CrossRef]
- Rai, M.; Pandit, R.; Gaikwad, S.; Yadav, A.; Gade, A. Potential applications of curcumin and curcumin nanoparticles: From traditional therapeutics to modern nanomedicine. Nanotechnol. Rev. 2015, 4, 161–172. [Google Scholar] [CrossRef]
- Bodhankar, M.M.; Chikhle, S. Various approaches towards enhancement of bioavailability of curcumin—A potent phytochemical. World J. Pharm. Res. 2018, 8, 606–626. [Google Scholar]
- Panzarini, E.; Mariano, S.; Tacconi, S.; Carata, E.; Tata, A.M.; Dini, L. Novel therapeutic delivery of nanocurcumin in central nervous system related disorders. Nanomaterials 2021, 11, 2. [Google Scholar] [CrossRef]
- Sacco, P.; Pedroso-Santana, S.; Kumar, Y.; Joly, N.; Martin, P.; Bocchetta, P. Ionotropic gelation of chitosan flat structures and potential applications. Molecules 2021, 26, 660. [Google Scholar] [CrossRef]
- Giri, T.K.; Thakur, D.; Alexander, A.; Badwaik, H.; Tripathy, M.; Tripathi, D.K. Biodegradable IPN hydrogel beads of pectin and grafted alginate for controlled delivery of diclofenac sodium. J. Mater. Sci. Mater. Med. 2013, 24, 1179–1190. [Google Scholar] [CrossRef]
- Sorasitthiyanukarn, F.; Muangnoi, C.; Ratnatilaka Na Bhuket, P.; Rojsitthisak, P.; Rojsitthisak, P. Chitosan/alginate nanoparticles as a promising approach for oral delivery of curcumin diglutaric acid for cancer treatment. Mater. Sci. Eng. C 2018, 93, 178–190. [Google Scholar] [CrossRef] [PubMed]
- Akbar, M.U.; Zia, K.M.; Malik, M.I.; Zahid, M.; Khera, R.A.; Khaliq, Z. Chapter 10-Curcumin-based bionanocomposites. In Bionanocomposites; Zia, K.M., Jabeen, F., Anjum, M.N., Ikram, S., Eds.; Elsevier: Amsterdam, The Netherlands, 2020; pp. 233–257. [Google Scholar]
- Bhunchu, S.; Muangnoi, C.; Rojsitthisak, P. Curcumin diethyl disuccinate encapsulated in chitosan/alginate nanoparticles for improvement of its in vitro cytotoxicity against MDA-MB-231 human breast cancer cells. Die Pharm. 2016, 71, 691–700. [Google Scholar] [CrossRef]
- Das, R.K.; Kasoju, N.; Bora, U. Encapsulation of curcumin in alginate-chitosan-pluronic composite nanoparticles for delivery to cancer cells. Nanomed. Nanotechnol. Biol. Med. 2010, 6, 153–160. [Google Scholar] [CrossRef] [PubMed]
- Akhtar, F.; Rizvi, M.M.A.; Kar, S.K. Oral delivery of curcumin bound to chitosan nanoparticles cured Plasmodium yoelii infected mice. Biotechnol. Adv. 2012, 30, 310–320. [Google Scholar] [CrossRef] [PubMed]
- Yadav, D.; Kumar, N. Nanonization of curcumin by antisolvent precipitation: Process development, characterization, freeze drying and stability performance. Int. J. Pharm. 2014, 477, 564–577. [Google Scholar] [CrossRef] [PubMed]
- Sadeghi, F.; Ashofteh, M.; Homayouni, A.; Abbaspour, M.; Nokhodchi, A.; Afrasiabi garekani, H. Antisolvent precipitation technique: A very promising approach to crystallize curcumin in presence of polyvinyl pyrrolidon for solubility and dissolution enhancement. Colloids Surf. B Biointerfaces 2016, 147, 258–264. [Google Scholar] [CrossRef]
- Kakran, M.; Sahoo, N.G.; Tan, I.-L.; Li, L. Preparation of nanoparticles of poorly water-soluble antioxidant curcumin by antisolvent precipitation methods. J. Nanoparticle Res. 2012, 14, 757. [Google Scholar] [CrossRef]
- Gessner, A.; Waicz, R.; Lieske, A.; Paulke, B.-R.; Mäder, K.; Müller, R. Nanoparticles with decreasing surface hydrophobicities: Influence on plasma protein adsorption. Int. J. Pharm. 2000, 196, 245–249. [Google Scholar] [CrossRef]
- Jones, M.-C.; Jones, S.A.; Riffo-Vasquez, Y.; Spina, D.; Hoffman, E.; Morgan, A.; Patel, A.; Page, C.; Forbes, B.; Dailey, L.A. Quantitative assessment of nanoparticle surface hydrophobicity and its influence on pulmonary biocompatibility. J. Control. Release 2014, 183, 94–104. [Google Scholar] [CrossRef]
- Hanafy, N.A.N.; El-Kemary, M.; Leporatti, S. Micelles structure development as a strategy to improve smart cancer therapy. Cancers 2018, 10, 238. [Google Scholar] [CrossRef]
- Smit, B. Molecular-dynamics simulations of amphiphilic molecules at a liquid-liquid interface. Phys. Rev. A 1988, 37, 3431. [Google Scholar] [CrossRef] [PubMed]
- Liu, L.; Sun, L.; Wu, Q.; Guo, W.; Li, L.; Chen, Y.; Li, Y.; Gong, C.; Qian, Z.; Wei, Y. Curcumin loaded polymeric micelles inhibit breast tumor growth and spontaneous pulmonary metastasis. Int. J. Pharm. 2013, 443, 175–182. [Google Scholar] [CrossRef] [PubMed]
- Gong, C.; Deng, S.; Wu, Q.; Xiang, M.; Wei, X.; Li, L.; Gao, X.; Wang, B.; Sun, L.; Chen, Y. Improving antiangiogenesis and anti-tumor activity of curcumin by biodegradable polymeric micelles. Biomaterials 2013, 34, 1413–1432. [Google Scholar] [CrossRef] [PubMed]
- Chang, T.; Trench, D.; Putnam, J.; Stenzel, M.H.; Lord, M.S. Curcumin-loading-dependent stability of PEGMEMA-based micelles affects endocytosis and exocytosis in colon carcinoma cells. Mol. Pharm. 2016, 13, 924–932. [Google Scholar] [CrossRef]
- Chen, S.; Li, Q.; Li, H.; Yang, L.; Yi, J.-Z.; Xie, M.; Zhang, L.-M. Long-circulating zein-polysulfobetaine conjugate-based nanocarriers for enhancing the stability and pharmacokinetics of curcumin. Mater. Sci. Eng. C 2020, 109, 110636. [Google Scholar] [CrossRef]
- Yang, X.; Li, Z.; Wang, N.; Li, L.; Song, L.; He, T.; Sun, L.; Wang, Z.; Wu, Q.; Luo, N.; et al. Curcumin-encapsulated polymeric micelles suppress the development of colon cancer in vitro and in vivo. Sci. Rep. 2015, 5, 10322. [Google Scholar] [CrossRef]
- Akbarzadeh, A.; Rezaei-Sadabady, R.; Davaran, S.; Joo, S.W.; Zarghami, N.; Hanifehpour, Y.; Samiei, M.; Kouhi, M.; Nejati-Koshki, K. Liposome: Classification, preparation, and applications. Nanoscale Res. Lett. 2013, 8, 102. [Google Scholar] [CrossRef]
- Bozzuto, G.; Molinari, A. Liposomes as nanomedical devices. Int. J. Nanomed. 2015, 10, 975–999. [Google Scholar] [CrossRef]
- Tiwari, G.; Tiwari, R.; Sriwastawa, B.; Bhati, L.; Pandey, S.; Pandey, P.; Bannerjee, S.K. Drug delivery systems: An updated review. Int. J. Pharm. Investig. 2012, 2, 2–11. [Google Scholar] [CrossRef]
- Çağdaş, M.; Sezer, A.D.; Bucak, S. Chapter 1—Liposomes as potential drug carrier systems for drug delivery. In Application of Nanotechnology in Drug Delivery; Sezer, A.D., Ed.; InTechOpen: London, UK, 2014; pp. 1–50. [Google Scholar]
- He, H.; Lu, Y.; Qi, J.; Zhu, Q.; Chen, Z.; Wu, W. Adapting liposomes for oral drug delivery. Acta Pharm. Sin. B 2019, 9, 36–48. [Google Scholar] [CrossRef]
- Chang, M.; Wu, M.; Li, H. Antitumor activities of novel glycyrrhetinic acid-modified curcumin-loaded cationic liposomes in vitro and in H22 tumor-bearing mice. Drug Deliv. 2018, 25, 1984–1995. [Google Scholar] [CrossRef] [PubMed]
- Zheng, B.; McClements, D.J. Formulation of more efficacious curcumin delivery systems using colloid science: Enhanced solubility, stability, and bioavailability. Molecules 2020, 25, 2791. [Google Scholar] [CrossRef] [PubMed]
- Yingchoncharoen, P.; Kalinowski, D.S.; Richardson, D.R. Lipid-based drug delivery systems in cancer therapy: What is available and what is yet to come. Pharmacol. Rev. 2016, 68, 701–787. [Google Scholar] [CrossRef] [PubMed]
- Immordino, M.L.; Dosio, F.; Cattel, L. Stealth liposomes: Review of the basic science, rationale, and clinical applications, existing and potential. Int. J. Nanomed. 2006, 1, 297–315. [Google Scholar]
- Dhule, S.S.; Penfornis, P.; Frazier, T.; Walker, R.; Feldman, J.; Tan, G.; He, J.; Alb, A.; John, V.; Pochampally, R. Curcumin-loaded γ-cyclodextrin liposomal nanoparticles as delivery vehicles for osteosarcoma. Nanomed. Nanotechnol. Biol. Med. 2012, 8, 440–451. [Google Scholar] [CrossRef]
- Tian, Y. Inhibitory effect of curcumin liposome on PC-3 human prostate cancer cells. Chin. J. Exp. Surg. 2014, 31, 1075–1078. [Google Scholar] [CrossRef]
- Tefas, L.R.; Sylvester, B.; Tomuta, I.; Sesarman, A.; Licarete, E.; Banciu, M.; Porfire, A. Development of antiproliferative long-circulating liposomes co-encapsulating doxorubicin and curcumin, through the use of a quality-by-design approach. Drug Des. Dev. Ther. 2017, 11, 1605–1621. [Google Scholar] [CrossRef]
- Huang, M.; Liang, C.; Tan, C.; Huang, S.; Ying, R.; Wang, Y.; Wang, Z.; Zhang, Y. Liposome co-encapsulation as a strategy for the delivery of curcumin and resveratrol. Food Funct. 2019, 10, 6447–6458. [Google Scholar] [CrossRef]
- Vetha, B.S.S.; Kim, E.-M.; Oh, P.-S.; Kim, S.H.; Lim, S.T.; Sohn, M.-H.; Jeong, H.-J. Curcumin encapsulated micellar nanoplatform for blue light emitting diode induced apoptosis as a new class of cancer therapy. Macromol. Res. 2019, 27, 1179–1184. [Google Scholar] [CrossRef]
- Chen, Y.; Wu, Q.; Zhang, Z.; Yuan, L.; Liu, X.; Zhou, L. Preparation of curcumin-loaded liposomes and evaluation of their skin permeation and pharmacodynamics. Molecules 2012, 17, 5972–5987. [Google Scholar] [CrossRef]
- Ndong Ntoutoume, G.M.A.; Granet, R.; Mbakidi, J.P.; Brégier, F.; Léger, D.Y.; Fidanzi-Dugas, C.; Lequart, V.; Joly, N.; Liagre, B.; Chaleix, V.; et al. Development of curcumin–cyclodextrin/cellulose nanocrystals complexes: New anticancer drug delivery systems. Bioorg. Med. Chem. Lett. 2016, 26, 941–945. [Google Scholar] [CrossRef] [PubMed]
- Guo, S. Encapsulation of curcumin into β-cyclodextrins inclusion: A review. E3S Web Conf. 2019, 131, 01100. [Google Scholar] [CrossRef]
- Zhang, L.; Man, S.; Qiu, H.; Liu, Z.; Zhang, M.; Ma, L.; Gao, W. Curcumin-cyclodextrin complexes enhanced the anti-cancer effects of curcumin. Environ. Toxicol. Pharmacol. 2016, 48, 31–38. [Google Scholar] [CrossRef] [PubMed]
- Abruzzo, A.; Zuccheri, G.; Belluti, F.; Provenzano, S.; Verardi, L.; Bigucci, F.; Cerchiara, T.; Luppi, B.; Calonghi, N. Chitosan nanoparticles for lipophilic anticancer drug delivery: Development, characterization and in vitro studies on HT29 cancer cells. Colloids Surf. B Biointerfaces 2016, 145, 362–372. [Google Scholar] [CrossRef]
- Lee, Y.-S.; Tarté, R.; Acevedo, N.C. Curcumin encapsulation in Pickering emulsions co-stabilized by starch nanoparticles and chitin nanofibers. RSC Adv. 2021, 11, 16275–16284. [Google Scholar] [CrossRef]
- Zhang, H.; Jiang, L.; Tong, M.; Lu, Y.; Ouyang, X.-K.; Ling, J. Encapsulation of curcumin using fucoidan stabilized zein nanoparticles: Preparation, characterization, and in vitro release performance. J. Mol. Liq. 2021, 329, 115586. [Google Scholar] [CrossRef]
- Maria, D.N.; Mishra, S.R.; Wang, L.; Abd-Elgawad, A.H.; Soliman, O.A.; El-Dahan, M.S.; Jablonski, M.M. Water-soluble complex of curcumin with cyclodextrins: Enhanced physical properties for ocular drug delivery. Curr. Drug Deliv. 2017, 14, 875–886. [Google Scholar] [CrossRef]
- Manju, S.; Sreenivasan, K. Conjugation of curcumin onto hyaluronic acid enhances its aqueous solubility and stability. J. Colloid Interface Sci. 2011, 359, 318–325. [Google Scholar] [CrossRef]
- Singh, D.V.; Agarwal, S.; Singh, P.; Godbole, M.M.; Misra, K. Curcumin conjugates induce apoptosis via a mitochondrion dependent pathway in MCF-7 and MDA-MB-231 cell lines. Asian Pac. J. Cancer Prev. 2013, 14, 5797–5804. [Google Scholar] [CrossRef]
- Muangnoi, C.; Jithavech, P.; Ratnatilaka Na Bhuket, P.; Supasena, W.; Wichitnithad, W.; Towiwat, P.; Niwattisaiwong, N.; Haworth, I.S.; Rojsitthisak, P. A curcumin-diglutaric acid conjugated prodrug with improved water solubility and antinociceptive properties compared to curcumin. Biosci. Biotechnol. Biochem. 2018, 82, 1301–1308. [Google Scholar] [CrossRef]
- Asha, A.B.; Narain, R. Chapter 15-Nanomaterials properties. In Polymer Science and Nanotechnology; Narain, R., Ed.; Elsevier: Amsterdam, The Netherlands, 2020; pp. 343–359. [Google Scholar]
- Jeevanandam, J.; Barhoum, A.; Chan, Y.S.; Dufresne, A.; Danquah, M.K. Review on nanoparticles and nanostructured materials: History, sources, toxicity and regulations. Beilstein J. Nanotechnol. 2018, 9, 1050–1074. [Google Scholar] [CrossRef] [PubMed]
- Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.D.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol. 2018, 16, 71. [Google Scholar] [CrossRef] [PubMed]
- Senapati, S.; Mahanta, A.K.; Kumar, S.; Maiti, P. Controlled drug delivery vehicles for cancer treatment and their performance. Signal Transduct. Target. Ther. 2018, 3, 7. [Google Scholar] [CrossRef] [PubMed]
- Gupta, S.; Kumar, P. Drug delivery using nanocarriers: Indian perspective. Proc. Natl. Acad. Sci. India Sect. B Biol. Sci. 2012, 82, 167–206. [Google Scholar] [CrossRef]
- Chen, Y.; Lu, Y.; Lee, R.J.; Xiang, G. Nano encapsulated curcumin: And its potential for biomedical applications. Int. J. Nanomed. 2020, 15, 3099–3120. [Google Scholar] [CrossRef]
- Kabir, M.; Rahman, M.; Akter, R.; Behl, T.; Kaushik, D.; Mittal, V.; Pandey, P.; Akhtar, M.F.; Saleem, A.; Albadrani, G.M. Potential role of curcumin and its nanoformulations to treat various types of cancers. Biomolecules 2021, 11, 392. [Google Scholar] [CrossRef]
- Sun, J.; Zhao, Y.; Hu, J. Curcumin inhibits imiquimod-induced psoriasis-like inflammation by inhibiting IL-1beta and IL-6 production in mice. PLoS ONE 2013, 8, e67078. [Google Scholar] [CrossRef]
- Bhatt, H.; Rompicharla, S.V.; Komanduri, N.; Aashma, S.; Paradkar, S.; Ghosh, B.; Biswas, S. Development of curcumin-loaded solid lipid nanoparticles utilizing glyceryl monostearate as single lipid using QbD approach: Characterization and evaluation of anticancer activity against human breast cancer cell line. Curr. Drug Deliv. 2018, 15, 1271–1283. [Google Scholar] [CrossRef]
- Abd-Ellatef, F.G.-E.; Gazzano, E.; Chirio, D.; Hamed, A.R.; Belisario, D.C.; Zuddas, C.; Peira, E.; Rolando, B.; Kopecka, J.; Assem Said Marie, M.; et al. Curcumin-loaded solid lipid nanoparticles bypass P-glycoprotein mediated doxorubicin resistance in triple negative breast cancer cells. Pharmaceutics 2020, 12, 96. [Google Scholar] [CrossRef]
- Shome, S.; Talukdar, A.D.; Choudhury, M.D.; Bhattacharya, M.K.; Upadhyaya, H. Curcumin as potential therapeutic natural product: A nanobiotechnological perspective. J. Pharm. Pharmacol. 2016, 68, 1481–1500. [Google Scholar] [CrossRef]
- Ferrari, R.; Sponchioni, M.; Morbidelli, M.; Moscatelli, D. Polymer nanoparticles for the intravenous delivery of anticancer drugs: The checkpoints on the road from the synthesis to clinical translation. Nanoscale 2018, 10, 22701–22719. [Google Scholar] [CrossRef] [PubMed]
- Chang, P.-Y.; Peng, S.-F.; Lee, C.-Y.; Lu, C.-C.; Tsai, S.-C.; Shieh, T.-M.; Wu, T.-S.; Tu, M.-G.; Chen, M.Y.; Yang, J.-S. Curcumin-loaded nanoparticles induce apoptotic cell death through regulation of the function of MDR1 and reactive oxygen species in cisplatin-resistant CAR human oral cancer cells. Int. J. Oncol. 2013, 43, 1141–1150. [Google Scholar] [CrossRef] [PubMed]
- Yallapu, M.; Gupta, B.; Jaggi, M.; Chauhan, S. Fabrication of curcumin encapsulated PLGA nanoparticles for improved therapeutic effects in metastatic cancer cells. J. Colloid Interface Sci. 2010, 351, 19–29. [Google Scholar] [CrossRef] [PubMed]
- Xie, M.; Fan, D.; Li, Y.; He, X.; Chen, X.; Chen, Y.; Zhu, J.; Xu, G.; Wu, X.; Lan, P. Supercritical carbon dioxide-developed silk fibroin nanoplatform for smart colon cancer therapy. Int. J. Nanomed. 2017, 12, 7751. [Google Scholar] [CrossRef]
- Chaurasia, S.; Chaubey, P.; Patel, R.R.; Kumar, N.; Mishra, B. Curcumin-polymeric nanoparticles against colon-26 tumor-bearing mice: Cytotoxicity, pharmacokinetic and anticancer efficacy studies. Drug Dev. Ind. Pharm. 2016, 42, 694–700. [Google Scholar] [CrossRef] [PubMed]
- Montalbán, M.G.; Coburn, J.M.; Lozano-Pérez, A.A.; Cenis, J.L.; Víllora, G.; Kaplan, D.L. Production of curcumin-loaded silk fibroin nanoparticles for cancer therapy. Nanomaterials 2018, 8, 126. [Google Scholar] [CrossRef]
- Kim, T.H.; Jiang, H.H.; Youn, Y.S.; Park, C.W.; Tak, K.K.; Lee, S.; Kim, H.; Jon, S.; Chen, X.; Lee, K.C. Preparation and characterization of water-soluble albumin-bound curcumin nanoparticles with improved antitumor activity. Int. J. Pharm. 2011, 403, 285–291. [Google Scholar] [CrossRef] [PubMed]
- Thadakapally, R.; Aafreen, A.; Aukunuru, J.; Habibuddin, M.; Jogala, S. Preparation and characterization of PEG-albumin-curcumin nanoparticles intended to treat breast cancer. Indian J. Pharm. Sci. 2016, 78, 65. [Google Scholar] [CrossRef]
- Dreaden, E.C.; Alkilany, A.M.; Huang, X.; Murphy, C.J.; El-Sayed, M.A. The golden age: Gold nanoparticles for biomedicine. Chem. Soc. Rev. 2012, 41, 2740–2779. [Google Scholar] [CrossRef]
- Yaqoob, S.B.; Adnan, R.; Rameez Khan, R.M.; Rashid, M. Gold, silver, and palladium nanoparticles: A chemical tool for biomedical applications. Front. Chem. 2020, 8, 376. [Google Scholar] [CrossRef]
- Nambiar, S.; Osei, E.; Fleck, A.; Darko, J.; Mutsaers, A.J.; Wettig, S. Synthesis of curcumin-functionalized gold nanoparticles and cytotoxicity studies in human prostate cancer cell line. Appl. Nanosci. 2018, 8, 347. [Google Scholar] [CrossRef]
- Liu, R.; Pei, Q.; Shou, T.; Zhang, W.; Hu, J.; Li, W. Apoptotic effect of green synthesized gold nanoparticles from Curcuma wenyujin extract against human renal cell carcinoma A498 cells. Int. J. Nanomed. 2019, 14, 4091–4103. [Google Scholar] [CrossRef] [PubMed]
- Elbialy, N.S.; Abdelfatah, E.A.; Khalil, W.A. Antitumor activity of curcumin-green synthesized gold nanoparticles: In vitro study. BioNanoScience 2019, 9, 813–820. [Google Scholar] [CrossRef]
- Beyene, A.M.; Moniruzzaman, M.; Karthikeyan, A.; Min, T. Curcumin nanoformulations with metal oxide nanomaterials for biomedical applications. Nanomaterials 2021, 11, 460. [Google Scholar] [CrossRef]
- Nikolova, M.P.; Chavali, M.S. Metal oxide nanoparticles as biomedical materials. Biomimetics 2020, 5, 27. [Google Scholar] [CrossRef]
- Yallapu, M.M.; Othman, S.F.; Curtis, E.T.; Bauer, N.A.; Chauhan, N.; Kumar, D.; Jaggi, M.; Chauhan, S.C. Curcumin-loaded magnetic nanoparticles for breast cancer therapeutics and imaging applications. Int. J. Nanomed. 2012, 7, 1761. [Google Scholar] [CrossRef]
- Saikia, C.; Das, M.K.; Ramteke, A.; Maji, T.K. Controlled release of curcumin from thiolated starch-coated iron oxide magnetic nanoparticles: An in vitro evaluation. Int. J. Polym. Mater. Polym. Biomater. 2017, 66, 349–358. [Google Scholar] [CrossRef]
- Aeineh, N.; Salehi, F.; Akrami, M.; Nemati, F.; Alipour, M.; Ghorbani, M.; Nikfar, B.; Salehian, F.; Riyahi Alam, N.; Sadat Ebrahimi, S.E. Glutathione conjugated polyethylenimine on the surface of Fe3O4 magnetic nanoparticles as a theranostic agent for targeted and controlled curcumin delivery. J. Biomater. Sci. 2018, 29, 1109–1125. [Google Scholar] [CrossRef]
- Moballegh Nasery, M.; Abadi, B.; Poormoghadam, D.; Zarrabi, A.; Keyhanvar, P.; Khanbabaei, H.; Ashrafizadeh, M.; Mohammadinejad, R.; Tavakol, S.; Sethi, G. Curcumin delivery mediated by bio-based nanoparticles: A review. Molecules 2020, 25, 689. [Google Scholar] [CrossRef]
- Hosu, O.; Tertis, M.; Cristea, C. Implication of magnetic nanoparticles in cancer detection, screening and treatment. Magnetochemistry 2019, 5, 55. [Google Scholar] [CrossRef]
- Mauri, E.; Giannitelli, S.M.; Trombetta, M.; Rainer, A. Synthesis of nanogels: Current trends and future outlook. Gels 2021, 7, 36. [Google Scholar] [CrossRef] [PubMed]
- Sabir, F.; Asad, M.I.; Qindeel, M.; Afzal, I.; Dar, M.J.; Shah, K.U.; Zeb, A.; Khan, G.M.; Ahmed, N.; Din, F.-U. Polymeric nanogels as versatile nanoplatforms for biomedical applications. J. Nanomater. 2019, 2019, 1526186. [Google Scholar] [CrossRef]
- Paramera, E.I.; Konteles, S.J.; Karathanos, V.T. Microencapsulation of curcumin in cells of Saccharomyces cerevisiae. Food Chem. 2011, 125, 892–902. [Google Scholar] [CrossRef]
- Paramera, E.I.; Konteles, S.J.; Karathanos, V.T. Stability and release properties of curcumin encapsulated in Saccharomyces cerevisiae, β-cyclodextrin and modified starch. Food Chem. 2011, 125, 913–922. [Google Scholar] [CrossRef]
- Singh, A.T.; Ghosh, M.; Forte, T.M.; Ryan, R.O.; Gordon, L.I. Curcumin nanodisk-induced apoptosis in mantle cell lymphoma. Leuk. Lymphoma 2011, 52, 1537–1543. [Google Scholar] [CrossRef]
- Ghosh, M.; Ryan, R.O. ApoE enhances nanodisk-mediated curcumin delivery to glioblastoma multiforme cells. Nanomedicine 2014, 9, 763–771. [Google Scholar] [CrossRef]
- Dandekar, P.P.; Jain, R.; Patil, S.; Dhumal, R.; Tiwari, D.; Sharma, S.; Vanage, G.; Patravale, V. Curcumin-loaded hydrogel nanoparticles: Application in anti-malarial therapy and toxicological evaluation. J. Pharm. Sci. 2010, 99, 4992–5010. [Google Scholar] [CrossRef]
- Amanlou, N.; Parsa, M.; Rostamizadeh, K.; Sadighian, S.; Moghaddam, F. Enhanced cytotoxic activity of curcumin on cancer cell lines by incorporating into gold/chitosan nanogels. Mater. Chem. Phys. 2019, 226, 151–157. [Google Scholar] [CrossRef]
- Priya, P.; Raj, R.M.; Vasanthakumar, V.; Raj, V. Curcumin-loaded layer-by-layer folic acid and casein coated carboxymethyl cellulose/casein nanogels for treatment of skin cancer. Arab. J. Chem. 2020, 13, 694–708. [Google Scholar] [CrossRef]
- Li, Y.; Gu, Z.; Zhang, C.; Li, S.; Zhang, L.; Zhou, G.; Wang, S.; Zhang, J. Synthesis, characterization and ROS-mediated antitumor effects of palladium (II) complexes of curcuminoids. Eur. J. Med. Chem. 2018, 144, 662–671. [Google Scholar] [CrossRef]
- Koohpar, Z.K.; Entezari, M.; Movafagh, A.; Hashemi, M. Anticancer activity of curcumin on human breast adenocarcinoma: Role of Mcl-1 gene. Iran. J. Cancer Prev. 2015, 8, e2331. [Google Scholar] [CrossRef] [PubMed]
- Hu, S.; Xu, Y.; Meng, L.; Huang, L.; Sun, H. Curcumin inhibits proliferation and promotes apoptosis of breast cancer cells. Exp. Ther. Med. 2018, 16, 1266–1272. [Google Scholar] [CrossRef] [PubMed]
- Koroth, J.; Nirgude, S.; Tiwari, S.; Gopalakrishnan, V.; Mahadeva, R.; Kumar, S.; Karki, S.S.; Choudhary, B. Investigation of anti-cancer and migrastatic properties of novel curcumin derivatives on breast and ovarian cancer cell lines. BMC Complement. Altern. Med. 2019, 19, 273. [Google Scholar] [CrossRef]
- Sharma, R.A.; Euden, S.A.; Platton, S.L.; Cooke, D.N.; Shafayat, A.; Hewitt, H.R.; Marczylo, T.H.; Morgan, B.; Hemingway, D.; Plummer, S.M. Phase I clinical trial of oral curcumin: Biomarkers of systemic activity and compliance. Clin. Cancer Res. 2004, 10, 6847–6854. [Google Scholar] [CrossRef] [PubMed]
- Dei Cas, M.; Ghidoni, R. Dietary curcumin: Correlation between bioavailability and health potential. Nutrients 2019, 11, 2147. [Google Scholar] [CrossRef] [PubMed]
- Desai, K. Curcumin Cyclodextrin Combination for Preventing or Treating Various Diseases. U.S. Patent No. US20100179103A1, 15 July 2010. [Google Scholar]
- Jäger, R.; Lowery, R.P.; Calvanese, A.V.; Joy, J.M.; Purpura, M.; Wilson, J.M. Comparative absorption of curcumin formulations. Nutr. J. 2014, 13, 11. [Google Scholar] [CrossRef] [PubMed]
- Karimpour, M.; Feizi, M.A.H.; Mahdavi, M.; Krammer, B.; Verwanger, T.; Najafi, F.; Babaei, E. Development of curcumin-loaded gemini surfactant nanoparticles: Synthesis, characterization and evaluation of anticancer activity against human breast cancer cell lines. Phytomedicine 2019, 57, 183–190. [Google Scholar] [CrossRef]
- Purpura, M.; Lowery, R.P.; Wilson, J.M.; Mannan, H.; Münch, G.; Razmovski-Naumovski, V. Analysis of different innovative formulations of curcumin for improved relative oral bioavailability in human subjects. Eur. J. Nutr. 2018, 57, 929–938. [Google Scholar] [CrossRef]
- Ireson, C.; Orr, S.; Jones, D.J.; Verschoyle, R.; Lim, C.-K.; Luo, J.-L.; Howells, L.; Plummer, S.; Jukes, R.; Williams, M. Characterization of metabolites of the chemopreventive agent curcumin in human and rat hepatocytes and in the rat in vivo, and evaluation of their ability to inhibit phorbol ester-induced prostaglandin E2 production. Cancer Res. 2001, 61, 1058–1064. [Google Scholar]
- Shoba, G.; Joy, D.; Joseph, T.; Majeed, M.; Rajendran, R.; Srinivas, P. Influence of piperine on the pharmacokinetics of curcumin in animals and human volunteers. Planta Med. 1998, 64, 353–356. [Google Scholar] [CrossRef]
- Zibaei, Z.; Babaei, E.; Rezaie Nezhad Zamani, A.; Rahbarghazi, R.; Azeez, H.J. Curcumin-enriched Gemini surfactant nanoparticles exhibited tumoricidal effects on human 3D spheroid HT-29 cells in vitro. Cancer Nanotechnol. 2021, 12, 3. [Google Scholar] [CrossRef]
- Al-Kinani, M.A.; Haider, A.J.; Al-Musawi, S. Design, construction and characterization of intelligence polymer coated core–shell nanocarrier for curcumin drug encapsulation and delivery in lung cancer therapy purposes. J. Inorg. Organomet. Polym. Mater. 2020, 31, 70–79. [Google Scholar] [CrossRef]
- Mahmoudi, R.; Hassandokht, F.; Ardakani, M.T.; Karimi, B.; Roustazadeh, A.; Tarvirdipour, S.; Barmak, M.J.; Nikseresht, M.; Baneshi, M.; Mousavizadeh, A. Intercalation of curcumin into liposomal chemotherapeutic agent augments apoptosis in breast cancer cells. J. Biomater. Appl. 2021, 35, 1005–1018. [Google Scholar] [CrossRef]
- Khan, A.Q.; Ahmed, E.I.; Elareer, N.; Fathima, H.; Prabhu, K.S.; Siveen, K.S.; Kulinski, M.; Azizi, F.; Dermime, S.; Ahmad, A. Curcumin-mediated apoptotic cell death in papillary thyroid cancer and cancer stem-like cells through targeting of the JAK/STAT3 signaling pathway. Int. J. Mol. Sci. 2020, 21, 438. [Google Scholar] [CrossRef] [PubMed]
- Kwon, Y. Curcumin as a cancer chemotherapy sensitizing agent. J. Korean Soc. Appl. Biol. Chem. 2014, 57, 273–280. [Google Scholar] [CrossRef]
- Wilken, R.; Veena, M.S.; Wang, M.B.; Srivatsan, E.S. Curcumin: A review of anti-cancer properties and therapeutic activity in head and neck squamous cell carcinoma. Mol. Cancer 2011, 10, 12. [Google Scholar] [CrossRef]
- Ravindran, J.; Prasad, S.; Aggarwal, B.B. Curcumin and cancer cells: How many ways can curry kill tumor cells selectively? AAPS J. 2009, 11, 495–510. [Google Scholar] [CrossRef]
- Zhou, H.; Beevers, C.S.; Huang, S. The targets of curcumin. Curr. Drug Targets 2011, 12, 332–347. [Google Scholar] [CrossRef]
- Lev-Ari, S.; Starr, A.; Vexler, A.; Karaush, V.; Loew, V.; Greif, J.; Fenig, E.; Aderka, D.; Ben-Yosef, R. Inhibition of pancreatic and lung adenocarcinoma cell survival by curcumin is associated with increased apoptosis, down-regulation of COX-2 and EGFR and inhibition of Erk1/2 activity. Anticancer Res. 2006, 26, 4423–4430. [Google Scholar]
- Ghaderi, S.; Babaei, E.; Hussen, B.M.; Mahdavi, M.; Azeez, H.J. Gemini curcumin suppresses proliferation of ovarian cancer OVCAR-3 cells via induction of apoptosis. Anticancer Agents Med. Chem. 2021, 21, 775–781. [Google Scholar] [CrossRef]
- Sobhkhizi, A.; Babaei, E.; Azeez, H.J.; Katiraee, F.; Hussen, B.M.; Hoseinpour Feizi, M.A. Dendrosomal nano-curcumin modulates P-glycoprotein activity and induces apoptosis in wild type and P53-mutant breast cancer cell lines. Jentashapir J. Cell. Mol. Biol. 2020, 11, e109143. [Google Scholar] [CrossRef]
- Ebrahimi, M.; Babaei, E.; Neri, F.; Feizi, M.A.H. Anti-proliferative and apoptotic effect of gemini curcumin in p53-wild type and p53-mutant colorectal cancer cell lines. Int. J. Pharm. 2021, 601, 120592. [Google Scholar] [CrossRef] [PubMed]
- Hajizadeh, M.; Hafez Ghoran, S.; Azeez, H.J.; Feizi, M.A.H.; Babaei, E. Apoptotic effects of Gemini curcumin on MDA-MB-468 breast cancer cell line. Anticancer Agents Med. Chem. 2021, 22, 2181–2188. [Google Scholar] [CrossRef] [PubMed]
- Jabbari, N.; Hafez Ghoran, S.; Semsari, H.; Hussen, B.; Babaei, E. Gemini Curcumin suppresses gastric cancer AGS cells proliferation through modulation of lncRNA CCAT2 and c-Myc genes. Turk. J. Pharm. Sci. 2022, 19, 239–245. [Google Scholar] [CrossRef]
- Tan, B.L.; Norhaizan, M.E. Curcumin combination chemotherapy: The implication and efficacy in cancer. Molecules 2019, 24, 2527. [Google Scholar] [CrossRef] [PubMed]
- Basniwal, R.K.; Khosla, R.; Jain, N. Improving the anticancer activity of curcumin using nanocurcumin dispersion in water. Nutr. Cancer 2014, 66, 1015–1022. [Google Scholar] [CrossRef]
- Yallapu, M.M.; Nagesh, P.K.B.; Jaggi, M.; Chauhan, S.C. Therapeutic applications of curcumin nanoformulations. AAPS J. 2015, 17, 1341–1356. [Google Scholar] [CrossRef]
- Baghi, N.; Bakhshinejad, B.; Keshavarz, R.; Babashah, S.; Sadeghizadeh, M. Dendrosomal nanocurcumin and exogenous p53 can act synergistically to elicit anticancer effects on breast cancer cells. Gene 2018, 670, 55–62. [Google Scholar] [CrossRef]
- D’Ignazio, L.; Batie, M.; Rocha, S. Hypoxia and inflammation in cancer, focus on HIF and NF-κB. Biomedicines 2017, 5, 21. [Google Scholar] [CrossRef]
- Khan, M.N.; Haggag, Y.A.; Lane, M.E.; McCarron, P.A.; Tambuwala, M.M. Polymeric nano-encapsulation of curcumin enhances its anti-cancer activity in breast (MDA-MB231) and lung (A549) cancer cells through reduction in expression of HIF-1α and nuclear p65 (REL A). Curr. Drug Deliv. 2018, 15, 286–295. [Google Scholar] [CrossRef]
- Monsuez, J.J.; Charniot, J.C.; Vignat, N.; Artigou, J.Y. Cardiac side-effects of cancer chemotherapy. Int. J. Cardiol. 2010, 144, 3–15. [Google Scholar] [CrossRef] [PubMed]
- Navya, P.N.; Kaphle, A.; Srinivas, S.P.; Bhargava, S.K.; Rotello, V.M.; Daima, H.K. Current trends and challenges in cancer management and therapy using designer nanomaterials. Nano Converg. 2019, 6, 23. [Google Scholar] [CrossRef] [PubMed]
- Yallapu, M.M.; Jaggi, M.; Chauhan, S.C. Curcumin nanoformulations: A future nanomedicine for cancer. Drug Discov. Today 2012, 17, 71–80. [Google Scholar] [CrossRef]
- Persano, F.; Gigli, G.; Leporatti, S. Lipid-polymer hybrid nanoparticles in cancer therapy: Current overview and future directions. Nano Express 2021, 2, 012006. [Google Scholar] [CrossRef]
- Yallapu, M.M.; Jaggi, M.; Chauhan, S.C. Curcumin nanomedicine: A road to cancer therapeutics. Curr. Pharm. Des. 2013, 19, 1994–2010. [Google Scholar] [CrossRef] [PubMed]
- Anand, P.; Nair, H.B.; Sung, B.; Kunnumakkara, A.B.; Yadav, V.R.; Tekmal, R.R.; Aggarwal, B.B. Design of curcumin-loaded PLGA nanoparticles formulation with enhanced cellular uptake, and increased bioactivity in vitro and superior bioavailability in vivo. Biochem. Pharmacol. 2010, 79, 330–338. [Google Scholar] [CrossRef]
- Nair, K.L.; Thulasidasan, A.K.; Deepa, G.; Anto, R.J.; Kumar, G.S. Purely aqueous PLGA nanoparticulate formulations of curcumin exhibit enhanced anticancer activity with dependence on the combination of the carrier. Int. J. Pharm. 2012, 425, 44–52. [Google Scholar] [CrossRef]
- Mukerjee, A.; Vishwanatha, J.K. Formulation, characterization and evaluation of curcumin-loaded PLGA nanospheres for cancer therapy. Anticancer Res. 2009, 29, 3867–3875. [Google Scholar]
- Gou, M.; Men, K.; Shi, H.; Xiang, M.; Zhang, J.; Song, J.; Long, J.; Wan, Y.; Luo, F.; Zhao, X.; et al. Curcumin-loaded biodegradable polymeric micelles for colon cancer therapy in vitro and in vivo. Nanoscale 2011, 3, 1558–1567. [Google Scholar] [CrossRef]
- Duan, J.; Zhang, Y.; Han, S.; Chen, Y.; Li, B.; Liao, M.; Chen, W.; Deng, X.; Zhao, J.; Huang, B. Synthesis and in vitro/in vivo anti-cancer evaluation of curcumin-loaded chitosan/poly(butyl cyanoacrylate) nanoparticles. Int. J. Pharm. 2010, 400, 211–220. [Google Scholar] [CrossRef]
- Babaei, E.; Sadeghizadeh, M.; Hassan, Z.M.; Feizi, M.A.; Najafi, F.; Hashemi, S.M. Dendrosomal curcumin significantly suppresses cancer cell proliferation in vitro and in vivo. Int. Immunopharmacol. 2012, 12, 226–234. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Zhu, W.; Yang, C.; Guo, H.; Yu, A.; Ji, J.; Gao, Y.; Sun, M.; Zhai, G. A novel folate-modified self-microemulsifying drug delivery system of curcumin for colon targeting. Int. J. Nanomed. 2012, 7, 151–162. [Google Scholar] [CrossRef]
- Rejinold, N.S.; Sreerekha, P.R.; Chennazhi, K.P.; Nair, S.V.; Jayakumar, R. Biocompatible, biodegradable and thermo-sensitive chitosan-g-poly(N-isopropylacrylamide) nanocarrier for curcumin drug delivery. Int. J. Biol. Macromol. 2011, 49, 161–172. [Google Scholar] [CrossRef] [PubMed]
- Chiu, S.S.; Lui, E.; Majeed, M.; Vishwanatha, J.K.; Ranjan, A.P.; Maitra, A.; Pramanik, D.; Smith, J.A.; Helson, L. Differential distribution of intravenous curcumin formulations in the rat brain. Anticancer Res. 2011, 31, 907–911. [Google Scholar]
- Lim, K.J.; Bisht, S.; Bar, E.E.; Maitra, A.; Eberhart, C.G. A polymeric nanoparticle formulation of curcumin inhibits growth, clonogenicity and stem-like fraction in malignant brain tumors. Cancer Biol. Ther. 2011, 11, 464–467. [Google Scholar] [CrossRef] [PubMed]
- Sahu, A.; Bora, U.; Kasoju, N.; Goswami, P. Synthesis of novel biodegradable and self-assembling methoxy poly(ethylene glycol)-palmitate nanocarrier for curcumin delivery to cancer cells. Acta Biomater. 2008, 4, 1752–1761. [Google Scholar] [CrossRef]
- Hettiarachchi, S.S.; Dunuweera, S.P.; Dunuweera, A.N.; Rajapakse, R.M.G. Synthesis of curcumin nanoparticles from raw turmeric rhizome. ACS Omega 2021, 6, 8246–8252. [Google Scholar] [CrossRef]
- Flora, G.; Gupta, D.; Tiwari, A. Nanocurcumin: A promising therapeutic advancement over native curcumin. Crit. Rev. Ther. Drug Carr. Syst. 2013, 30, 331–368. [Google Scholar] [CrossRef]
- Rahimi, H.R.; Nedaeinia, R.; Sepehri Shamloo, A.; Nikdoust, S.; Kazemi Oskuee, R. Novel delivery system for natural products: Nano-curcumin formulations. Avicenna J. Phytomedicine 2016, 6, 383–398. [Google Scholar]
- Bisht, S.; Feldmann, G.; Soni, S.; Ravi, R.; Karikar, C.; Maitra, A.; Maitra, A. Polymeric nanoparticle-encapsulated curcumin (“nanocurcumin”): A novel strategy for human cancer therapy. J. Nanobiotechnology 2007, 5, 3. [Google Scholar] [CrossRef]
- Ma, Z.; Shayeganpour, A.; Brocks, D.R.; Lavasanifar, A.; Samuel, J. High-performance liquid chromatography analysis of curcumin in rat plasma: Application to pharmacokinetics of polymeric micellar formulation of curcumin. Biomed. Chromatogr. 2007, 21, 546–552. [Google Scholar] [CrossRef]
- Dende, C.; Meena, J.; Nagarajan, P.; Nagaraj, V.A.; Panda, A.K.; Padmanaban, G. Nanocurcumin is superior to native curcumin in preventing degenerative changes in Experimental Cerebral Malaria. Sci. Rep. 2017, 7, 10062. [Google Scholar] [CrossRef] [PubMed]
- Mythri, R.B.; Jagatha, B.; Pradhan, N.; Andersen, J.; Bharath, M.S. Mitochondrial complex I inhibition in Parkinson’s disease: How can curcumin protect mitochondria? Antioxid. Redox Signal. 2007, 9, 399–408. [Google Scholar] [CrossRef] [PubMed]
- Mohanty, C.; Sahoo, S.K. The in vitro stability and in vivo pharmacokinetics of curcumin prepared as an aqueous nanoparticulate formulation. Biomaterials 2010, 31, 6597–6611. [Google Scholar] [CrossRef] [PubMed]
- Muller, R.H.; Keck, C.M. Challenges and solutions for the delivery of biotech drugs—A review of drug nanocrystal technology and lipid nanoparticles. J. Biotechnol. 2004, 113, 151–170. [Google Scholar] [CrossRef]
- No, D.S.; Algburi, A.; Huynh, P.; Moret, A.; Ringard, M.; Comito, N.; Drider, D.; Takhistov, P.; Chikindas, M.L. Antimicrobial efficacy of curcumin nanoparticles against Listeria monocytogenes is mediated by surface charge. J. Food Saf. 2017, 37, e12353. [Google Scholar] [CrossRef]
- Beatty, G.L.; Gladney, W.L. Immune escape mechanisms as a guide for cancer immunotherapy. Clin. Cancer Res. 2015, 21, 687–692. [Google Scholar] [CrossRef]
- Von Rundstedt, F.-C.; Necchi, A. Current markers and their value in the era of immuno-oncology. Transl. Androl. Urol. 2017, 6, 1111. [Google Scholar] [CrossRef]
- Dustin, M.L. Killers on sterols. Nature 2016, 531, 583–584. [Google Scholar] [CrossRef]
- Zhang, H.; Chen, J. Current status and future directions of cancer immunotherapy. J. Cancer 2018, 9, 1773. [Google Scholar] [CrossRef]
- Vanneman, M.; Dranoff, G. Combining immunotherapy and targeted therapies in cancer treatment. Nat. Rev. Cancer 2012, 12, 237–251. [Google Scholar] [CrossRef] [PubMed]
- Hernández, Á.-P.; Juanes-Velasco, P.; Landeira-Viñuela, A.; Bareke, H.; Montalvillo, E.; Góngora, R.; Fuentes, M. Restoring the immunity in the tumor microenvironment: Insights into immunogenic cell death in onco-therapies. Cancers 2021, 13, 2821. [Google Scholar] [CrossRef]
- Koury, J.; Lucero, M.; Cato, C.; Chang, L.; Geiger, J.; Henry, D.; Hernandez, J.; Hung, F.; Kaur, P.; Teskey, G. Immunotherapies: Exploiting the immune system for cancer treatment. J. Immunol. Res. 2018, 2018, 9585614. [Google Scholar] [CrossRef] [PubMed]
- Paul, S.; Sa, G. Curcumin as an adjuvant to cancer immunotherapy. Front. Oncol. 2021, 11, 675923. [Google Scholar] [CrossRef] [PubMed]
- Zhu, X.; Yu, Z.; Feng, L.; Deng, L.; Fang, Z.; Liu, Z.; Li, Y.; Wu, X.; Qin, L.; Guo, R. Chitosan-based nanoparticle co-delivery of docetaxel and curcumin ameliorates anti-tumor chemoimmunotherapy in lung cancer. Carbohydr. Polym. 2021, 268, 118237. [Google Scholar] [CrossRef]
- Venditto, V.J.; Simanek, E.E. Cancer therapies utilizing the camptothecins: A review of the in vivo literature. Mol. Pharm. 2010, 7, 307–349. [Google Scholar] [CrossRef]
- Wang, Z.; Wang, X.; Yu, H.; Chen, M. Glioma-targeted multifunctional nanoparticles to co-deliver camptothecin and curcumin for enhanced chemo-immunotherapy. Biomater. Sci. 2022, 10, 1292–1303. [Google Scholar] [CrossRef]
- Bhattacharyya, S.; Md Sakib Hossain, D.; Mohanty, S.; Sankar Sen, G.; Chattopadhyay, S.; Banerjee, S.; Chakraborty, J.; Das, K.; Sarkar, D.; Das, T. Curcumin reverses T cell-mediated adaptive immune dysfunctions in tumor-bearing hosts. Cell. Mol. Immunol. 2010, 7, 306–315. [Google Scholar] [CrossRef]
- Zhao, H.-M.; Xu, R.; Huang, X.-Y.; Cheng, S.-M.; Huang, M.-F.; Yue, H.-Y.; Wang, X.; Zou, Y.; Lu, A.-P.; Liu, D.-Y. Curcumin improves regulatory T cells in gut-associated lymphoid tissue of colitis mice. World J. Gastroenterol. 2016, 22, 5374. [Google Scholar] [CrossRef]
- Milano, F.; Mari, L.; van de Luijtgaarden, W.; Parikh, K.; Calpe, S.; Krishnadath, K. Nano-curcumin inhibits proliferation of esophageal adenocarcinoma cells and enhances the T cell mediated immune response. Front. Oncol. 2013, 3, 137. [Google Scholar] [CrossRef]
- Rosenberg, S.A.; Restifo, N.P.; Yang, J.C.; Morgan, R.A.; Dudley, M.E. Adoptive cell transfer: A clinical path to effective cancer immunotherapy. Nat. Rev. Cancer 2008, 8, 299–308. [Google Scholar] [CrossRef] [PubMed]
- Hawkins, R.E.; Gilham, D.E.; Debets, R.; Eshhar, Z.; Taylor, N.; Abken, H.; Schumacher, T.N. Development of adoptive cell therapy for cancer: A clinical perspective. Hum. Gene Ther. 2010, 21, 665–672. [Google Scholar] [CrossRef] [PubMed]
- Chang, Y.-F.; Chuang, H.-Y.; Hsu, C.-H.; Liu, R.-S.; Gambhir, S.S.; Hwang, J.-J. Immunomodulation of curcumin on adoptive therapy with T cell functional imaging in mice. Cancer Prev. Res. 2012, 5, 444–452. [Google Scholar] [CrossRef] [PubMed]
- Hossen, S.; Hossain, M.K.; Basher, M.; Mia, M.; Rahman, M.; Uddin, M.J. Smart nanocarrier-based drug delivery systems for cancer therapy and toxicity studies: A review. J. Adv. Res. 2019, 15, 1–18. [Google Scholar] [CrossRef]
- Zhang, J.; Li, J.; Shi, Z.; Yang, Y.; Xie, X.; Lee, S.M.; Wang, Y.; Leong, K.W.; Chen, M. pH-sensitive polymeric nanoparticles for co-delivery of doxorubicin and curcumin to treat cancer via enhanced pro-apoptotic and anti-angiogenic activities. Acta Biomater. 2017, 58, 349–364. [Google Scholar] [CrossRef]
- Kamble, S.; Utage, B.; Mogle, P.; Kamble, R.; Hese, S.; Dawane, B.; Gacche, R. Evaluation of curcumin capped copper nanoparticles as possible inhibitors of human breast cancer cells and angiogenesis: A comparative study with native curcumin. AAPS PharmSciTech 2016, 17, 1030–1041. [Google Scholar] [CrossRef]
- Ali, I.; Ahmed, S.; Elhaj, B.M.; Ali, H.S.; Alsubaie, A.; Almalki, A.S. Enhanced anticancer activities of curcumin-loaded green gum acacia-based silver nanoparticles against melanoma and breast cancer cells. Appl. Nanosci. 2021, 11, 2679–2687. [Google Scholar] [CrossRef]
- Van Der Vlies, A.J.; Morisaki, M.; Neng, H.I.; Hansen, E.M.; Hasegawa, U. Framboidal nanoparticles containing a curcumin–phenylboronic acid complex with antiangiogenic and anticancer activities. Bioconjugate Chem. 2019, 30, 861–870. [Google Scholar] [CrossRef]
- Chang, A.H.; Parsonnet, J. Role of bacteria in oncogenesis. Clin. Microbiol. Rev. 2010, 23, 837–857. [Google Scholar] [CrossRef]
- Weitzman, M.D.; Fradet-Turcotte, A. Virus DNA replication and the host DNA damage response. Annu. Rev. Virol. 2018, 5, 141. [Google Scholar] [CrossRef]
- Zella, D.; Gallo, R.C. Viruses and bacteria associated with cancer: An overview. Viruses 2021, 13, 1039. [Google Scholar] [CrossRef] [PubMed]
- Zheng, D.; Huang, C.; Huang, H.; Zhao, Y.; Khan, M.R.U.; Zhao, H.; Huang, L. Antibacterial mechanism of curcumin: A review. Chem. Biodivers. 2020, 17, e2000171. [Google Scholar] [CrossRef] [PubMed]
- Dai, C.; Lin, J.; Li, H.; Shen, Z.; Wang, Y.; Velkov, T.; Shen, J. The natural product curcumin as an antibacterial agent: Current achievements and problems. Antioxidants 2022, 11, 459. [Google Scholar] [CrossRef] [PubMed]
- Alam, J.; Dilnawaz, F.; Sahoo, S.K.; Singh, D.V.; Mukhopadhyay, A.K.; Hussain, T.; Pati, S. Curcumin encapsulated into biocompatible co-polymer PLGA nanoparticle enhanced anti-gastric cancer and anti-Helicobacter Pylori effect. Asian Pac. J. Cancer Prev. 2022, 23, 61–70. [Google Scholar] [CrossRef]
- Jennings, M.R.; Parks, R.J. Curcumin as an antiviral agent. Viruses 2020, 12, 1242. [Google Scholar] [CrossRef]
- Mathew, D.; Hsu, W.L. Antiviral potential of curcumin. J. Funct. Foods 2018, 40, 692–699. [Google Scholar] [CrossRef]
- Prusty, B.K.; Das, B.C. Constitutive activation of transcription factor AP-1 in cervical cancer and suppression of human papillomavirus (HPV) transcription and AP-1 activity in HeLa cells by curcumin. Int. J. Cancer 2005, 113, 951–960. [Google Scholar] [CrossRef]
- Mishra, A.; Kumar, R.; Tyagi, A.; Kohaar, I.; Hedau, S.; Bharti, A.C.; Sarker, S.; Dey, D.; Saluja, D.; Das, B. Curcumin modulates cellular AP-1, NF-kB, and HPV16 E6 proteins in oral cancer. Ecancermedicalscience 2015, 9, 525. [Google Scholar] [CrossRef]
- Mitra, S.; Lami, M.S.; Ghosh, A.; Das, R.; Tallei, T.E.; Islam, F.; Dhama, K.; Begum, M.Y.; Aldahish, A.; Chidambaram, K.; et al. Hormonal therapy for gynecological cancers: How far has science progressed toward clinical applications? Cancers 2022, 14, 759. [Google Scholar] [CrossRef]
- Li, H.; Zhong, C.; Wang, Q.; Chen, W.; Yuan, Y. Curcumin is an APE1 redox inhibitor and exhibits an antiviral activity against KSHV replication and pathogenesis. Antivir. Res. 2019, 167, 98–103. [Google Scholar] [CrossRef]
- Bollen, L.J.; Tjong-A-Hung, S.P.; Van Der Velden, J.; Mol, B.W.; Boer, K.; Kate, F.J.T.; Bleker, O.P.; Schegget, J.T. Clearance of cervical human papillomavirus infection by treatment for cervical dysplasia. Sex. Transm. Dis. 1997, 24, 456–460. [Google Scholar] [CrossRef] [PubMed]
- Debata, P.R.; Castellanos, M.R.; Fata, J.E.; Baggett, S.; Rajupet, S.; Szerszen, A.; Begum, S.; Mata, A.; Murty, V.V.; Opitz, L.M.; et al. A novel curcumin-based vaginal cream Vacurin selectively eliminates apposed human cervical cancer cells. Gynecol. Oncol. 2013, 129, 145–153. [Google Scholar] [CrossRef] [PubMed]
- Talwar, G.P.; Dar, S.A.; Rai, M.K.; Reddy, K.V.R.; Mitra, D.; Kulkarni, S.V.; Doncel, G.F.; Buck, C.B.; Schiller, J.T.; Muralidhar, S.; et al. A novel polyherbal microbicide with inhibitory effect on bacterial, fungal and viral genital pathogens. Int. J. Antimicrob. Agents 2008, 32, 180–185. [Google Scholar] [CrossRef]
- Maher, D.M.; Bell, M.C.; O’Donnell, E.A.; Gupta, B.K.; Jaggi, M.; Chauhan, S.C. Curcumin suppresses human papillomavirus oncoproteins, restores p53, rb, and ptpn13 proteins and inhibits benzo[a]pyrene-induced upregulation of HPV E7. Mol. Carcinog. 2011, 50, 47–57. [Google Scholar] [CrossRef] [PubMed]
- Dang, Y.P.; Yuan, X.Y.; Tian, R.; Li, D.G.; Liu, W. Curcumin improves the paclitaxel-induced apoptosis of HPV-positive human cervical cancer cells via the NF-κB-p53-caspase-3 pathway. Exp. Ther. Med. 2015, 9, 1470–1476. [Google Scholar] [CrossRef]
- Mudduluru, G.; George-William, J.N.; Muppala, S.; Asangani, I.A.; Kumarswamy, R.; Nelson, L.D.; Allgayer, H. Curcumin regulates miR-21 expression and inhibits invasion and metastasis in colorectal cancer. Biosci. Rep. 2011, 31, 185–197. [Google Scholar] [CrossRef]
- Ahmadi, M.; Agah, E.; Nafissi, S.; Jaafari, M.R.; Harirchian, M.H.; Sarraf, P.; Faghihi-Kashani, S.; Hosseini, S.J.; Ghoreishi, A.; Aghamollaii, V. Safety and efficacy of nanocurcumin as add-on therapy to riluzole in patients with amyotrophic lateral sclerosis: A pilot randomized clinical trial. Neurotherapeutics 2018, 15, 430–438. [Google Scholar] [CrossRef]
- Dolati, S.; Babaloo, Z.; Ayromlou, H.; Ahmadi, M.; Rikhtegar, R.; Rostamzadeh, D.; Roshangar, L.; Nouri, M.; Mehdizadeh, A.; Younesi, V. Nanocurcumin improves regulatory T-cell frequency and function in patients with multiple sclerosis. J. Neuroimmunol. 2019, 327, 15–21. [Google Scholar] [CrossRef]
- Braden, A.R.; Vishwanatha, J.K.; Kafka, E. Formulation of Active Agent Loaded Activated PLGA Nanoparticles for Targeted Cancer Nano-Therapeutics. U.S. Patent No. US20080253961A1, 16 October 2008. [Google Scholar]
- Ranjan, A.P.; Mukerjee, A.; Vishwanatha, J.K. Solid in Oil/Water Emulsion-Diffusion-Evaporation Formulation for Preparing Curcumin-Loaded PLGA Nanoparticles. U.S. Patent No. US20100290982A1, 18 November 2010. [Google Scholar]
- Pathak, Y.; Tran, H.T. Nanoemulsions Containing Antioxidants and Other Health-Promoting Compounds. U.S. Patent No. US20120052126A1, 1 March 2012. [Google Scholar]
- Kumar, K.S.; Gnanaprakash, D.; Mayilvaganan, K.; Arunraj, C.; Mohankumar, S. Chitosan-gold nanoparticles as delivery systems for curcumin. Int. J. Pharm. Sci. Res. 2012, 3, 4533. [Google Scholar] [CrossRef]
- Khamar, B.M.; Gogia, A.P.; Goda, C.C.; Shenoy, D.B.; Shrivastava, R.R.; Patravale, V.B.; Modi, I.A.; Laddha, R.N.; Khan, I.A. Pharmaceutical Compositions of Curcumin. U.S. Patent No. US9474727B2, 25 October 2016. [Google Scholar]
- Shen, Y.; Tang, H.; Van Kirk, E.; Murdoch, W.; Radosz, M. Curcumin-Containing Polymers and Water-Soluble Curcumin Derivatives as Prodrugs of Prodrug Carriers. U.S. Patent No. US20120003177A1, 5 January 2012. [Google Scholar]
- Kurzrock, R.; Li, L.; Mehta, K.; Aggarawal, B.B. Liposomal Curcumin for Treatment of Cancer. U.S. Patent No. US20060067998A1, 15 March 2006. [Google Scholar]
- Chauhan, S.; Jaggi, M.; Yallapu, M.M. Magnetic Nanoparticle Formulations, Methods for Making Such Formulations, and Methods for Their Use. U.S. Patent No. US20130245357A1, 19 September 2013. [Google Scholar]
- Kumar, A.; Mohapatra, S.S.; Cameron, D.F. Nanoparticle Targeted Drug Delivery to the Lungs Using Extra-Testicular Sertoli Cells. WIPO Patent No. US10272053B2, 30 April 2019. [Google Scholar]
- Chaniyilparampu, R.N.; Mungala, M.; Kapoor, A.; Gokaraju, G.R.; Gokaraju, R.R.; Bhupathiraju, K.; Golakoti, T.; Nair, A.K.; Murali, M.R.; Parthasarathy, K. Topical Formulation(s) For the Treatment of Inflammation, Skin and Mucosal Disorders and Other Diseases. U.S. Patent No. US8535693B2, 17 September 2013. [Google Scholar]
- Di Martino, R.M.C.; Luppi, B.; Bisi, A.; Gobbi, S.; Rampa, A.; Abruzzo, A.; Belluti, F. Recent progress on curcumin-based therapeutics: A patent review (2012–2016). Part I: Curcumin. Expert Opin. Ther. Pat. 2017, 27, 579–590. [Google Scholar] [CrossRef]
- Xianwang, W.; Hufuqiang, K.; Jian, H. Preparation Method and Application of Curcumin Chitosan-Stearic Acid Graft Micelle. Chinese Patent No. CN102743336A, 24 October 2012. [Google Scholar]
- Bansal, A.K.; Dantuluri, A.K.R.; Bhaskarao, S.G.; Bapurao, P.Y. Nanocrystalline Solid Dispersion Compositions and Process of Preparation. WIPO Patent No. WO2013132457A2, 12 September 2013. [Google Scholar]
- Pattayil, A.J.; Jayaprabha, K.N. Curcumin Coated Magnetite Nanoparticles for Biomedical Applications. WIPO Patent No. WO2013108270A1, 25 July 2013. [Google Scholar]
- Dhar, S.; Marrache, S.M. Nanoparticles for Mitochondrial Trafficking of Agents. WIPO Patent No. WO2013123298A1, 22 August 2013. [Google Scholar]
- Ranjan, A.P.; Mukerjee, A.; Vishwanatha, J.K.; Helson, L. Curcumin-Er, a Liposomal-PLGA Sustained Release Nanocurcumin for Minimizing QT Prolongation for Cancer Therapy. U.S. Patent No. US9138411B2, 22 September 2015. [Google Scholar]
- Sripathy, R.; Mandapati, V.N.S.R.R.; Gopaal, A.; Somashekara, N.; Chaniyilparampu, R.N.; Gokaraju, R.R.; Gokaraju, G.R.; Bhupathiraju, K.; Anjana, D. Highly Bioavailable, Water Soluble and Sustained Release Nanoformulations Hydrophobic Plant Derived Compounds and Extracts. U.S. Patent No. US20150072012A1, 12 March 2015. [Google Scholar]
- Liu, Y.; Wang, W.; Dong, W.; Hao, H.; Xia, X.; Huang, Y.; Jin, Y.; Jiang, L.; Zhou, J. Phospholipid/Chitosan Drug Delivery System, Preparation Method and Uses Thereof. Chinese Patent No. WO2017186065A1, 12 November 2017. [Google Scholar]
- Sezgin, V.C.; Bayraktar, O. Preparation of Curcumin- and Piperine Loaded Biopolymer Based Nano-Delivery Systems Using Electrospray/Coating Techniques. European Patent No. EP3142702B1, 3 January 2018. [Google Scholar]
Curcumin Nanoforms | In Vitro Cytotoxic Activity | Molecular Mechanism | In Vivo Results | Ref. |
---|---|---|---|---|
Poly(lactide-co-glycolide); PLGA | Cytotoxicity against HCT116, DU145, MDA-MB-231, SEG-1, Jurkat, and KBM-5 cells with IC50 < 5 μM. | NF-κB-induced inactivation of and decrease in cyclin D1, MMP-9, and VEGF production. | The half-life of curcumin nanoparticles was 1.75 times longer than curcumin. | [184] |
Poly(lactide-co-glycolide); PLGA | Equal cytotoxicity of nanocurcumin and curcumin toward SKBr3, HeLa, and A549 cells. | Increase in Annexin V staining, cleaved PARP expression. Decrease in NF-κB activation. | Not available. | [185] |
Poly(lactide-co-glycolide); PLGA | Cytotoxicity against PC-3, LNCaP, and DU145 cells; curcumin-loaded PLGA nanostructures: IC50 = 20–22.5 μM; free curcumin: IC50 = 32–34 μM. | Inhibition of NF-κB function. | Not available. | [186] |
β-cyclodextrin self-assembly of curcumin | In C4-2 and DU145 cells, the curcumin self-assembly concentration was 16.8 μM and 17.6 μM, respectively, which is slightly less than the free curcumin concentration. | Increase in cleaved PARP expression. | Increased curcumin levels in serum concentrations by up to twofold (Unpublished data with Subhash Chauhan Lab) | [17] |
MPEG-PCL micelle | Cytotoxicity against C-26 colon cancer cells; Cur-MPEG-PCL micelles: IC50 = 5.78 mg·mL−1. Free curcumin: IC50 = 3.95 mg·mL−1. | Not available. | Increase in curcumin concentrations in rat plasma (>2 times) and suppression of subcutaneous C-26 colon cancer development in a xenograft mice model. | [187] |
Poly(butyl cyanoacrylate) nanomateriales | Cytotoxicity against Bel7402, HepG2, and Huh7 cells (IC50 ≈ 15 μg/mL). | Suppression of VEGF and downregulation of COX-2 expression. | A 2.2-fold reduction in HepG2 tumor volume in a xenograft mice model. | [188] |
Dendrosomal curcumin | Cytotoxicity against WEHI-164 cells; IC50 = 16.8 & 7.5 μM after 24 & 48 h. Cytotoxicity against A431 cells: (IC50 = 19.2 and 14.3 μM after 24 & 48 h. | Increase in cleaved PARP expression and further Annexin V staining (apoptosis). | Reduction in tumor development. | [189] |
Self-microemulsifying medication delivery device enhanced with folic acid. | Effective cytotoxicity of folate curcumin-nanoemulsion, curcumin-emulsion, and free curcumin against HeLa cells at concentrations of 18.27, 36.69, and 30.4 μM, respectively. Effective cytotoxicity of folate curcumin-nanoemulsion, curcumin-emulsion, and free curcumin against HT-29 cells at concentrations of 20.57, 38.59, and 25.62 μM, respectively. | Not available. | Increase in folate curcumin-nanoemulsion adsorbsion from 58.41% to 73.38% in 6 h (in situ colon-perfused rats) | [190] |
Thermo-sensitive nanocarrier | Showing particular toxic effects on cancer cell lines (KB, MCF-7, and PC-3 cells) while being nontoxic to the L929 cell line. | Increase in apoptosis due to Annexin-A and PI binding. | Not available. | [191] |
NanoCurc™ | Little or inhibited growth of JHH-GBM14, D283Med, DAOY, and glioblastoma neurosphere lines HSR-GBM1. | G(2)/M arrest and apoptosis induction via the inhibition of STAT3 and Hedgehog signaling pathways. | ~0.5% localization of the injected drug within the brain. | [192,193] |
Amphiphilic mPEG-palmitic acid polymer | Cytotoxicity against HeLa cells; nanocurcumin; IC50 = 15.58 μM, curcumin; IC50 = 14.32 μM. | Increasing the anticancer activity in vitro by enzyme-catalyzed release. | Not available. | [194] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Hafez Ghoran, S.; Calcaterra, A.; Abbasi, M.; Taktaz, F.; Nieselt, K.; Babaei, E. Curcumin-Based Nanoformulations: A Promising Adjuvant towards Cancer Treatment. Molecules 2022, 27, 5236. https://doi.org/10.3390/molecules27165236
Hafez Ghoran S, Calcaterra A, Abbasi M, Taktaz F, Nieselt K, Babaei E. Curcumin-Based Nanoformulations: A Promising Adjuvant towards Cancer Treatment. Molecules. 2022; 27(16):5236. https://doi.org/10.3390/molecules27165236
Chicago/Turabian StyleHafez Ghoran, Salar, Andrea Calcaterra, Milad Abbasi, Fatemeh Taktaz, Kay Nieselt, and Esmaeil Babaei. 2022. "Curcumin-Based Nanoformulations: A Promising Adjuvant towards Cancer Treatment" Molecules 27, no. 16: 5236. https://doi.org/10.3390/molecules27165236
APA StyleHafez Ghoran, S., Calcaterra, A., Abbasi, M., Taktaz, F., Nieselt, K., & Babaei, E. (2022). Curcumin-Based Nanoformulations: A Promising Adjuvant towards Cancer Treatment. Molecules, 27(16), 5236. https://doi.org/10.3390/molecules27165236