Locoregional Lymphatic Delivery Systems Using Nanoparticles and Hydrogels for Anticancer Immunotherapy
Abstract
:1. Introduction
2. Advanced NPs for LNs Targeting and Adaptive Cancer Immunotherapy
2.1. Basic Criteria for the Lymphatic Delivery of NPs and Immunomodulatory Drugs
2.2. Advanced NPs to Exploit Passive Transport to the LNs
2.2.1. Physical Load; Hydrophobic/Hydrophilic Interactions
2.2.2. Physical Load; Electrostatic Interactions
2.2.3. DNA/RNA Engineering; Spherical Nucleic Acids (SNA) and DNA/RNA Origami
2.2.4. Self-Assembled or Recombinant Protein
2.2.5. Chemical Conjugation
2.3. Advanced NPs for Active Transport to the LNs
2.4. Advanced NPs for Deeper Access to the LNs
3. Advanced HGs for LNs Accumulation and Adaptive Cancer Immunotherapy
4. Hybrid HGs and NPs for LNs Targeting and Adaptive Cancer Immunotherapy
5. Conclusions and Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Nam, J.; Son, S.; Park, K.S.; Zou, W.; Shea, L.D.; Moon, J.J. Cancer nanomedicine for combination cancer immunotherapy. Nat. Rev. Mater. 2019, 4, 398–414. [Google Scholar] [CrossRef]
- Zhang, J.; Huang, D.; Saw, P.E.; Song, E. Turning cold tumors hot: From molecular mechanisms to clinical applications. Trends Immunol. 2022, 43, 524–545. [Google Scholar] [CrossRef] [PubMed]
- Francis, D.M.; Thomas, S.N. Progress and opportunities for enhancing the delivery and efficacy of checkpoint inhibitors for cancer immunotherapy. Adv. Drug Deliv. Rev. 2017, 114, 33–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, J.; Manspeaker, M.P.; Thomas, S.N. Augmenting the synergies of chemotherapy and immunotherapy through drug delivery. Acta Biomater. 2019, 88, 1–4. [Google Scholar] [CrossRef] [PubMed]
- Manspeaker, M.P.; O’Melia, M.J.; Thomas, S.N. Elicitation of stem-like CD8+ T cell responses via lymph node-targeted chemoimmunotherapy evokes systemic tumor control. J. Immunother. Cancer 2022, 10, e005079. [Google Scholar] [CrossRef] [PubMed]
- Fucikova, J.; Kepp, O.; Kasikova, L.; Petroni, G.; Yamazaki, T.; Liu, P.; Zhao, L.; Spisek, R.; Kroemer, G.; Galluzzi, L. Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis. 2020, 11, 1013. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Zhao, Z.; Qiu, N.; Zhou, Q.; Wang, G.; Jiang, H.; Piao, Y.; Zhou, Z.; Tang, J.; Shen, Y. Co-delivery of IOX1 and doxorubicin for antibodyindependent cancer chemo-immunotherapy. Nat. Commun. 2021, 12, 2425. [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.; et al. Nano-enabled pancreas cancer immunotherapy using immunogenic cell death and reversing immunosuppression. Nat. Commun. 2017, 8, 1811. [Google Scholar] [CrossRef] [Green Version]
- Liu, P.; Zhao, L.; Pol, J.; Levesque, S.; Petrazzuolo, A.; Pfirschke, C.; Engblom, C.; Rickelt, S.; Yamazaki, T.; Iribarren, K.; et al. Crizotinib-induced immunogenic cell death in non-small cell lung cancer. Nat. Commun. 2019, 10, 1486. [Google Scholar] [CrossRef] [Green Version]
- Imbert, C.; Montfort, A.; Fraisse, M.; Marcheteau, E.; Gilhodes, J.; Martin, E.; Bertrand, F.; Marcellin, M.; Burlet-Schiltz, O.; de Peredo, A.G.; et al. Resistance of melanoma to immune checkpoint inhibitors is overcome by targeting the sphingosine kinase-1. Nat. Commun. 2020, 11, 437. [Google Scholar] [CrossRef]
- Rivas, E.I.; Linares, J.; Zwick, M.; Gomez-Llonin, A.; Guiu, M.; Labernadie, A.; Badia-Ramentol, J.; Llado, A.; Bardia, L.; Perez-Nunez, I.; et al. Targeted immunotherapy against distinct cancer-associated fibroblasts overcomes treatment resistance in refractory HER2+ breast tumors. Nat. Commun. 2022, 13, 5310. [Google Scholar] [CrossRef] [PubMed]
- Ma, S.; Zhao, Y.; Lee, W.C.; Ong, L.-T.; Lee, P.L.; Jiang, Z.; Oguz, G.; Niu, Z.; Liu, M.; Goh, J.Y.; et al. Hypoxia induces HIF1α-dependent epigenetic vulnerability in triple negative breast cancer to confer immune effector dysfunction and resistance to anti-PD-1 immunotherapy. Nat. Commun. 2022, 13, 4118. [Google Scholar] [CrossRef] [PubMed]
- Deng, Y.; Xia, X.; Zhao, Y.; Zhao, Z.; Martinez, C.; Yin, W.; Yao, J.; Hang, Q.; Wu, W.; Zhang, J.; et al. Glucocorticoid receptor regulates PD-L1 and MHCI in pancreatic cancer cells to promote immune evasion and immunotherapy resistance. Nat. Commun. 2021, 12, 7041. [Google Scholar] [CrossRef] [PubMed]
- Kang, T.H.; Park, J.H.; Yang, A.; Park, H.J.; Lee, S.E.; Kim, Y.S.; Jang, G.-Y.; Farmer, E.; Lam, B.; Park, Y.-M.; et al. Annexin A5 as an immune checkpoint inhibitor and tumor-homing molecule for cancer treatment. Nat. Commun. 2020, 11, 1137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maeda, H. Toward a full understanding of the EPR effect in primary and metastatic tumors as well as issues related to its heterogeneity. Adv. Drug Deliver. Rev. 2015, 91, 3–6. [Google Scholar] [CrossRef] [PubMed]
- Musetti, S.; Huang, L. Nanoparticle-Mediated Remodeling of the Tumor Microenvironment to Enhance Immunotherapy. ACS Nano 2018, 12, 11740–11755. [Google Scholar] [CrossRef]
- Yang, F.; Shi, K.; Jia, Y.-P.; Hao, Y.; Peng, J.-R.; Qian, Z.-Y. Advanced biomaterials for cancer immunotherapy. Acta Pharmacol. Sin. 2020, 41, 911–927. [Google Scholar] [CrossRef]
- Kim, J.; Archer, P.A.; Thomas, S.N. Innovations in lymph node targeting nanocarriers. Semin. Immunol. 2021, 56, 101534. [Google Scholar] [CrossRef]
- Schudel, A.; Francis, D.M.; Thomas, S.N. Material design for lymph node drug delivery. Nat. Rev. Mater. 2019, 4, 415–428. [Google Scholar] [CrossRef]
- Manspeaker, M.P.; Thomas, S.N. Lymphatic immunomodulation using engineered drug delivery systems for cancer immunotherapy. Adv. Drug Deliver. Rev. 2020, 160, 19–35. [Google Scholar] [CrossRef]
- Thomas, S.N.; Rohner, N.A.; Edwards, E.E. Implications of Lymphatic Transport to Lymph Nodes in Immunity and Immunotherapy. Annu. Rev. Biomed. Eng. 2016, 18, 207–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sestito, L.F.; Thomas, S.N. Biomaterials for Modulating Lymphatic Function in Immunoengineering. ACS Pharmacol. Transl. Sci. 2019, 2, 293–310. [Google Scholar] [CrossRef] [PubMed]
- Cai, S.; Yang, Q.; Bagby, T.R.; Forrest, L. Lymphatic drug delivery using engineered liposomes and solid lipid nanoparticles. Adv. Drug Deliver Rev. 2011, 63, 901–908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ravizzini, G.; Turkbey, B.; Barrett, T.; Kobayashi, H.; Choyke, P.L. Nanoparticles in sentinel lymph node mapping. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2009, 1, 610–623. [Google Scholar] [CrossRef]
- Niu, G.; Chen, X. Lymphatic Imaging: Focus on Imaging Probes. Theranostics 2015, 5, 686–697. [Google Scholar] [CrossRef] [Green Version]
- Hameed, S.; Chen, H.; Irfan, M.; Bajwa, S.Z.; Khan, W.S.; Baig, S.M.; Dai, Z. Fluorescence Guided Sentinel Lymph Node Mapping: From Current Molecular Probes to Future Multimodal Nanoprobes. Bioconjugate Chem. 2019, 30, 13–28. [Google Scholar] [CrossRef]
- Jiang, H.; Wang, Q.; Sun, X. Lymph node targeting strategies to improve vaccination efficacy. J. Control. Release 2017, 267, 47–56. [Google Scholar] [CrossRef]
- Trac, N.; Chung, E.J. Overcoming physiological barriers by nanoparticles for intravenous drug delivery to the lymph nodes. Exp. Biol. Med. 2021, 246, 2358–2371. [Google Scholar] [CrossRef]
- Gutjahr, A.; Phelip, C.; Collen, A.-L.; Monge, C.; Boisgard, A.-S.; Paul, S.; Verrier, B. Biodegradable Polymeric Nanoparticles-Based Vaccine Adjuvants for Lymph Nodes Targeting. Vaccines 2016, 4, 34. [Google Scholar] [CrossRef] [Green Version]
- Sahdev, P.; Ochyl, L.J.; Moon, J.J. Biomaterials for Nanoparticle Vaccine Delivery Systems. Pharm. Res. 2014, 31, 2563–2582. [Google Scholar] [CrossRef]
- Francis, D.M.; Manspeaker, M.P.; Schudel, A.; Sestito, L.F.; O’Melia, M.J.; Kissick, H.T.; Pollack, B.P.; Waller, E.K.; Thomas, S.N. Blockade of immune checkpoints in lymph nodes through locoregional delivery augments cancer immunotherapy. Sci. Transl. Med. 2020, 12, eaay3575. [Google Scholar] [CrossRef]
- Yu, S.; Wang, C.; Yu, J.; Wang, J.; Lu, Y.; Zhang, Y.; Zhang, X.; Quanyin, H.; Sun, W.; He, C.; et al. Injectable Bioresponsive Gel Depot for Enhanced Immune Checkpoint Blockade. Adv. Mater. 2018, 30, 1801527. [Google Scholar] [CrossRef] [PubMed]
- Ruan, H.; Hu, Q.; Wen, D.; Chen, Q.; Chen, G.; Lu, Y.; Wang, J.; Cheng, H.; Lu, W.; Gu, Z. A Dual-Bioresponsive Drug-Delivery Depot for Combination of Epigenetic Modulation and Immune Checkpoint Blockade. Adv. Mater. 2019, 31, 1806957. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Zhou, J.; Hu, L.; Han, X.; Zou, X.; Chen, Q.; Chen, Y.; Liu, Z.; Wang, C. In Situ Formed Fibrin Scaffold with Cyclophosphamide to Synergize with Immune Checkpoint Blockade for Inhibition of Cancer Recurrence after Surgery. Adv. Funct. Mater. 2019, 30, 1906922. [Google Scholar] [CrossRef]
- Kim, J.; Francis, D.M.; Thomas, S.N. In Situ Crosslinked Hydrogel Depot for Sustained Antibody Release Improves Immune Checkpoint Blockade Cancer Immunotherapy. Nanomaterials 2021, 11, 471. [Google Scholar] [CrossRef]
- Leach, D.G.; Young, S.; Hartgerink, J.D. Advances in immunotherapy delivery from implantable and injectable biomaterials. Acta Biomater. 2019, 88, 15–31. [Google Scholar] [CrossRef]
- Cai, L.; Xu, J.; Yang, Z.; Tong, R.; Dong, Z.; Wang, C.; Leong, K.W. Engineered biomaterials for cancer immunotherapy. MeComm 2020, 1, 35–46. [Google Scholar] [CrossRef]
- Kim, J.; Choi, Y.; Kim, D.-H.; Yoon, H.Y.; Kim, K. Injectable Hydrogel-Based Combination Cancer Immunotherapy for Overcoming Localized Therapeutic Efficacy. Pharmaceutics 2022, 14, 1908. [Google Scholar] [CrossRef]
- Swartz, M.A.; Fleury, M.E. Interstitial Flow and Its Effects in Soft Tissues. Annu. Rev. Biomed. Eng. 2007, 9, 229–256. [Google Scholar] [CrossRef] [Green Version]
- Rohner, N.A.; Thomas, S.N. Flexible Macromolecule versus Rigid Particle Retention in the Injected Skin and Accumulation in Draining Lymph Nodes Are Differentially Influenced by Hydrodynamic Size. ACS Biomater. Sci. Eng. 2017, 3, 153–159. [Google Scholar] [CrossRef]
- Rohner, N.A.; Thomas, S.N. Melanoma growth effects on molecular clearance from tumors and biodistribution into systemic tissues versus draining lymph nodes. J. Control. Release 2016, 223, 99–108. [Google Scholar] [CrossRef] [Green Version]
- O’Melia, M.J.; Rohner, N.A.; Manspeaker, M.P.; Francis, D.M.; Kissick, H.T.; Thomas, S.N. Quality of CD8+ T cell immunity evoked in lymph nodes is compartmentalized by route of antigen transport and functional in tumor context. Sci. Adv. 2020, 6, eabd7134. [Google Scholar] [CrossRef]
- Reddy, S.T.; Berk, D.A.; Jain, R.K.; Swartz, M.A. A sensitive in vivo model for quantifying interstitial convective transport of injected macromolecules and nanoparticles. J. Appl. Physiol. 2006, 101, 1162–1169. [Google Scholar] [CrossRef] [Green Version]
- Archer, P.A.; Sestito, L.F.; Manspeaker, M.P.; O’Melia, M.J.; Rohner, N.A.; Schudel, A.; Mei, Y.; Thomas, S.N. Quantitation of lymphatic transport mechanism and barrier influences on lymph node-resident leukocyte access to lymph-borne macromolecules and drug delivery systems. Drug Deliv. Transl. Res. 2021, 11, 2328–2343. [Google Scholar] [CrossRef]
- Howard, G.P.; Verma, G.; Ke, X.; Thayer, W.M.; Hamerly, T.; Baxter, V.K.; Lee, J.E.; Dinglasan, R.R.; Mao, H.-Q. Critical size limit of biodegradable nanoparticles for enhanced lymph node trafficking and paracortex penetration. Nano Res. 2019, 12, 837–844. [Google Scholar] [CrossRef]
- Zhang, Y.-N.; Lazarovits, J.; Poon, W.; Ouyang, B.; Nguyen, L.N.M.; Kingston, B.R.; Chan, W.C.W. Nanoparticle Size Influences Antigen Retention and Presentation in Lymph Node Follicles for Humoral Immunity. Nano Lett. 2019, 19, 7226–7235. [Google Scholar] [CrossRef]
- Nakamura, T.; Kawai, M.; Sato, Y.; Maeki, M.; Tokeshi, M.; Harashima, H. The Effect of Size and Charge of Lipid Nanoparticles Prepared by Microfluidic Mixing on Their Lymph Node Transitivity and Distribution. Mol. Pharm. 2020, 17, 944–953. [Google Scholar] [CrossRef]
- Wu, X.; Li, Y.; Chen, X.; Zhou, Z.; Pang, J.; Luo, X.; Kong, M. A surface charge dependent enhanced Th1 antigen-specific immune response in lymph nodes by transfersome-based nanovaccine-loaded dissolving microneedle-assisted transdermal immunization. J. Mater. Chem. B 2019, 7, 4854–4866. [Google Scholar] [CrossRef]
- Baek, J.-S.; Cho, C.-W. Surface modification of solid lipid nanoparticles for oral delivery of curcumin: Improvement of bioavailability through enhanced cellular uptake, and lymphatic uptake. Eur. J. Pharm. Biopharm. 2017, 117, 132–140. [Google Scholar] [CrossRef]
- Nishimoto, Y.; Nagashima, S.; Nakajima, K.; Ohira, T.; Sato, T.; Izawa, T.; Yamate, J.; Higashikawa, K.; Kuge, Y.; Ogawa, M.; et al. Carboxyl-, sulfonyl-, and phosphate-terminal dendrimers as a nanoplatform with lymph node targeting. Int. J. Pharm. 2020, 576, 119021. [Google Scholar] [CrossRef]
- Knop, K.; Hoogenboom, R.; Fischer, D.; Schubert, U.S. Poly(ethylene glycol) in Drug Delivery: Pros and Cons as Well as Potential Alternatives. Angew. Chem. Int. Ed. 2010, 49, 6288–6308. [Google Scholar] [CrossRef]
- Tomasetti, L.; Liebl, R.; Wastl, D.S.; Breunig, M. Influence of PEGylation on nanoparticle mobility in different models of the extracellular matrix. Eur. J. Pharm. Biopharm. 2016, 108, 145–155. [Google Scholar] [CrossRef]
- Mueller, S.N.; Tian, S.; DeSimone, J.M. Rapid and Persistent Delivery of Antigen by Lymph Node Targeting PRINT Nanoparticle Vaccine Carrier To Promote Humoral Immunity. Mol. Pharm. 2015, 12, 1356–1365. [Google Scholar] [CrossRef] [Green Version]
- Zukancic, D.; Suys, E.J.; Pilkington, E.H.; Algarni, A.; Al-Wassiti, H.; Truong, N.P. The Importance of Poly(ethylene glycol) and Lipid Structure in Targeted Gene Delivery to Lymph Nodes by Lipid Nanoparticles. Pharmaceutics 2020, 12, 1068. [Google Scholar] [CrossRef]
- McCright, J.; Skeen, C.; Yarmovsky, J.; Maisel, K. Nanoparticles with dense poly(ethylene glycol) coatings with near neutral charge are maximally transported across lymphatics and to the lymph nodes. Acta Biomater. 2022, 145, 146–158. [Google Scholar] [CrossRef]
- Kim, D.; Wu, Y.; Shim, G.; Oh, Y.-K. Lipid Nanoparticle-Mediated Lymphatic Delivery of Immunostimulatory Nucleic Acids. Pharmaceutics 2021, 13, 490. [Google Scholar] [CrossRef]
- Uthaman, S.; Kim, H.S.; Revuri, V.; Min, J.-J.; Lee, Y.-K.; Huh, K.M.; Park, I.-K. Green synthesis of bioactive polysaccharide-capped gold nanoparticles for lymph node CT imaging. Carbohydr. Polym. 2018, 181, 27–33. [Google Scholar] [CrossRef]
- Vu-Quang, H.; Yoo, M.-K.; Jeong, H.-J.; Lee, H.-J.; Muthiah, M.; Rhee, J.H.; Lee, J.-H.; Cho, C.-S.; Jeong, Y.Y.; Park, I.-K. Targeted delivery of mannan-coated superparamagnetic iron oxide nanoparticles to antigen-presenting cells for magnetic resonance-based diagnosis of metastatic lymph nodes in vivo. Acta Biomater. 2011, 7, 3935–3945. [Google Scholar] [CrossRef]
- Vu-Quang, H.; Muthiah, M.; Kim, Y.-K.; Cho, C.-S.; Namgung, R.; Kim, W.J.; Rhee, J.H.; Kang, S.H.; Jun, S.Y.; Choi, Y.-J.; et al. Carboxylic mannan-coated iron oxide nanoparticles targeted to immune cells for lymph node-specific MRI in vivo. Carbohydr. Polym. 2012, 88, 780–788. [Google Scholar] [CrossRef]
- Muthiah, M.; Vu-Quang, H.; Kim, Y.-K.; Rhee, J.H.; Kang, S.H.; Jun, S.Y.; Choi, Y.-J.; Jeong, Y.Y.; Cho, C.-S.; Park, I.-K. Mannose-poly(ethylene glycol)-linked SPION targeted to antigen presenting cells for magnetic resonance imaging on lymph node. Carbohydr. Polym. 2013, 92, 1586–1595. [Google Scholar] [CrossRef]
- Son, S.; Nam, J.; Zenkov, I.; Ochyl, L.J.; Xu, Y.; Scheetz, L.; Shi, J.; Farokhzad, O.C.; Moon, J.J. Sugar-Nanocapsules Imprinted with Microbial Molecular Patterns for mRNA Vaccination. Nano Lett. 2020, 20, 1499–1509. [Google Scholar] [CrossRef]
- Liang, X.; Li, X.; Duan, J.; Chen, Y.; Wang, X.; Pang, L.; Kong, D.; Song, B.; Li, C.; Yang, J. Nanoparticles with CD44 Targeting and ROS Triggering Properties as Effective in Vivo Antigen Delivery System. Mol. Pharm. 2018, 15, 508–518. [Google Scholar] [CrossRef]
- Kim, S.-Y.; Heo, M.B.; Hwang, G.-S.; Jung, Y.; Choi, D.Y.; Park, Y.-M.; Lim, Y.T. Multivalent Polymer Nanocomplex Targeting Endosomal Receptor of Immune Cells for Enhanced Antitumor and Systemic Memory Response. Angew. Chem. 2015, 127, 8257–8261. [Google Scholar] [CrossRef]
- Rosalia, R.A.; Cruz, L.J.; van Duikeren, S.; Tromp, A.T.; Silva, A.L.; Jiskoot, W.; de Gruijl, T.; Lowik, C.; Oostendorp, J.; van der Burg, S.H.; et al. CD40-targeted dendritic cell delivery of PLGA-nanoparticle vaccines induce potent anti-tumor responses. Biomaterials 2015, 40, 88–97. [Google Scholar] [CrossRef]
- Baekkevold, E.S.; Yamanaka, T.; Palframan, R.T.; Carlsen, H.S.; Reinholt, F.P.; von Andrian, U.H.; Brandtzaeg, P.; Haraldsen, G. The CCR7 Ligand ELC (CCL19) Is Transcytosed in High Endothelial Venules and Mediates T Cell Recruitment. J. Exp. Med. 2001, 193, 1105–1111. [Google Scholar] [CrossRef] [Green Version]
- Palframan, R.T.; Jung, S.; Cheng, G.; Weninger, W.; Luo, Y.; Dorf, M.; Littman, D.R.; Rollins, B.J.; Zweerink, H.; Rot, A.; et al. Inflammatory Chemokine Transport and Presentation in HEV: A Remote Control Mechanism for Monocyte Recruitment to Lymph Nodes in Inflamed Tissues. J. Exp. Med. 2001, 194, 1361–1373. [Google Scholar] [CrossRef] [Green Version]
- Bahmani, B.; Uehara, M.; Jiang, L.; Ordikhani, F.; Banouni, N.; Ichimura, T.; Solhjou, Z.; Furtmuller, G.J.; Brandacher, G.; Alvarez, D.; et al. Targeted delivery of immune therapeutics to lymph nodes prolongs cardiac allograft survival. J. Clin. Investig. 2018, 128, 4770–4786. [Google Scholar] [CrossRef]
- Ager, A. High endothelial venules and Other Blood vessels: Critical Regulators of Lymphoid Organ Development and Function. Front. Immunol. 2017, 8, 45. [Google Scholar] [CrossRef]
- Yoneyama, H.; Matsuno, K.; Zhang, Y.; Nishiwaki, T.; Kitabatake, M.; Ueha, S.; Narumi, S.; Morikawa, S.; Ezaki, T.; Lu, B.; et al. Evidence for recruitment of plasmacytoid dendritic cell precursors to inflamed lymph nodes through high endothelial venules. Int. Immunol. 2004, 16, 915–928. [Google Scholar] [CrossRef]
- Jiang, L.; Jung, S.; Zhao, J.; Kasinath, V.; Ichimura, T.; Joseph, J.; Fiorina, P.; Liss, A.S.; Shah, K.; Annabi, N.; et al. Simultaneous targeting of primary tumor, draining lymph node, and distant metastases through high endothelial venule-targeted delivery. Nano Today 2021, 36, 101045. [Google Scholar] [CrossRef]
- Azzi, J.; Yin, Q.; Uehara, M.; Ohori, S.; Tang, L.; Cai, K.; Ichimura, T.; McGrath, M.; Maarouf, O.; Kefaloyianni, E.; et al. Targeted Delivery of Immunomodulators to Lymph Nodes. Cell Rep. 2016, 15, 1202–1213. [Google Scholar] [CrossRef] [Green Version]
- Bahmani, B.; Uehara, M.; Ordikhani, F.; Li, X.; Jiang, L.; Banouni, N.; Ichimura, T.; Kasinath, V.; Eskandari, S.K.; Annabi, N.; et al. Ectopic high endothelial venules in pancreatic ductal adenocarcinoma: A unique site for targeted delivery. EBioMedicine 2018, 38, 79–99. [Google Scholar] [CrossRef] [Green Version]
- Roozendaal, R.; Mempel, T.R.; Pitcher, L.A.; Gonzalez, S.F.; Verschoor, A.; Mebius, R.E.; von Andrian, U.H.; Carroll, M.C. Conduits Mediate Transport of Low-Molecular-Weight Antigen to Lymph Node Follicles. Immunity 2009, 30, 264–276. [Google Scholar] [CrossRef] [Green Version]
- De Koker, S.; Cui, J.; Vanparijs, N.; Albertazzi, L.; Grooten, J.; Caruso, F.; De Geest, B.G. Engineering Polymer Hydrogel Nanoparticles for Lymph Node-Targeted Delivery. Angew. Chem. 2016, 128, 1356–1361. [Google Scholar] [CrossRef] [Green Version]
- Klechevsky, E.; Morita, R.; Liu, M.; Cao, Y.; Coquery, S.; Thompson-Snipes, L.; Briere, F.; Chaussabel, D.; Zurawski, G.; Palucka, A.K.; et al. Functional Specializations of Human Epidermal Langerhans Cells and CD14+ Dermal Dendritic Cells. Immunity 2008, 29, 497–510. [Google Scholar] [CrossRef] [Green Version]
- Chandrasekaran, S.; Chan, M.F.; Li, J.; King, M.R. Super natural killer cells that target metastases in the tumor draining lymph nodes. Biomaterials 2016, 77, 66–76. [Google Scholar] [CrossRef] [Green Version]
- Schmid, D.; Park, C.G.; Hartl, C.A.; Subedi, N.; Cartwright, A.N.; Bou Puerto, R.; Zheng, Y.; Maiarana, J.; Freeman, G.J.; Wucherpfennig, K.W.; et al. T cell-targeting nanoparticles focus delivery of immunotherapy to improve antitumor immunity. Nat. Commun. 2017, 8, 1747. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Wang, C.; Wang, J.; Hu, Q.; Longworthy, B.; Ye, Y.; Sun, W.; Lin, J.; Wang, T.; Fine, J.; et al. PD-1 Blockade Cellular Vesicles for Cancer Immunotherapy. Adv. Mater. 2018, 30, 1707112. [Google Scholar] [CrossRef]
- Kataoka, K.; Harada, A.; Nagasaki, Y. Block copolymer micelles for drug delivery: Design, characterization and biological significance. Adv. Drug Deliver. Rev. 2012, 64, 37–48. [Google Scholar] [CrossRef]
- Thomas, S.N.; Vokali, E.; Lund, A.W.; Hubbell, J.A.; Swartz, M.A. Targeting the tumor-draining lymph node with adjuvanted nanoparticles reshapes the anti-tumor immune response. Biomaterials 2014, 35, 814–824. [Google Scholar] [CrossRef]
- Kim, J.; Sestito, L.F.; Im, S.; Kim, W.J.; Thomas, S.N. Poly(cyclodextrin)-Polydrug Nanocomplexes as Synthetic Oncolytic Virus for Locoregional Melanoma Chemoimmunotherapy. Adv. Funct. Mater. 2020, 30, 1908788. [Google Scholar] [CrossRef]
- Horejs, C. From lipids to lipid nanoparticles to mRNA vaccines. Nat. Rev. Mater. 2021, 6, 1075–1076. [Google Scholar] [CrossRef]
- Hanson, M.C.; Crespo, M.P.; Abraham, W.; Moynihan, K.D.; Szeto, G.L.; Chen, S.H.; Melo, M.B.; Mueller, S.; Irvine, D.J. Nanoparticulate STING agonists are potent lymph node–targeted vaccine adjuvants. J. Clin. Investig. 2015, 125, 2532–2546. [Google Scholar] [CrossRef] [Green Version]
- Oberli, M.A.; Reichmuth, A.M.; Dorkin, J.R.; Mitchell, M.J.; Fenton, O.S.; Jaklenec, A.; Anderson, D.G.; Langer, R.; Blankschtein, D. Lipid Nanoparticle Assisted mRNA Delivery for Potent Cancer Immunotherapy. Nano Lett. 2017, 17, 1326–1335. [Google Scholar] [CrossRef]
- Bourquin, C.; Anz, D.; Zwiorek, K.; Lanz, A.-L.; Fuchs, S.; Wigel, S.; Wurzenberger, C.; von der Borch, P.; Golic, M.; Moder, S.; et al. Targeting CpG Oligonucleotides to the Lymph Node by Nanoparticles Elicits Efficient Antitumoral Immunity. J. Immunol. 2008, 181, 2990–2998. [Google Scholar] [CrossRef] [Green Version]
- Zeng, Q.; Li, H.; Jiang, H.; Yu, J.; Wang, Y.; Ke, H.; Gong, T.; Zhang, Z.; Sun, X. Tailoring polymeric hybrid micelles with lymph node targeting ability to improve the potency of cancer vaccines. Biomaterials 2017, 122, 105–113. [Google Scholar] [CrossRef]
- Kim, S.-Y.; Noh, Y.-W.; Kang, T.H.; Kim, J.-E.; Kim, S.; Um, S.H.; Oh, D.-B.; Park, Y.-M.; Lim, Y.T. Synthetic vaccine nanoparticles target to lymph node triggering enhanced innate and adaptive antitumor immunity. Biomaterials 2017, 130, 56–66. [Google Scholar] [CrossRef]
- An, M.; Li, M.; Xi, J.; Liu, H. Silica Nanoparticle as a Lymph Node Targeting Platform for Vaccine Delivery. ACS Appl. Mater. Interfaces 2017, 9, 23466–23475. [Google Scholar] [CrossRef]
- Lu, Y.; Yang, Y.; Gu, Z.; Zhang, J.; Song, H.; Xiang, G.; Yu, C. Glutathione-depletion mesoporous organosilica nanoparticles as a self-adjuvant and Co-delivery platform for enhanced cancer immunotherapy. Biomaterials 2018, 175, 82–92. [Google Scholar] [CrossRef]
- Jiang, H.; Wang, Q.; Li, L.; Zeng, Q.; Li, H.; Gong, T.; Zhang, Z.; Sun, X. Turning the Old Adjuvant from Gel to Nanoparticles to Amplify CD8+ T Cell Responses. Adv. Sci. 2018, 5, 1700426. [Google Scholar] [CrossRef]
- Mody, K.T.; Popat, A.; Mahony, D.; Cavallaro, A.S.; Yu, C.; Mitter, N. Mesoporous silica nanoparticles as antigen carriers and adjuvants for vaccine delivery. Nanoscale 2013, 5, 5167–5179. [Google Scholar] [CrossRef]
- Mok, H.; Lee, S.H.; Park, J.W.; Park, T.G. Multimeric small interfering ribonucleic acid for highly efficient sequence-specific gene silencing. Nat. Mater. 2010, 9, 272–278. [Google Scholar] [CrossRef]
- Zhu, G.; Mei, L.; Vishwasrao, H.D.; Jacobson, O.; Wang, Z.; Liu, Y.; Yung, B.C.; Fu, X.; Jin, A.; Niu, G.; et al. Intertwining DNA-RNA nanocapsules loaded with tumor neoantigens as synergistic nanovaccines for cancer immunotherapy. Nat. Commun. 2017, 8, 1482. [Google Scholar] [CrossRef] [Green Version]
- Ni, Q.; Zhang, F.; Liu, Y.; Wang, Z.; Yu, G.; Liang, B.; Niu, G.; Su, T.; Zhu, G.; Lu, G.; et al. A bi-adjuvant nanovaccine that potentiates immunogenicity of neoantigen for combination immunotherapy of colorectal cancer. Sci. Adv. 2020, 6, eaaw6071. [Google Scholar] [CrossRef] [Green Version]
- Sun, X.; Liu, H. Nucleic Acid Nanostructure Assisted Immune Modulation. ACS Appl. Bio Mater. 2020, 3, 2765–2778. [Google Scholar] [CrossRef]
- Cutler, J.I.; Auyeung, E.; Mirkin, C.A. Spherical Nucleic Acids. J. Am. Chem. Soc. 2012, 134, 1376–1391. [Google Scholar] [CrossRef]
- Radovic-Moreno, A.F.; Chernyak, N.; Mader, C.C.; Nallagatla, S.; Kang, R.S.; Hao, L.; Walker, D.A.; Halo, T.L.; Merkel, T.J.; Rische, C.H.; et al. Immunomodulatory spherical nucleic acids. Proc. Natl. Acad. Sci. USA 2015, 112, 3892–3897. [Google Scholar] [CrossRef] [Green Version]
- Callmann, C.E.; Kusmeierz, C.D.; Dittmar, J.W.; Broger, L.; Mirkin, C.A. Impact of Liposomal Spherical Nucleic Acid Structure on Immunotherapeutic Function. ACS Cent. Sci. 2021, 7, 892–899. [Google Scholar] [CrossRef]
- Kuai, R.; Ochyl, L.J.; Bahjat, K.S.; Schwendeman, A.; Moon, J.J. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nat. Mater. 2017, 16, 489–496. [Google Scholar] [CrossRef]
- Callmann, C.E.; Cole, L.E.; Kusmierz, C.D.; Huang, Z.; Horiuchi, D.; Mirkin, C.A. Tumor cell lysate-loaded immunostimulatory spherical nucleic acids as therapeutics for triple-negative breast cancer. Proc. Natl. Acad. Sci. USA 2020, 117, 17543–17550. [Google Scholar] [CrossRef]
- Chen, P.; Wang, D.; Wang, Y.; Zhang, L.; Wang, Q.; Liu, L.; Li, J.; Sun, X.; Ren, M.; Wang, R.; et al. Maximizing TLR9 Activation in Cancer Immunotherapy with DualAdjuvanted Spherical Nucleic Acids. Nano Lett. 2022, 22, 4058–4066. [Google Scholar] [CrossRef]
- Wang, P.; Meyer, T.A.; Pan, V.; Dutta, P.K.; Ke, Y. The Beauty and Utility of DNA Origami. Chem 2017, 2, 359–382. [Google Scholar] [CrossRef] [Green Version]
- Kim, K.-R.; Lee, Y.-D.; Lee, T.; Kim, B.-S.; Kim, S.; Ahn, D.-R. Sentinel lymph node imaging by a fluorescently labeled DNA tetrahedron. Biomaterials 2013, 34, 5226–5235. [Google Scholar] [CrossRef]
- Liu, S.; Jiang, Q.; Zhao, X.; Zhao, R.; Wang, Y.; Wang, Y.; Liu, J.; Shang, Y.; Zhao, S.; Wu, T.; et al. A DNA nanodevice-based vaccine for cancer immunotherapy. Nat. Mater. 2021, 20, 421–430. [Google Scholar] [CrossRef]
- Xi, X.; Zhang, L.; Lu, G.; Gao, X.; Wei, W.; Ma, G. Lymph Node-Targeting Nanovaccine through Antigen-CpG Self-Assembly Potentiates Cytotoxic T Cell Activation. J. Immunol. Res. 2018, 2018, 3714960. [Google Scholar] [CrossRef]
- Zhao, H.; Xu, J.; Li, Y.; Guan, X.; Han, X.; Xu, Y.; Zhou, H.; Peng, R.; Wang, J.; Liu, Z. Nanoscale Coordination Polymer Based Nanovaccine for Tumor Immunotherapy. ACS Nano 2019, 13, 13127–13135. [Google Scholar] [CrossRef]
- Lee, B.-R.; Ko, H.K.; Ryu, J.H.; Ahn, K.Y.; Lee, Y.-H.; Oh, S.J.; Na, J.H.; Kim, T.W.; Byun, Y.; Kwon, I.C.; et al. Engineered Human Ferritin Nanoparticles for Direct Delivery of Tumor Antigens to Lymph Node and Cancer Immunotherapy. Sci. Rep. 2016, 6, 35182. [Google Scholar] [CrossRef] [Green Version]
- Kim, G.B.; Sung, H.-D.; Nam, G.-H.; Kim, W.; Kim, S.; Kang, D.; Lee, E.J.; Kim, I.-S. Design of PD-1-decorated nanocages targeting tumor-draining lymph node for promoting T cell activation. J. Control. Release 2021, 333, 328–338. [Google Scholar] [CrossRef]
- Kim, J.H.; Noh, Y.-W.; Heo, M.B.; Cho, M.Y.; Lim, Y.T. Multifunctional Hybrid Nanoconjugates for Efficient In Vivo Delivery of Immunomodulating Oligonucleotides and Enhanced Antitumor Immunity. Angew. Chem. Int. Ed. 2012, 51, 9670–9673. [Google Scholar] [CrossRef]
- Ruiz-de-Angulo, A.; Zabaleta, A.; Gomez-Vallejo, V.; Llop, J.; Mareque-Rivas, J.C. Microdosed Lipid-Coated 67 Ga-Magnetite Enhances Antigen-Specific Immunity by Image Tracked Delivery of Antigen and CpG to Lymph Nodes. ACS Nano 2016, 10, 1602–1618. [Google Scholar] [CrossRef]
- Xu, C.; Hong, H.; Lee, Y.; Park, K.S.; Sun, M.; Wang, T.; Aikins, M.E.; Xu, Y.; Moon, J.J. Efficient Lymph Node-Targeted Delivery of Personalized Cancer Vaccines with Reactive Oxygen Species-Inducing Reduced Graphene Oxide Nanosheets. ACS Nano 2020, 14, 13268–13278. [Google Scholar] [CrossRef] [PubMed]
- Chu, Y.; Qian, L.; Ke, Y.; Feng, X.; Chen, X.; Liu, F.; Yu, L.; Zhang, L.; Tao, Y.; Xu, R.; et al. Lymph node-targeted neoantigen nanovaccines potentiate anti-tumor immune responses of post-surgical melanoma. J. Nanobiotechnology 2022, 20, 190. [Google Scholar] [CrossRef] [PubMed]
- Jiang, D.; Gao, T.; Liang, S.; Mu, W.; Fu, S.; Liu, Y.; Yang, R.; Zhang, Z.; Liu, Y.; Zhang, N. Lymph Node Delivery Strategy Enables the Activation of Cytotoxic T Lymphocytes and Natural Killer Cells to Augment Cancer Immunotherapy. ACS Appl. Mater. Interfaces 2021, 13, 22213–22224. [Google Scholar] [CrossRef]
- Shi, G.-N.; Zhang, C.-N.; Xu, R.; Niu, J.-F.; Song, H.-J.; Zhang, X.-Y.; Wang, W.-W.; Wang, Y.-M.; Li, C.; Wei, X.-Q.; et al. Enhanced antitumor immunity by targeting dendritic cells with tumor cell lysate-loaded chitosan nanoparticles vaccine. Biomaterials 2017, 113, 191–202. [Google Scholar] [CrossRef]
- Yang, X.; Yu, T.; Zeng, Y.; Lian, K.; Zhou, X.; Li, S.; Qiu, G.; Jin, X.; Yuan, H.; Hu, F. Tumor-draining lymph node targeting chitosan micelles as antigen-capturing adjuvants for personalized immunotherapy. Carbohydr. Polym. 2020, 240, 116270. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.; Shi, G.; Zhang, J.; Song, H.; Niu, J.; Shi, S.; Huang, P.; Wang, Y.; Wang, W.; Li, C.; et al. Targeted antigen delivery to dendritic cell via functionalized alginate nanoparticles for cancer immunotherapy. J. Control. Release 2017, 256, 170–181. [Google Scholar] [CrossRef] [PubMed]
- Lv, S.; Song, K.; Yen, A.; Peeler, D.J.; Nguyen, D.C.; Olshefsky, A.; Sylvestre, M.; Srinivasan, S.; Stayton, P.S.; Pun, S.H. Well-Defined Mannosylated Polymer for Peptide Vaccine Delivery with Enhanced Antitumor Immunity. Adv. Healthc. Mater. 2022, 11, 2101651. [Google Scholar] [CrossRef]
- Zeng, Z.; Pu, K. Improving Cancer Immunotherapy by Cell Membrane-Camouflaged Nanoparticles. Adv. Funct. Mater. 2020, 30, 2004397. [Google Scholar] [CrossRef]
- Guo, Y.; Wang, D.; Song, Q.; Wu, T.; Zhuang, X.; Bao, Y.; Kong, M.; Qi, Y.; Tan, S.; Zhang, Z. Erythrocyte Membrane-Enveloped Polymeric Nanoparticles as Nanovaccine for Induction of Antitumor Immunity against Melanoma. ACS Nano 2015, 9, 6918–6933. [Google Scholar] [CrossRef]
- Yang, R.; Xu, J.; Xu, L.; Sun, X.; Chen, Q.; Zhao, Y.; Peng, R.; Liu, Z. Cancer Cell Membrane-Coated Adjuvant Nanoparticles with Mannose Modification for Effective Anticancer Vaccination. ACS Nano 2018, 12, 5121–5129. [Google Scholar] [CrossRef]
- Yang, X.; Yu, T.; Zeng, Y.; Lian, K.; Zhou, X.; Ke, J.; Li, Y.; Yuan, H.; Hu, F. pH-Responsive Biomimetic Polymeric Micelles as Lymph NodeTargeting Vaccines for Enhanced Antitumor Immune Responses. Biomacromolecules 2020, 21, 2818–2828. [Google Scholar] [CrossRef]
- Xiao, P.; Wang, J.; Zhao, Z.; Liu, X.; Sun, X.; Wang, D.; Li, Y. Engineering Nanoscale Artificial Antigen-Presenting Cells by Metabolic Dendritic Cell Labeling to Potentiate Cancer Immunotherapy. Nano Lett. 2021, 21, 2094–2103. [Google Scholar] [CrossRef] [PubMed]
- Liu, W.-L.; Zou, M.-Z.; Liu, T.; Zeng, J.-Y.; Li, X.; Yu, W.-Y.; Li, C.-X.; Ye, J.-J.; Song, W.; Feng, J.; et al. Cytomembrane nanovaccines show therapeutic effects by mimicking tumor cells and antigen presenting cells. Nat. Commun. 2019, 10, 3199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, J.; Liu, F.; Sheu, W.C.; Meng, Z.; Xie, Y.; Xu, H.; Li, M.; Chen, A.T.; Liu, J.; Bao, Y.; et al. Copresentation of Tumor Antigens and Costimulatory Molecules via Biomimetic Nanoparticles for Effective Cancer Immunotherapy. Nano Lett. 2020, 20, 4084–4094. [Google Scholar] [CrossRef] [PubMed]
- Herrmann, I.K.; Wood, M.J.A.; Fuhrmann, G. Extracellular vesicles as a next-generation drug delivery platform. Nat. Nanotech. 2021, 16, 748–759. [Google Scholar] [CrossRef] [PubMed]
- Phung, C.D.; Pham, T.T.; Nguyen, H.T.; Nguyen, T.T.; Ou, W.; Jeong, J.-H.; Choi, H.-G.; Ku, S.K.; Yong, C.S.; Kim, J.O. Anti-CTLA-4 antibody-functionalized dendritic cell-derived exosomes targeting tumor-draining lymph nodes for effective induction of antitumor T-cell responses. Acta Biomater. 2020, 115, 371–382. [Google Scholar] [CrossRef]
- Cheng, L.; Wang, Y.; Huang, L. Exosomes from M1-Polarized Macrophages Potentiate the Cancer Vaccine by Creating a Proinflammatory Microenvironment in the Lymph Node. Mol. Ther. 2017, 25, 1665–1675. [Google Scholar] [CrossRef] [Green Version]
- Schudel, A.; Chapman, A.P.; Yau, M.-K.; Higginson, C.J.; Francis, D.M.; Manspeaker, M.P.; Avecilla, A.R.C.; Rohner, N.A.; Finn, M.G.; Thomas, S.N. Programmable multistage drug delivery to lymph nodes. Nat. Nanotech. 2020, 15, 491–499. [Google Scholar] [CrossRef]
- Xue, X.; Huang, Y.; Bo, R.; Jia, B.; Wu, H.; Yuan, Y.; Wang, Z.; Ma, Z.; Jing, D.; Xu, X.; et al. Trojan Horse nanotheranostics with dual transformability and multifunctionality for highly effective cancer treatment. Nat. Commun. 2018, 9, 3653. [Google Scholar] [CrossRef] [Green Version]
- Li, H.-J.; Du, J.-Z.; Liu, J.; Du, X.-J.; Shen, S.; Zhu, Y.-H.; Wang, X.; Ye, X.; Nie, S.; Wang, J. Smart Superstructures with Ultrahigh pHSensitivity for Targeting Acidic Tumor Microenvironment: Instantaneous Size Switching and Improved Tumor Penetration. ACS Nano 2016, 10, 6753–6761. [Google Scholar] [CrossRef]
- Chen, B.; Dai, W.; He, B.; Zhang, H.; Wang, X.; Wang, Y.; Zhang, Q. Current Multistage Drug Delivery Systems Based on the Tumor Microenvironment. Theranostics 2017, 7, 538–558. [Google Scholar] [CrossRef] [PubMed]
- Huynh, V.; Jesmer, A.H.; Shoaib, M.M.; D’Angelo, A.D.; Rullo, A.F.; Wylie, R.G. Improved Efficacy of Antibody Cancer Immunotherapeutics through Local and Sustained Delivery. ChemBioChem 2019, 20, 747–753. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Ye, T.; Ye, C.; Wan, C.; Yuan, S.; Liu, Y.; Li, T.; Jiang, F.; Lovell, J.F.; Jin, H.; et al. Secretions from hypochlorous acid-treated tumor cells delivered in a melittin hydrogel potentiate cancer immunotherapy. Bioact. Mater. 2022, 9, 541–553. [Google Scholar] [CrossRef] [PubMed]
- Cao, Y.; Zhou, Y.; Pan, J.; Zhong, X.; Ding, J.; Jing, X.; Sun, S.K. A general strategy towards an injectable microwave-sensitive immune hydrogel for combined percutaneous microwave ablation and immunotherapy. Chem. Eng. J. 2021, 422, 130111. [Google Scholar] [CrossRef]
- Chen, Q.; Zhou, S.; Ding, Y.; Chen, D.; Dahiru, N.S.; Tang, H.; Xu, H.; Ji, M.; Wang, X.; Li, Z.; et al. A bio-responsive, cargo-catchable gel for postsurgical tumor treatment via ICD-based immunotherapy. J. Control. Release 2022, 346, 212–225. [Google Scholar] [CrossRef]
- Jin, H.; Wan, C.; Zou, Z.; Zhao, G.; Zhang, L.; Geng, Y.; Chen, T.; Huang, A.; Jiang, F.; Feng, J.-P.; et al. Tumor Ablation and Therapeutic Immunity Induction by an Injectable Peptide Hydrogel. ACS Nano 2018, 12, 3295–3310. [Google Scholar] [CrossRef]
- Yang, F.; Shi, K.; Hao, Y.; Jia, Y.; Liu, Q.; Chen, Y.; Pan, M.; Yuan, L.; Yu, Y.; Qian, Z. Cyclophosphamide loaded thermo-responsive hydrogel system synergize with a hydrogel cancer vaccine to amplify cancer immunotherapy in a prime-boost manner. Bioact. Mater. 2021, 6, 3036–3048. [Google Scholar] [CrossRef]
- Lee, J.; Le, Q.-V.; Yang, G.; Oh, Y.-K. Improved Efficacy of Antibody Cancer Immunotherapeutics through Local and Sustained Delivery. Biomaterials 2019, 218, 119359. [Google Scholar] [CrossRef]
- He, H.; Fei, Z.; Guo, T.; Hou, Y.; Li, D.; Wang, K.; Ren, F.; Fan, K.; Zhou, D.; Xie, C.; et al. Bioadhesive injectable hydrogel with phenolic carbon quantum dot supported Pd single atom nanozymes as a localized immunomodulation niche for cancer catalytic immunotherapy. Biomaterials 2022, 280, 121272. [Google Scholar] [CrossRef]
- Huo, W.; Yang, X.; Wang, B.; Cao, L.; Fang, Z.; Li, Z.; Liu, H.; Liang, X.-j.; Zhang, J.; Jin, Y. Biomineralized hydrogel DC vaccine for cancer immunotherapy: A boosting strategy via improving immunogenicity and reversing immune-inhibitory microenvironment. Biomaterials 2022, 288, 121722. [Google Scholar] [CrossRef]
- Wang, X.; Ye, L.; He, W.; Teng, C.; Sun, S.; Lu, H.; Li, S.; Lv, L.; Cao, X.; Yin, H.; et al. In situ targeting nanoparticles-hydrogel hybrid system for combined chemo-immunotherapy of glioma. J. Control. Release 2022, 345, 786–797. [Google Scholar] [CrossRef] [PubMed]
- Ren, X.; Wang, N.; Zhou, Y.; Song, A.; Jin, G.; Li, Z.; Luan, Y. An injectable hydrogel using an immunomodulating gelator for amplified tumor immunotherapy by blocking the arginase pathway. Acta Biomater. 2021, 124, 179–190. [Google Scholar] [CrossRef] [PubMed]
- Ding, M.; Fan, Y.; Lv, Y.; Liu, J.; Yu, N.; Kong, D.; Sun, H.; Li, J. A prodrug hydrogel with tumor microenvironment and near-infrared light dual-responsive action for synergistic cancer immunotherapy. Acta Biomater. 2022, 149, 334–346. [Google Scholar] [CrossRef] [PubMed]
- Wan, C.; Sun, Y.; Hu, Y.; Huang, J.; Lu, L.; Gao, Y.; Zi, H.; He, Q.; Sun, J.; Lovell, J.F.; et al. Peptide hydrogels loaded with irradiated tumor cell secretions enhance cancer immunotherapy. Nano Today 2021, 41, 101323. [Google Scholar] [CrossRef]
- Xiao, Z.; Wang, D.; Wang, C.; Chen, Z.; Huang, C.; Yang, Y.; Xie, L.; Zhang, L.; Xu, L.; Zhang, M.-R.; et al. PEIGel: A biocompatible and injectable scaffold with innate immune adjuvanticity for synergized local immunotherapy. Mater. Today Bio 2022, 15, 100297. [Google Scholar] [CrossRef]
- Qi, J.; Ding, T.; Liu, T.; Xia, X.; Wu, S.; Liu, J.; Chen, Q.; Zhang, D.; Zhao, H. Inosine-Based Supramolecular Hydrogel for Highly Efficient PD-L1 Blockade Therapy via Mediating CD8+ T Cells. Adv. Funct. Mater. 2022, 32, 2204273. [Google Scholar] [CrossRef]
- Sun, S.; Gu, W.; Wu, H.; Zhao, Q.; Qian, S.; Xiao, H.; Yang, K.; Liu, J.; Jin, Y.; Hu, C.; et al. Immunostimulant In Situ Hydrogel Improves Synergetic Radioimmunotherapy of Malignant Glioblastoma Relapse Post-Resection. Adv. Funct. Mater. 2022, 32, 2205038. [Google Scholar] [CrossRef]
- Si, X.; Ji, G.; Ma, S.; Xu, Y.; Zhao, J.; Zhang, Y.; Huang, Z.; Tang, Z.; Song, W.; Chen, X. In–Situ-Sprayed Dual-Functional Immunotherapeutic Gel for Colorectal Cancer Postsurgical Treatment. Adv. Healthc. Mater. 2021, 10, 2100862. [Google Scholar] [CrossRef]
- Meng, Z.; Zhou, X.; Xu, J.; Han, X.; Dong, Z.; Wang, H.; Zhang, Y.; She, J.; Xu, L.; Wang, C.; et al. Light-Triggered In Situ Gelation to Enable Robust Photodynamic-Immunotherapy by Repeated Stimulations. Adv. Mater. 2019, 31, 1900927. [Google Scholar] [CrossRef]
- Sun, L.; Shen, F.; Tian, L.; Tao, H.; Xiong, Z.; Xu, J.; Liu, Z. ATP-Responsive Smart Hydrogel Releasing Immune Adjuvant Synchronized with Repeated Chemotherapy or Radiotherapy to Boost Antitumor Immunity. Adv. Mater. 2021, 33, 2007910. [Google Scholar] [CrossRef]
- Li, S.; Zhu, C.; Zhou, X.; Chen, L.; Bo, X.; Shen, Y.; Guan, X.; Han, X.; Shan, D.; Sun, L.; et al. Engineering ROS-Responsive Bioscaffolds for Disrupting Myeloid Cell-Driven Immunosuppressive Niche to Enhance PD-L1 Blockade-Based Postablative Immunotherapy. Adv. Sci. 2022, 9, 2104619. [Google Scholar] [CrossRef] [PubMed]
- Wang, F.; Xu, D.; Su, H.; Zhang, W.; Sun, X.; Monroe, M.K.; Chakroun, R.W.; Wang, Z.; Dai, W.; Oh, R.; et al. Supramolecular prodrug hydrogelator as an immune booster for checkpoint blocker–based immunotherapy. Sci. Adv. 2020, 6, eaaz8985. [Google Scholar] [CrossRef] [PubMed]
- Gu, J.; Zhao, G.; Yu, J.; Xu, P.; Yan, J.; Jin, Z.; Chen, S.; Wang, Y.; Zhang, L.W.; Wang, Y. Injectable pH-responsive hydrogel for combinatorial chemoimmunotherapy tailored to the tumor microenvironment. J. Nanobiotechnology 2022, 20, 372. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Ding, Y.; Liu, J.; Wang, J.; Mo, F.; Wang, Y.; Chen-Mayfield, T.-J.; Sondel, P.M.; Hong, S.; Hu, Q. Depletion of tumor associated macrophages enhances local and systemic platelet-mediated anti-PD-1 delivery for post-surgery tumor recurrence treatment. Nat. Commun. 2022, 13, 1845. [Google Scholar] [CrossRef] [PubMed]
- Wang, F.; Su, H.; Xu, D.; Dai, W.; Zhang, W.; Wang, Z.; Anderson, C.F.; Zheng, M.; Oh, R.; Wan, F.; et al. Tumour sensitization via the extended intratumoural release of a STING agonist and camptothecin from a self-assembled hydrogel. Nat. Biomed. Eng. 2020, 4, 1090–1101. [Google Scholar] [CrossRef]
- Park, J.; Wrzesinski, S.H.; Stern, E.; Look, M.; Criscione, J.; Ragheb, R.; Jay, S.M.; Demento, S.L.; Agawu, A.; Limon, P.L.; et al. Combination delivery of TGF-β inhibitor and IL-2 by nanoscale liposomal polymeric gels enhances tumour immunotherapy. Nat. Mater. 2012, 11, 895–905. [Google Scholar] [CrossRef]
- Hou, X.-l.; Dai, X.; Yang, J.; Zhang, B.; Zhao, D.-h.; Li, C.-q.; Yin, Z.-y.; Zhao, Y.-d.; Liu, B. Injectable polypeptide-engineered hydrogel depot for amplifying the anti-tumor immune effect induced by chemo-photothermal therapy. J. Mater. Chem. B 2020, 8, 8623–8633. [Google Scholar] [CrossRef]
- Yan, J.; Zhang, Z.; Zhan, X.; Chen, K.; Pu, Y.; Liang, Y.; He, B. In situ injection of dual-delivery PEG based MMP-2 sensitive hydrogels for enhanced tumor penetration and chemo-immune combination therapy. Nanoscale 2021, 13, 9577–9589. [Google Scholar] [CrossRef]
- Song, H.; Yang, P.; Huang, P.; Zhang, C.; Kong, D.; Wang, W. Injectable polypeptide hydrogel-based co-delivery of vaccine and immune checkpoint inhibitors improves tumor immunotherapy. Theranostics 2019, 9, 2299–2314. [Google Scholar] [CrossRef]
- Dai, X.; Meng, J.; Deng, S.; Zhang, L.; Wan, C.; Lu, L.; Huang, J.; Hu, Y.; Zhang, Z.; Li, Y.; et al. Targeting CAMKII to reprogram tumor-associated macrophages and inhibit tumor cells for cancer immunotherapy with an injectable hybrid peptide hydrogel. Theranostics 2020, 10, 3049–3063. [Google Scholar] [CrossRef]
- Liang, X.; Li, L.; Li, X.; He, T.; Gong, S.; Zhu, S.; Zhang, M.; Wu, Q.; Gong, C. A spontaneous multifunctional hydrogel vaccine amplifies the innate immune response to launch a powerful antitumor adaptive immune response. Theranostics 2021, 11, 6936–6949. [Google Scholar] [CrossRef] [PubMed]
- Li, C.; Liu, Y.; Li, D.; Wang, Q.; Zhou, S.; Zhang, H.; Wang, Y.; He, Z.; Liu, H.; Sun, J. Promising alternatives of CD47 monoclonal antibody: An injectable degradable hydrogel loaded with PQ912 for postoperative immunotherapy effectively blocks CD47-SIRPα signal. Theranostics 2022, 12, 4581–4598. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Feng, Z.; Liu, J.; Li, H.; Su, Q.; Zhang, J.; Huang, P.; Wang, W.; Liu, J. Polarization of tumor-associated macrophages by TLR7/8 conjugated radiosensitive peptide hydrogel for overcoming tumor radioresistance. Bioact. Mater. 2022, 16, 359–371. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.; Cao, Z.; Zhang, R.; Chen, Y.; Yang, X. Injectable Supramolecular Hydrogel for Locoregional Immune Checkpoint Blockade and Enhanced Cancer Chemo-Immunotherapy. ACS Appl. Mater. Interfaces 2021, 13, 33874–33884. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Liu, K.; Gong, Y.; Zhu, W.; Zhu, J.; Pan, F.; Chao, Y.; Xiao, Z.; Liu, Y.; Wang, X.; et al. Vitamin C supramolecular hydrogel for enhanced cancer immunotherapy. Biomaterials 2022, 287, 121673. [Google Scholar] [CrossRef]
- Wu, Y.; Li, Q.; Shim, G.; Oh, Y.-K. Melanin-loaded CpG DNA hydrogel for modulation of tumor immune microenvironment. J.Control. Release 2021, 330, 540–553. [Google Scholar] [CrossRef]
- Kremenovic, M.; Chan, A.A.; Feng, B.; Bariswyl, L.; Robatel, S.; Gruber, T.; Tang, L.; Lee, D.J.; Schenk, M. BCG hydrogel promotes CTSS-mediated antigen processing and presentation, thereby suppressing metastasis and prolonging survival in melanoma. J. Immunother. Cancer 2022, 10, e004133. [Google Scholar] [CrossRef]
- Li, Y.; Fang, M.; Zhang, J.; Wang, J.; Song, Y.; Shi, J.; Li, W.; Wu, G.; Ren, J.; Wang, Z.; et al. Hydrogel dual delivered celecoxib and anti-PD-1 synergistically improve antitumor immunity. Oncoimmunology 2016, 5, e1074374. [Google Scholar] [CrossRef]
- Wang, B.; Chen, J.; Caserto, J.S.; Wang, X.; Ma, M. An in situ hydrogel-mediated chemo-immunometabolic cancer therapy. Nat. Commun. 2022, 13, 3821. [Google Scholar] [CrossRef]
- He, T.; Liang, X.; Li, L.; Gong, S.; Li, X.; Zhang, M.; Zhu, S.; Xiao, H.; Wu, Q.; Gong, C. A spontaneously formed and self-adjuvanted hydrogel vaccine triggers strong immune responses. Mater. Des. 2021, 197, 109232. [Google Scholar] [CrossRef]
- Song, H.; Huang, P.; Niu, J.; Shi, G.; Zhang, C.; Kong, D.; Wang, W. Injectable polypeptide hydrogel for dual-delivery of antigen and TLR3 agonist to modulate dendritic cells in vivo and enhance potent cytotoxic T-lymphocyte response against melanoma. Biomaterials 2018, 159, 119–129. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Q.; Wang, Y.; Zhao, B.; Chen, H.; Cai, Z.; Zheng, Y.; Zeng, Y.; Zhang, D.; Liu, X. Neoantigen Immunotherapeutic-Gel Combined with TIM-3 Blockade Effectively Restrains Orthotopic Hepatocellular Carcinoma Progression. Nano Lett. 2022, 22, 2048–2058. [Google Scholar] [CrossRef] [PubMed]
- Yin, Y.; Li, X.; Ma, H.; Zhang, J.; Yu, D.; Zhao, R.; Yu, S.; Nie, G.; Wang, H. In Situ Transforming RNA Nanovaccines from Polyethylenimine Functionalized Graphene Oxide Hydrogel for Durable Cancer Immunotherapy. Nano Lett. 2021, 21, 2224–2231. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.; Francis, D.M.; Sestito, L.F.; Archer, P.A.; Manspeaker, M.P.; O’Melia, M.J.; Thomas, S.N. Thermosensitive hydrogel releasing nitric oxide donor and anti-CTLA-4 micelles for anti-tumor immunotherapy. Nat. Commun. 2022, 13, 1479. [Google Scholar] [CrossRef]
- O’Melia, M.J.; Manspeaker, M.P.; Thomas, S.N. Tumor-draining lymph nodes are survival niches that support T cell priming against lymphatic transported tumor antigen and effects of immune checkpoint blockade in TNBC. Cancer Immunol. Immunother. 2021, 70, 2179. [Google Scholar] [CrossRef] [PubMed]
- Engelhard, V.H.; Rodriguez, A.B.; Mauldin, I.S.; Woods, A.N.; Peske, J.D.; Slingluff, C.L., Jr. Immune Cell Infiltration and Tertiary Lymphoid Structures as Determinants of Antitumor Immunity. J. Immunol. 2018, 200, 432–442. [Google Scholar] [CrossRef] [Green Version]
- Joshi, N.S.; Akama-Garren, E.H.; Lu, Y.; Lee, D.-Y.; Chang, G.P.; Li, A.; DuPage, M.; Tammela, T.; Kerper, N.R.; Farago, A.F.; et al. Regulatory T Cells in Tumor-Associated Tertiary Lymphoid Structures Suppress Anti-tumor T Cell Responses. Immunity 2015, 43, 579–590. [Google Scholar] [CrossRef] [Green Version]
- Mohamed, L.; Elsaka, A.; Zamzam, Y.; Elkady, A. Tumor Infiltrating Lymphocytes and Tertiary Lymphoid Structure as Prognostic and Predictive Factor for Neoadjuvant Chemotherapy in Stage II & III Breast Cancer. Arch. Can. Res. 2018, 6, 17. [Google Scholar]
- Tseng, Y.-C.; Xu, Z.; Guley, K.; Yuan, H.; Huang, L. Lipidecalcium phosphate nanoparticles for delivery to the lymphatic system and SPECT/CT imaging of lymph node metastases. Biomaterials 2014, 35, 4688–4698. [Google Scholar] [CrossRef] [Green Version]
- Videira, M.A.; Botelho, M.F.; Santos, A.C.; Gouveia, L.F.; Pedroso De Lima, J.J.; Almeida, A.J. Lymphatic Uptake of Pulmonary Delivered Radiolabelled Solid Lipid Nanoparticles. J. Drug Target. 2002, 10, 607–613. [Google Scholar] [CrossRef]
- Mohammad, A.K.; Amayreh, L.K.; Mazzara, J.M.; Reineke, J.J. Rapid Lymph Accumulation of Polystyrene Nanoparticles Following Pulmonary Administration. Pharm. Res. 2013, 30, 424–434. [Google Scholar] [CrossRef] [PubMed]
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
Cho, K.J.; Cho, Y.-E.; Kim, J. Locoregional Lymphatic Delivery Systems Using Nanoparticles and Hydrogels for Anticancer Immunotherapy. Pharmaceutics 2022, 14, 2752. https://doi.org/10.3390/pharmaceutics14122752
Cho KJ, Cho Y-E, Kim J. Locoregional Lymphatic Delivery Systems Using Nanoparticles and Hydrogels for Anticancer Immunotherapy. Pharmaceutics. 2022; 14(12):2752. https://doi.org/10.3390/pharmaceutics14122752
Chicago/Turabian StyleCho, Kyeong Jin, Young-Eun Cho, and Jihoon Kim. 2022. "Locoregional Lymphatic Delivery Systems Using Nanoparticles and Hydrogels for Anticancer Immunotherapy" Pharmaceutics 14, no. 12: 2752. https://doi.org/10.3390/pharmaceutics14122752
APA StyleCho, K. J., Cho, Y. -E., & Kim, J. (2022). Locoregional Lymphatic Delivery Systems Using Nanoparticles and Hydrogels for Anticancer Immunotherapy. Pharmaceutics, 14(12), 2752. https://doi.org/10.3390/pharmaceutics14122752