Advances in the Application of Nanomaterials to the Treatment of Melanoma
Abstract
:1. Introduction
2. Cutaneous Melanoma
2.1. Nanoparticles for Drug-Targeted Therapy
2.1.1. Passive Targeting
2.1.2. Active Targeting
2.1.3. Stimuli-Responsive Nanomaterials
2.2. Nanoparticles for Immunotherapy
2.2.1. Immune Checkpoint Blockade
2.2.2. Adoptive T Cell Therapy
2.2.3. Cytokine-Based Drugs
2.2.4. Cancer Vaccines
2.3. Photothermal and Photodynamic Therapy
2.3.1. Photothermal Therapy
2.3.2. Photodynamic Therapy
2.3.3. Combination Therapy
3. Uveal Melanoma
3.1. Nanoparticles for Drug Delivery
3.2. Nanoparticles for Radiotherapy
3.3. Nanoparticles for PDT and PTT
3.4. Nanoparticles for Imaging
4. Mucosal Melanoma
5. Discussion
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
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]
- Bolick, N.L.; Geller, A.C. Epidemiology of Melanoma. Hematol. Oncol. Clin. N. Am. 2021, 35, 57–72. [Google Scholar] [CrossRef] [PubMed]
- Caini, S.; Gandini, S.; Sera, F.; Raimondi, S.; Fargnoli, M.C.; Boniol, M.; Armstrong, B.K. Meta-analysis of risk factors for cutaneous melanoma according to anatomical site and clinico-pathological variant. Eur. J. Cancer 2009, 45, 3054–3063. [Google Scholar] [CrossRef] [PubMed]
- Elder, D.E.; Bastian, B.C.; Cree, I.A.; Massi, D.; Scolyer, R.A. The 2018 World Health Organization Classification of Cutaneous, Mucosal, and Uveal Melanoma: Detailed Analysis of 9 Distinct Subtypes Defined by Their Evolutionary Pathway. Arch. Pathol. Lab. Med. 2020, 144, 500–522. [Google Scholar] [CrossRef]
- Rabbie, R.; Ferguson, P.; Molina-Aguilar, C.; Adams, D.J.; Robles-Espinoza, C.D. Melanoma subtypes: Genomic profiles, prognostic molecular markers and therapeutic possibilities. J. Pathol. 2019, 247, 539–551. [Google Scholar] [CrossRef]
- Cress, R.D.; Holly, E.A. Incidence of cutaneous melanoma among non-Hispanic whites, Hispanics, Asians, and blacks: An analysis of california cancer registry data, 1988–1993. Cancer Causes Control 1997, 8, 246–252. [Google Scholar] [CrossRef]
- Raigani, S.; Cohen, S.; Boland, G.M. The Role of Surgery for Melanoma in an Era of Effective Systemic Therapy. Curr. Oncol. Rep. 2017, 19, 17. [Google Scholar] [CrossRef]
- Seth, R.; Messersmith, H.; Kaur, V.; Kirkwood, J.M.; Kudchadkar, R.; McQuade, J.L.; Provenzano, A.; Swami, U.; Weber, J.; Alluri, K.C.; et al. Systemic Therapy for Melanoma: ASCO Guideline. J. Clin. Oncol. 2020, 38, 3947–3970. [Google Scholar] [CrossRef]
- Gershenwald, J.E.; Scolyer, R.A.; Hess, K.R.; Sondak, V.K.; Long, G.V.; Ross, M.I.; Lazar, A.J.; Faries, M.B.; Kirkwood, J.M.; McArthur, G.A.; et al. Melanoma staging: Evidence-based changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer J. Clin. 2017, 67, 472–492. [Google Scholar] [CrossRef]
- Parveen, S.; Misra, R.; Sahoo, S.K. Nanoparticles: A boon to drug delivery, therapeutics, diagnostics and imaging. Nanomedicine 2012, 8, 147–166. [Google Scholar] [CrossRef]
- Gharpure, K.M.; Wu, S.Y.; Li, C.; Lopez-Berestein, G.; Sood, A.K. Nanotechnology: Future of Oncotherapy. Clin. Cancer Res. 2015, 21, 3121–3130. [Google Scholar] [CrossRef] [PubMed]
- Eddy, K.; Chen, S. Overcoming Immune Evasion in Melanoma. Int. J. Mol. Sci. 2020, 21, 8984. [Google Scholar] [CrossRef] [PubMed]
- Schadendorf, D.; van Akkooi, A.C.J.; Berking, C.; Griewank, K.G.; Gutzmer, R.; Hauschild, A.; Stang, A.; Roesch, A.; Ugurel, S. Melanoma. Lancet 2018, 392, 971–984. [Google Scholar] [CrossRef]
- Lawrence, M.S.; Stojanov, P.; Polak, P.; Kryukov, G.V.; Cibulskis, K.; Sivachenko, A.; Carter, S.L.; Stewart, C.; Mermel, C.H.; Roberts, S.A.; et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 2013, 499, 214–218. [Google Scholar] [CrossRef]
- Naidoo, C.; Kruger, C.A.; Abrahamse, H. Photodynamic Therapy for Metastatic Melanoma Treatment: A Review. Technol. Cancer Res. Treat. 2018, 17, 1533033818791795. [Google Scholar] [CrossRef]
- Sahu, T.; Ratre, Y.K.; Chauhan, S.; Bhaskar, L.V.K.S.; Nair, M.P.; Verma, H.K. Nanotechnology based drug delivery system: Current strategies and emerging therapeutic potential for medical science. J. Drug Deliv. Sci. Technol. 2021, 63, 102487. [Google Scholar] [CrossRef]
- Jeong, K.; Kang, C.S.; Kim, Y.; Lee, Y.D.; Kwon, I.C.; Kim, S. Development of highly efficient nanocarrier-mediated delivery approaches for cancer therapy. Cancer Lett. 2016, 374, 31–43. [Google Scholar] [CrossRef]
- Kumbham, S.; Paul, M.; Itoo, A.; Ghosh, B.; Biswas, S. Oleanolic acid-conjugated human serum albumin nanoparticles encapsulating doxorubicin as synergistic combination chemotherapy in oropharyngeal carcinoma and melanoma. Int. J. Pharm. 2022, 614, 121479. [Google Scholar] [CrossRef]
- Ashley, C.E.; Carnes, E.C.; Phillips, G.K.; Padilla, D.; Durfee, P.N.; Brown, P.A.; Hanna, T.N.; Liu, J.; Phillips, B.; Carter, M.B.; et al. The targeted delivery of multicomponent cargos to cancer cells by nanoporous particle-supported lipid bilayers. Nat. Mater. 2011, 10, 389–397. [Google Scholar] [CrossRef]
- Li, Y.; Chen, X.; Ji, J.; Li, L.; Zhai, G. Redox-responsive nanoparticles based on Chondroitin Sulfate and Docetaxel prodrug for tumor targeted delivery of Docetaxel. Carbohydr. Polym. 2021, 255, 117393. [Google Scholar] [CrossRef]
- Matsumura, Y.; Maeda, H. A new concept for macromolecular therapeutics in cancer chemotherapy: Mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res. 1986, 46, 6387–6392. [Google Scholar] [PubMed]
- Sindhwani, S.; Syed, A.M.; Ngai, J.; Kingston, B.R.; Maiorino, L.; Rothschild, J.; MacMillan, P.; Zhang, Y.; Rajesh, N.U.; Hoang, T.; et al. The entry of nanoparticles into solid tumours. Nat. Mater. 2020, 19, 566–575. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Wang, Y.; Liang, R.; An, X.; Wang, K.; Shen, G.; Tu, Y.; Zhu, J.; Tao, J. Recent advances in targeted nanoparticles drug delivery to melanoma. Nanomedicine 2015, 11, 769–794. [Google Scholar] [CrossRef] [PubMed]
- Makhani, E.Y.; Zhang, A.; Haun, J.B. Quantifying and controlling bond multivalency for advanced nanoparticle targeting to cells. Nano Converg. 2021, 8, 38. [Google Scholar] [CrossRef]
- Shi, J.; Kantoff, P.W.; Wooster, R.; Farokhzad, O.C. Cancer nanomedicine: Progress, challenges and opportunities. Nat. Rev. Cancer 2017, 17, 20–37. [Google Scholar] [CrossRef]
- Wilhelm, S.; Tavares, A.J.; Dai, Q.; Ohta, S.; Audet, J.; Dvorak, H.F.; Chan, W.C.W. Analysis of nanoparticle delivery to tumours. Nat. Rev. Mater. 2016, 1, 16014. [Google Scholar] [CrossRef]
- Sadauskas, E.; Wallin, H.; Stoltenberg, M.; Vogel, U.; Doering, P.; Larsen, A.; Danscher, G. Kupffer cells are central in the removal of nanoparticles from the organism. Part. Fibre Toxicol. 2007, 4, 10. [Google Scholar] [CrossRef]
- Scott, C.C.; Vacca, F.; Gruenberg, J. Endosome maturation, transport and functions. Semin. Cell Dev. Biol. 2014, 31, 2–10. [Google Scholar] [CrossRef]
- Rosenblum, D.; Joshi, N.; Tao, W.; Karp, J.M.; Peer, D. Progress and challenges towards targeted delivery of cancer therapeutics. Nat. Commun. 2018, 9, 1410. [Google Scholar] [CrossRef]
- Chauhan, V.P.; Jain, R.K. Strategies for advancing cancer nanomedicine. Nat. Mater. 2013, 12, 958–962. [Google Scholar] [CrossRef] [Green Version]
- Lee, H.; Hoang, B.; Fonge, H.; Reilly, R.M.; Allen, C. In Vivo Distribution of Polymeric Nanoparticles at the Whole-Body, Tumor, and Cellular Levels. Pharm. Res. 2010, 27, 2343–2355. [Google Scholar] [CrossRef] [PubMed]
- Tang, L.; Fan, T.M.; Borst, L.B.; Cheng, J. Synthesis and Biological Response of Size-Specific, Monodisperse Drug–Silica Nanoconjugates. ACS Nano 2012, 6, 3954–3966. [Google Scholar] [CrossRef] [PubMed]
- Toy, R.; Peiris, P.M.; Ghaghada, K.B.; Karathanasis, E. Shaping cancer nanomedicine: The effect of particle shape on the in vivo journey of nanoparticles. Nanomedicine 2013, 9, 121–134. [Google Scholar] [CrossRef] [PubMed]
- Anselmo, A.C.; Zhang, M.; Kumar, S.; Vogus, D.R.; Menegatti, S.; Helgeson, M.E.; Mitragotri, S. Elasticity of Nanoparticles Influences Their Blood Circulation, Phagocytosis, Endocytosis, and Targeting. ACS Nano 2015, 9, 3169–3177. [Google Scholar] [CrossRef]
- van Vlerken, L.E.; Duan, Z.; Little, S.R.; Seiden, M.V.; Amiji, M.M. Biodistribution and Pharmacokinetic Analysis of Paclitaxel and Ceramide Administered in Multifunctional Polymer-Blend Nanoparticles in Drug Resistant Breast Cancer Model. Mol. Pharm. 2008, 5, 516–526. [Google Scholar] [CrossRef]
- Hu Che-Ming, J.; Zhang, L.; Aryal, S.; Cheung, C.; Fang Ronnie, H.; Zhang, L. Erythrocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc. Natl. Acad. Sci. USA 2011, 108, 10980–10985. [Google Scholar] [CrossRef]
- Aryal, S.; Hu, C.-M.J.; Fang, R.H.; Dehaini, D.; Carpenter, C.; Zhang, D.-E.; Zhang, L. Erythrocyte membrane-cloaked polymeric nanoparticles for controlled drug loading and release. Nanomedicine 2013, 8, 1271–1280. [Google Scholar] [CrossRef]
- Hu, C.-M.J.; Fang, R.H.; Luk, B.T.; Chen, K.N.H.; Carpenter, C.; Gao, W.; Zhang, K.; Zhang, L. ‘Marker-of-self’ functionalization of nanoscale particles through a top-down cellular membrane coating approach. Nanoscale 2013, 5, 2664–2668. [Google Scholar] [CrossRef]
- Song, M.; Dong, S.; An, X.; Zhang, W.; Shen, N.; Li, Y.; Guo, C.; Liu, C.; Li, X.; Chen, S. Erythrocyte-biomimetic nanosystems to improve antitumor effects of paclitaxel on epithelial cancers. J. Control. Release 2022, 345, 744–754. [Google Scholar] [CrossRef]
- Zhou, X.; Yu, R.; Cao, X.; Zhang, Z.R.; Deng, L. Bio-Mimicking Nanoparticles for Targeted Therapy of Malignant Melanoma. J. Biomed. Nanotechnol. 2019, 15, 993–1004. [Google Scholar] [CrossRef]
- Zhao, P.; Qiu, L.; Zhou, S.; Li, L.; Qian, Z.; Zhang, H. Cancer Cell Membrane Camouflaged Mesoporous Silica Nanoparticles Combined with Immune Checkpoint Blockade for Regulating Tumor Microenvironment and Enhancing Antitumor Therapy. Int. J. Nanomed. 2021, 16, 2107–2121. [Google Scholar] [CrossRef]
- Filipczak, N.; Pan, J.; Yalamarty, S.S.K.; Torchilin, V.P. Recent advancements in liposome technology. Adv. Drug Deliv. Rev. 2020, 156, 4–22. [Google Scholar] [CrossRef]
- Vecchione, R.; Quagliariello, V.; Giustetto, P.; Calabria, D.; Sathya, A.; Marotta, R.; Profeta, M.; Nitti, S.; Silvestri, N.; Pellegrino, T.; et al. Oil/water nano-emulsion loaded with cobalt ferrite oxide nanocubes for photo-acoustic and magnetic resonance dual imaging in cancer: In vitro and preclinical studies. Nanomedicine 2017, 13, 275–286. [Google Scholar] [CrossRef] [PubMed]
- Sercombe, L.; Veerati, T.; Moheimani, F.; Wu, S.Y.; Sood, A.K.; Hua, S. Advances and Challenges of Liposome Assisted Drug Delivery. Front. Pharmacol. 2015, 6, 286. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Xie, X.; Wang, W.; Jiang, S.; Mei, W.; Zhang, Y.; Liu, S.; Yu, X. Choline phosphate lipid as an intra-crosslinker in liposomes for drug and antibody delivery under guard. Nanoscale 2022, 14, 2277–2286. [Google Scholar] [CrossRef]
- Wang, K.; Jiang, H.; Li, W.; Qiang, M.; Dong, T.; Li, H. Role of Vitamin C in Skin Diseases. Front. Physiol. 2018, 9, 819. [Google Scholar] [CrossRef] [PubMed]
- Berretta, M.; Quagliariello, V.; Maurea, N.; Di Francia, R.; Sharifi, S.; Facchini, G.; Rinaldi, L.; Piezzo, M.; Manuela, C.; Nunnari, G.; et al. Multiple Effects of Ascorbic Acid against Chronic Diseases: Updated Evidence from Preclinical and Clinical Studies. Antioxidants 2020, 9, 1182. [Google Scholar] [CrossRef]
- Cha, J.; Roomi, M.W.; Ivanov, V.; Kalinovsky, T.; Niedzwiecki, A.; Rath, M. Ascorbate supplementation inhibits growth and metastasis of B16FO melanoma and 4T1 breast cancer cells in vitamin C-deficient mice. Int. J. Oncol. 2013, 42, 55–64. [Google Scholar] [CrossRef]
- Li, J.; Guo, C.; Feng, F.; Fan, A.; Dai, Y.; Li, N.; Zhao, D.; Chen, X.; Lu, Y. Co-delivery of docetaxel and palmitoyl ascorbate by liposome for enhanced synergistic antitumor efficacy. Sci. Rep. 2016, 6, 38787. [Google Scholar] [CrossRef]
- Sawant, R.R.; Vaze, O.S.; Rockwell, K.; Torchilin, V.P. Palmitoyl ascorbate-modified liposomes as nanoparticle platform for ascorbate-mediated cytotoxicity and paclitaxel co-delivery. Eur. J. Pharm. Biopharm. 2010, 75, 321–326. [Google Scholar] [CrossRef]
- Zhou, M.; Li, X.; Li, Y.; Yao, Q.; Ming, Y.; Li, Z.; Lu, L.; Shi, S. Ascorbyl palmitate-incorporated paclitaxel-loaded composite nanoparticles for synergistic anti-tumoral therapy. Drug Deliv. 2017, 24, 1230–1242. [Google Scholar] [CrossRef] [PubMed]
- Langer, R. Drug delivery and targeting. Nature 1998, 392, 5–10. [Google Scholar] [PubMed]
- Guo, J.; Gao, X.; Su, L.; Xia, H.; Gu, G.; Pang, Z.; Jiang, X.; Yao, L.; Chen, J.; Chen, H. Aptamer-functionalized PEG-PLGA nanoparticles for enhanced anti-glioma drug delivery. Biomaterials 2011, 32, 8010–8020. [Google Scholar] [CrossRef] [PubMed]
- Barjasteh, M.; Vossoughi, M.; Bagherzadeh, M.; Pooshang Bagheri, K. Green synthesis of PEG-coated MIL-100(Fe) for controlled release of dacarbazine and its anticancer potential against human melanoma cells. Int. J. Pharm. 2022, 618, 121647. [Google Scholar] [CrossRef]
- Arfin, S.M.; Kendall, R.L.; Hall, L.; Weaver, L.H.; Stewart, A.E.; Matthews, B.W.; Bradshaw, R.A. Eukaryotic methionyl aminopeptidases: Two classes of cobalt-dependent enzymes. Proc. Natl. Acad. Sci. USA 1995, 92, 7714–7718. [Google Scholar] [CrossRef]
- Esa, R.; Steinberg, E.; Dagan, A.; Yekhtin, Z.; Tischenko, K.; Benny, O. Newly synthesized methionine aminopeptidase 2 inhibitor hinders tumor growth. Drug Deliv. Transl. Res. 2022. [Google Scholar] [CrossRef]
- Malaguarnera, L. Influence of Resveratrol on the Immune Response. Nutrients 2019, 11, 946. [Google Scholar] [CrossRef]
- Yee, Y.J.; Benson, H.A.E.; Dass, C.R.; Chen, Y. Evaluation of novel conjugated resveratrol polymeric nanoparticles in reduction of plasma degradation, hepatic metabolism and its augmentation of anticancer activity in vitro and in vivo. Int. J. Pharm. 2022, 615, 121499. [Google Scholar] [CrossRef]
- Tang, F.; Li, L.; Chen, D. Mesoporous silica nanoparticles: Synthesis, biocompatibility and drug delivery. Adv. Mater. 2012, 24, 1504–1534. [Google Scholar] [CrossRef]
- Vallet-Regí, M.; Schüth, F.; Lozano, D.; Colilla, M.; Manzano, M. Engineering mesoporous silica nanoparticles for drug delivery: Where are we after two decades? Chem. Soc. Rev. 2022, 51, 5365–5451. [Google Scholar] [CrossRef]
- Ferreira, N.H.; Ribeiro, A.B.; Rinaldi-Neto, F.; Fernandes, F.S.; do Nascimento, S.; Braz, W.R.; Nassar, E.J.; Tavares, D.C. Anti-Melanoma Activity of Indomethacin Incorporated into Mesoporous Silica Nanoparticles. Pharm. Res. 2020, 37, 172. [Google Scholar] [CrossRef] [PubMed]
- Liu, Q.; Wang, C.; Zheng, Y.; Zhao, Y.; Wang, Y.; Hao, J.; Zhao, X.; Yi, K.; Shi, L.; Kang, C.; et al. Virus-like nanoparticle as a co-delivery system to enhance efficacy of CRISPR/Cas9-based cancer immunotherapy. Biomaterials 2020, 258, 120275. [Google Scholar] [CrossRef] [PubMed]
- Skrott, Z.; Mistrik, M.; Andersen, K.K.; Friis, S.; Majera, D.; Gursky, J.; Ozdian, T.; Bartkova, J.; Turi, Z.; Moudry, P.; et al. Alcohol-abuse drug disulfiram targets cancer via p97 segregase adaptor NPL4. Nature 2017, 552, 194–199. [Google Scholar] [CrossRef]
- Oakes, S.A. Endoplasmic Reticulum Stress Signaling in Cancer Cells. Am. J. Pathol. 2020, 190, 934–946. [Google Scholar] [CrossRef] [PubMed]
- Paun, R.A.; Dumut, D.C.; Centorame, A.; Thuraisingam, T.; Hajduch, M.; Mistrik, M.; Dzubak, P.; De Sanctis, J.B.; Radzioch, D.; Tabrizian, M. One-Step Synthesis of Nanoliposomal Copper Diethyldithiocarbamate and Its Assessment for Cancer Therapy. Pharmaceutics 2022, 14, 640. [Google Scholar] [CrossRef]
- Kim, J.H.; Moon, M.J.; Kim, D.Y.; Heo, S.H.; Jeong, Y.Y. Hyaluronic Acid-Based Nanomaterials for Cancer Therapy. Polymers 2018, 10, 1133. [Google Scholar] [CrossRef]
- Mishra, H.; Mishra, P.K.; Iqbal, Z.; Jaggi, M.; Madaan, A.; Bhuyan, K.; Gupta, N.; Gupta, N.; Vats, K.; Verma, R.; et al. Co-Delivery of Eugenol and Dacarbazine by Hyaluronic Acid-Coated Liposomes for Targeted Inhibition of Survivin in Treatment of Resistant Metastatic Melanoma. Pharmaceutics 2019, 11, 163. [Google Scholar] [CrossRef]
- Zhang, C.; Xie, C.; Lu, Y. Local Anesthetic Lidocaine and Cancer: Insight Into Tumor Progression and Recurrence. Front. Oncol. 2021, 11, 669746. [Google Scholar] [CrossRef]
- Liu, H.; Dilger, J.P.; Lin, J. Effects of local anesthetics on cancer cells. Pharmacol. Ther. 2020, 212, 107558. [Google Scholar] [CrossRef]
- Peng, F.; Zhang, W.; Qiu, F. Self-assembling Peptides in Current Nanomedicine: Versatile Nanomaterials for Drug Delivery. Curr. Med. Chem. 2020, 27, 4855–4881. [Google Scholar] [CrossRef]
- Yang, Y.; Sun, J.; Peng, F.; Liu, H.; Zhao, G.; Chen, J.; Zhang, W.; Qiu, F. Enhanced Antitumor Activity of Lidocaine Nanoparticles Encapsulated by a Self-Assembling Peptide. Front. Pharmacol. 2022, 13, 770892. [Google Scholar] [CrossRef] [PubMed]
- Hu, L.; Yang, X.; Yin, J.; Rong, X.; Huang, X.; Yu, P.; He, Z.; Liu, Y. Combination of AgNPs and Domiphen is Antimicrobial Against Biofilms of Common Pathogens. Int. J. Nanomed. 2021, 16, 7181–7194. [Google Scholar] [CrossRef] [PubMed]
- Barbasz, A.; Czyżowska, A.; Piergies, N.; Oćwieja, M. Design cytotoxicity: The effect of silver nanoparticles stabilized by selected antioxidants on melanoma cells. J. Appl. Toxicol. 2022, 42, 570–587. [Google Scholar] [CrossRef] [PubMed]
- Bertrand, N.; Wu, J.; Xu, X.; Kamaly, N.; Farokhzad, O.C. Cancer nanotechnology: The impact of passive and active targeting in the era of modern cancer biology. Adv. Drug Deliv. Rev. 2014, 66, 2–25. [Google Scholar] [CrossRef]
- Florence, A.T. “Targeting” nanoparticles: The constraints of physical laws and physical barriers. J. Control. Release 2012, 164, 115–124. [Google Scholar] [CrossRef]
- Kirpotin, D.B.; Drummond, D.C.; Shao, Y.; Shalaby, M.R.; Hong, K.; Nielsen, U.B.; Marks, J.D.; Benz, C.C.; Park, J.W. Antibody Targeting of Long-Circulating Lipidic Nanoparticles Does Not Increase Tumor Localization but Does Increase Internalization in Animal Models. Cancer Res. 2006, 66, 6732–6740. [Google Scholar] [CrossRef]
- Xiong, W.; Guo, Z.; Zeng, B.; Wang, T.; Zeng, X.; Cao, W.; Lian, D. Dacarbazine-Loaded Targeted Polymeric Nanoparticles for Enhancing Malignant Melanoma Therapy. Front. Bioeng. Biotechnol. 2022, 10, 847901. [Google Scholar] [CrossRef]
- Liu, Y.; Tao, J.; Li, Y.; Yang, J.; Yu, Y.; Wang, M.; Xu, X.; Huang, C.; Huang, W.; Dong, J.; et al. Targeting hypoxia-inducible factor-1alpha with Tf-PEI-shRNA complex via transferrin receptor-mediated endocytosis inhibits melanoma growth. Mol. Ther. 2009, 17, 269–277. [Google Scholar] [CrossRef]
- Filipczak, N.; Jaromin, A.; Piwoni, A.; Mahmud, M.; Sarisozen, C.; Torchilin, V.; Gubernator, J. A Triple Co-Delivery Liposomal Carrier That Enhances Apoptosis via an Intrinsic Pathway in Melanoma Cells. Cancers 2019, 11, 1982. [Google Scholar] [CrossRef]
- Sakpakdeejaroen, I.; Somani, S.; Laskar, P.; Mullin, M.; Dufès, C. Regression of Melanoma Following Intravenous Injection of Plumbagin Entrapped in Transferrin-Conjugated, Lipid-Polymer Hybrid Nanoparticles. Int. J. Nanomed. 2021, 16, 2615–2631. [Google Scholar] [CrossRef]
- Yuan, M.; Qiu, Y.; Zhang, L.; Gao, H.; He, Q. Targeted delivery of transferrin and TAT co-modified liposomes encapsulating both paclitaxel and doxorubicin for melanoma. Drug Deliv. 2016, 23, 1171–1183. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Tezcan, O.; Li, D.; Beztsinna, N.; Lou, B.; Etrych, T.; Ulbrich, K.; Metselaar, J.M.; Lammers, T.; Hennink, W.E. Overcoming multidrug resistance using folate receptor-targeted and pH-responsive polymeric nanogels containing covalently entrapped doxorubicin. Nanoscale 2017, 9, 10404–10419. [Google Scholar] [CrossRef] [PubMed]
- Elechalawar, C.K.; Sridharan, K.; Pal, A.; Ahmed, M.T.; Yousuf, M.; Adhikari, S.S.; Banerjee, R. Cationic folate-mediated liposomal delivery of bis-arylidene oxindole induces efficient melanoma tumor regression. Biomater. Sci. 2017, 5, 1898–1909. [Google Scholar] [CrossRef] [PubMed]
- Qiu, Y.; Ren, K.; Zhao, W.; Yu, Q.; Guo, R.; He, J.; Mei, L.; Liu, Y.; Tang, J.; Xu, S.; et al. A “dual-guide” bioinspired drug delivery strategy of a macrophage-based carrier against postoperative triple-negative breast cancer recurrence. J. Control. Release 2021, 329, 191–204. [Google Scholar] [CrossRef] [PubMed]
- Ukidve, A.; Zhao, Z.; Fehnel, A.; Krishnan, V.; Pan, D.C.; Gao, Y.; Mandal, A.; Muzykantov, V.; Mitragotri, S. Erythrocyte-driven immunization via biomimicry of their natural antigen-presenting function. Proc. Natl. Acad. Sci. USA 2020, 117, 17727–17736. [Google Scholar] [CrossRef]
- Saber, S.H.; Ali, H.E.A.; Gaballa, R.; Gaballah, M.; Ali, H.I.; Zerfaoui, M.; Abd Elmageed, Z.Y. Exosomes are the Driving Force in Preparing the Soil for the Metastatic Seeds: Lessons from the Prostate Cancer. Cells 2020, 9, 564. [Google Scholar] [CrossRef]
- Whiteside, T.L. Tumor-Derived Exosomes and Their Role in Cancer Progression. Adv. Clin. Chem. 2016, 74, 103–141. [Google Scholar] [CrossRef]
- Kotmakçı, M.; Bozok Çetintaş, V. Extracellular Vesicles as Natural Nanosized Delivery Systems for Small-Molecule Drugs and Genetic Material: Steps towards the Future Nanomedicines. J. Pharm. Pharm. Sci. 2015, 18, 396–413. [Google Scholar] [CrossRef]
- Patras, L.; Ionescu, A.E.; Munteanu, C.; Hajdu, R.; Kosa, A.; Porfire, A.; Licarete, E.; Rauca, V.F.; Sesarman, A.; Luput, L.; et al. Trojan horse treatment based on PEG-coated extracellular vesicles to deliver doxorubicin to melanoma in vitro and in vivo. Cancer Biol. Ther. 2022, 23, 1–16. [Google Scholar] [CrossRef]
- Lanza, R.; Russell, D.W.; Nagy, A. Engineering universal cells that evade immune detection. Nat. Rev. Immunol. 2019, 19, 723–733. [Google Scholar] [CrossRef]
- Gao, C.; Wang, Q.; Li, J.; Kwong, C.H.T.; Wei, J.; Xie, B.; Lu, S.; Lee, S.M.Y.; Wang, R. In vivo hitchhiking of immune cells by intracellular self-assembly of bacteria-mimetic nanomedicine for targeted therapy of melanoma. Sci. Adv. 2022, 8, eabn1805. [Google Scholar] [CrossRef] [PubMed]
- He, W.; Zhang, Z.; Yang, W.; Zheng, X.; You, W.; Yao, Y.; Yan, J.; Liu, W. Turing milk into pro-apoptotic oral nanotherapeutic: De novo bionic chiral-peptide supramolecule for cancer targeted and immunological therapy. Theranostics 2022, 12, 2322–2334. [Google Scholar] [CrossRef] [PubMed]
- Herringson, T.P.; Altin, J.G. Effective tumor targeting and enhanced anti-tumor effect of liposomes engrafted with peptides specific for tumor lymphatics and vasculature. Int. J. Pharm. 2011, 411, 206–214. [Google Scholar] [CrossRef] [PubMed]
- Altin, J.G.; Zhao, Y. Using peptides to promote delivery and improve anti-tumour efficacy of liposomal drug. J. Drug Target. 2022, 30, 544–556. [Google Scholar] [CrossRef]
- Corbo, C.; Molinaro, R.; Parodi, A.; Toledano Furman, N.E.; Salvatore, F.; Tasciotti, E. The impact of nanoparticle protein corona on cytotoxicity, immunotoxicity and target drug delivery. Nanomedicine 2015, 11, 81–100. [Google Scholar] [CrossRef] [PubMed]
- Meewan, J.; Somani, S.; Laskar, P.; Irving, C.; Mullin, M.; Woods, S.; Roberts, C.W.; Alzahrani, A.R.; Ferro, V.A.; McGill, S.; et al. Limited Impact of the Protein Corona on the Cellular Uptake of PEGylated Zein Micelles by Melanoma Cancer Cells. Pharmaceutics 2022, 14, 439. [Google Scholar] [CrossRef]
- Feng, X.; Liu, H.; Pan, J.; Xiong, Y.; Zhu, X.; Yan, X.; Duan, Y.; Huang, Y. Liposome-Encapsulated Tiancimycin A Is Active against Melanoma and Metastatic Breast Tumors: The Effect of cRGD Modification of the Liposomal Carrier and Tiancimycin A Dose on Drug Activity and Toxicity. Mol. Pharm. 2022, 19, 1078–1090. [Google Scholar] [CrossRef]
- Zhou, G.; Jin, X.; Zhu, P.; Yao, J.U.; Zhang, Y.; Teng, L.; Lee, R.J.; Zhang, X.; Hong, W. Human Serum Albumin Nanoparticles as a Novel Delivery System for Cabazitaxel. Anticancer Res. 2016, 36, 1649–1656. [Google Scholar]
- Mandalà, M.; Voit, C. Targeting BRAF in melanoma: Biological and clinical challenges. Crit. Rev. Oncol. Hematol. 2013, 87, 239–255. [Google Scholar] [CrossRef]
- Imlimthan, S.; Khng, Y.C.; Keinänen, O.; Zhang, W.; Airaksinen, A.J.; Kostiainen, M.A.; Zeglis, B.M.; Santos, H.A.; Sarparanta, M. A Theranostic Cellulose Nanocrystal-Based Drug Delivery System with Enhanced Retention in Pulmonary Metastasis of Melanoma. Small 2021, 17, e2007705. [Google Scholar] [CrossRef]
- Tran, M.A.; Gowda, R.; Sharma, A.; Park, E.J.; Adair, J.; Kester, M.; Smith, N.B.; Robertson, G.P. Targeting V600EB-Raf and Akt3 using nanoliposomal-small interfering RNA inhibits cutaneous melanocytic lesion development. Cancer Res. 2008, 68, 7638–7649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mi, Y.; Mu, C.; Wolfram, J.; Deng, Z.; Hu, T.Y.; Liu, X.; Blanco, E.; Shen, H.; Ferrari, M. A Micro/Nano Composite for Combination Treatment of Melanoma Lung Metastasis. Adv. Healthc. Mater. 2016, 5, 936–946. [Google Scholar] [CrossRef] [PubMed]
- Tham, H.P.; Xu, K.; Lim, W.Q.; Chen, H.; Zheng, M.; Thng, T.G.S.; Venkatraman, S.S.; Xu, C.; Zhao, Y. Microneedle-Assisted Topical Delivery of Photodynamically Active Mesoporous Formulation for Combination Therapy of Deep-Seated Melanoma. ACS Nano 2018, 12, 11936–11948. [Google Scholar] [CrossRef] [PubMed]
- Fu, Y.; Rathod, D.; Patel, K. Protein kinase C inhibitor anchored BRD4 PROTAC PEGylated nanoliposomes for the treatment of vemurafenib-resistant melanoma. Exp. Cell Res. 2020, 396, 112275. [Google Scholar] [CrossRef] [PubMed]
- Yatvin Milton, B.; Weinstein John, N.; Dennis Warren, H.; Blumenthal, R. Design of Liposomes for Enhanced Local Release of Drugs by Hyperthermia. Science 1978, 202, 1290–1293. [Google Scholar] [CrossRef]
- Mura, S.; Nicolas, J.; Couvreur, P. Stimuli-responsive nanocarriers for drug delivery. Nat. Mater. 2013, 12, 991–1003. [Google Scholar] [CrossRef]
- Di Cicco, C.; Vecchione, R.; Quagliariello, V.; Busato, A.; Tufano, I.; Bedini, E.; Gerosa, M.; Sbarbati, A.; Boschi, F.; Marzola, P.; et al. Biocompatible, photo-responsive layer-by-layer polymer nanocapsules with an oil core: In vitro and in vivo study. J. R. Soc. Interface 2022, 19, 20210800. [Google Scholar] [CrossRef]
- Yuan, H.; Zhang, L.; Ma, T.; Huang, J.; Nie, C.; Cao, S.; Xiang, X.; Ma, L.; Cheng, C.; Qiu, L. Spiky Cascade Biocatalysts as Peroxisome-Mimics for Ultrasound-Augmented Tumor Ablation. ACS Appl. Mater. Interfaces 2022, 14, 15970–15981. [Google Scholar] [CrossRef]
- Lee, J.; Wang, Y.; Xue, C.; Chen, Y.; Qu, M.; Thakor, J.; Zhou, X.; Barros, N.R.; Falcone, N.; Young, P.; et al. pH-Responsive doxorubicin delivery using shear-thinning biomaterials for localized melanoma treatment. Nanoscale 2022, 14, 350–360. [Google Scholar] [CrossRef]
- Boedtkjer, E.; Pedersen, S.F. The Acidic Tumor Microenvironment as a Driver of Cancer. Annu. Rev. Physiol. 2020, 82, 103–126. [Google Scholar] [CrossRef]
- Tomás, H.; Alves, C.S.; Rodrigues, J. Laponite®: A key nanoplatform for biomedical applications? Nanomed. Nanotechnol. Biol. Med. 2018, 14, 2407–2420. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Wu, Y.; Guo, R.; Huang, Y.; Wen, S.; Shen, M.; Wang, J.; Shi, X. Laponite nanodisks as an efficient platform for Doxorubicin delivery to cancer cells. Langmuir 2013, 29, 5030–5036. [Google Scholar] [CrossRef]
- Pourmanouchehri, Z.; Ebrahimi, S.; Limoee, M.; Jalilian, F.; Janfaza, S.; Vosoughi, A.; Behbood, L. Controlled release of 5-fluorouracil to melanoma cells using a hydrogel/micelle composites based on deoxycholic acid and carboxymethyl chitosan. Int. J. Biol. Macromol. 2022, 206, 159–166. [Google Scholar] [CrossRef] [PubMed]
- Mellman, I.; Coukos, G.; Dranoff, G. Cancer immunotherapy comes of age. Nature 2011, 480, 480–489. [Google Scholar] [CrossRef]
- Heshmati Aghda, N.; Torres Hurtado, S.; Abdulsahib, S.M.; Lara, E.J.; Tunnell, J.W.; Betancourt, T. Dual Photothermal/Chemotherapy of Melanoma Cells with Albumin Nanoparticles Carrying Indocyanine Green and Doxorubicin Leads to Immunogenic Cell Death. Macromol. Biosci. 2022, 22, e2100353. [Google Scholar] [CrossRef] [PubMed]
- Leach, D.R.; Krummel, M.F.; Allison, J.P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 1996, 271, 1734–1736. [Google Scholar] [CrossRef]
- Okazaki, T.; Honjo, T. The PD-1-PD-L pathway in immunological tolerance. Trends Immunol. 2006, 27, 195–201. [Google Scholar] [CrossRef]
- Hogan, S.A.; Levesque, M.P.; Cheng, P.F. Melanoma immunotherapy: Next-generation biomarkers. Front. Oncol. 2018, 8, 178. [Google Scholar] [CrossRef]
- Marzagalli, M.; Ebelt, N.D.; Manuel, E.R. Unraveling the crosstalk between melanoma and immune cells in the tumor microenvironment. Semin. Cancer Biol. 2019, 59, 236–250. [Google Scholar] [CrossRef]
- Sheen, M.R.; Fiering, S. In situ vaccination: Harvesting low hanging fruit on the cancer immunotherapy tree. WIREs Nanomed. Nanobiotechnology 2019, 11, e1524. [Google Scholar] [CrossRef]
- Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef] [PubMed]
- Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.M.; Hwu, W.J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [PubMed]
- Tang, J.; Yu, J.X.; Hubbard-Lucey, V.M.; Neftelinov, S.T.; Hodge, J.P.; Lin, Y. The clinical trial landscape for PD1/PDl1 immune checkpoint inhibitors. Nat. Rev. Drug Discov. 2018, 17, 854–855. [Google Scholar] [CrossRef] [PubMed]
- Zou, W.; Wolchok Jedd, D.; Chen, L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci. Transl. Med. 2016, 8, 328rv324. [Google Scholar] [CrossRef]
- Munhoz, R.R.; Postow, M.A. Clinical Development of PD-1 in Advanced Melanoma. Cancer J. 2018, 24, 7–14. [Google Scholar] [CrossRef]
- Liu, B.; Cao, W.; Qiao, G.; Yao, S.; Pan, S.; Wang, L.; Yue, C.; Ma, L.; Liu, Y.; Cui, D. Effects of gold nanoprism-assisted human PD-L1 siRNA on both gene down-regulation and photothermal therapy on lung cancer. Acta Biomater. 2019, 99, 307–319. [Google Scholar] [CrossRef]
- Wei, S.; Shao, X.; Liu, Y.; Xiong, B.; Cui, P.; Liu, Z.; Li, Q. Genome editing of PD-L1 mediated by nucleobase-modified polyamidoamine for cancer immunotherapy. J. Mater. Chem. B 2022, 10, 1291–1300. [Google Scholar] [CrossRef]
- Kwak, G.; Kim, D.; Nam, G.H.; Wang, S.Y.; Kim, I.S.; Kim, S.H.; Kwon, I.C.; Yeo, Y. Programmed Cell Death Protein Ligand-1 Silencing with Polyethylenimine-Dermatan Sulfate Complex for Dual Inhibition of Melanoma Growth. ACS Nano 2017, 11, 10135–10146. [Google Scholar] [CrossRef]
- Rosenberg Steven, A.; Restifo Nicholas, P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015, 348, 62–68. [Google Scholar] [CrossRef]
- Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2020, 20, 7–24. [Google Scholar] [CrossRef]
- Xiong, X.; Zhao, J.; Su, R.; Liu, C.; Guo, X.; Zhou, S. Double enhancement of immunogenic cell death and antigen presentation for cancer immunotherapy. Nano Today 2021, 39, 101225. [Google Scholar] [CrossRef]
- Yang, G.; Xu, L.; Xu, J.; Zhang, R.; Song, G.; Chao, Y.; Feng, L.; Han, F.; Dong, Z.; Li, B.; et al. Smart Nanoreactors for pH-Responsive Tumor Homing, Mitochondria-Targeting, and Enhanced Photodynamic-Immunotherapy of Cancer. Nano Lett. 2018, 18, 2475–2484. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Li, L.; Lyu, J.; Xiang, Y.; Chen, L.; Zhou, Z.; Huang, Y. Split bullets loaded nanoparticles for amplified immunotherapy. J. Control. Release 2022, 347, 199–210. [Google Scholar] [CrossRef]
- Su, T.; Zhang, Y.; Valerie, K.; Wang, X.-Y.; Lin, S.; Zhu, G. STING activation in cancer immunotherapy. Theranostics 2019, 9, 7759–7771. [Google Scholar] [CrossRef]
- Corrales, L.; Glickman, L.H.; McWhirter, S.M.; Kanne, D.B.; Sivick, K.E.; Katibah, G.E.; Woo, S.R.; Lemmens, E.; Banda, T.; Leong, J.J.; et al. Direct Activation of STING in the Tumor Microenvironment Leads to Potent and Systemic Tumor Regression and Immunity. Cell Rep. 2015, 11, 1018–1030. [Google Scholar] [CrossRef]
- Zheng, H.; Guo, B.; Qiu, X.; Xia, Y.; Qu, Y.; Cheng, L.; Meng, F.; Zhong, Z. Polymersome-mediated cytosolic delivery of cyclic dinucleotide STING agonist enhances tumor immunotherapy. Bioact. Mater. 2022, 16, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Han, D.; Liu, J.; Chen, C.; Dong, L.; Liu, Y.; Chang, R.; Huang, X.; Liu, Y.; Wang, J.; Dougherty, U.; et al. Anti-tumour immunity controlled through mRNA m6A methylation and YTHDF1 in dendritic cells. Nature 2019, 566, 270–274. [Google Scholar] [CrossRef]
- Ouyang, Z.; Gao, Y.; Yang, R.; Shen, M.; Shi, X. Genetic Engineering of Dendritic Cells Using Partially Zwitterionic Dendrimer-Entrapped Gold Nanoparticles Boosts Efficient Tumor Immunotherapy. Biomacromolecules 2022, 23, 1326–1336. [Google Scholar] [CrossRef]
- Ichikawa, J.; Yoshida, T.; Isser, A.; Laino, A.S.; Vassallo, M.; Woods, D.; Kim, S.; Oelke, M.; Jones, K.; Schneck, J.P.; et al. Rapid Expansion of Highly Functional Antigen-Specific T Cells from Patients with Melanoma by Nanoscale Artificial Antigen-Presenting Cells. Clin. Cancer Res. 2020, 26, 3384–3396. [Google Scholar] [CrossRef]
- Song, R.; Li, T.; Ye, J.; Sun, F.; Hou, B.; Saeed, M.; Gao, J.; Wang, Y.; Zhu, Q.; Xu, Z.; et al. Acidity-Activatable Dynamic Nanoparticles Boosting Ferroptotic Cell Death for Immunotherapy of Cancer. Adv. Mater. 2021, 33, e2101155. [Google Scholar] [CrossRef]
- Kuang, X.; Wang, Z.; Luo, Z.; He, Z.; Liang, L.; Gao, Q.; Li, Y.; Xia, K.; Xie, Z.; Chang, R.; et al. Ag nanoparticles enhance immune checkpoint blockade efficacy by promoting of immune surveillance in melanoma. J. Colloid Interface Sci. 2022, 616, 189–200. [Google Scholar] [CrossRef]
- Liu, T.; Yan, J.; He, C.; You, W.; Ma, F.; Chang, Z.; Li, Y.; Han, S.; He, W.; Liu, W. A Tumor-Targeting Metal-Organic Nanoparticle Constructed by Dynamic Combinatorial Chemistry toward Accurately Redressing Carcinogenic Wnt Cascade. Small 2022, 18, e2104849. [Google Scholar] [CrossRef]
- Mullard, A. Restoring IL-2 to its cancer immunotherapy glory. Nat. Rev. Drug Discov. 2021, 20, 163–165. [Google Scholar] [CrossRef]
- Liu, J.Q.; Zhang, C.; Zhang, X.; Yan, J.; Zeng, C.; Talebian, F.; Lynch, K.; Zhao, W.; Hou, X.; Du, S.; et al. Intratumoral delivery of IL-12 and IL-27 mRNA using lipid nanoparticles for cancer immunotherapy. J. Control. Release 2022, 345, 306–313. [Google Scholar] [CrossRef]
- Lee, D.W.; Gardner, R.; Porter, D.L.; Louis, C.U.; Ahmed, N.; Jensen, M.; Grupp, S.A.; Mackall, C.L. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014, 124, 188–195. [Google Scholar] [CrossRef]
- Dimitriou, F.; Matter, A.V.; Mangana, J.; Urosevic-Maiwald, M.; Micaletto, S.; Braun, R.P.; French, L.E.; Dummer, R. Cytokine Release Syndrome During Sequential Treatment with Immune Checkpoint Inhibitors and Kinase Inhibitors for Metastatic Melanoma. J. Immunother. 2019, 42, 29–32. [Google Scholar] [CrossRef]
- Sivakumar, P.V.; Garcia, R.; Waggie, K.S.; Anderson-Haley, M.; Nelson, A.; Hughes, S.D. Comparison of vascular leak syndrome in mice treated with IL21 or IL2. Comp. Med. 2013, 63, 13–21. [Google Scholar]
- Kim, J.; Kang, S.; Kim, K.W.; Heo, M.G.; Park, D.I.; Lee, J.H.; Lim, N.J.; Min, D.H.; Won, C. Nanoparticle delivery of recombinant IL-2 (BALLkine-2) achieves durable tumor control with less systemic adverse effects in cancer immunotherapy. Biomaterials 2022, 280, 121257. [Google Scholar] [CrossRef]
- Chen, J.; Fang, H.; Hu, Y.; Wu, J.; Zhang, S.; Feng, Y.; Lin, L.; Tian, H.; Chen, X. Combining mannose receptor mediated nanovaccines and gene regulated PD-L1 blockade for boosting cancer immunotherapy. Bioact. Mater. 2022, 7, 167–180. [Google Scholar] [CrossRef]
- Nguyen, T.L.; Cha, B.G.; Choi, Y.; Im, J.; Kim, J. Injectable dual-scale mesoporous silica cancer vaccine enabling efficient delivery of antigen/adjuvant-loaded nanoparticles to dendritic cells recruited in local macroporous scaffold. Biomaterials 2020, 239, 119859. [Google Scholar] [CrossRef]
- Berti, C.; Graciotti, M.; Boarino, A.; Yakkala, C.; Kandalaft, L.E.; Klok, H.A. Polymer Nanoparticle-Mediated Delivery of Oxidized Tumor Lysate-Based Cancer Vaccines. Macromol. Biosci. 2022, 22, e2100356. [Google Scholar] [CrossRef] [PubMed]
- Lee, A.; Dadhwal, S.; Gamble, A.; Hook, S. Liposomes with cyclodextrin channels and polyethyleneimine (PEI) improves cytoplasmic vaccine delivery and induces anti-cancer immune activity in mice. J. Liposome Res. 2022, 32, 22–31. [Google Scholar] [CrossRef] [PubMed]
- Song, H.; Su, Q.; Shi, W.; Huang, P.; Zhang, C.; Zhang, C.; Liu, Q.; Wang, W. Antigen epitope-TLR7/8a conjugate as self-assembled carrier-free nanovaccine for personalized immunotherapy. Acta Biomater. 2022, 141, 398–407. [Google Scholar] [CrossRef]
- Song, X.; Jiang, Y.; Zhang, W.; Elfawal, G.; Wang, K.; Jiang, D.; Hong, H.; Wu, J.; He, C.; Mo, X.; et al. Transcutaneous tumor vaccination combined with anti-programmed death-1 monoclonal antibody treatment produces a synergistic antitumor effect. Acta Biomater. 2022, 140, 247–260. [Google Scholar] [CrossRef]
- Beiss, V.; Mao, C.; Fiering, S.N.; Steinmetz, N.F. Cowpea Mosaic Virus Outperforms Other Members of the Secoviridae as In Situ Vaccine for Cancer Immunotherapy. Mol. Pharm. 2022, 19, 1573–1585. [Google Scholar] [CrossRef]
- Gromeier, M.; Nair, S.K. Recombinant Poliovirus for Cancer Immunotherapy. Annu. Rev. Med. 2018, 69, 289–299. [Google Scholar] [CrossRef]
- Koudelka, K.J.; Destito, G.; Plummer, E.M.; Trauger, S.A.; Siuzdak, G.; Manchester, M. Endothelial targeting of cowpea mosaic virus (CPMV) via surface vimentin. PLoS Pathog. 2009, 5, e1000417. [Google Scholar] [CrossRef]
- Jiang, X.; Wang, J.; Zheng, X.; Liu, Z.; Zhang, X.; Li, Y.; Wilhelm, J.; Cao, J.; Huang, G.; Zhang, J.; et al. Intratumoral administration of STING-activating nanovaccine enhances T cell immunotherapy. J. Immunother. Cancer 2022, 10, e003960. [Google Scholar] [CrossRef]
- Jackson, N.A.C.; Kester, K.E.; Casimiro, D.; Gurunathan, S.; DeRosa, F. The promise of mRNA vaccines: A biotech and industrial perspective. NPJ Vaccines 2020, 5, 11. [Google Scholar] [CrossRef]
- Pardi, N.; Hogan, M.J.; Porter, F.W.; Weissman, D. mRNA vaccines—A new era in vaccinology. Nat. Rev. Drug Discov. 2018, 17, 261–279. [Google Scholar] [CrossRef]
- Li, L.; He, W.; You, W.; Yan, J.; Liu, W. Turing miRNA into infinite coordination supermolecule: A general and enabling nanoengineering strategy for resurrecting nuclear acid therapeutics. J. Nanobiotechnology 2022, 20, 10. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Ma, X.; Yue, Y.; Zhang, K.; Cheng, K.; Feng, Q.; Ma, N.; Liang, J.; Zhang, T.; Zhang, L.; et al. Rapid Surface Display of mRNA Antigens by Bacteria-Derived Outer Membrane Vesicles for a Personalized Tumor Vaccine. Adv. Mater. 2022, 34, e2109984. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Kim, S.A.; Nam, G.H.; Hong, Y.; Kim, G.B.; Choi, Y.; Lee, S.; Cho, Y.; Kwon, M.; Jeong, C.; et al. In situ immunogenic clearance induced by a combination of photodynamic therapy and rho-kinase inhibition sensitizes immune checkpoint blockade response to elicit systemic antitumor immunity against intraocular melanoma and its metastasis. J. Immunother. Cancer 2021, 9, e001481. [Google Scholar] [CrossRef]
- He, L.; Mao, C.; Brasino, M.; Harguindey, A.; Park, W.; Goodwin, A.P.; Cha, J.N. TiO2-Capped Gold Nanorods for Plasmon-Enhanced Production of Reactive Oxygen Species and Photothermal Delivery of Chemotherapeutic Agents. ACS Appl. Mater. Interfaces 2018, 10, 27965–27971. [Google Scholar] [CrossRef]
- Chen, W.; Qin, M.; Chen, X.; Wang, Q.; Zhang, Z.; Sun, X. Combining photothermal therapy and immunotherapy against melanoma by polydopamine-coated Al2O3 nanoparticles. Theranostics 2018, 8, 2229–2241. [Google Scholar] [CrossRef]
- Gobin, A.M.; Lee, M.H.; Halas, N.J.; James, W.D.; Drezek, R.A.; West, J.L. Near-infrared resonant nanoshells for combined optical imaging and photothermal cancer therapy. Nano Lett. 2007, 7, 1929–1934. [Google Scholar] [CrossRef]
- Li, X.; Pan, Y.; Chen, C.; Gao, Y.; Liu, X.; Yang, K.; Luan, X.; Zhou, D.; Zeng, F.; Han, X.; et al. Hypoxia-Responsive Gene Editing to Reduce Tumor Thermal Tolerance for Mild-Photothermal Therapy. Angew. Chem. Int. Ed. Engl. 2021, 60, 21200–21204. [Google Scholar] [CrossRef]
- Hu, J.J.; Cheng, Y.J.; Zhang, X.Z. Recent advances in nanomaterials for enhanced photothermal therapy of tumors. Nanoscale 2018, 10, 22657–22672. [Google Scholar] [CrossRef]
- Bobo, D.; Robinson, K.J.; Islam, J.; Thurecht, K.J.; Corrie, S.R. Nanoparticle-Based Medicines: A Review of FDA-Approved Materials and Clinical Trials to Date. Pharm. Res. 2016, 33, 2373–2387. [Google Scholar] [CrossRef]
- Estelrich, J.; Busquets, M.A. Iron Oxide Nanoparticles in Photothermal Therapy. Molecules 2018, 23, 1567. [Google Scholar] [CrossRef]
- Wang, X.; Xuan, L.; Pan, Y. Photothermal ablation of murine melanomas by Fe3O4 nanoparticle clusters. Beilstein J. Nanotechnol. 2022, 13, 255–264. [Google Scholar] [CrossRef] [PubMed]
- Jenke, R.; Reßing, N.; Hansen, F.K.; Aigner, A.; Büch, T. Anticancer Therapy with HDAC Inhibitors: Mechanism-Based Combination Strategies and Future Perspectives. Cancers 2021, 13, 634. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Ma, Z.; Shi, Z.; Huang, Y.; Chen, T.; Hou, L.; Jiang, T.; Yang, F. Chidamide stacked in magnetic polypyrrole nano-composites counter thermotolerance and metastasis for visualized cancer photothermal therapy. Drug Deliv. 2022, 29, 1312–1325. [Google Scholar] [CrossRef]
- Kwiatkowski, S.; Knap, B.; Przystupski, D.; Saczko, J.; Kędzierska, E.; Knap-Czop, K.; Kotlińska, J.; Michel, O.; Kotowski, K.; Kulbacka, J. Photodynamic therapy—Mechanisms, photosensitizers and combinations. Biomed. Pharmacother. 2018, 106, 1098–1107. [Google Scholar] [CrossRef]
- Nkune, N.W.; Abrahamse, H. Nanoparticle-Based Drug Delivery Systems for Photodynamic Therapy of Metastatic Melanoma: A Review. Int. J. Mol. Sci. 2021, 22, 12549. [Google Scholar] [CrossRef]
- Hamblin, M.R.; Chiang, L.Y.; Lakshmanan, S.; Huang, Y.Y.; Garcia-Diaz, M.; Karimi, M.; De Souza Rastelli, A.N.; Chandran, R. Nanotechnology for photodynamic therapy: A perspective from the Laboratory of Dr. Michael R. Hamblin in the Wellman Center for Photomedicine at Massachusetts General Hospital and Harvard Medical School. Nanotechnol. Rev. 2015, 4, 359–372. [Google Scholar] [CrossRef]
- Kou, J.; Dou, D.; Yang, L. Porphyrin photosensitizers in photodynamic therapy and its applications. Oncotarget 2017, 8, 81591–81603. [Google Scholar] [CrossRef]
- Hong, E.J.; Choi, D.G.; Shim, M.S. Targeted and effective photodynamic therapy for cancer using functionalized nanomaterials. Acta Pharm. Sin. B 2016, 6, 297–307. [Google Scholar] [CrossRef]
- Trindade, A.C.; de Castro, P.; Pinto, B.; Ambrósio, J.A.R.; de Oliveira Junior, B.M.; Beltrame Junior, M.; Gonçalves, E.P.; Pinto, J.G.; Ferreira-Strixino, J.; Simioni, A.R. Gelatin nanoparticles via template polymerization for drug delivery system to photoprocess application in cells. J. Biomater. Sci. Polym. Ed. 2022, 33, 551–568. [Google Scholar] [CrossRef]
- Rong, P.; Yang, K.; Srivastan, A.; Kiesewetter, D.; Yue, X.; Wang, F.; Nie, L.; Bhirde, A.; Wang, Z.; Liu, Z.; et al. Photosensitizer loaded nano-graphene for multimodality imaging guided tumor photodynamic therapy. Theranostics 2014, 4, 229–239. [Google Scholar] [CrossRef]
- Huang, X.; Mu, N.; Ding, Y.; Lam, H.W.; Yue, L.; Gao, C.; Chen, T.; Yuan, Z.; Wang, R. Targeted delivery and enhanced uptake of chemo-photodynamic nanomedicine for melanoma treatment. Acta Biomater. 2022, 147, 356–365. [Google Scholar] [CrossRef]
- Shan, Y.; Tan, B.; Zhang, M.; Xie, X.; Liao, J. Restorative biodegradable two-layered hybrid microneedles for melanoma photothermal/chemo co-therapy and wound healing. J. Nanobiotechnology 2022, 20, 238. [Google Scholar] [CrossRef]
- Yang, Z.; Zhu, Y.; Dong, Z.; Hao, Y.; Wang, C.; Li, Q.; Wu, Y.; Feng, L.; Liu, Z. Engineering bioluminescent bacteria to boost photodynamic therapy and systemic anti-tumor immunity for synergistic cancer treatment. Biomaterials 2022, 281, 121332. [Google Scholar] [CrossRef] [PubMed]
- Clemente, N.; Miletto, I.; Gianotti, E.; Invernizzi, M.; Marchese, L.; Dianzani, U.; Renò, F. Verteporfin-loaded mesoporous silica nanoparticles inhibit mouse melanoma proliferation in vitro and in vivo. J. Photochem. Photobiol. B 2019, 197, 111533. [Google Scholar] [CrossRef] [PubMed]
- Rizzi, M.; Tonello, S.; Estevão, B.M.; Gianotti, E.; Marchese, L.; Renò, F. Verteporfin based silica nanoparticle for in vitro selective inhibition of human highly invasive melanoma cell proliferation. J. Photochem. Photobiol. B 2017, 167, 1–6. [Google Scholar] [CrossRef]
- Clemente, N.; Miletto, I.; Gianotti, E.; Sabbatini, M.; Invernizzi, M.; Marchese, L.; Dianzani, U.; Renò, F. Verteporfin-Loaded Mesoporous Silica Nanoparticles’ Topical Applications Inhibit Mouse Melanoma Lymphangiogenesis and Micrometastasis In Vivo. Int. J. Mol. Sci. 2021, 22, 13443. [Google Scholar] [CrossRef]
- Lai, X.; Liu, X.L.; Pan, H.; Zhu, M.H.; Long, M.; Yuan, Y.; Zhang, Z.; Dong, X.; Lu, Q.; Sun, P.; et al. Light-Triggered Efficient Sequential Drug Delivery of Biomimetic Nanosystem for Multimodal Chemo-, Antiangiogenic, and Anti-MDSC Therapy in Melanoma. Adv. Mater. 2022, 34, e2106682. [Google Scholar] [CrossRef]
- Xie, J.; Liang, R.; Li, Q.; Wang, K.; Hussain, M.; Dong, L.; Shen, C.; Li, H.; Shen, G.; Zhu, J.; et al. Photosensitizer-loaded gold nanocages for immunogenic phototherapy of aggressive melanoma. Acta Biomater. 2022, 142, 264–273. [Google Scholar] [CrossRef]
- Li, W.; Tang, J.; Terry, R.N.; Li, S.; Brunie, A.; Callahan, R.L.; Noel, R.K.; Rodríguez, C.A.; Schwendeman, S.P.; Prausnitz, M.R. Long-acting reversible contraception by effervescent microneedle patch. Sci. Adv. 2019, 5, eaaw8145. [Google Scholar] [CrossRef]
- Sutrisno, L.; Chen, H.; Yoshitomi, T.; Kawazoe, N.; Yang, Y.; Chen, G. PLGA-collagen-BPNS Bifunctional composite mesh for photothermal therapy of melanoma and skin tissue engineering. J. Mater. Chem. B 2022, 10, 204–213. [Google Scholar] [CrossRef]
- Wei, Z.; Sun, J.; Lu, S.; Liu, Y.; Wang, B.; Zhao, L.; Wang, Z.; Liu, K.; Li, J.; Su, J.; et al. An Engineered Protein-Au Bioplaster for Efficient Skin Tumor Therapy. Adv. Mater. 2022, 34, e2110062. [Google Scholar] [CrossRef] [PubMed]
- Shields, C.L.; Furuta, M.; Thangappan, A.; Nagori, S.; Mashayekhi, A.; Lally, D.R.; Kelly, C.C.; Rudich, D.S.; Nagori, A.V.; Wakade, O.A.; et al. Metastasis of uveal melanoma millimeter-by-millimeter in 8033 consecutive eyes. Arch. Ophthalmol. 2009, 127, 989–998. [Google Scholar] [CrossRef]
- Andreoli, M.T.; Mieler, W.F.; Leiderman, Y.I. Epidemiological trends in uveal melanoma. Br. J. Ophthalmol. 2015, 99, 1550–1553. [Google Scholar] [CrossRef]
- Singh, A.D.; Topham, A. Incidence of uveal melanoma in the United States: 1973-1997. Ophthalmology 2003, 110, 956–961. [Google Scholar] [CrossRef]
- The Collaborative Ocular Melanoma Study, G. Assessment of Metastatic Disease Status at Death in 435 Patients with Large Choroidal Melanoma in the Collaborative Ocular Melanoma Study (COMS): COMS Report No. 15. Arch. Ophthalmol. 2001, 119, 670–676. [Google Scholar] [CrossRef]
- Amirouchene-Angelozzi, N.; Schoumacher, M.; Stern, M.H.; Cassoux, N.; Desjardins, L.; Piperno-Neumann, S.; Lantz, O.; Roman-Roman, S. Upcoming translational challenges for uveal melanoma. Br. J. Cancer 2015, 113, 1746. [Google Scholar] [CrossRef]
- Damato, B. Does ocular treatment of uveal melanoma influence survival? Br. J. Cancer 2010, 103, 285–290. [Google Scholar] [CrossRef]
- Pons, F.; Plana, M.; Caminal, J.M.; Pera, J.; Fernandes, I.; Perez, J.; Garcia-Del-Muro, X.; Marcoval, J.; Penin, R.; Fabra, A.; et al. Metastatic uveal melanoma: Is there a role for conventional chemotherapy?—A single center study based on 58 patients. Melanoma Res. 2011, 21, 217–222. [Google Scholar] [CrossRef]
- Sánchez-López, E.; Espina, M.; Doktorovova, S.; Souto, E.B.; García, M.L. Lipid nanoparticles (SLN, NLC): Overcoming the anatomical and physiological barriers of the eye—Part I—Barriers and determining factors in ocular delivery. Eur. J. Pharm. Biopharm. 2017, 110, 70–75. [Google Scholar] [CrossRef]
- Spataro, G.; Malecaze, F.; Turrin, C.O.; Soler, V.; Duhayon, C.; Elena, P.P.; Majoral, J.P.; Caminade, A.M. Designing dendrimers for ocular drug delivery. Eur. J. Med. Chem. 2010, 45, 326–334. [Google Scholar] [CrossRef]
- González-Fernández, F.M.; Bianchera, A.; Gasco, P.; Nicoli, S.; Pescina, S. Lipid-Based Nanocarriers for Ophthalmic Administration: Towards Experimental Design Implementation. Pharmaceutics 2021, 13, 447. [Google Scholar] [CrossRef]
- Augsburger, J.J.; Corrêa, Z.M.; Shaikh, A.H. Effectiveness of Treatments for Metastatic Uveal Melanoma. Am. J. Ophthalmol. 2009, 148, 119–127. [Google Scholar] [CrossRef]
- Bhatia, S.; Moon, J.; Margolin, K.A.; Weber, J.S.; Lao, C.D.; Othus, M.; Aparicio, A.M.; Ribas, A.; Sondak, V.K. Phase II trial of sorafenib in combination with carboplatin and paclitaxel in patients with metastatic uveal melanoma: SWOG S0512. PLoS ONE 2012, 7, e48787. [Google Scholar] [CrossRef]
- Santonocito, M.; Zappulla, C.; Viola, S.; La Rosa, L.R.; Solfato, E.; Abbate, I.; Tarallo, V.; Apicella, I.; Platania, C.B.; Maugeri, G.; et al. Assessment of a New Nanostructured Microemulsion System for Ocular Delivery of Sorafenib to Posterior Segment of the Eye. Int. J. Mol. Sci. 2021, 22, 4404. [Google Scholar] [CrossRef]
- Bonaccorso, A.; Pepe, V.; Zappulla, C.; Cimino, C.; Pricoco, A.; Puglisi, G.; Giuliano, F.; Pignatello, R.; Carbone, C. Sorafenib Repurposing for Ophthalmic Delivery by Lipid Nanoparticles: A Preliminary Study. Pharmaceutics 2021, 13, 1956. [Google Scholar] [CrossRef]
- Wu, T.; Sun, L.; Wu, Y.; Xiang, R.; Li, Y.; Rong, W.; Sun, F.; Wang, N. Prognostic value of legumain in uveal melanoma. Mol. Med. Rep. 2016, 13, 2377–2384. [Google Scholar] [CrossRef]
- Luo, M.; Li, Q.; Wang, D.; Ge, C.; Wang, J.; Nan, K.; Lin, S. Fabrication of chitosan based nanocomposite with legumain sensitive properties using charge driven self-assembly strategy. J. Mater. Sci. Mater. Med. 2018, 29, 142. [Google Scholar] [CrossRef]
- Xie, L.; Yue, W.; Ibrahim, K.; Shen, J. A long-acting curcumin nanoparticle/in situ hydrogel composite for the treatment of uveal melanoma. Pharmaceutics 2021, 13, 1335. [Google Scholar] [CrossRef]
- Hanna, S.C.; Krishnan, B.; Bailey, S.T.; Moschos, S.J.; Kuan, P.F.; Shimamura, T.; Osborne, L.D.; Siegel, M.B.; Duncan, L.M.; O’Brien, E.T., 3rd; et al. HIF1α and HIF2α independently activate SRC to promote melanoma metastases. J. Clin. Investig. 2013, 123, 2078–2093. [Google Scholar] [CrossRef]
- Mouriaux, F.; Sanschagrin, F.; Diorio, C.; Landreville, S.; Comoz, F.; Petit, E.; Bernaudin, M.; Rousseau, A.P.; Bergeron, D.; Morcos, M. Increased HIF-1α expression correlates with cell proliferation and vascular markers CD31 and VEGF-A in uveal melanoma. Investig. Ophthalmol. Vis. Sci. 2014, 55, 1277–1283. [Google Scholar] [CrossRef]
- Xie, L.; Yang, Y.; Shen, J. Efficient inhibition of uveal melanoma via ternary siRNA complexes. Int. J. Pharm. 2020, 573, 118894. [Google Scholar] [CrossRef] [PubMed]
- Shields, C.L.; Lim, L.S.; Dalvin, L.A.; Shields, J.A. Small choroidal melanoma: Detection with multimodal imaging and management with plaque radiotherapy or AU-011 nanoparticle therapy. Curr. Opin. Ophthalmol. 2019, 30, 206–214. [Google Scholar] [CrossRef] [PubMed]
- Butterworth, K.T.; McMahon, S.J.; Currell, F.J.; Prise, K.M. Physical basis and biological mechanisms of gold nanoparticle radiosensitization. Nanoscale 2012, 4, 4830–4838. [Google Scholar] [CrossRef]
- Ruan, J.; Wang, Y.; Li, F.; Jia, R.; Zhou, G.; Shao, C.; Zhu, L.; Cui, M.; Yang, D.P.; Ge, S. Graphene Quantum Dots for Radiotherapy. ACS Appl. Mater. Interfaces 2018, 10, 14342–14355. [Google Scholar] [CrossRef]
- Hainfeld, J.F.; Dilmanian, F.A.; Zhong, Z.; Slatkin, D.N.; Kalef-Ezra, J.A.; Smilowitz, H.M. Gold nanoparticles enhance the radiation therapy of a murine squamous cell carcinoma. Phys. Med. Biol. 2010, 55, 3045–3059. [Google Scholar] [CrossRef]
- Chang, M.Y.; Shiau, A.L.; Chen, Y.H.; Chang, C.J.; Chen, H.H.; Wu, C.L. Increased apoptotic potential and dose-enhancing effect of gold nanoparticles in combination with single-dose clinical electron beams on tumor-bearing mice. Cancer Sci. 2008, 99, 1479–1484. [Google Scholar] [CrossRef]
- Berbeco, R.I.; Ngwa, W.; Makrigiorgos, G.M. Localized dose enhancement to tumor blood vessel endothelial cells via megavoltage X-rays and targeted gold nanoparticles: New potential for external beam radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 2011, 81, 270–276. [Google Scholar] [CrossRef]
- Ahijado-Guzmán, R.; Sánchez-Arribas, N.; Martínez-Negro, M.; González-Rubio, G.; Santiago-Varela, M.; Pardo, M.; Piñeiro, A.; López-Montero, I.; Junquera, E.; Guerrero-Martínez, A. Intercellular Trafficking of Gold Nanostars in Uveal Melanoma Cells for Plasmonic Photothermal Therapy. Nanomaterials 2020, 10, 590. [Google Scholar] [CrossRef]
- Li, J.; Xue, Y.; Tian, J.; Liu, Z.; Zhuang, A.; Gu, P.; Zhou, H.; Zhang, W.; Fan, X. Fluorinated-functionalized hyaluronic acid nanoparticles for enhanced photodynamic therapy of ocular choroidal melanoma by ameliorating hypoxia. Carbohydr. Polym. 2020, 237, 116119. [Google Scholar] [CrossRef]
- Li, L.; Zeng, Z.; Chen, Z.; Gao, R.; Pan, L.; Deng, J.; Ye, X.; Zhang, J.; Zhang, S.; Mei, C.; et al. Microenvironment-Triggered Degradable Hydrogel for Imaging Diagnosis and Combined Treatment of Intraocular Choroidal Melanoma. ACS Nano 2020, 14, 15403–15416. [Google Scholar] [CrossRef]
- Zhang, Y.; Du, Y.; Le, W.; Wang, K.; Kieffer, N.; Zhang, J. Redox control of the survival of healthy and diseased cells. Antioxid. Redox Signal. 2011, 15, 2867–2908. [Google Scholar] [CrossRef] [PubMed]
- Ding, Y.; Yu, J.; Chen, X.; Wang, S.; Tu, Z.; Shen, G.; Wang, H.; Jia, R.; Ge, S.; Ruan, J.; et al. Dose-Dependent Carbon-Dot-Induced ROS Promote Uveal Melanoma Cell Tumorigenicity via Activation of mTOR Signaling and Glutamine Metabolism. Adv. Sci. 2021, 8, 2002404. [Google Scholar] [CrossRef] [PubMed]
- Eskelin, S.; Kivelä, T. Mode of presentation and time to treatment of uveal melanoma in Finland. Br. J. Ophthalmol. 2002, 86, 333–338. [Google Scholar] [CrossRef]
- Raveendran, S.; Lim, H.T.; Maekawa, T.; Vadakke Matham, M.; Sakthi Kumar, D. Gold nanocages entering into the realm of high-contrast photoacoustic ocular imaging. Nanoscale 2018, 10, 13959–13968. [Google Scholar] [CrossRef]
- Krause, M.; Kwong, K.K.; Xiong, J.; Gragoudas, E.S.; Young, L.H. MRI of blood volume and cellular uptake of superparamagnetic iron in an animal model of choroidal melanoma. Ophthalmic Res. 2002, 34, 241–250. [Google Scholar] [CrossRef] [PubMed]
- Postow, M.A.; Hamid, O.; Carvajal, R.D. Mucosal melanoma: Pathogenesis, clinical behavior, and management. Curr. Oncol. Rep. 2012, 14, 441–448. [Google Scholar] [CrossRef]
- McLaughlin, C.C.; Wu, X.C.; Jemal, A.; Martin, H.J.; Roche, L.M.; Chen, V.W. Incidence of noncutaneous melanomas in the U.S. Cancer 2005, 103, 1000–1007. [Google Scholar] [CrossRef]
- Patrick, R.J.; Fenske, N.A.; Messina, J.L. Primary mucosal melanoma. J. Am. Acad. Dermatol. 2007, 56, 828–834. [Google Scholar] [CrossRef]
- Ascierto, P.A.; Accorona, R.; Botti, G.; Farina, D.; Fossati, P.; Gatta, G.; Gogas, H.; Lombardi, D.; Maroldi, R.; Nicolai, P.; et al. Mucosal melanoma of the head and neck. Crit. Rev. Oncol./Hematol. 2017, 112, 136–152. [Google Scholar] [CrossRef]
- Lazarev, S.; Gupta, V.; Hu, K.; Harrison, L.B.; Bakst, R. Mucosal melanoma of the head and neck: A systematic review of the literature. Int. J. Radiat. Oncol. Biol. Phys. 2014, 90, 1108–1118. [Google Scholar] [CrossRef]
- Mao, L.; Qi, Z.; Zhang, L.; Guo, J.; Si, L. Immunotherapy in Acral and Mucosal Melanoma: Current Status and Future Directions. Front. Immunol. 2021, 12, 680407. [Google Scholar] [CrossRef] [PubMed]
- Kaunitz, G.J.; Cottrell, T.R.; Lilo, M.; Muthappan, V.; Esandrio, J.; Berry, S.; Xu, H.; Ogurtsova, A.; Anders, R.A.; Fischer, A.H.; et al. Melanoma subtypes demonstrate distinct PD-L1 expression profiles. Lab. Investig. 2017, 97, 1063–1071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lima, I.A.; Khalil, N.M.; Tominaga, T.T.; Lechanteur, A.; Sarmento, B.; Mainardes, R.M. Mucoadhesive chitosan-coated PLGA nanoparticles for oral delivery of ferulic acid. Artif. Cells Nanomed. Biotechnol. 2018, 46, 993–1002. [Google Scholar] [CrossRef] [PubMed]
- Long, P.; Zhang, Q.; Xue, M.; Cao, G.; Li, C.; Chen, W.; Jin, F.; Li, Z.; Li, R.; Wang, X.; et al. Tomato lectin-modified nanoemulsion-encapsulated MAGE1-HSP70/SEA complex protein vaccine: Targeting intestinal M cells following peroral administration. Biomed. Pharmacother. 2019, 115, 108886. [Google Scholar] [CrossRef]
- Liao, W.; Xiang, W.; Wang, F.F.; Wang, R.; Ding, Y. Curcumin inhibited growth of human melanoma A375 cells via inciting oxidative stress. Biomed. Pharmacother. 2017, 95, 1177–1186. [Google Scholar] [CrossRef]
- Yucel, C.; Quagliariello, V.; Iaffaioli, R.V.; Ferrari, G.; Donsì, F. Submicron complex lipid carriers for curcumin delivery to intestinal epithelial cells: Effect of different emulsifiers on bioaccessibility and cell uptake. Int. J. Pharm. 2015, 494, 357–369. [Google Scholar] [CrossRef]
- Pan, Y.; Xie, Q.T.; Zhu, J.; Li, X.M.; Meng, R.; Zhang, B.; Chen, H.Q.; Jin, Z.Y. Study on the fabrication and in vitro digestion behavior of curcumin-loaded emulsions stabilized by succinylated whey protein hydrolysates. Food Chem. 2019, 287, 76–84. [Google Scholar] [CrossRef]
- Song, W.-M.; Agrawal, P.; Von Itter, R.; Fontanals-Cirera, B.; Wang, M.; Zhou, X.; Mahal, L.K.; Hernando, E.; Zhang, B. Network models of primary melanoma microenvironments identify key melanoma regulators underlying prognosis. Nat. Commun. 2021, 12, 1214. [Google Scholar] [CrossRef]
- Chan, K.K.-W.; Chan, R.C.-L.; Ho, R.S.-L.; Chan, J.Y.-W. Clinical Patterns of Melanoma in Asians: 11-Year Experience in a Tertiary Referral Center. Ann. Plast. Surg. 2016, 77 (Suppl. 1), S6–S11. [Google Scholar] [CrossRef]
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
Wang, Z.; Yu, Y.; Wang, C.; Li, J.; Pang, Y. Advances in the Application of Nanomaterials to the Treatment of Melanoma. Pharmaceutics 2022, 14, 2090. https://doi.org/10.3390/pharmaceutics14102090
Wang Z, Yu Y, Wang C, Li J, Pang Y. Advances in the Application of Nanomaterials to the Treatment of Melanoma. Pharmaceutics. 2022; 14(10):2090. https://doi.org/10.3390/pharmaceutics14102090
Chicago/Turabian StyleWang, Zeqi, Yu Yu, Chuqiao Wang, Jin Li, and Yan Pang. 2022. "Advances in the Application of Nanomaterials to the Treatment of Melanoma" Pharmaceutics 14, no. 10: 2090. https://doi.org/10.3390/pharmaceutics14102090
APA StyleWang, Z., Yu, Y., Wang, C., Li, J., & Pang, Y. (2022). Advances in the Application of Nanomaterials to the Treatment of Melanoma. Pharmaceutics, 14(10), 2090. https://doi.org/10.3390/pharmaceutics14102090