Pardoll DM. The blockade of immune checkpoints in most cancers immunotherapy. Nat Rev Most cancers. 2012;12(4):252–64.
Shah NN, Fry TJ. Mechanisms of resistance to CAR T cell remedy. Nat Rev Clin Oncol. 2019;16(6):372–85.
Saxena M, et al. Therapeutic most cancers vaccines. Nat Rev Most cancers. 2021;21(6):360–78.
Zhao Y, et al. Nanotechnology-based immunotherapies to fight most cancers metastasis. Mol Biol Rep. 2021;48(9):6563–80.
Chen F, et al. Nanobiomaterial-based vaccination immunotherapy of most cancers. Biomaterials. 2021;270: 120709.
Wang Q, et al. Lymph node-targeting nanovaccines for most cancers immunotherapy. J Management Launch. 2022;351:102–22.
Hollingsworth RE, Jansen Ok. Turning the nook on therapeutic most cancers vaccines. NPJ Vaccines. 2019;4:7.
Cai T, et al. Supply of nanovaccine in the direction of lymphoid organs: latest methods in enhancing most cancers immunotherapy. J Nanobiotechnol. 2021;19(1):389.
Najibi AJ, Mooney DJ. Cell and tissue engineering in lymph nodes for most cancers immunotherapy. Adv Drug Deliv Rev. 2020;161–162:42–62.
Goldberg MS. Enhancing most cancers immunotherapy by nanotechnology. Nat Rev Most cancers. 2019;19(10):587–602.
Zhang Y, et al. Nanovaccines for most cancers immunotherapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2019;11(5): e1559.
Li SR, et al. Current advances in porous nanomaterials-based drug supply programs for most cancers immunotherapy. J Nanobiotechnol. 2022;20(1):277.
Sharma R, et al. Growth, characterization and ex vivo evaluation of lipid-polymer primarily based nanocomposite(s) as a possible service for site-specific supply of immunogenic molecules. J Drug Deliv Sci Technol. 2019;51:310–9.
Dong H, et al. Biomaterials facilitating dendritic cell-mediated most cancers immunotherapy. Adv Sci (Weinh). 2023;10(18): e2301339.
Tian R, et al. Multimodal stratified imaging of nanovaccines in lymph nodes for enhancing most cancers immunotherapy. Adv Drug Deliv Rev. 2020;161–162:145–60.
Sestito LF, et al. Lymphatic-draining nanoparticles ship Bay K8644 payload to lymphatic vessels and improve their pumping operate. Sci Adv. 2023;9(8): eabq0435.
Zahin N, et al. Nanoparticles and its biomedical purposes in well being and ailments: particular concentrate on drug supply. Environ Sci Pollut Res Int. 2020;27(16):19151–68.
Girard JP, Moussion C, Förster R. HEVs, lymphatics and homeostatic immune cell trafficking in lymph nodes. Nat Rev Immunol. 2012;12(11):762–73.
Sainte-Marie G. The lymph node revisited: improvement, morphology, functioning, and function in triggering major immune responses. Anat Rec (Hoboken). 2010;293(2):320–37.
Jalkanen S, Salmi M. Lymphatic endothelial cells of the lymph node. Nat Rev Immunol. 2020;20(9):566–78.
Davis MJ, et al. Determinants of valve gating in accumulating lymphatic vessels from rat mesentery. Am J Physiol Coronary heart Circ Physiol. 2011;301(1):H48-60.
Scallan JP, et al. Lymphatic pumping: mechanics, mechanisms and malfunction. J Physiol. 2016;594(20):5749–68.
Kim J, Archer PA, Thomas SN. Improvements in lymph node concentrating on nanocarriers. Semin Immunol. 2021;56: 101534.
Roozendaal R, Mebius RE, Kraal G. The conduit system of the lymph node. Int Immunol. 2008;20(12):1483–7.
Ager A. Excessive endothelial venules and different blood vessels: important regulators of lymphoid organ improvement and performance. Entrance Immunol. 2017;8:45.
Baekkevold ES, et al. The CCR7 ligand elc (CCL19) is transcytosed in excessive endothelial venules and mediates T cell recruitment. J Exp Med. 2001;193(9):1105–12.
Drayton DL, et al. Lymphoid organ improvement: from ontogeny to neogenesis. Nat Immunol. 2006;7(4):344–53.
Gretz JE, et al. Lymph-borne chemokines and different low molecular weight molecules attain excessive endothelial venules by way of specialised conduits whereas a useful barrier limits entry to the lymphocyte microenvironments in lymph node cortex. J Exp Med. 2000;192(10):1425–40.
du Bois H, Heim TA, Lund AW. Tumor-draining lymph nodes: On the crossroads of metastasis and immunity. Sci Immunol. 2021;6(63): eabg3551.
Koukourakis MI, Giatromanolaki A. Tumor draining lymph nodes, immune response, and radiotherapy: in the direction of a revisal of therapeutic ideas. Biochim Biophys Acta Rev Most cancers. 2022;1877(3): 188704.
Sautes-Fridman C, et al. Tertiary lymphoid buildings within the period of most cancers immunotherapy. Nat Rev Most cancers. 2019;19(6):307–25.
Lin L, et al. Tertiary lymphoid organs in most cancers immunology: mechanisms and the brand new technique for immunotherapy. Entrance Immunol. 2019;10:1398.
Picker LJ, Butcher EC. Physiological and molecular mechanisms of lymphocyte homing. Annu Rev Immunol. 1992;10:561–91.
Ngo VN, et al. Lymphotoxin alpha/beta and tumor necrosis issue are required for stromal cell expression of homing chemokines in B and T cell areas of the spleen. J Exp Med. 1999;189(2):403–12.
Joshi NS, et al. Regulatory T cells in tumor-associated tertiary lymphoid buildings suppress anti-tumor T cell responses. Immunity. 2015;43(3):579–90.
Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39(1):1–10.
Johanns TM, et al. Endogenous neoantigen-specific CD8 T cells recognized in two glioblastoma fashions utilizing a most cancers immunogenomics strategy. Most cancers Immunol Res. 2016;4(12):1007–15.
Roberts EW, et al. Essential function for CD103(+)/CD141(+) dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma. Most cancers Cell. 2016;30(2):324–36.
Salmon H, et al. Growth and activation of CD103(+) dendritic cell progenitors on the tumor website enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity. 2016;44(4):924–38.
Motz GT, Coukos G. Deciphering and reversing tumor immune suppression. Immunity. 2013;39(1):61–73.
Chen DS, Mellman I. Components of most cancers immunity and the cancer-immune set level. Nature. 2017;541(7637):321–30.
Workel HH, et al. A transcriptionally distinct CXCL13(+)CD103(+)CD8(+) T-cell inhabitants is related to B-cell recruitment and neoantigen load in human most cancers. Most cancers Immunol Res. 2019;7(5):784–96.
Cabrita R, et al. Tertiary lymphoid buildings enhance immunotherapy and survival in melanoma. Nature. 2020;577(7791):561–5.
Maldonado L, et al. Intramuscular therapeutic vaccination concentrating on HPV16 induces T cell responses that localize in mucosal lesions. Sci Transl Med. 2014;6(221):221ra13.
Rafiq S, Hackett CS, Brentjens RJ. Engineering methods to beat the present roadblocks in CAR T cell remedy. Nat Rev Clin Oncol. 2020;17(3):147–67.
Liu H, et al. Therapeutic nanovaccines sensitize EBV-associated tumors to checkpoint blockade remedy. Biomaterials. 2020;255: 120158.
Palucka Ok, Banchereau J. Dendritic-cell-based therapeutic most cancers vaccines. Immunity. 2013;39(1):38–48.
Wang J, et al. Alternative of nanovaccine supply mode has profound impacts on the intralymph node spatiotemporal distribution and immunotherapy efficacy. Adv Sci (Weinh). 2020;7(19):2001108.
Tumeh PC, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515(7528):568–71.
Siddiqui I, et al. Intratumoral Tcf1(+)PD-1(+)CD8(+) T cells with stem-like properties promote tumor management in response to vaccination and checkpoint blockade immunotherapy. Immunity. 2019;50(1):195–211.
Jewell CM, Lopez SC, Irvine DJ. In situ engineering of the lymph node microenvironment by way of intranodal injection of adjuvant-releasing polymer particles. Proc Natl Acad Sci U S A. 2011;108(38):15745–50.
Tagawa ST, et al. Section I research of intranodal supply of a plasmid DNA vaccine for sufferers with Stage IV melanoma. Most cancers. 2003;98(1):144–54.
Spaner DE, et al. Enhanced viral and tumor immunity with intranodal injection of canary pox viruses expressing the melanoma antigen, gp100. Most cancers. 2006;106(4):890–9.
Mohanan D, et al. Administration routes have an effect on the standard of immune responses: a cross-sectional analysis of particulate antigen-delivery programs. J Management Launch. 2010;147(3):342–9.
Jiang H, Wang Q, Solar X. Lymph node concentrating on methods to enhance vaccination efficacy. J Management Launch. 2017;267:47–56.
Chen Y, De Koker S, De Geest BG. Engineering methods for lymph node focused immune activation. Acc Chem Res. 2020;53(10):2055–67.
Schudel A, Francis DM, Thomas SN. Materials design for lymph node drug supply. Nat Rev Mater. 2019;4(6):415–28.
Zhang L, et al. Focused codelivery of an antigen and twin agonists by hybrid nanoparticles for enhanced most cancers immunotherapy. Nano Lett. 2019;19(7):4237–49.
Kramer S, et al. HPMA-based nanocarriers for efficient immune system stimulation. Macromol Biosci. 2019;19(6): e1800481.
Irvine DJ, et al. Artificial nanoparticles for vaccines and immunotherapy. Chem Rev. 2015;115(19):11109–46.
Nakamura T, et al. The impact of measurement and cost of lipid nanoparticles ready by microfluidic mixing on their lymph node transitivity and distribution. Mol Pharm. 2020;17(3):944–53.
Zeng Q, et al. Cationic micelle supply of Trp2 peptide for environment friendly lymphatic draining and enhanced cytotoxic T-lymphocyte responses. J Management Launch. 2015;200:1–12.
Kiss E, Bertoti I, Vargha-Butler EI. XPS and wettability characterization of modified poly(lactic acid) and poly(lactic/glycolic acid) movies. J Colloid Interface Sci. 2002;245(1):91–8.
Luo M, et al. Artificial nanovaccines for immunotherapy. J Management Launch. 2017;263:200–10.
Randolph GJ, Angeli V, Swartz MA. Dendritic-cell trafficking to lymph nodes by lymphatic vessels. Nat Rev Immunol. 2005;5(8):617–28.
Niikura Ok, et al. Gold nanoparticles as a vaccine platform: affect of measurement and form on immunological responses in vitro and in vivo. ACS Nano. 2013;7(5):3926–38.
Li Q, et al. Elastic nanovaccine enhances dendritic cell-mediated tumor immunotherapy. Small. 2022;18(32): e2201108.
Wculek SK, et al. Dendritic cells in most cancers immunology and immunotherapy. Nat Rev Immunol. 2020;20(1):7–24.
Sharma R, Vyas SP. Mannose functionalized plain and endosomolytic nanocomposite(s)-based strategy for the induction of efficient antitumor immune response in C57BL/6 mice melanoma mannequin. Drug Dev Ind Pharm. 2019;45(7):1089–100.
Sharma R, et al. Launch promoter-based systematically designed nanocomposite(s): a novel strategy for site-specific supply of tumor-associated antigen(s) (TAAs). Artif Cells Nanomed Biotechnol. 2018;46(sup2):776–89.
Ke X, et al. Bodily and chemical profiles of nanoparticles for lymphatic concentrating on. Adv Drug Deliv Rev. 2019;151–152:72–93.
Wiig H, Swartz MA. Interstitial fluid and lymph formation and transport: physiological regulation and roles in irritation and most cancers. Physiol Rev. 2012;92(3):1005–60.
Stylianopoulos T, et al. Diffusion of particles within the extracellular matrix: the impact of repulsive electrostatic interactions. Biophys J. 2010;99(5):1342–9.
Dong H, et al. Polyethyleneimine modification of aluminum hydroxide nanoparticle enhances antigen transportation and cross-presentation of dendritic cells. Int J Nanomed. 2018;13:3353–65.
Zhang Y, et al. Endogenous/exogenous nanovaccines synergistically improve dendritic cell-mediated tumor immunotherapy. Adv Healthc Mater. 2023;584: e2203028.
Nishimoto Y, et al. Carboxyl-, sulfonyl-, and phosphate-terminal dendrimers as a nanoplatform with lymph node concentrating on. Int J Pharm. 2020;576: 119021.
Pasut G, Veronese FM. Polymer–drug conjugation, latest achievements and common methods. Prog Polym Sci. 2007;32(8–9):933–61.
Knop Ok, et al. Poly(ethylene glycol) in drug supply: execs and cons in addition to potential options. Angew Chem Int Ed Engl. 2010;49(36):6288–308.
Stroh M, et al. Multiphoton microscopy guides neurotrophin modification with poly(ethylene glycol) to reinforce interstitial diffusion. Nat Mater. 2004;3(7):489–94.
Nance EA, et al. A dense poly(ethylene glycol) coating improves penetration of huge polymeric nanoparticles inside mind tissue. Sci Transl Med. 2012;4(149): 149ra119.
Nam J, et al. Modularly programmable nanoparticle vaccine primarily based on polyethyleneimine for customized most cancers immunotherapy. Adv Sci (Weinh). 2021;8(5):2002577.
Moghimi SM. The impact of methoxy-PEG chain size and molecular structure on lymph node concentrating on of immuno-PEG liposomes. Biomaterials. 2006;27(1):136–44.
Rao DA, et al. Biodegradable PLGA primarily based nanoparticles for sustained regional lymphatic drug supply. J Pharm Sci. 2010;99(4):2018–31.
Kourtis IC, et al. Peripherally administered nanoparticles goal monocytic myeloid cells, secondary lymphoid organs and tumors in mice. PLoS ONE. 2013;8(4): e61646.
Reddy ST, et al. In vivo concentrating on of dendritic cells in lymph nodes with poly(propylene sulfide) nanoparticles. J Management Launch. 2006;112(1):26–34.
Reddy ST, et al. Exploiting lymphatic transport and complement activation in nanoparticle vaccines. Nat Biotechnol. 2007;25(10):1159–64.
Ryan GM, Kaminskas LM, Porter CJ. Nano-chemotherapeutics: maximising lymphatic drug publicity to enhance the remedy of lymph-metastatic cancers. J Management Launch. 2014;193:241–56.
O’Melia MJ, et al. High quality of CD8(+) T cell immunity evoked in lymph nodes is compartmentalized by route of antigen transport and useful in tumor context. Sci Adv. 2020;6(50): eabd7134.
Kim J, et al. Injectable, spontaneously assembling, inorganic scaffolds modulate immune cells in vivo and improve vaccine efficacy. Nat Biotechnol. 2015;33(1):64–72.
Huang X, et al. The impact of the form of mesoporous silica nanoparticles on mobile uptake and cell operate. Biomaterials. 2010;31(3):438–48.
Hinde E, et al. Pair correlation microscopy reveals the function of nanoparticle form in intracellular transport and website of drug launch. Nat Nanotechnol. 2017;12(1):81–9.
Cha BG, Jeong JH, Kim J. Further-large pore mesoporous silica nanoparticles enabling co-delivery of excessive quantities of protein antigen and toll-like receptor 9 agonist for enhanced most cancers vaccine efficacy. ACS Cent Sci. 2018;4(4):484–92.
Xia Y, et al. Exploiting the pliability and lateral mobility of Pickering emulsion for enhanced vaccination. Nat Mater. 2018;17(2):187–94.
Tune T, et al. Engineering the deformability of albumin-stabilized emulsions for lymph-node vaccine supply. Adv Mater. 2021;33(26): e2100106.
Torchilin VP. Current advances with liposomes as pharmaceutical carriers. Nat Rev Drug Discov. 2005;4(2):145–60.
Kaur R, et al. Manipulation of the floor pegylation together with decreased vesicle measurement of cationic liposomal adjuvants modifies their clearance kinetics from the injection website, and the speed and kind of T cell response. J Management Launch. 2012;164(3):331–7.
Zhuang Y, et al. PEGylated cationic liposomes robustly increase vaccine-induced immune responses: function of lymphatic trafficking and biodistribution. J Management Launch. 2012;159(1):135–42.
Yu X, et al. Melittin-lipid nanoparticles goal to lymph nodes and elicit a systemic anti-tumor immune response. Nat Commun. 2020;11(1):1110.
Kuai R, et al. Designer vaccine nanodiscs for customized most cancers immunotherapy. Nat Mater. 2017;16(4):489–96.
Oberli MA, et al. Lipid nanoparticle assisted mRNA supply for potent most cancers immunotherapy. Nano Lett. 2017;17(3):1326–35.
Kim M, et al. Engineered ionizable lipid nanoparticles for focused supply of RNA therapeutics into several types of cells within the liver. Sci Adv. 2021;7(9): eabf4398.
Maeta M, et al. Vitamin E scaffolds of pH-responsive lipid nanoparticles as DNA vaccines in most cancers and protozoan an infection. Mol Pharm. 2020;17(4):1237–47.
Miao L, Zhang Y, Huang L. mRNA vaccine for most cancers immunotherapy. Mol Most cancers. 2021;20(1):41.
Bhardwaj P, et al. Developments in prophylactic and therapeutic nanovaccines. Acta Biomater. 2020;108:1–21.
Sahdev P, Ochyl LJ, Moon JJ. Biomaterials for nanoparticle vaccine supply programs. Pharm Res. 2014;31(10):2563–82.
Liu XY, et al. The traits and transfection effectivity of cationic poly (ester-co-urethane) – brief chain PEI conjugates self-assembled with DNA. Biomaterials. 2009;30(34):6665–73.
Xu J, et al. A common technique in the direction of customized nanovaccines primarily based on fluoropolymers for post-surgical most cancers immunotherapy. Nat Nanotechnol. 2020;15(12):1043–52.
Zeng Q, et al. Tailoring polymeric hybrid micelles with lymph node concentrating on capacity to enhance the efficiency of most cancers vaccines. Biomaterials. 2017;122:105–13.
Shae D, et al. Co-delivery of peptide neoantigens and stimulator of interferon genes agonists enhances response to most cancers vaccines. ACS Nano. 2020;14(8):9904–16.
Lv M, et al. Redox-responsive hyperbranched poly(amido amine) and polymer dots as a vaccine supply system for most cancers immunotherapy. J Mater Chem B. 2017;5(48):9532–45.
Shi GN, et al. Enhanced antitumor immunity by concentrating on dendritic cells with tumor cell lysate-loaded chitosan nanoparticles vaccine. Biomaterials. 2017;113:191–202.
Aranaz I, Mengibar M, Harris R, Panos I, Miralles B, Acosta N, Galed G, Heras A. Useful characterization of chitin and chitosan. Curr Chem Biol. 2009;3(2):203.
Wang X, et al. Inorganic nanomaterials with fast clearance for biomedical purposes. Chem Soc Rev. 2021;50(15):8669–742.
Hess KL, Medintz IL, Jewell CM. Designing inorganic nanomaterials for vaccines and immunotherapies. Nano At present. 2019;27:73–98.
Almeida JPM, et al. In vivo gold nanoparticle supply of peptide vaccine induces anti-tumor immune response in prophylactic and therapeutic tumor fashions. Small. 2015;11(12):1453–9.
Cao F, et al. Photothermally managed MHC Class I Restricted CD8(+) T-cell responses elicited by hyaluronic acid embellished gold nanoparticles as a vaccine for most cancers immunotherapy. Adv Healthc Mater. 2018;7(10): e1701439.
Guo Y, et al. Magnetic-responsive and focused most cancers nanotheranostics by PA/MR bimodal imaging-guided photothermally triggered immunotherapy. Biomaterials. 2019;219: 119370.
Hassan HA, et al. Twin stimulation of antigen presenting cells utilizing carbon nanotube-based vaccine supply system for most cancers immunotherapy. Biomaterials. 2016;104:310–22.
Wagner J, et al. mesoporous silica nanoparticles as pH-responsive service for the immune-activating drug resiquimod improve the native immune response in mice. ACS Nano. 2021;15(3):4450–66.
Li Y, et al. Designing and engineering of nanocarriers for bioapplication in most cancers immunotherapy. ACS Appl Bio Mater. 2020;3(12):8321–37.
Li F, et al. Engineering magnetosomes for high-performance most cancers vaccination. ACS Cent Sci. 2019;5(5):796–807.
Liu H, et al. SPIO improve the cross-presentation and migration of DCs and anionic SPIO affect the nanoadjuvant results associated to interleukin-1beta. Nanoscale Res Lett. 2018;13(1):409.
Yang Y, et al. Silica-based nanoparticles for biomedical purposes: from nanocarriers to biomodulators. Acc Chem Res. 2020;53(8):1545–56.
Zhang W, et al. Preparation and software of mesoporous core-shell nanosilica utilizing leucine by-product as template in efficient drug supply. Chin Chem Lett. 2020;31(5):1165–7.
Chen L, et al. Simultaneous T cell activation and macrophage polarization to advertise potent tumor suppression by iron oxide-embedded large-pore mesoporous organosilica core-shell nanospheres. Adv Healthc Mater. 2019;8(9): e1900039.
Hong X, et al. The pore measurement of mesoporous silica nanoparticles regulates their antigen supply effectivity. Sci Adv. 2020;6(25): eaaz4462.
Hassan H, et al. Software of carbon nanotubes in most cancers vaccines: achievements, challenges and probabilities. J Management Launch. 2019;297:79–90.
Xu C, et al. Environment friendly lymph node-targeted supply of customized most cancers vaccines with reactive oxygen species-inducing decreased graphene oxide nanosheets. ACS Nano. 2020;14(10):13268–78.
Zeng Z, Pu Ok. Enhancing most cancers immunotherapy by cell membrane-camouflaged nanoparticles. Adv Funct Mater. 2020;30(43):2004397.
Yang X, et al. pH-responsive biomimetic polymeric micelles as lymph node-targeting vaccines for enhanced antitumor immune responses. Biomacromol. 2020;21(7):2818–28.
Liu WL, et al. Cytomembrane nanovaccines present therapeutic results by mimicking tumor cells and antigen presenting cells. Nat Commun. 2019;10(1):3199.
Morishita M, et al. Exosome-based tumor antigens-adjuvant co-delivery using genetically engineered tumor cell-derived exosomes with immunostimulatory CpG DNA. Biomaterials. 2016;111:55–65.
Phung CD, et al. Anti-CTLA-4 antibody-functionalized dendritic cell-derived exosomes concentrating on tumor-draining lymph nodes for efficient induction of antitumor T-cell responses. Acta Biomater. 2020;115:371–82.
Wang L, et al. Lymph node-targeted immune-activation mediated by imiquimod-loaded mesoporous polydopamine based-nanocarriers. Biomaterials. 2020;255: 120208.
Guo Y, et al. Erythrocyte membrane-enveloped polymeric nanoparticles as nanovaccine for induction of antitumor immunity in opposition to melanoma. ACS Nano. 2015;9(7):6918–33.
Forster R, Braun A, Worbs T. Lymph node homing of T cells and dendritic cells by way of afferent lymphatics. Traits Immunol. 2012;33(6):271–80.
Santos P, Almeida F. Exosome-based vaccines: historical past, present state, and scientific trials. Entrance Immunol. 2021;12: 711565.
Harari A, et al. Antitumour dendritic cell vaccination in a priming and boosting strategy. Nat Rev Drug Discov. 2020;19(9):635–52.
Chaput N, et al. Exosomes as potent cell-free peptide-based vaccine. II. Exosomes in CpG adjuvants effectively prime naive Tc1 lymphocytes resulting in tumor rejection. J Immunol. 2004;172(4):2137–46.
Andre F, et al. Exosomes as potent cell-free peptide-based vaccine. I. Dendritic cell-derived exosomes switch useful MHC class I/peptide complexes to dendritic cells. J Immunol. 2004;172(4):2126–36.
d’Ischia M, et al. Polydopamine and eumelanin: from structure-property relationships to a unified tailoring technique. Acc Chem Res. 2014;47(12):3541–50.
Liu Y, Ai Ok, Lu L. Polydopamine and its by-product supplies: synthesis and promising purposes in vitality, environmental, and biomedical fields. Chem Rev. 2014;114(9):5057–115.
Zhu G, et al. Albumin/vaccine nanocomplexes that assemble in vivo for mixture most cancers immunotherapy. Nat Commun. 2017;8(1):1954.
Lee BR, et al. Engineered human ferritin nanoparticles for direct supply of tumor antigens to lymph node and most cancers immunotherapy. Sci Rep. 2016;6:35182.
Wang T, et al. A most cancers vaccine-mediated postoperative immunotherapy for recurrent and metastatic tumors. Nat Commun. 2018;9(1):1532.
Qian Y, et al. Concentrating on dendritic cells in lymph node with an antigen peptide-based nanovaccine for most cancers immunotherapy. Biomaterials. 2016;98:171–83.
Liu S, et al. A DNA nanodevice-based vaccine for most cancers immunotherapy. Nat Mater. 2021;20(3):421–30.
Lei C, et al. Hyaluronic acid and albumin primarily based nanoparticles for drug supply. J Management Launch. 2021;331:416–33.
Zhang W, et al. In vivo irreversible albumin-binding near-infrared dye conjugate as a naked-eye and fluorescence dual-mode imaging agent for lymph node tumor metastasis prognosis. Biomaterials. 2019;217: 119279.
Abdallah M, et al. Lymphatic concentrating on by albumin-hitchhiking: purposes and optimisation. J Management Launch. 2020;327:117–28.
Liu H, et al. Construction-based programming of lymph-node concentrating on in molecular vaccines. Nature. 2014;507(7493):519–22.
Rudra JS, et al. A self-assembling peptide performing as an immune adjuvant. Proc Natl Acad Sci U S A. 2010;107(2):622–7.
Abdullah T, et al. Supramolecular self-assembled peptide-based vaccines: present state and future views. Entrance Chem. 2020;8: 598160.
Veneziano R, et al. Function of nanoscale antigen group on B-cell activation probed utilizing DNA origami. Nat Nanotechnol. 2020;15(8):716–23.
Zhu G, et al. Intertwining DNA-RNA nanocapsules loaded with tumor neoantigens as synergistic nanovaccines for most cancers immunotherapy. Nat Commun. 2017;8(1):1482.
Qi S, et al. The brilliant way forward for nanotechnology in lymphatic system imaging and imaging-guided surgical procedure. J Nanobiotechnol. 2022;20(1):24.
Steeg PS. Tumor metastasis: mechanistic insights and scientific challenges. Nat Med. 2006;12(8):895–904.
Suhail Y, et al. Programs biology of most cancers metastasis. Cell Syst. 2019;9(2):109–27.
Moncayo VM, et al. Sentinel lymph node biopsy procedures. Semin Nucl Med. 2017;47(6):595–617.
Bieniasz-Krzywiec P, et al. Podoplanin-expressing macrophages promote lymphangiogenesis and lymphoinvasion in breast most cancers. Cell Metab. 2019;30(5):917–36.
Halabi WJ, et al. Ureteral accidents in colorectal surgical procedure: an evaluation of tendencies, outcomes, and danger elements over a 10-year interval in the US. Dis Colon Rectum. 2014;57(2):179–86.
Frankman EA, et al. Decrease urinary tract harm in girls in the US, 1979–2006. Am J Obstet Gynecol. 2010;202(5):495.
Wei Z, et al. Peroxidase-mimicking evodiamine/indocyanine inexperienced nanoliposomes for multimodal imaging-guided theranostics for oral squamous cell carcinoma. Bioact Mater. 2021;6(7):2144–57.
Wang B, et al. Picture-guided dendritic cell-based vaccine immunotherapy in murine carcinoma fashions. Am J Transl Res. 2017;9(10):4564–73.
Modo M, Hoehn M, Bulte JW. Mobile MR imaging. Mol Imaging. 2005;4(3):143–64.
Pan D, et al. Photoacoustic sentinel lymph node imaging with self-assembled copper neodecanoate nanoparticles. ACS Nano. 2012;6(2):1260–7.
Hong G, Antaris AL, Dai H. Close to-infrared fluorophores for biomedical imaging. Nat Biomed Eng. 2017;1(0010):1–22.
Fan X, et al. Nanoprobes-assisted multichannel NIR-II fluorescence imaging-guided resection and photothermal ablation of lymph nodes. Adv Sci (Weinh). 2021;8(9):2003972.
Cao Z, et al. Thrombus-targeted nano-agents for NIR-II diagnostic fluorescence imaging-guided flap thromboembolism multi-model remedy. J Nanobiotechnol. 2022;20(1):447.
Cai Y, et al. Fused-ring small-molecule-based bathochromic nano-agents for tumor NIR-II fluorescence imaging-guided photothermal/photodynamic remedy. ACS Appl Bio Mater. 2021;4(2):1942–9.
Dai Y, et al. Metastatic standing of sentinel lymph nodes in breast most cancers decided with photoacoustic microscopy by way of dual-targeting nanoparticles. Gentle Sci Appl. 2020;9:164.
Wang Z, et al. pH-amplified CRET nanoparticles for in vivo imaging of tumor metastatic lymph nodes. Angew Chem Int Ed Engl. 2021;60(26):14512–20.
Xiong L, Shuhendler AJ, Rao J. Self-luminescing BRET-FRET near-infrared dots for in vivo lymph-node mapping and tumour imaging. Nat Commun. 2012;3:1193.
Zhang C, et al. Close to-infrared upconversion multimodal nanoparticles for focused radionuclide remedy of breast most cancers lymphatic metastases. Entrance Immunol. 2022;13:1063678.
Zhao D, et al. Particular prognosis of lymph node micrometastasis in breast most cancers by concentrating on activatable near-infrared fluorescence imaging. Biomaterials. 2022;282: 121388.
Wang F, et al. In vivo non-invasive confocal fluorescence imaging past 1,700 nm utilizing superconducting nanowire single-photon detectors. Nat Nanotechnol. 2022;17(6):653–60.
Aime S, et al. Tunable imaging of cells labeled with MRI-PARACEST brokers. Angew Chem Int Ed Engl. 2005;44(12):1813–5.
de Vries IJ, et al. Magnetic resonance monitoring of dendritic cells in melanoma sufferers for monitoring of mobile remedy. Nat Biotechnol. 2005;23(11):1407–13.
Go Y, et al. Tumor-associated macrophages prolong alongside lymphatic movement within the pre-metastatic lymph nodes of human gastric most cancers. Ann Surg Oncol. 2016;23(Suppl 2):S230–5.
Lu Y, et al. A therapeutic DC vaccine with maintained immunological exercise displays sturdy anti-tumor efficacy. J Management Launch. 2022;349:254–68.
Perrin J, et al. Cell monitoring in most cancers immunotherapy. Entrance Med (Lausanne). 2020;7:34.
Kang SW, et al. Cell labeling and monitoring technique with out distorted indicators by phagocytosis of macrophages. Theranostics. 2014;4(4):420–31.
Jendelova P, et al. Magnetic resonance monitoring of transplanted bone marrow and embryonic stem cells labeled by iron oxide nanoparticles in rat mind and spinal twine. J Neurosci Res. 2004;76(2):232–43.
Hoehn M, et al. Monitoring of implanted stem cell migration in vivo: a extremely resolved in vivo magnetic resonance imaging investigation of experimental stroke in rat. Proc Natl Acad Sci U S A. 2002;99(25):16267–72.
Crich SG, et al. Improved route for the visualization of stem cells labeled with a Gd-/Eu-chelate as twin (MRI and fluorescence) agent. Magn Reson Med. 2004;51(5):938–44.
Tian R, et al. Multiplexed NIR-II probes for lymph node-invaded most cancers detection and imaging-guided surgical procedure. Adv Mater. 2020;32(11): e1907365.
Kircher MF, Gambhir SS, Grimm J. Noninvasive cell-tracking strategies. Nat Rev Clin Oncol. 2011;8(11):677–88.
Bulte JW, Kraitchman DL. Iron oxide MR distinction brokers for molecular and mobile imaging. NMR Biomed. 2004;17(7):484–99.
Stephan MT, et al. Therapeutic cell engineering with surface-conjugated artificial nanoparticles. Nat Med. 2010;16(9):1035–41.
Swartz MA, Fleury ME. Interstitial movement and its results in smooth tissues. Annu Rev Biomed Eng. 2007;9:229–56.
Nicolas JF, Man B. Intradermal, epidermal and transcutaneous vaccination: from immunology to scientific apply. Professional Rev Vaccines. 2008;7(8):1201–14.