Advances in Engineered Polymer Nanoparticle Tracking Platforms towards Cancer Immunotherapy—Current Status and Future Perspectives
Abstract
:1. Introduction
2. Evolution of Polymeric Nanomaterials and Their Engineering Strategies in Cancer Immunotherapies
3. Functional Immunotherapies Delivered to Cancer Cells via Polymeric Nanoparticles
4. Multifunctional Nanoparticulate Systems for Cancer Immunotherapies
5. Antigen-Delivering Strategies Using Polymeric Nanomaterials to Ensure the Immuno-Therapeutic Values
6. Engineered Polymeric Nanomaterials to Target and Modulate Tumor Microenvironments (TMEs)
7. Conclusion and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Brubaker, S.W.; Bonham, K.S.; Zanoni, I.; Kagan, J.C. Innate immune pattern recognition: A cell biological perspective. Annu. Rev. Immunol. 2015, 33, 257–290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vetter, V.; Denizer, G.; Friedland, L.R.; Krishnan, J.; Shapiro, M. Understanding modern-day vaccines: What you need to know. Ann. Med. 2018, 50, 110–120. [Google Scholar] [CrossRef]
- Marshall, J.S.; Warrington, R.; Watson, W.; Kim, H.L. An introduction to immunology and immunopathology. Allergy Asthma Clin. Immunol. 2018, 14, 49. [Google Scholar] [CrossRef] [Green Version]
- June, C.H.; Warshauer, J.T.; Bluestone, J.A. Is autoimmunity the Achilles’ heel of cancer immunotherapy? Nat. Med. 2017, 23, 540–547. [Google Scholar] [CrossRef] [Green Version]
- Jo, S.D.; Nam, G.-H.; Kwak, G.; Yang, Y.; Kwon, I.C. Harnessing designed nanoparticles: Current strategies and future perspectives in cancer immunotherapy. Nano Today 2017, 17, 23–37. [Google Scholar] [CrossRef]
- Sun, Q.; Barz, M.; De Geest, B.G.; Diken, M.; Hennink, W.E.; Kiessling, F.; Lammers, T.; Shi, Y. Nanomedicine and macroscale materials in immuno-oncology. Chem. Soc. Rev. 2019, 48, 351–381. [Google Scholar] [CrossRef] [PubMed]
- 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. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef]
- Siriwon, N.; Kim, Y.J.; Siegler, E.; Chen, X.; Rohrs, J.A.; Liu, Y.; Wang, P. CAR-T cells surface-engineered with drug-encapsulated nanoparticles can ameliorate intratumoral T-cell hypofunction. Cancer Immunol. Res. 2018, 6, 812–824. [Google Scholar] [CrossRef] [Green Version]
- Gondan, A.I.B.; Ruiz-de-Angulo, A.; Zabaleta, A.; Blanco, N.G.; Cobaleda-Siles, B.M.; García-Granda, M.J.; Padro, D.; Llop, J.; Arnaiz, B.; Gato, M. Effective cancer immunotherapy in mice by polyIC-imiquimod complexes and engineered magnetic nanoparticles. Biomaterials 2018, 170, 95–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pierini, F.; Nakielski, P.; Urbanek, O.; Pawłowska, S.; Lanzi, M.; De Sio, L.; Kowalewski, T.A. Polymer-based nanomaterials for photothermal therapy: From light-responsive to multifunctional nanoplatforms for synergistically combined technologies. Biomacromolecules 2018, 19, 4147–4167. [Google Scholar] [CrossRef]
- O’Donnell, J.S.; Teng, M.W.; Smyth, M.J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef]
- Shao, F.; Zhang, M.; Xu, L.; Yin, D.; Li, M.; Jiang, Q.; Zhang, Q.; Yang, Y. Multiboosting of Cancer Immunotherapy by a Core–Shell Delivery System. Mol. Pharm. 2019, 17, 338–348. [Google Scholar] [CrossRef] [PubMed]
- Alshamsan, A.; Hamdy, S.; Haddadi, A.; Samuel, J.; El-Kadi, A.O.; Uludağ, H.; Lavasanifar, A. STAT3 knockdown in B16 melanoma by siRNA lipopolyplexes induces bystander immune response in vitro and in vivo. Transl. Oncol. 2011, 4, 178–188. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Lu, K.; He, C.; Guo, N.; Chan, C.; Ni, K.; Lan, G.; Tang, H.; Pelizzari, C.; Fu, Y.-X.; Spiotto, M.T. Low-dose X-ray radiotherapy–radiodynamic therapy via nanoscale metal–organic frameworks enhances checkpoint blockade immunotherapy. Nat. Biomed. Eng. 2018, 2, 600–610. [Google Scholar] [CrossRef]
- Duan, X.; Chan, C.; Guo, N.; Han, W.; Weichselbaum, R.R.; Lin, W. Photodynamic therapy mediated by nontoxic core–shell nanoparticles synergizes with immune checkpoint blockade to elicit antitumor immunity and antimetastatic effect on breast cancer. J. Am. Chem. Soc. 2016, 138, 16686–16695. [Google Scholar] [CrossRef] [Green Version]
- Lan, G.; Ni, K.; Xu, Z.; Veroneau, S.S.; Song, Y.; Lin, W. Nanoscale metal–organic framework overcomes hypoxia for photodynamic therapy primed cancer immunotherapy. J. Am. Chem. Soc. 2018, 140, 5670–5673. [Google Scholar] [CrossRef]
- Fan, Q.; Chen, Z.; Wang, C.; Liu, Z. Toward biomaterials for enhancing immune checkpoint blockade therapy. Adv. Funct. Mater. 2018, 28, 1802540. [Google Scholar] [CrossRef]
- Chang, M.; Wang, M.; Wang, M.; Shu, M.; Ding, B.; Li, C.; Pang, M.; Cui, S.; Hou, Z.; Lin, J. A multifunctional Cascade bioreactor based on hollow-structured Cu2MoS4 for synergetic cancer chemo-dynamic therapy/starvation therapy/phototherapy/immunotherapy with remarkably enhanced efficacy. Adv. Mater. 2019, 31, 1905271. [Google Scholar] [CrossRef]
- Aikins, M.E.; Xu, C.; Moon, J.J. Engineered Nanoparticles for Cancer Vaccination and Immunotherapy. Acc. Chem. Res. 2020, 53, 2094–2105. [Google Scholar] [CrossRef]
- Zhang, X.; Xia, L.-Y.; Chen, X.; Chen, Z.; Wu, F.-G. Hydrogel-based phototherapy for fighting cancer and bacterial infection. Sci. China Mater. 2017, 60, 487–503. [Google Scholar] [CrossRef]
- Liu, Y.; Bhattarai, P.; Dai, Z.; Chen, X. Photothermal therapy and photoacoustic imaging via nanotheranostics in fighting cancer. Chem. Soc. Rev. 2019, 48, 2053–2108. [Google Scholar] [CrossRef] [PubMed]
- Lu, J.; Liu, X.; Liao, Y.-P.; Salazar, F.; Sun, B.; Jiang, W.; Chang, C.H.; Jiang, J.; Wang, X.; Wu, A.M. Nano-enabled pancreas cancer immunotherapy using immunogenic cell death and reversing immunosuppression. Nat. Commun. 2017, 8, 1811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, S.; Sun, Z.; Hou, Y. Engineering Nanoparticles toward the Modulation of Emerging Cancer Immunotherapy. Adv. Healthc. Mater. 2020, 10, 2000845. [Google Scholar] [CrossRef]
- Zhao, Y.-D.; Muhetaerjiang, M.; An, H.-W.; Fang, X.; Zhao, Y.; Wang, H. Nanomedicine Enables Spatiotemporally Regulating Macrophage-based Cancer Immunotherapy. Biomaterials 2020, 268, 120552. [Google Scholar] [CrossRef] [PubMed]
- He, C.; Duan, X.; Guo, N.; Chan, C.; Poon, C.; Weichselbaum, R.R.; Lin, W. Core-shell nanoscale coordination polymers combine chemotherapy and photodynamic therapy to potentiate checkpoint blockade cancer immunotherapy. Nat. Commun. 2016, 7, 12499. [Google Scholar] [CrossRef] [Green Version]
- Cabrita, R.; Lauss, M.; Sanna, A.; Donia, M.; Larsen, M.S.; Mitra, S.; Johansson, I.; Phung, B.; Harbst, K.; Vallon-Christersson, J. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 2020, 577, 561–565. [Google Scholar] [CrossRef] [PubMed]
- Apetoh, L.; Ladoire, S.; Coukos, G.; Ghiringhelli, F. Combining immunotherapy and anticancer agents: The right path to achieve cancer cure? Ann. Oncol. 2015, 26, 1813–1823. [Google Scholar] [CrossRef]
- Pei, M.; Xu, R.; Zhang, C.; Wang, X.; Li, C.; Hu, Y. Mannose-functionalized antigen nanoparticles for targeted dendritic cells, accelerated endosomal escape and enhanced MHC-I antigen presentation. Colloids Surf. B Biointerfaces 2021, 197, 111378. [Google Scholar] [CrossRef]
- Harari, A.; Graciotti, M.; Bassani-Sternberg, M.; Kandalaft, L.E. Antitumour dendritic cell vaccination in a priming and boosting approach. Nat. Rev. Drug Discov. 2020, 19, 635–652. [Google Scholar] [CrossRef]
- Scheetz, L.; Park, K.S.; Li, Q.; Lowenstein, P.R.; Castro, M.G.; Schwendeman, A.; Moon, J.J. Engineering patient-specific cancer immunotherapies. Nat. Biomed. Eng. 2019, 3, 768–782. [Google Scholar] [CrossRef]
- Cho, N.-H.; Cheong, T.-C.; Min, J.H.; Wu, J.H.; Lee, S.J.; Kim, D.; Yang, J.-S.; Kim, S.; Kim, Y.K.; Seong, S.-Y. A multifunctional core–shell nanoparticle for dendritic cell-based cancer immunotherapy. Nat. Nanotechnol. 2011, 6, 675–682. [Google Scholar] [CrossRef]
- Akita, H.; Kogure, K.; Moriguchi, R.; Nakamura, Y.; Higashi, T.; Nakamura, T.; Serada, S.; Fujimoto, M.; Naka, T.; Futaki, S. Nanoparticles for ex vivo siRNA delivery to dendritic cells for cancer vaccines: Programmed endosomal escape and dissociation. J. Control. Release 2010, 143, 311–317. [Google Scholar] [CrossRef]
- Demaria, O.; Cornen, S.; Daëron, M.; Morel, Y.; Medzhitov, R.; Vivier, E. Harnessing innate immunity in cancer therapy. Nature 2019, 574, 45–56. [Google Scholar] [CrossRef]
- Eyquem, J.; Mansilla-Soto, J.; Giavridis, T.; van der Stegen, S.J.; Hamieh, M.; Cunanan, K.M.; Odak, A.; Gönen, M.; Sadelain, M. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 2017, 543, 113–117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- van der Meel, R.; Sulheim, E.; Shi, Y.; Kiessling, F.; Mulder, W.J.; Lammers, T. Smart cancer nanomedicine. Nat. Nanotechnol. 2019, 14, 1007–1017. [Google Scholar] [CrossRef] [PubMed]
- Shang, T.; Yu, X.; Han, S.; Yang, B. Nanomedicine-based tumor photothermal therapy synergized immunotherapy. Biomater. Sci. 2020, 8, 5241–5259. [Google Scholar] [CrossRef]
- Lynn, G.M.; Laga, R.; Jewell, C.M. Induction of anti-cancer T cell immunity by in situ vaccination using systemically administered nanomedicines. Cancer Lett. 2019, 459, 192–203. [Google Scholar] [CrossRef] [PubMed]
- Wilson, D.R.; Sen, R.; Sunshine, J.C.; Pardoll, D.M.; Green, J.J.; Kim, Y.J. Biodegradable STING agonist nanoparticles for enhanced cancer immunotherapy. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 237–246. [Google Scholar] [CrossRef] [PubMed]
- Han, H.D.; Byeon, Y.; Kang, T.H.; Jung, I.D.; Lee, J.-W.; Shin, B.C.; Lee, Y.J.; Sood, A.K.; Park, Y.-M. Toll-like receptor 3-induced immune response by poly(d,l-lactide-co-glycolide) nanoparticles for dendritic cell-based cancer immunotherapy. Int. J. Nanomed. 2016, 11, 5729–5742. [Google Scholar] [CrossRef] [Green Version]
- Duan, F.; Feng, X.; Yang, X.; Sun, W.; Jin, Y.; Liu, H.; Ge, K.; Li, Z.; Zhang, J. A simple and powerful co-delivery system based on pH-responsive metal-organic frameworks for enhanced cancer immunotherapy. Biomaterials 2017, 122, 23–33. [Google Scholar] [CrossRef] [PubMed]
- Warashina, S.; Nakamura, T.; Harashima, H. A20 silencing by lipid envelope-type nanoparticles enhances the efficiency of lipopolysaccharide-activated dendritic cells. Biol. Pharm. Bull. 2011, 34, 1348–1351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sau, S.; Alsaab, H.O.; Bhise, K.; Alzhrani, R.; Nabil, G.; Iyer, A.K. Multifunctional nanoparticles for cancer immunotherapy: A groundbreaking approach for reprogramming malfunctioned tumor environment. J. Control. Release 2018, 274, 24–34. [Google Scholar] [CrossRef] [PubMed]
- Han, S.; Huang, K.; Gu, Z.; Wu, J. Tumor immune microenvironment modulation-based drug delivery strategies for cancer immunotherapy. Nanoscale 2020, 12, 413–436. [Google Scholar] [CrossRef] [PubMed]
- Korangath, P.; Barnett, J.D.; Sharma, A.; Henderson, E.T.; Stewart, J.; Yu, S.-H.; Kandala, S.K.; Yang, C.-T.; Caserto, J.S.; Hedayati, M. Nanoparticle interactions with immune cells dominate tumor retention and induce T cell–mediated tumor suppression in models of breast cancer. Sci. Adv. 2020, 6, eaay1601. [Google Scholar] [CrossRef] [Green Version]
- Prakash, S.; Kumbhojkar, N.; Clegg, J.R.; Mitragotri, S. Cell-bound Nanoparticles for Tissue Targeting and Immunotherapy: Engineering of the Particle-Membrane Interface. Curr. Opin. Colloid Interface Sci. 2020, 52, 101408. [Google Scholar] [CrossRef]
- Chen, F.; Hableel, G.; Zhao, E.R.; Jokerst, J.V. Multifunctional nanomedicine with silica: Role of silica in nanoparticles for theranostic, imaging, and drug monitoring. J. Colloid Interface Sci. 2018, 521, 261–279. [Google Scholar] [CrossRef]
- Ulbrich, K.; Hola, K.; Subr, V.; Bakandritsos, A.; Tucek, J.; Zboril, R. Targeted drug delivery with polymers and magnetic nanoparticles: Covalent and noncovalent approaches, release control, and clinical studies. Chem. Rev. 2016, 116, 5338–5431. [Google Scholar] [CrossRef]
- Xie, Y.Q.; Wei, L.; Tang, L. Immunoengineering with biomaterials for enhanced cancer immunotherapy. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2018, 10, e1506. [Google Scholar] [CrossRef]
- Su, S.; Hu, B.; Shao, J.; Shen, B.; Du, J.; Du, Y.; Zhou, J.; Yu, L.; Zhang, L.; Chen, F. CRISPR-Cas9 mediated efficient PD-1 disruption on human primary T cells from cancer patients. Sci. Rep. 2016, 6, 20070. [Google Scholar] [CrossRef] [Green Version]
- Ren, J.; Liu, X.; Fang, C.; Jiang, S.; June, C.H.; Zhao, Y. Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clin. Cancer Res. 2017, 23, 2255–2266. [Google Scholar] [CrossRef] [Green Version]
- Zeng, Z.; Pu, K. Improving Cancer Immunotherapy by Cell Membrane-Camouflaged Nanoparticles. Adv. Funct. Mater. 2020, 30, 2004397. [Google Scholar] [CrossRef]
- Singh, A.P.; Biswas, A.; Shukla, A.; Maiti, P. Targeted therapy in chronic diseases using nanomaterial-based drug delivery vehicles. Signal. Transduct. Target. Ther. 2019, 4, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sukumar, U.K.; Rajendran, J.C.B.; Gambhir, S.S.; Massoud, T.F.; Paulmurugan, R. SP94-Targeted Triblock Copolymer Nanoparticle Delivers Thymidine Kinase–p53–Nitroreductase Triple Therapeutic Gene and Restores Anticancer Function against Hepatocellular Carcinoma in Vivo. ACS Appl. Mater. Interfaces 2020, 12, 11307–11319. [Google Scholar] [CrossRef]
- Irvine, D.J.; Hanson, M.C.; Rakhra, K.; Tokatlian, T. Synthetic nanoparticles for vaccines and immunotherapy. Chem. Rev. 2015, 115, 11109–11146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zu, M.; Ma, L.; Zhang, X.; Xie, D.; Kang, Y.; Xiao, B. Chondroitin sulfate-functionalized polymeric nanoparticles for colon cancer-targeted chemotherapy. Colloids Surf. B Biointerfaces 2019, 177, 399–406. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.; Pu, K. Molecular and nanoengineering approaches towards activatable cancer immunotherapy. Chem. Soc. Rev. 2020, 49, 4234–4253. [Google Scholar] [CrossRef]
- Chen, Q.; Chen, M.; Liu, Z. Local biomaterials-assisted cancer immunotherapy to trigger systemic antitumor responses. Chem. Soc. Rev. 2019, 48, 5506–5526. [Google Scholar] [CrossRef]
- Rajendrakumar, S.K.; Uthaman, S.; Cho, C.-S.; Park, I.-K. Nanoparticle-based phototriggered cancer immunotherapy and its domino effect in the tumor microenvironment. Biomacromolecules 2018, 19, 1869–1887. [Google Scholar] [CrossRef]
- Wu, Y.; Gu, W.; Li, L.; Chen, C.; Xu, Z.P. Enhancing PD-1 gene silence in T lymphocytes by comparing the delivery performance of two inorganic nanoparticle platforms. Nanomaterials 2019, 9, 159. [Google Scholar] [CrossRef] [Green Version]
- Thangam, R.; Kim, M.S.; Bae, G.; Kim, Y.; Kang, N.; Lee, S.; Jung, H.J.; Jang, J.; Choi, H.; Li, N. Remote Switching of Elastic Movement of Decorated Ligand Nanostructures Controls the Adhesion-Regulated Polarization of Host Macrophages. Adv. Funct. Mater. 2021, 31, 2008698. [Google Scholar] [CrossRef]
- Bose, R.J.; Tharmalingam, N.; Garcia Marques, F.J.; Sukumar, U.K.; Natarajan, A.; Zeng, Y.; Robinson, E.; Bermudez, A.; Chang, E.; Habte, F. Reconstructed apoptotic bodies as targeted “nano decoys” to treat intracellular bacterial infections within macrophages and cancer cells. ACS Nano 2020, 14, 5818–5835. [Google Scholar] [CrossRef]
- García-Aranda, M.; Redondo, M. Targeting protein kinases to enhance the response to anti-PD-1/PD-L1 immunotherapy. Int. J. Mol. Sci. 2019, 20, 2296. [Google Scholar] [CrossRef] [Green Version]
- Conde, J.; Bao, C.; Tan, Y.; Cui, D.; Edelman, E.R.; Azevedo, H.S.; Byrne, H.J.; Artzi, N.; Tian, F. Dual targeted immunotherapy via in vivo delivery of biohybrid RNAi-peptide nanoparticles to tumor-associated macrophages and cancer cells. Adv. Funct. Mater. 2015, 25, 4183–4194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, Y.-L.; Xu, C.-F.; Li, H.-J.; Cao, Z.-T.; Liu, J.; Wang, J.-L.; Du, X.-J.; Yang, X.-Z.; Gu, Z.; Wang, J. Macrophage-specific in vivo gene editing using cationic lipid-assisted polymeric nanoparticles. ACS Nano 2018, 12, 994–1005. [Google Scholar] [CrossRef]
- Tu, K.; Deng, H.; Kong, L.; Wang, Y.; Yang, T.; Hu, Q.; Hu, M.; Yang, C.; Zhang, Z. Reshaping tumor immune microenvironment through acidity-responsive nanoparticles featured with CRISPR/Cas9-mediated programmed death-ligand 1 attenuation and chemotherapeutics-induced immunogenic cell death. ACS Appl. Mater. Interfaces 2020, 12, 16018–16030. [Google Scholar] [CrossRef] [PubMed]
- Singh, R.K.; Patel, K.D.; Leong, K.W.; Kim, H.-W. Progress in Nanotheranostics Based on Mesoporous Silica Nanomaterial Platforms. ACS Appl. Mater. Interfaces 2017, 9, 10309–10337. [Google Scholar] [CrossRef]
- Sundarraj, S.; Thangam, R.; Sujitha, M.V.; Vimala, K.; Kannan, S. Ligand-conjugated mesoporous silica nanorattles based on enzyme targeted prodrug delivery system for effective lung cancer therapy. Toxicol. Appl. Pharmacol. 2014, 275, 232–243. [Google Scholar] [CrossRef]
- Mukherjee, S.; Liang, L.; Veiseh, O. Recent Advancements of Magnetic Nanomaterials in Cancer Therapy. Pharmaceutics 2020, 12, 147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vivek, R.; Thangam, R.; Kumar, S.R.; Rejeeth, C.; Sivasubramanian, S.; Vincent, S.; Gopi, D.; Kannan, S. HER2 targeted breast cancer therapy with switchable “Off/On” multifunctional “Smart” magnetic polymer core–shell nanocomposites. ACS Appl. Mater. Interfaces 2016, 8, 2262–2279. [Google Scholar] [CrossRef] [PubMed]
- Singh, R.K.; Patel, K.D.; Mahapatra, C.; Parthiban, S.P.; Kim, T.-H.; Kim, H.-W. Combinatory Cancer Therapeutics with Nanoceria-Capped Mesoporous Silica Nanocarriers through pH-triggered Drug Release and Redox Activity. ACS Appl. Mater. Interfaces 2019, 11, 288–299. [Google Scholar] [CrossRef] [PubMed]
- Patel, K.D.; Singh, R.K.; Kim, H.-W. Carbon-based nanomaterials as an emerging platform for theranostics. Mater. Horiz. 2019, 6, 434–469. [Google Scholar] [CrossRef]
- Mukherjee, S.; Madamsetty, V.S.; Bhattacharya, D.; Roy Chowdhury, S.; Paul, M.K.; Mukherjee, A. Recent advancements of nanomedicine in neurodegenerative disorders theranostics. Adv. Funct. Mater. 2020, 30, 2003054. [Google Scholar] [CrossRef]
- Liu, X.; Feng, Z.; Wang, C.; Su, Q.; Song, H.; Zhang, C.; Huang, P.; Liang, X.-J.; Dong, A.; Kong, D. Co-localized delivery of nanomedicine and nanovaccine augments the postoperative cancer immunotherapy by amplifying T-cell responses. Biomaterials 2020, 230, 119649. [Google Scholar] [CrossRef]
- Byeon, Y.; Lee, J.W.; Choi, W.S.; Won, J.E.; Kim, G.H.; Kim, M.G.; Wi, T.I.; Lee, J.M.; Kang, T.H.; Jung, I.D.; et al. CD44-targeting PLGA nanoparticles incorporating paclitaxel and FAK siRNA overcome chemoresistance in epithelial ovarian cancer. Cancer Res. 2018, 78, 6247–6256. [Google Scholar] [CrossRef] [Green Version]
- Dey, A.; Manna, S.; Kumar, S.; Chattopadhyay, S.; Saha, B.; Roy, S. Immunostimulatory effect of chitosan conjugated green copper oxide nanoparticles in tumor immunotherapy. Cytokine 2020, 127, 154958. [Google Scholar] [CrossRef]
- Maiyo, F.; Singh, M. Folate-Targeted mRNA Delivery Using Chitosan-Functionalized Selenium Nanoparticles: Potential in Cancer Immunotherapy. Pharmaceuticals 2019, 12, 164. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Cai, C.; Li, J.; Li, J.; Li, J.; Sun, T.; Wang, L.; Wu, H.; Yu, G. Chitosan-based nanomaterials for drug delivery. Molecules 2018, 23, 2661. [Google Scholar] [CrossRef] [Green Version]
- Rafiq, S.; Hackett, C.S.; Brentjens, R.J. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 2020, 17, 147–167. [Google Scholar] [CrossRef] [PubMed]
- Kumari, P.; Ghosh, B.; Biswas, S. Nanocarriers for cancer-targeted drug delivery. J. Drug Target. 2016, 24, 179–191. [Google Scholar] [CrossRef] [PubMed]
- Yu, W.; He, X.; Yang, Z.; Yang, X.; Xiao, W.; Liu, R.; Xie, R.; Qin, L.; Gao, H. Sequentially responsive biomimetic nanoparticles with optimal size in combination with checkpoint blockade for cascade synergetic treatment of breast cancer and lung metastasis. Biomaterials 2019, 217, 119309. [Google Scholar] [CrossRef]
- Hoshyar, N.; Gray, S.; Han, H.B.; Bao, G. The effect of nanoparticle size on in vivo pharmacokinetics and cellular interaction. Nanomedicine 2016, 11, 673–692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, F.; Stephan, S.B.; Ene, C.I.; Smith, T.T.; Holland, E.C.; Stephan, M.T. Nanoparticles that reshape the tumor milieu create a therapeutic window for effective T-cell therapy in solid malignancies. Cancer Res. 2018, 78, 3718–3730. [Google Scholar] [CrossRef] [Green Version]
- Rosalia, R.A.; Cruz, L.J.; van Duikeren, S.; Tromp, A.T.; Silva, A.L.; Jiskoot, W.; de Gruijl, T.; Löwik, C.; Oostendorp, J.; van der Burg, S.H. CD40-targeted dendritic cell delivery of PLGA-nanoparticle vaccines induce potent anti-tumor responses. Biomaterials 2015, 40, 88–97. [Google Scholar] [CrossRef] [PubMed]
- Jia, L.; Lu, Y.; Shao, J.; Liang, X.-J.; Xu, Y. Nanoproteomics: A new sprout from emerging links between nanotechnology and proteomics. Trends Biotechnol. 2013, 31, 99–107. [Google Scholar] [CrossRef]
- Yoon, H.Y.; Selvan, S.T.; Yang, Y.; Kim, M.J.; Yi, D.K.; Kwon, I.C.; Kim, K. Engineering nanoparticle strategies for effective cancer immunotherapy. Biomaterials 2018, 178, 597–607. [Google Scholar] [CrossRef] [PubMed]
- Jadidi-Niaragh, F.; Atyabi, F.; Rastegari, A.; Kheshtchin, N.; Arab, S.; Hassannia, H.; Ajami, M.; Mirsanei, Z.; Habibi, S.; Masoumi, F. CD73 specific siRNA loaded chitosan lactate nanoparticles potentiate the antitumor effect of a dendritic cell vaccine in 4T1 breast cancer bearing mice. J. Control. Release 2017, 246, 46–59. [Google Scholar] [CrossRef]
- Almalik, A.; Benabdelkamel, H.; Masood, A.; Alanazi, I.O.; Alradwan, I.; Majrashi, M.A.; Alfadda, A.A.; Alghamdi, W.M.; Alrabiah, H.; Tirelli, N. Hyaluronic acid coated chitosan nanoparticles reduced the immunogenicity of the formed protein corona. Sci. Rep. 2017, 7, 10542. [Google Scholar] [CrossRef]
- Monopoli, M.P.; Åberg, C.; Salvati, A.; Dawson, K.A. Biomolecular coronas provide the biological identity of nano-sized materials. Nat. Nanotechnol. 2012, 7, 779–786. [Google Scholar] [CrossRef]
- Almalik, A.; Donno, R.; Cadman, C.J.; Cellesi, F.; Day, P.J.; Tirelli, N. Hyaluronic acid-coated chitosan nanoparticles: Molecular weight-dependent effects on morphology and hyaluronic acid presentation. J. Control. Release 2013, 172, 1142–1150. [Google Scholar] [CrossRef]
- Zhuang, J.; Holay, M.; Park, J.H.; Fang, R.H.; Zhang, J.; Zhang, L. Nanoparticle delivery of immunostimulatory agents for cancer immunotherapy. Theranostics 2019, 9, 7826. [Google Scholar] [CrossRef]
- Ni, J.; Song, J.; Wang, B.; Hua, H.; Zhu, H.; Guo, X.; Xiong, S.; Zhao, Y. Dendritic cell vaccine for the effective immunotherapy of breast cancer. Biomed. Pharmacother. 2020, 126, 110046. [Google Scholar] [CrossRef]
- Dijkstra, K.K.; Voabil, P.; Schumacher, T.N.; Voest, E.E. Genomics-and Transcriptomics-Based Patient Selection for Cancer Treatment With Immune Checkpoint Inhibitors A Review. JAMA Oncol. 2016, 2, 1490–1495. [Google Scholar] [CrossRef] [PubMed]
- Cheng, M.A.; Farmer, E.; Huang, C.; Lin, J.; Hung, C.F.; Wu, T.C. Therapeutic DNA Vaccines for Human Papillomavirus and Associated Diseases. Hum. Gene 2018, 29, 971–996. [Google Scholar] [CrossRef] [PubMed]
- Schlom, J.; Hodge, J.W.; Palena, C.; Tsang, K.Y.; Jochems, C.; Greiner, J.W.; Farsaci, B.; Madan, R.A.; Heery, C.R.; Gulley, J.L. Therapeutic cancer vaccines. Adv. Cancer Res. 2014, 121, 67–124. [Google Scholar] [CrossRef] [PubMed]
- Kantoff, P.W.; Higano, C.S.; Shore, N.D.; Berger, E.R.; Small, E.J.; Penson, D.F.; Redfern, C.H.; Ferrari, A.C.; Dreicer, R.; Sims, R.B. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N. Engl. J. Med. 2010, 363, 411–422. [Google Scholar] [CrossRef] [Green Version]
- Pandolfi, F.; Cianci, R.; Pagliari, D.; Casciano, F.; Bagala, C.; Astone, A.; Landolfi, R.; Barone, C. The immune response to tumors as a tool toward immunotherapy. Clin. Dev. Immunol. 2011, 2011, 894704. [Google Scholar] [CrossRef] [PubMed]
- Neoptolemos, J.P.; Stocken, D.D.; Bassi, C.; Ghaneh, P.; Cunningham, D.; Goldstein, D.; Padbury, R.; Moore, M.J.; Gallinger, S.; Mariette, C. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: A randomized controlled trial. JAMA 2010, 304, 1073–1081. [Google Scholar] [CrossRef]
- Fan, Y.; Moon, J.J. Nanoparticle drug delivery systems designed to improve cancer vaccines and immunotherapy. Vaccines 2015, 3, 662–685. [Google Scholar] [CrossRef] [Green Version]
- Liu, J.; Miao, L.; Sui, J.; Hao, Y.; Huang, G. Nanoparticle cancer vaccines: Design considerations and recent advances. Asian J. Pharm. Sci. 2019, 15, 576–590. [Google Scholar] [CrossRef]
- Dougan, M.; Dougan, S.K. Programmable bacteria as cancer therapy. Nat. Med. 2019, 25, 1030–1031. [Google Scholar] [CrossRef]
- Hu, Z.; Ott, P.A.; Wu, C.J. Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat. Rev. Immunol. 2018, 18, 168–182. [Google Scholar] [CrossRef]
- Zhu, G.Z.; Zhang, F.W.; Ni, Q.Q.; Niu, G.; Chen, X.Y. Efficient Nanovaccine Delivery in Cancer Immunotherapy. ACS Nano 2017, 11, 2387–2392. [Google Scholar] [CrossRef]
- Yan, S.; Zeng, X.; Tang, Y.a.; Liu, B.F.; Wang, Y.; Liu, X. Activating antitumor immunity and antimetastatic effect through polydopamine-encapsulated core–shell upconversion nanoparticles. Adv. Mater. 2019, 31, 1905825. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Cheng, Y.; Xu, Y.; Wang, Z.; Du, X.; Li, C.; Peng, J.; Gao, L.; Liang, X.; Ma, C. Increased expression of programmed cell death protein 1 on NK cells inhibits NK-cell-mediated anti-tumor function and indicates poor prognosis in digestive cancers. Oncogene 2017, 36, 6143–6153. [Google Scholar] [CrossRef] [Green Version]
- Rafiq, S.; Yeku, O.O.; Jackson, H.J.; Purdon, T.J.; van Leeuwen, D.G.; Drakes, D.J.; Song, M.; Miele, M.M.; Li, Z.; Wang, P. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat. Biotechnol. 2018, 36, 847–856. [Google Scholar] [CrossRef] [PubMed]
- Blondin-Ladrie, L.; Aranguren, M.; Doyon-Laliberté, K.; Poudrier, J.; Roger, M. The Importance of Regulation in Natural Immunity to HIV. Vaccines 2021, 9, 271. [Google Scholar] [CrossRef]
- Heo, M.B.; Cho, M.Y.; Lim, Y.T. Polymer nanoparticles for enhanced immune response: Combined delivery of tumor antigen and small interference RNA for immunosuppressive gene to dendritic cells. Acta Biomater. 2014, 10, 2169–2176. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.; Wang, J.; Zhang, X.; Yu, S.; Wen, D.; Hu, Q.; Ye, Y.; Bomba, H.; Hu, X.; Liu, Z. In situ formed reactive oxygen species–responsive scaffold with gemcitabine and checkpoint inhibitor for combination therapy. Sci. Transl. Med. 2018, 10, eaan3682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qiu, H.; Min, Y.; Rodgers, Z.; Zhang, L.; Wang, A.Z. Nanomedicine approaches to improve cancer immunotherapy. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2017, 9, e1456. [Google Scholar] [CrossRef]
- Phuengkham, H.; Song, C.; Lim, Y.T. A designer scaffold with immune nanoconverters for reverting immunosuppression and enhancing immune checkpoint blockade therapy. Adv. Mater. 2019, 31, 1903242. [Google Scholar] [CrossRef]
- Smith, W.M.; Purvis, I.J.; Bomstad, C.N.; Labak, C.M.; Velpula, K.K.; Tsung, A.J.; Regan, J.N.; Venkataraman, S.; Vibhakar, R.; Asuthkar, S. Therapeutic targeting of immune checkpoints with small molecule inhibitors. Am. J. Transl. Res. 2019, 11, 529. [Google Scholar]
- Park, J.-J.; Thi, E.P.; Carpio, V.H.; Bi, Y.; Cole, A.G.; Dorsey, B.D.; Fan, K.; Harasym, T.; Iott, C.L.; Kadhim, S. Checkpoint inhibition through small molecule-induced internalization of programmed death-ligand 1. Nat. Commun. 2021, 12, 1222. [Google Scholar] [CrossRef]
- Gurbatri, C.R.; Lia, I.; Vincent, R.; Coker, C.; Castro, S.; Treuting, P.M.; Hinchliffe, T.E.; Arpaia, N.; Danino, T. Engineered probiotics for local tumor delivery of checkpoint blockade nanobodies. Sci. Transl. Med. 2020, 12, eaax0876. [Google Scholar] [CrossRef]
- Duan, X.; Chan, C.; Han, W.; Guo, N.; Weichselbaum, R.R.; Lin, W. Immunostimulatory nanomedicines synergize with checkpoint blockade immunotherapy to eradicate colorectal tumors. Nat. Commun. 2019, 10, 1899. [Google Scholar] [CrossRef] [Green Version]
- Kuai, R.; Yuan, W.; Son, S.; Nam, J.; Xu, Y.; Fan, Y.; Schwendeman, A.; Moon, J.J. Elimination of established tumors with nanodisc-based combination chemoimmunotherapy. Sci. Adv. 2018, 4, eaao1736. [Google Scholar] [CrossRef] [PubMed]
- Gubin, M.M.; Zhang, X.; Schuster, H.; Caron, E.; Ward, J.P.; Noguchi, T.; Ivanova, Y.; Hundal, J.; Arthur, C.D.; Krebber, W.-J. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 2014, 515, 577–581. [Google Scholar] [CrossRef]
- Jiang, Y.; Li, Y.; Zhu, B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis. 2015, 6, e1792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.; Lu, Y.; Zhu, X.; Li, C.; Yan, M.; Pan, J.; Ma, G. Tumor microenvironment-responsive prodrug nanoplatform via co-self-assembly of photothermal agent and IDO inhibitor for enhanced tumor penetration and cancer immunotherapy. Biomaterials 2020, 242, 119933. [Google Scholar] [CrossRef]
- Shields IV, C.W.; Wang, L.L.W.; Evans, M.A.; Mitragotri, S. Materials for immunotherapy. Adv. Mater. 2020, 32, 1901633. [Google Scholar] [CrossRef] [PubMed]
- Ye, Y.; Wang, J.; Hu, Q.; Hochu, G.M.; Xin, H.; Wang, C.; Gu, Z. Synergistic transcutaneous immunotherapy enhances antitumor immune responses through delivery of checkpoint inhibitors. ACS Nano 2016, 10, 8956–8963. [Google Scholar] [CrossRef]
- Sun, L.Z.; Wang, X.W.; Saredy, J.; Yuan, Z.Y.; Yang, X.F.; Wang, H. Innate-adaptive immunity interplay and redox regulation in immune response. Redox Biol. 2020, 37, 101759. [Google Scholar] [CrossRef] [PubMed]
- Merkley, S.D.; Chock, C.J.; Yang, X.X.O.; Harris, J.; Castillo, E.F. Modulating T Cell Responses via Autophagy: The Intrinsic Influence Controlling the Function of Both Antigen-Presenting Cells and T Cells. Front. Immunol. 2018, 9, 2914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, H.; Mooney, D.J. Biomaterial-assisted targeted modulation of immune cells in cancer treatment. Nat. Mater. 2018, 17, 761–772. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.; Niu, L.; Larson, P.; Kucaba, T.A.; Murphy, K.A.; James, B.R.; Ferguson, D.M.; Griffith, T.S.; Panyam, J. Polymeric nanoparticles encapsulating novel TLR7/8 agonists as immunostimulatory adjuvants for enhanced cancer immunotherapy. Biomaterials 2018, 164, 38–53. [Google Scholar] [CrossRef]
- Yang, Z.G.; Ma, Y.F.; Zhao, H.; Yuan, Y.; Kim, B.Y.S. Nanotechnology platforms for cancer immunotherapy. Wires Nanomed. Nanobiotechnol. 2020, 12, e1590. [Google Scholar] [CrossRef]
- Cao, J.; Huang, D.; Peppas, N.A. Advanced engineered nanoparticulate platforms to address key biological barriers for delivering chemotherapeutic agents to target sites. Adv. Drug Deliv. Rev. 2020, 167, 170–188. [Google Scholar] [CrossRef]
- Wang, D.; Wang, T.; Yu, H.; Feng, B.; Zhou, L.; Zhou, F.; Hou, B.; Zhang, H.; Luo, M.; Li, Y. Engineering nanoparticles to locally activate T cells in the tumor microenvironment. Sci. Immunol. 2019, 4, eaau6584. [Google Scholar] [CrossRef]
- Wang, L.; Wang, Z.; Qin, Y.; Liang, W. Delivered antigen peptides to resident CD8alpha(+) DCs in lymph node by micelle-based vaccine augment antigen-specific CD8(+) effector T cell response. Eur. J. Pharm. Biopharm. 2020, 147, 76–86. [Google Scholar] [CrossRef]
- Jin, Z.; Gao, S.; Cui, X.L.; Sun, D.J.; Zhao, K. Adjuvants and delivery systems based on polymeric nanoparticles for mucosal vaccines. Int. J. Pharm. 2019, 572, 118731. [Google Scholar] [CrossRef] [PubMed]
- Galstyan, A.; Markman, J.L.; Shatalova, E.S.; Chiechi, A.; Korman, A.J.; Patil, R.; Klymyshyn, D.; Tourtellotte, W.G.; Israel, L.L.; Braubach, O. Blood–brain barrier permeable nano immunoconjugates induce local immune responses for glioma therapy. Nat. Commun. 2019, 10, 3850. [Google Scholar] [CrossRef] [PubMed]
- Tian, Y.; Jiang, X.; Chen, X.; Shao, Z.; Yang, W. Doxorubicin-Loaded Magnetic Silk Fibroin Nanoparticles for Targeted Therapy of Multidrug-Resistant Cancer. Adv. Mater. 2014, 26, 7393–7398. [Google Scholar] [CrossRef]
- Han, H.D.; Byeon, Y.; Jang, J.H.; Jeon, H.N.; Kim, G.H.; Kim, M.G.; Pack, C.G.; Kang, T.H.; Jung, I.D.; Lim, Y.T.; et al. In vivo stepwise immunomodulation using chitosan nanoparticles as a platform nanotechnology for cancer immunotherapy. Sci. Rep. 2016, 6, 39348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mudgal, J.; Mudgal, P.P.; Kinra, M.; Raval, R. Immunomodulatory role of chitosan-based nanoparticles and oligosaccharides in cyclophosphamide-treated mice. Scand. J. Immunol. 2019, 89, e12749. [Google Scholar] [CrossRef] [PubMed]
- Munakata, L.; Tanimoto, Y.; Osa, A.; Meng, J.; Haseda, Y.; Naito, Y.; Machiyama, H.; Kumanogoh, A.; Omata, D.; Maruyama, K.; et al. Lipid nanoparticles of Type-A CpG D35 suppress tumor growth by changing tumor immune-microenvironment and activate CD8 T cells in mice. J. Control. Release 2019, 313, 106–119. [Google Scholar] [CrossRef] [PubMed]
- Shirai, S.; Shibuya, M.; Kawai, A.; Tamiya, S.; Munakata, L.; Omata, D.; Suzuki, R.; Aoshi, T.; Yoshioka, Y. Lipid Nanoparticles Potentiate CpG-Oligodeoxynucleotide-Based Vaccine for Influenza Virus. Front. Immunol. 2020, 10, 3018. [Google Scholar] [CrossRef] [PubMed]
- Verbeke, R.; Lentacker, I.; Wayteck, L.; Breckpot, K.; Van Bockstal, M.; Descamps, B.; Vanhove, C.; De Smedt, S.C.; Dewitte, H. Co-delivery of nucleoside-modified mRNA and TLR agonists for cancer immunotherapy: Restoring the immunogenicity of immunosilent mRNA. J. Control. Release 2017, 266, 287–300. [Google Scholar] [CrossRef]
- Klauber, T.C.B.; Laursen, J.M.; Zucker, D.; Brix, S.; Jensen, S.S.; Andresen, T.L. Delivery of TLR7 agonist to monocytes and dendritic cells by DCIR targeted liposomes induces robust production of anti-cancer cytokines. Acta Biomater. 2017, 53, 367–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jin, J.-O.; Park, H.; Zhang, W.; de Vries, J.W.; Gruszka, A.; Lee, M.W.; Ahn, D.-R.; Herrmann, A.; Kwak, M. Modular delivery of CpG-incorporated lipid-DNA nanoparticles for spleen DC activation. Biomaterials 2017, 115, 81–89. [Google Scholar] [CrossRef]
- Lee, K.; Kim, S.Y.; Seo, Y.; Kim, M.H.; Chang, J.; Lee, H. Adjuvant incorporated lipid nanoparticles for enhanced mRNA-mediated cancer immunotherapy. Biomater. Sci. 2020, 8, 1101–1105. [Google Scholar] [CrossRef]
- Wu, Z.; Chen, C.; Luo, J.; Davis, J.R.J.; Zhang, B.; Tang, L.; Shi, W.; Liao, D. EGFP-EGF1-conjugated poly(lactic-co-glycolic acid) nanoparticles as a carrier for the delivery of CCR2− shRNA to atherosclerotic macrophage in vitro. Sci. Rep. 2020, 10, 19636. [Google Scholar] [CrossRef]
- Hafner, A.M.; Corthésy, B.; Textor, M.; Merkle, H.P. Surface-assembled poly(I:C) on PEGylated PLGA microspheres as vaccine adjuvant: APC activation and bystander cell stimulation. Int. J. Pharm. 2016, 514, 176–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Q.; Xu, L.; Liang, C.; Wang, C.; Peng, R.; Liu, Z. Photothermal therapy with immune-adjuvant nanoparticles together with checkpoint blockade for effective cancer immunotherapy. Nat. Commun. 2016, 7, 13193. [Google Scholar] [CrossRef]
- Chen, Q.; Chen, J.; Yang, Z.; Xu, J.; Xu, L.; Liang, C.; Han, X.; Liu, Z. Nanoparticle-Enhanced Radiotherapy to Trigger Robust Cancer Immunotherapy. Adv. Mater. 2019, 31, 1802228. [Google Scholar] [CrossRef] [PubMed]
- Yang, W.; Zhang, F.; Deng, H.; Lin, L.; Wang, S.; Kang, F.; Yu, G.; Lau, J.; Tian, R.; Zhang, M.; et al. Smart Nanovesicle-Mediated Immunogenic Cell Death through Tumor Microenvironment Modulation for Effective Photodynamic Immunotherapy. ACS Nano 2020, 14, 620–631. [Google Scholar] [CrossRef]
- Kim, W.G.; Choi, B.; Yang, H.-J.; Han, J.-A.; Jung, H.; Cho, H.; Kang, S.; Hong, S.Y. Covalent Conjugation of Small-Molecule Adjuvants to Nanoparticles Induces Robust Cytotoxic T Cell Responses via DC Activation. Bioconjugate Chem. 2016, 27, 2007–2013. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Qiao, L.; Zhang, S.; Wan, G.; Chen, B.; Zhou, P.; Zhang, N.; Wang, Y. Dual pH-responsive multifunctional nanoparticles for targeted treatment of breast cancer by combining immunotherapy and chemotherapy. Acta Biomater. 2018, 66, 310–324. [Google Scholar] [CrossRef]
- Li, H.; Tian, J.; Wu, A.; Wang, J.; Ge, C.; Sun, Z. Self-assembled silk fibroin nanoparticles loaded with binary drugs in the treatment of breast carcinoma. Int. J. Nanomed. 2016, 11, 4373–4380. [Google Scholar] [CrossRef] [Green Version]
- Kumar, S.; Anselmo, A.C.; Banerjee, A.; Zakrewsky, M.; Mitragotri, S. Shape and size-dependent immune response to antigen-carrying nanoparticles. J. Control. Release 2015, 220, 141–148. [Google Scholar] [CrossRef] [PubMed]
- Habibi, N.; Christau, S.; Ochyl, L.J.; Fan, Z.; Hassani Najafabadi, A.; Kuehnhammer, M.; Zhang, M.; Helgeson, M.; von Klitzing, R.; Moon, J.J.; et al. Engineered Ovalbumin Nanoparticles for Cancer Immunotherapy. Adv. Ther. 2020, 3, 2000100. [Google Scholar] [CrossRef]
- Higgins, S.G.; Becce, M.; Belessiotis-Richards, A.; Seong, H.; Sero, J.E.; Stevens, M.M. High-Aspect-Ratio Nanostructured Surfaces as Biological Metamaterials. Adv. Mater. 2020, 32, 1903862. [Google Scholar] [CrossRef]
- Wang, Y.; Deng, L.; Gonzalez, G.X.; Luthra, L.; Dong, C.; Ma, Y.; Zou, J.; Kang, S.-M.; Wang, B.-Z. Double-Layered M2e-NA Protein Nanoparticle Immunization Induces Broad Cross-Protection against Different Influenza Viruses in Mice. Adv. Healthc. Mater. 2020, 9, 1901176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Armand, P.; Shipp, M.A.; Ribrag, V.; Michot, J.M.; Zinzani, P.L.; Kuruvilla, J.; Snyder, E.S.; Ricart, A.D.; Balakumaran, A.; Rose, S.; et al. Programmed Death-1 Blockade With Pembrolizumab in Patients with Classical Hodgkin Lymphoma After Brentuximab Vedotin Failure. J. Clin. Oncol. 2016, 34, 3733. [Google Scholar] [CrossRef] [PubMed]
- Garg, A.D.; Coulie, P.G.; Van den Eynde, B.J.; Agostinis, P. Integrating Next-Generation Dendritic Cell Vaccines into the Current Cancer Immunotherapy Landscape. Trends Immunol. 2017, 38, 577–593. [Google Scholar] [CrossRef] [PubMed]
- Kabiljo, J.; Harpain, F.; Carotta, S.; Bergmann, M. Radiotherapy as a Backbone for Novel Concepts in Cancer Immunotherapy. Cancers 2020, 12, 79. [Google Scholar] [CrossRef] [Green Version]
- Min, Y.; Roche, K.C.; Tian, S.; Eblan, M.J.; McKinnon, K.P.; Caster, J.M.; Chai, S.; Herring, L.E.; Zhang, L.; Zhang, T. Antigen-capturing nanoparticles improve the abscopal effect and cancer immunotherapy. Nat. Nanotechnol. 2017, 12, 877. [Google Scholar] [CrossRef] [Green Version]
- Jin, J.; Krishnamachary, B.; Barnett, J.D.; Chatterjee, S.; Chang, D.; Mironchik, Y.; Wildes, F.; Jaffee, E.M.; Nimmagadda, S.; Bhujwalla, Z.M. Human cancer cell membrane-coated biomimetic nanoparticles reduce fibroblast-mediated invasion and metastasis and induce T-cells. ACS Appl. Mater. Interfaces 2019, 11, 7850–7861. [Google Scholar] [CrossRef]
- Xie, Z.J.; Fan, T.J.; An, J.; Choi, W.; Duo, Y.H.; Ge, Y.Q.; Zhang, B.; Nie, G.H.; Xie, N.; Zheng, T.T.; et al. Emerging combination strategies with phototherapy in cancer nanomedicine. Chem. Soc. Rev. 2020, 49, 8065–8087. [Google Scholar] [CrossRef]
- Mir, M.; Ahmed, N.; Rehman, A.U. Recent applications of PLGA based nanostructures in drug delivery. Colloid Surf. B 2017, 159, 217–231. [Google Scholar] [CrossRef]
- Muhammad, Q.; Jang, Y.; Kang, S.H.; Moon, J.; Kim, W.J.; Park, H. Modulation of immune responses with nanoparticles and reduction of their immunotoxicity. Biomater. Sci. 2020, 8, 1490–1501. [Google Scholar] [CrossRef]
- Feng, X.R.; Xu, W.G.; Li, Z.M.; Song, W.T.; Ding, J.X.; Chen, X.S. Immunomodulatory Nanosystems. Adv. Sci. 2019, 6, 1900101. [Google Scholar] [CrossRef]
- Thangam, R.; Sundarraj, S.; Vivek, R.; Suresh, V.; Sivasubramanian, S.; Paulpandi, M.; Karthick, S.V.; Ragavi, A.S.; Kannan, S. Theranostic potentials of multifunctional chitosan–silver–phycoerythrin nanocomposites against triple negative breast cancer cells. RSC Adv. 2015, 5, 12209–12223. [Google Scholar] [CrossRef]
- Wang, S.; Wang, Z.; Yu, G.; Zhou, Z.; Jacobson, O.; Liu, Y.; Ma, Y.; Zhang, F.; Chen, Z.Y.; Chen, X. Tumor-specific drug release and reactive oxygen species generation for cancer chemo/chemodynamic combination therapy. Adv. Sci. 2019, 6, 1801986. [Google Scholar] [CrossRef] [PubMed]
- Grimaldi, A.M.; Simeone, E.; Giannarelli, D.; Muto, P.; Falivene, S.; Borzillo, V.; Giugliano, F.M.; Sandomenico, F.; Petrillo, A.; Curvietto, M. Abscopal effects of radiotherapy on advanced melanoma patients who progressed after ipilimumab immunotherapy. Oncoimmunology 2014, 3, e28780. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez-Ruiz, M.E.; Rodriguez, I.; Garasa, S.; Barbes, B.; Solorzano, J.L.; Perez-Gracia, J.L.; Labiano, S.; Sanmamed, M.F.; Azpilikueta, A.; Bolaños, E. Abscopal effects of radiotherapy are enhanced by combined immunostimulatory mAbs and are dependent on CD8 T cells and crosspriming. Cancer Res. 2016, 76, 5994–6005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.-X.; Zhao, Y.-Y.; Shen, J.; Sun, X.; Liu, Y.; Liu, H.; Wang, Y.; Wang, J. Nanoenabled modulation of acidic tumor microenvironment reverses anergy of infiltrating T cells and potentiates anti-PD-1 therapy. Nano Lett. 2019, 19, 2774–2783. [Google Scholar] [CrossRef]
- Zhang, H.; Zhang, J.; Li, Q.; Song, A.; Tian, H.; Wang, J.; Li, Z.; Luan, Y. Site-specific MOF-based immunotherapeutic nanoplatforms via synergistic tumor cells-targeted treatment and dendritic cells-targeted immunomodulation. Biomaterials 2020, 245, 119983. [Google Scholar] [CrossRef]
- Hu, Q.Y.; Sun, W.J.; Wang, C.; Gu, Z. Recent advances of cocktail chemotherapy by combination drug delivery systems. Adv. Drug Deliv. Rev. 2016, 98, 19–34. [Google Scholar] [CrossRef] [Green Version]
- Asadzadeh, Z.; Safarzadeh, E.; Safaei, S.; Baradaran, A.; Mohammadi, A.; Hajiasgharzadeh, K.; Derakhshani, A.; Argentiero, A.; Silvestris, N.; Baradaran, B. Current Approaches for Combination Therapy of Cancer: The Role of Immunogenic Cell Death. Cancers 2020, 12, 1047. [Google Scholar] [CrossRef] [Green Version]
- Vivek, R.; Thangam, R.; NipunBabu, V.; Rejeeth, C.; Sivasubramanian, S.; Gunasekaran, P.; Muthuchelian, K.; Kannan, S. Multifunctional HER2-antibody conjugated polymeric nanocarrier-based drug delivery system for multi-drug-resistant breast cancer therapy. ACS Appl. Mater. Interfaces 2014, 6, 6469–6480. [Google Scholar] [CrossRef]
- Liu, Y.; Schiff, M.; Czymmek, K.; Talloczy, Z.; Levine, B.; Dinesh-Kumar, S.P. Autophagy regulates programmed cell death during the plant innate immune response. Cell 2005, 121, 567–577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.; Thangam, R.; You, S.-H.; Sultonova, R.D.; Venu, A.; Min, J.-J.; Hong, Y. Engineering Calreticulin-Targeting Monobodies to Detect Immunogenic Cell Death in Cancer Chemotherapy. Cancers 2021, 13, 2801. [Google Scholar] [CrossRef]
- Wemeau, M.; Kepp, O.; Tesniere, A.; Panaretakis, T.; Flament, C.; De Botton, S.; Zitvogel, L.; Kroemer, G.; Chaput, N. Calreticulin exposure on malignant blasts predicts a cellular anticancer immune response in patients with acute myeloid leukemia. Cell Death Dis. 2010, 1, e104. [Google Scholar] [CrossRef] [Green Version]
- Fan, Y.; Kuai, R.; Xu, Y.; Ochyl, L.J.; Irvine, D.J.; Moon, J.J. Immunogenic cell death amplified by co-localized adjuvant delivery for cancer immunotherapy. Nano Lett. 2017, 17, 7387–7393. [Google Scholar] [CrossRef]
- Kim, D.-Y.; Pyo, A.; Yun, M.; Thangam, R.; You, S.-H.; Zhang, Y.; Jung, Y.-R.; Nguyen, D.-H.; Venu, A.; Kim, H.S. Imaging calreticulin for early detection of immunogenic cell death during anticancer treatment. J. Nucl. Med. 2021, 62, 956–960. [Google Scholar] [CrossRef] [PubMed]
- Najafi, M.; Goradel, N.H.; Farhood, B.; Salehi, E.; Solhjoo, S.; Toolee, H.; Kharazinejad, E.; Mortezaee, K. Tumor microenvironment: Interactions and therapy. J. Cell. Physiol. 2019, 234, 5700–5721. [Google Scholar] [CrossRef] [PubMed]
- Guo, S.; Deng, C.X. Effect of Stromal Cells in Tumor Microenvironment on Metastasis Initiation. Int. J. Biol. Sci. 2018, 14, 2083–2093. [Google Scholar] [CrossRef]
- Schietinger, A.; Philip, M.; Krisnawan, V.E.; Chiu, E.Y.; Delrow, J.J.; Basom, R.S.; Lauer, P.; Brockstedt, D.G.; Knoblaugh, S.E.; Hammerling, G.J.; et al. Tumor-Specific T Cell Dysfunction Is a Dynamic Antigen-Driven Differentiation Program Initiated Early during Tumorigenesis. Immunity 2016, 45, 389–401. [Google Scholar] [CrossRef] [Green Version]
- Gonzatti, M.B.; Sousa, M.E.P.; Tunissi, A.S.; Mortara, R.A.; de Oliveira, A.M.; Pereira Cerize, N.N.; Keller, A.C. Nano spray dryer for vectorizing alpha-galactosylceramide in polymeric nanoparticles: A single step process to enhance invariant Natural Killer T lymphocyte responses. Int. J. Pharm. 2019, 565, 123–132. [Google Scholar] [CrossRef]
- Polo, E.; Collado, M.; Pelaz, B.; del Pino, P. Advances toward More Efficient Targeted Delivery of Nanoparticles In Vivo: Understanding Interactions between Nanoparticles and Cells. ACS Nano 2017, 11, 2397–2402. [Google Scholar] [CrossRef]
- Huang, K.-W.; Hsu, F.-F.; Qiu, J.T.; Chern, G.-J.; Lee, Y.-A.; Chang, C.-C.; Huang, Y.-T.; Sung, Y.-C.; Chiang, C.-C.; Huang, R.-L. Highly efficient and tumor-selective nanoparticles for dual-targeted immunogene therapy against cancer. Sci. Adv. 2020, 6, eaax5032.s. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Nanocarriers | Drugs/Biomolecules | Targeted Therapeutics | Reference |
---|---|---|---|
Chitosan nanoparticles (CS NPs) | Polyinosinic-polycytidylic acid sodium salts | Next-generation vaccines to bypass the ex vivo manipulation and induce immune responses by targeting toll-like receptor 3 (TLR3) in endosomes. | [133] |
Chitosan-conjugated copper oxide nanoparticles (CS@CuONPs) | Specific antigen conjugated vehicle for antigen delivery | To activate macrophages and trigger CTLs to induce cell death. | [76] |
Chitosan-coated selenium nanoparticles (CS@SeNPs) | Folic acid-functionalized CS@SeNPs targeting moiety for mRNA delivery | To act as tumor vaccine and immunotherapy with amplified immune response. | [77] |
Chitosan and gallic acid grafted chitosan nanoparticles (GACS NPs) | Cyclophosphamide (CPA) | To exhibit significant immune stimulation in CPA-treated mice in vivo. | [134] |
Lipid nanoparticles (LNPs) | Type A CpG oligodeoxynucleotides (ODNs) | To induce Th1 and CD8 T-cells skewed immune environment and without toxicity via activating Th1 and CD8+ T-cells-skewed immune modulation. | [135] |
Functionalized LNPs | Oligodeoxynucleotides (ODNs) with unmethylated cytosine-phosphate-guanine (CpG) motifs (CpG ODNs) | Augmented the adjuvant vaccine effects of CpG-ODN and increased protective spectrum of conventional influenza split vaccine. | [136] |
mRNA-LNPs | TLR agonist mono-phosphoryl lipid A (MPLA) | Induced T-cell immunity without the strong induction of type I-IFNs under the reduced DCs activated TMEs. | [137] |
Lipid-base nanoparticles (immunoliposomes) | Toll-like receptor 7 (TLR7) agonist TMX-202 | To display high specificity over lymphocytes that showed adequate TLR-specific secretion of the anti-cancer cytokines viz IL-12p70, INF-α 2a, and INF-γ. | [138] |
Lipid-modified DNA NPs | Immune adjuvant CpG motifs | To show up-regulation of a co-stimulatory molecule, cytokine expression and production of the pro-inflammatory cytokine, and activation of DCs in TME. | [139] |
Modified LNPs | Pam3 and OVA-mRNA | To exhibit expression of tumor antigens with enhanced immune stimulation via improving the tumor prevention efficacy by administered mRNA vaccines. | [140] |
Mannose-functionalized antigen nanoparticles (MAN-OVA/PEI NPs) | Antigen ovalbumin (OVA) vaccine delivery system | To accelerate endosomal/lysosomal escape and increase MHC-I antigen presentation to B3Z T-cell hybridoma. | [29] |
Poly(lactic-co-glycolic acid) (PLGA NPs) | Chemokine (C-C motif) receptor-2-shRNA (CCR2-shRNA- EGFP-EGF1) and Coumarin-6 | To be effectively taken up by an atherosclerotic cellular model of macrophages and target-silence corresponding CCR2 mRNA expression for the use of developed NPs in the therapy of atherosclerosis. | [141] |
PEGylated PLGA microsphere | Self-assembled poly, tetanus toxoid (tt) | Up-regulation of expressions of IL-6, TNF-α, IL-12p70, and IL-10 concludes the surface assembled microspheres as vaccine adjuvant. | [142] |
Combinatory PLGA NPs | Combined with TLR7/8 bi-specific agonists | PLGA NPs triggered DC activation, which led to enhanced maturation, induction of CD8+ T-cells, and significant anti-cancer efficacy in vivo. | [125] |
Functionalized PLGA NPs | OVA antigen, and polyinosinic-polycytidylic acid sodium salt (toll-like receptor 3; TLR3) | Achieved a vaccination with PLGA NP-treated DCs that helped generate OVA-specific CD8+ T-cells and enhanced anti-tumor efficacy via delivering tumor-specific antigen and adjuvants to DCs. | [40] |
Modified PLGA NPs | Indocyanine green (ICG), imiquimod (R837), and toll-like receptor 7 (TLR7) agonist | Clinically approved components showed NIR light-triggered photothermal ablation, generating tumor-associated antigens, and this adjuvant showed vaccine-like functions. | [143] |
Core-shell PLGA NPs | Imiquimod (R837) and TLR7 agonist | To significantly improve the radiotherapy efficacy by reducing the tumor hypoxia and modulating the immune-suppressive TMEs, which enables synergistic local treatments for clinical translation. | [144] |
pH-responsive nanovesicles self-assembled of block copolymer polyethylene glycol-b-cationic polypeptide (pRNVs) | Photosensitizer 2-(1-hexayloxyethyl)-2-devinyl pyropheophorbide-a (HPPH), and indoleamine 2,3-dioxygenase inhibitor, indoximod (IND) | To serve as nanocarriers and immunogenic cell death (ICD) via pre-apoptotic exposure of calreticulin, which further enabled the photodynamic cancer therapy and increased dendritic cell (DC) recruitment and immune response provocation after ICD induction. | [145] |
Adjuvant NPs | Imidazoquinoline moiety | Delivered NPs were effectively internalized by immature DCs and exhibited enhanced in vivo activation, facilitating multivalent interactions between delivered moieties and endosomal TLR7. | [146] |
Poly(L-histidine and hyaluronic acid) NPs | Immune regulator 848 and doxorubicin (DOX) | Dual pH-responsive NPs exhibited enhanced tumor-targeting ability and growth inhibition via regulating anti-tumor immunity and killing cancer cells. The release of R848 and DOX achieved synergistic effects of immunotherapy and chemotherapy against breast cancer. | [147] |
Silk-fibroin nanoparticles (SF NPs) | Doxorubicin (DOX) | Magnetic NPs and DOX-loaded SF NPs exhibited magnetic-field-induced tumor-targeting ability and effective chemotherapy of multidrug resistance and thereby imaging and drug delivery carriers in vivo. | [132] |
SF NPs | 5-fluorouracil (5-FU) and curcumin | SF NPs with drug 5-FU and curcumin showed controlled release and anti-cancer effects induced by apoptosis of cancer cells via generation of cellular reactive oxygen species in vitro. | [148] |
Carboxylated polystyrene particles | Ovalbumin (OVA) | Shape- and size-controlled nanocarrier importance is demonstrated in modulating immune responses. This is a highly relevant physical attribute for antigen-presenting nanocarriers for immune modulation. | [149] |
Protein NPs made of polymerized OVA and chemically linked poly(ethylene) glycol (PEG) | Antigen OVA | To show activity in dendritic cells, resulting in higher OT-I CD8+ cells proliferation in vitro. NPs enhanced lymphatic drainage in vivo and increased uptake by lymph node macrophages, dendritic cells, and B cells thereby expressed higher anti-OVA antibody titers to signify the improved humoral immune responses in vivo. | [150] |
Polylactic acid (PLA) gigaporous microsphere | Antigens such as OVA, mucin 1 (MUC1) and neoantigen | Self-healing microsphere-based vaccine showed a potent T-cell response, combined with immune checkpoints inhibitors that facilitated improved performance in cancer vaccination. | [151] |
Matrix protein 2 ectodomain (M2e) and M2e-neuraminidase (M2e-NA) fusion protein NPs | M2e and M2e-NA proteins | Double-layered protein NPs containing structure-stabilized M2e and NA can be utilized to develop into universal influenza virus vaccines. Moreover, layered protein NPs can be a vaccine platform for pathogens. | [152] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Thangam, R.; Patel, K.D.; Kang, H.; Paulmurugan, R. Advances in Engineered Polymer Nanoparticle Tracking Platforms towards Cancer Immunotherapy—Current Status and Future Perspectives. Vaccines 2021, 9, 935. https://doi.org/10.3390/vaccines9080935
Thangam R, Patel KD, Kang H, Paulmurugan R. Advances in Engineered Polymer Nanoparticle Tracking Platforms towards Cancer Immunotherapy—Current Status and Future Perspectives. Vaccines. 2021; 9(8):935. https://doi.org/10.3390/vaccines9080935
Chicago/Turabian StyleThangam, Ramar, Kapil D. Patel, Heemin Kang, and Ramasamy Paulmurugan. 2021. "Advances in Engineered Polymer Nanoparticle Tracking Platforms towards Cancer Immunotherapy—Current Status and Future Perspectives" Vaccines 9, no. 8: 935. https://doi.org/10.3390/vaccines9080935
APA StyleThangam, R., Patel, K. D., Kang, H., & Paulmurugan, R. (2021). Advances in Engineered Polymer Nanoparticle Tracking Platforms towards Cancer Immunotherapy—Current Status and Future Perspectives. Vaccines, 9(8), 935. https://doi.org/10.3390/vaccines9080935