375
Views
2
CrossRef citations to date
0
Altmetric
Review

Anticancer nanomedicines harnessing tumor microenvironmental components

, , , , , , , , & show all
Pages 337-354 | Received 22 Nov 2021, Accepted 03 Mar 2022, Published online: 14 Mar 2022

References

  • Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.
  • Matsumura Y, Maeda H. A new concept for macromolecular therape utics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res. 1986;46:6387–6392.
  • Morikawa S, Baluk P, Kaidoh T, et al. Abnormalities in pericytes on blood vessels and endothelial sprouts in tumors. Am J Pathol. 2002;160:985–1000.
  • Baluk P, Morikawa S, Haskell A, et al. Abnormalities of basement membrane on blood vessels and endothelial sprouts in tumors. Am J Pathol. 2003;163:1801–1815.
  • Kalluri R. Basement membranes: structure, assembly and role in tumour angiogenesis. Nat Rev Cancer. 2003;3:422–433.
  • Hashizume H, Baluk P, Morikawa S, et al. Openings between defective endothelial cells explain tumor vessel leakiness. Am J Pathol. 2000;156:1363–1380.
  • Loureiroa B A, Azoiaa NG, Gomesb AC, et al. Albumin-based nanodevices as drug carriers. Curr Pharm Des. 2016;10:1371–1390.
  • Maeda H, Islam W. Overcoming barriers for tumor-targeted drug delivery. Polym Protein Conjugates. 2020;3:41–58.
  • Hare JI, Lammers T, Ashford MB, et al. Challenges and strategies in anti-cancer nanomedicine development: an industry perspective. Adv Drug Deliv Rev. 2017;108:25–38.
  • Thomas RG, Surendran SP, Jeong YY. Tumor microenvironment-stimuli responsive nanoparticles for anticancer therapy. Front Mol Biosci. 2020;7:610533.
  • Yoo J, Park C, Yi G, et al. Active targeting strategies using biological ligands for nanoparticle drug delivery systems. Cancers (Basel). 2019;11:640.
  • Salahpour Anarjan F. Active targeting drug delivery nanocarriers: ligands. Nano Struct Nano Obj. 2019;19:100370.
  • Spicer CD, Jumeaux C, Gupta B, et al. Peptide and protein nanoparticle conjugates: versatile platforms for biomedical applications. Chem Soc Rev. 2018;47:3574–3620.
  • Li CM, Haratipour P, Lingeman RG, et al. Novel peptide therapeutic approaches for cancer treatment. Cells. 2021;10:2908.
  • Song KM, Lee S, Ban C. Aptamers and their biological applications. Sensors (Basel). 2012;12:612–631.
  • Marie-Cécile M, Monique D, Laurence C. Selection of nucleic acid aptamers targeting tumor cell-surface protein biomarkers. Cancers (Basel). 2017;9:69.
  • Han J, Gao L, Wang J, et al. Application and development of aptamer in cancer: from clinical diagnosis to cancer therapy. J Cancer. 2020;11:6902–6915.
  • Habibzadeh Mashatooki M, Jahanbin Sardroodi J. A molecular dynamics study proposing the existence of structural interaction between cancer cell receptor and RNA aptamer. J Inorg Organomet Polym Mater. 2020;30:4520–4532.
  • Yazdian-Robati R, Bayat P, Oroojalian F, et al. Therapeutic applications of AS1411 aptamer, an update review. Int J Biol Macromol. 2020;155:1420–1431.
  • Torres-Perez SA, Torres-Perez CE, Pedraza-Escalona M, et al. Glycosylated nanoparticles for cancer-targeted drug delivery. Front Oncol. 2020;10:605037.
  • Samadian H, Hosseini-Nami S, Kamrava SK, et al. Folate-conjugated gold nanoparticle as a new nanoplatform for targeted cancer therapy. J Cancer Res Clin Oncol. 2016;142:2217–2229.
  • Danhier F. To exploit the tumor microenvironment: since the EPR effect fails in the clinic, what is the future of nanomedicine? J Control Release. 2016;244:108–121.
  • Yang H, Tong Z, Sun S, et al. Enhancement of tumour penetration by nanomedicines through strategies based on transport processes and barriers. J Control Release. 2020;328:28–44.
  • Berraondo P, Sanmamed MF, Ochoa MC, et al. Cytokines in clinical cancer immunotherapy. Br J Cancer. 2019;120:6–15.
  • Riley RS, June CH, Langer R, et al. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov. 2019;18:175–196.
  • Vikas P, Borcherding N, Zhang W. The clinical promise of immunotherapy in triple-negative breast cancer. Cancer Manag Res. 2018;10:6823–6833.
  • Whiteside TL, Demaria S, Rodriguez-Ruiz ME, et al. Emerging Opportunities and Challenges in Cancer Immunotherapy. Clin Cancer Res. 2016;22:1845–1855.
  • Mao Y, Keller ET, Garfield DH, et al. Stromal cells in tumor microenvironment and breast cancer. Cancer Metastasis Rev. 2013;32:303–315.
  • Swartz MA, Iida N, Roberts EW, et al. Tumor microenvironment complexity: emerging roles in cancer therapy. Cancer Res. 2012;72:2473–2480.
  • Zhong S, Jeong JH, Chen Z, et al. Targeting tumor microenvironment by small-molecule inhibitors. Transl Oncol. 2020;13:57–69.
  • Lugano R, Ramachandran M, Dimberg A. Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell Mol Life Sci. 2020;77:1745–1770.
  • Dai Q, Wilhelm S, Ding D, et al. Quantifying the ligand-coated nanoparticle delivery to cancer cells in solid tumors. ACS Nano. 2018;12:8423–8435.
  • Ruoslahti E, Bhatia SN, Sailor MJ. Targeting of drugs and nanoparticles to tumors. J Cell Biol. 2010;188:759–768.
  • Yang M, Li J, Gu P, et al. The application of nanoparticles in cancer immunotherapy: targeting tumor microenvironment. Bioact Mater. 2021;6:1973–1987.
  • den Breems Ny, Eftimie R. The re-polarisation of M2 and M1 macrophages and its role on cancer outcomes. J Theor Biol. 2016;390:23–39.
  • Wang Y, Lin Y-X, Qiao S-L, et al. Polymeric nanoparticles promote macrophage reversal from M2 to M1 phenotypes in the tumor microenvironment. Biomaterials. 2017;112:153–163.
  • Pal R, Chakraborty B, Nath A, et al. Noble metal nanoparticle-induced oxidative stress modulates tumor associated macrophages (TAMs) from an M2 to M1 phenotype: an in vitro approach. Int Immunopharmacol. 2016;38:332–341.
  • Figueiredo P, Lepland A, Scodeller P, et al. Peptide-guided resiquimod-loaded lignin nanoparticles convert tumor-associated macrophages from M2 to M1 phenotype for enhanced chemotherapy. Acta Biomater. 2020;133:231–243.
  • Norton J, Foster D, Chinta M, et al. Pancreatic cancer associated fibroblasts (CAF): under-explored target for pancreatic cancer treatment. Cancers (Basel). 2020;12:1347.
  • Ren J, Smid M, Iaria J, et al. Cancer-associated fibroblast-derived Gremlin 1 promotes breast cancer progression. Breast Cancer Res. 2019;21:109.
  • Arneth B. Tumor microenvironment. Medicina (Kaunas). 2019;56:15.
  • Holle AW, Young JL, Spatz JP. In vitro cancer cell-ECM interactions inform in vivo cancer treatment. Adv Drug Deliv Rev. 2016;97:270–279.
  • Harisi R, Jeney A. Extracellular matrix as target for antitumor therapy. Onco Targets Ther. 2015;8:1387–1398.
  • Danhier F, Feron O, Preat V. To exploit the tumor microenvironment: passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J Control Release. 2010;148:135–146.
  • Huai Y, Hossen MN, Wilhelm S, et al. Nanoparticle interactions with the tumor microenvironment. Bioconjug Chem. 2019;30:2247–2263.
  • Feng L, Dong Z, Tao D, et al. The acidic tumor microenvironment: a target for smart cancer nano-theranostics. Natl Sci Rev. 2018;5:269–286.
  • Wang Y, Shang W, Niu M, et al. Hypoxia-active nanoparticles used in tumor theranostic. Int J Nanomedicine. 2019;14:3705–3722.
  • Challapalli A, Carroll L, Aboagye EO. Molecular mechanisms of hypoxia in cancer. Clin Transl Imaging. 2017;5:225–253.
  • Kim SJ, Kim HS, Seo YR. Understanding of ROS-inducing strategy in anticancer therapy. Oxid Med Cell Longev. 2019;2019:5381692.
  • Yang B, Chen Y, Shi J. Reactive oxygen species (ROS)-based nanomedicine. Chem Rev. 2019;119:4881–4985.
  • Cook JA, Gius D, Wink DA, et al. Oxidative stress, redox, and the tumor microenvironment. Semin Radiat Oncol. 2004;14:259–266.
  • Yang B, Yao H, Tian H, et al. Intratumoral synthesis of nano-metalchelate for tumor catalytic therapy by ligand field-enhanced coordination. Nat Commun. 2021;12:3393.
  • Zhao M, Liu Q, Gong Y, et al. GSH-dependent antioxidant defense contributes to the acclimation of colon cancer cells to acidic microenvironment. Cell Cycle. 2016;15:1125–1133.
  • Yang B, Shi J. Ascorbate tumor chemotherapy by an iron-engineered nanomedicine-catalyzed tumor-specific pro-oxidation. J Am Chem Soc. 2020;142:21775–21785.
  • Wu W, Chen M, Luo T, et al. ROS and GSH-responsive S-nitrosoglutathione functionalized polymeric nanoparticles to overcome multidrug resistance in cancer. Acta Biomater. 2020;103:259–271.
  • L-y Y, Shen Y-A, Chen M-H, et al. The feasibility of ROS- and GSH-responsive micelles for treating tumor-initiating and metastatic cancer stem cells. J Mat Chem B. 2019;7:3109–3118.
  • Shay G, Lynch CC, Fingleton B. Moving targets: emerging roles for MMPs in cancer progression and metastasis. Matrix Biol. 2015;44-46:200–206.
  • Jablonska-Trypuc A, Matejczyk M, Rosochacki S. Matrix metalloproteinases (MMPs), the main extracellular matrix (ECM) enzymes in collagen degradation, as a target for anticancer drugs. J Enzyme Inhib Med Chem. 2016;31:177–183.
  • Rundhaug JE. Matrix metalloproteinases and angiogenesis. J Cell Mol Med. 2005;9:267–285.
  • Shimoda M, Ohtsuka T, Okada Y, et al. Stromal metalloproteinases: crucial contributors to the tumor microenvironment. Pathol Int. 2021;71:1–14.
  • Alaseem A, Alhazzani K, Dondapati P, et al. Matrix metalloproteinases: a challenging paradigm of cancer management. Semin Cancer Biol. 2019;56:100–115.
  • MacEwan SR, Callahan DJ, Chilkoti A. Stimulus-responsive macromolecules and nanoparticles for cancer drug delivery. Nanomedicine. 2010;5:793–806.
  • Gulzar A, Xu J, Wang C, et al. Tumour microenvironment responsive nanoconstructs for cancer theranostic. Nano Today. 2019;26:16–56.
  • Uthaman S, Huh KM, Park IK. Tumor microenvironment-responsive nanoparticles for cancer theragnostic applications. Biomater Res. 2018;22:22.
  • Wang L, Huo M, Chen Y, et al. Tumor microenvironment-enabled nanotherapy. Adv Healthc Mater. 2018;7:e1701156.
  • Kundu M, Sadhukhan P, Ghosh N, et al. pH-responsive and targeted delivery of curcumin via phenylboronic acid-functionalized ZnO nanoparticles for breast cancer therapy. J Adv Res. 2019;18:161–172.
  • Schmitt S, Tahk S, Lohner A, et al. Fusion of bacterial flagellin to a dendritic cell-targeting alphaCD40 antibody construct coupled with viral or leukemia-specific antigens enhances dendritic cell maturation and activates peptide-responsive T cells. Front Immunol. 2020;11:602802.
  • Pan D, She W, Guo C, et al. PEGylated dendritic diaminocyclohexyl-platinum (II) conjugates as pH-responsive drug delivery vehicles with enhanced tumor accumulation and antitumor efficacy. Biomaterials. 2014;35:10080–10092.
  • Li N, Li N, Yi Q, et al. Amphiphilic peptide dendritic copolymer-doxorubicin nanoscale conjugate self-assembled to enzyme-responsive anti-cancer agent. Biomaterials. 2014;35:9529–9545. .
  • Yang Y, Pan D, Luo K, et al. Biodegradable and amphiphilic block copolymer-doxorubicin conjugate as polymeric nanoscale drug delivery vehicle for breast cancer therapy. Biomaterials. 2013;34:8430–8443.
  • She W, Li N, Luo K, et al. Dendronized heparin-doxorubicin conjugate based nanoparticle as pH-responsive drug delivery system for cancer therapy. Biomaterials. 2013;34:2252–2264.
  • Li HJ, Du JZ, Liu J, et al. Smart superstructures with ultrahigh pH-sensitivity for targeting acidic tumor microenvironment: instantaneous size switching and improved tumor penetration. ACS Nano. 2016;10:6753–6761.
  • Yang W, Yang S, Jiang L, et al. Tumor microenvironment triggered biodegradation of inorganic nanoparticles for enhanced tumor theranostics. RSC Adv. 2020;10:26742–26751.
  • Shi Z, Li Q, Mei L. pH-sensitive nanoscale materials as robust drug delivery systems for cancer therapy. Chin Chem Lett. 2020;31:1345–1356.
  • He Y, Zeng B, Liang S, et al. Synthesis of pH-responsive biodegradable mesoporous silica-calcium phosphate hybrid nanoparticles as a high potential drug carrier. ACS Appl Mater Interfaces. 2017;9:44402–44409.
  • Kumari R, Sunil D, Ningthoujam RS. Hypoxia-responsive nanoparticle based drug delivery systems in cancer therapy: an up-to-date review. J Control Release. 2020;319:135–156.
  • Khatoon S, Han HS, Jeon J, et al. Hypoxia-responsive mesoporous nanoparticles for doxorubicin delivery. Polymers (Basel). 2018;10:390.
  • Zhou M, Xie Y, Xu S, et al. Hypoxia-activated nanomedicines for effective cancer therapy. Eur J Med Chem. 2020;195:112274.
  • Phung CD, Tran TH, Pham LM, et al. Current developments in nanotechnology for improved cancer treatment, focusing on tumor hypoxia. J Control Release. 2020;324:413–429.
  • Yang G, Phua SZF, Lim WQ, et al. A hypoxia‐responsive albumin‐based nanosystem for deep tumor penetration and excellent therapeutic efficacy. Adv Mater. 2019;31:e1901513.
  • Luo L, Qi Y, Zhong H, et al. GSH-sensitive polymeric prodrug: synthesis and loading with photosensitizers as nanoscale chemo-photodynamic anti-cancer nanomedicine. Acta Pharm Sin B. 2022;12:424–436.
  • Xu X, Saw PE, Tao W, et al. ROS-responsive polyprodrug nanoparticles for triggered drug delivery and effective cancer therapy. Adv Mater. 2017;29:1700141.
  • Iyer R, Nguyen T, Padanilam D, et al. Glutathione-responsive biodegradable polyurethane nanoparticles for lung cancer treatment. J Control Release. 2020;321:363–371.
  • Chen R, Ma Z, Xiang Z, et al. Hydrogen peroxide and glutathione dual redox-responsive nanoparticles for controlled DOX release. Macromol Biosci. 2020;20:e1900331.
  • Wu Y, Li F, Zhang X, et al. Tumor microenvironment-responsive PEGylated heparin-pyropheophorbide-a nanoconjugates for photodynamic therapy. Carbohydr Polym. 2021;255:117490.
  • Wang W, Zhang X, Li Z, et al. Dendronized hyaluronic acid-docetaxel conjugate as a stimuli-responsive nano-agent for breast cancer therapy. Carbohydr Polym. 2021;267:118160.
  • Luo Q, Xiao X, Dai X, et al. Cross-linked and biodegradable polymeric system as a safe magnetic resonance imaging contrast agent. ACS Appl Mater Interfaces. 2018;10:1575–1588.
  • Wu Y, Zhong D, Li Y, et al. A tumor-activatable peptide supramolecular nanoplatform for the delivery of dual-gene targeted siRNAs for drug-resistant cancer treatment. Nanoscale. 2021;13:4887–4898.
  • Duan Z, Cai H, Zhang H, et al. PEGylated multistimuli-responsive dendritic prodrug-based nanoscale system for enhanced anticancer activity. ACS Appl Mater Interfaces. 2018;10:35770–35783.
  • Chen WH, Luo GF, Lei Q, et al. MMP-2 responsive polymeric micelles for cancer-targeted intracellular drug delivery. Chem Commun (Camb). 2015;51:465–468.
  • Yao Q, Kou L, Tu Y, et al. MMP-responsive ‘smart’ drug delivery and tumor targeting. Trends Pharmacol Sci. 2018;39:766–781.
  • Battistella C, Callmann CE, Thompson MP, et al. Delivery of immunotherapeutic nanoparticles to tumors via enzyme-directed assembly. Adv Healthc Mater. 2019;8:e1901105.
  • Han M, Huang-Fu MY, Guo WW, et al. MMP-2-sensitive HA end-conjugated poly(amidoamine) dendrimers via click reaction to enhance drug penetration into solid tumor. ACS Appl Mater Interfaces. 2017;9:42459–42470.
  • Zhang C, Pan D, Luo K, et al. Dendrimer–doxorubicin conjugate as enzyme-sensitive and polymeric nanoscale drug delivery vehicle for ovarian cancer therapy. Polym Chem. 2014;5:5227–5235.
  • Wang J, Li N, Cao L, et al. DOX-loaded peptide dendritic copolymer nanoparticles for combating multidrug resistance by regulating the lysosomal pathway of apoptosis in breast cancer cells. J Mater Chem B. 2020;8:1157–1170.
  • Cai H, Wang X, Zhang H, et al. Enzyme-sensitive biodegradable and multifunctional polymeric conjugate as theranostic nanomedicine. Appl Mater Today. 2018;11:207–218.
  • Chen K, Liao S, Guo S, et al. Enzyme/pH-sensitive dendritic polymer-DOX conjugate for cancer treatment. Sci China Mater. 2018;61:1462–1474.
  • Tan P, Cai H, Wei Q, et al. Enhanced chemo-photodynamic therapy of an enzyme-responsive prodrug in bladder cancer patient-derived xenograft models. Biomaterials. 2021;277:121061.
  • Ou Y, Chen K, Cai H, et al. Enzyme/pH-sensitive polyHPMA–DOX conjugate as a biocompatible and efficient anticancer agent. Biomater Sci. 2018;6:1177–1188.
  • Cai H, Tan P, Chen X, et al. Stimuli-sensitive linear-dendritic block copolymer-drug prodrug as a nanoplatform for tumor combination therapy. Adv Mater. 2021;32:33e2108049.
  • Duan Z, Luo Q, Dai X, et al. Synergistic therapy of a naturally inspired glycopolymer-based biomimetic nanomedicine harnessing tumor genomic instability. Adv Mater. 2021;33:e2104594.
  • Luo Q, Duan Z, Li X, et al. Branched polymer‐based redox/enzyme‐activatable photodynamic nanoagent to trigger sting‐dependent immune responses for enhanced therapeutic effect. Advanced Functional Materials. 2021: 2110408.
  • He X, Yang Y, Li L, et al. Engineering extracellular matrix to improve drug delivery for cancer therapy. Drug Discov Today. 2020;25:1727–1734.
  • Zinger A, Koren L, Adir O, et al. Collagenase nanoparticles enhance the penetration of drugs into pancreatic tumors. ACS Nano. 2019;13:11008–11021.
  • Zhou H, Fan Z, Deng J, et al. Hyaluronidase embedded in nanocarrier peg shell for enhanced tumor penetration and highly efficient antitumor efficacy. Nano Lett. 2016;16:3268–3277.
  • Estrella V, Chen T, Lloyd M, et al. Acidity generated by the tumor microenvironment drives local invasion. Cancer Res. 2013;73:1524–1535.
  • Horsman MR, Vaupel P. Pathophysiological basis for the formation of the tumor microenvironment. Front Oncol. 2016;6:66.
  • Michiels C, Tellier C, Feron O. Cycling hypoxia: a key feature of the tumor microenvironment. Biochim Biophys Acta. 2016;1866:76–86.
  • Gorrini C, Harris IS, Mak TW. Modulation of oxidative stress as an anticancer strategy. Nat Rev Drug Discov. 2013;12:931–947.
  • Zhong X, Wang X, Li J, et al. ROS-based dynamic therapy synergy with modulating tumor cell-microenvironment mediated by inorganic nanomedicine. Coord Chem Rev. 2021;437:213828
  • Bansal A, Simon MC. Glutathione metabolism in cancer progression and treatment resistance. J Cell Biol. 2018;217:2291–2298.
  • Lee MH, Yang Z, Lim CW, et al. Disulfide-cleavage-triggered chemosensors and their biological applications. Chem Rev. 2013;113:5071–5109.
  • Park H, Saravanakumar G, Kim J, et al. Tumor microenvironment sensitive nanocarriers for bioimaging and therapeutics. Adv Healthc Mater. 2021;10:e2000834.
  • Reid SE, Kay EJ, Neilson LJ, et al. Tumor matrix stiffness promotes metastatic cancer cell interaction with the endothelium. EMBO J. 2017;36:2373–2389.
  • Liotta LAA. Degrade, and move: the three-step model of invasion. Cancer Res. 2016;76:3115–3117.
  • Wagner M, Wiig H. Tumor Interstitial Fluid Formation, Characterization, and Clinical Implications. Front Oncol. 2015;5:115.
  • Yu T, Liu K, Wu Y, et al. High interstitial fluid pressure promotes tumor cell proliferation and invasion in oral squamous cell carcinoma. Int J Mol Med. 2013;32:1093–1100.
  • Yang S, Chen C, Qiu Y, et al. Paying attention to tumor blood vessels: cancer phototherapy assisted with nano delivery strategies. Biomaterials. 2021;268:120562.
  • Pankova D, Chen Y, Terajima M, et al. Cancer-associated fibroblasts induce a collagen cross-link switch in tumor stroma. Mol Cancer Res. 2016;14:287–295.
  • Paijens ST, Vledder A, de Bruyn M, et al. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol. 2021;18:842–859.
  • Batlle E, Clevers H. Cancer stem cells revisited. Nat Med. 2017;23:1124–1134.
  • Gerhard GM, Bill R, Messemaker M, et al. Tumor-infiltrating dendritic cell states are conserved across solid human cancers. J Exp Med. 2021;218:e20200264.
  • Chen D, Zhang X, Li Z, et al. Metabolic regulatory crosstalk between tumor microenvironment and tumor-associated macrophages. Theranostics. 2021;11:1016–1030.
  • Muntimadugu E, Kommineni N, Khan W. Exploring the potential of nanotherapeutics in targeting tumor microenvironment for cancer therapy. Pharmacol Res. 2017;126:109–122.
  • Sakurai Y, Akita H, Harashima H. Targeting tumor endothelial cells with nanoparticles. Int J Mol Sci. 2019;20:5819.
  • Liu M, Song W, Huang L. Drug delivery systems targeting tumor-associated fibroblasts for cancer immunotherapy. Cancer Lett. 2019;448:31–39.
  • Arezumand R, Alibakhshi A, Ranjbari J, et al. Nanobodies as novel agents for targeting angiogenesis in solid cancers. Front Immunol. 2017;8:1746.
  • Ding X, Su Y, Wang C, et al. Synergistic suppression of tumor angiogenesis by the co-delivering of vascular endothelial growth factor targeted siRNA and candesartan mediated by functionalized carbon nanovectors. ACS Appl Mater Interfaces. 2017;9:23353–23369.
  • Su Y, Hu Y, Wang Y, et al. A precision-guided MWNT mediated reawakening the sunk synergy in RAS for anti-angiogenesis lung cancer therapy. Biomaterials. 2017;139:75–90.
  • Guo D, Xu S, Wang N, et al. Prodrug-embedded angiogenic vessel-targeting nanoparticle: a positive feedback amplifier in hypoxia-induced chemo-photo therapy. Biomaterials. 2017;144:188–198.
  • Shamay Y, Golan M, Tyomkin D, et al. Assessing the therapeutic efficacy of VEGFR-1-targeted polymer drug conjugates in mouse tumor models. J Control Release. 2016;229:192–199.
  • Chantarasrivong C, Higuchi Y, Tsuda M, et al. Sialyl LewisX mimic-decorated liposomes for anti-angiogenic everolimus delivery to E-selectin expressing endothelial cells. RSC Adv. 2019;9:20518–20527.
  • Xu J, Lee SS, Seo H, et al. Quinic acid-conjugated nanoparticles enhance drug delivery to solid tumors via interactions with endothelial selectins. Small. 2018;14:e1803601.
  • Paraiso KH, Smalley KS. Fibroblast-mediated drug resistance in cancer. Biochem Pharmacol. 2013;85:1033–1041.
  • Ji T, Zhao Y, Ding Y, et al. Transformable peptide nanocarriers for expeditious drug release and effective cancer therapy via cancer-associated fibroblast activation. Angew Chem Int Ed Engl. 2016;55:1050–1055.
  • Yu Q, Qiu Y, Li J, et al. Targeting cancer-associated fibroblasts by dual-responsive lipid-albumin nanoparticles to enhance drug perfusion for pancreatic tumor therapy. J Control Release. 2020;321:564–575.
  • Chen B, Wang Z, Sun J, et al. A tenascin C targeted nanoliposome with navitoclax for specifically eradicating of cancer-associated fibroblasts. Nanomedicine. 2016;12:131–141.
  • Wang Y, Zhang Q, Chen Y, et al. Antitumor effects of immunity-enhancing traditional Chinese medicine. Biomed Pharmacother. 2020;121:109570.
  • He Y, de Araujo Junior RF, Cruz LJ, et al. Functionalized nanoparticles targeting tumor-associated macrophages as cancer therapy. Pharmaceutics. 2021;13:1670.
  • Ding J, Zhao D, Hu Y, et al. Terminating the renewal of tumor-associated macrophages: a sialic acid-based targeted delivery strategy for cancer immunotherapy. Int J Pharm. 2019;571:118706.
  • Han S, Wang W, Wang S, et al. Multifunctional biomimetic nanoparticles loading baicalin for polarizing tumor-associated macrophages. Nanoscale. 2019;11:20206–20220.
  • Kim MS, Lee JS, Kim JE, et al. Enhancement of antitumor effect of radiotherapy via combination with Au@SiO2 nanoparticles targeted to tumor-associated macrophages. J Ind Eng Chem. 2020;84:349–357.
  • Rong L, Zhang Y, Li WS, et al. Iron chelated melanin-like nanoparticles for tumor-associated macrophage repolarization and cancer therapy. Biomaterials. 2019;225:119515.
  • Mastelic-Gavillet B, Balint K, Boudousquie C, et al. Personalized dendritic cell vaccines-recent breakthroughs and encouraging clinical results. Front Immunol. 2019;10:766.
  • Saxena M, Bhardwaj N. Re-Emergence of dendritic cell vaccines for cancer treatment. Trends in Cancer. 2018;4:119–137.
  • Wculek SK, Cueto FJ, Mujal AM, et al. Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol. 2020;20:7–24.
  • Tran TH, Tran TTP, Nguyen HT, et al. Nanoparticles for dendritic cell-based immunotherapy. Int J Pharm. 2018;542:253–265.
  • El-Sayed N, Korotchenko E, Scheiblhofer S, et al. Functionalized multifunctional nanovaccine for targeting dendritic cells and modulation of immune response. Int J Pharm. 2021;593:120123.
  • Zhang C, Shi G, Zhang J, et al. Targeted antigen delivery to dendritic cell via functionalized alginate nanoparticles for cancer immunotherapy. J Control Release. 2017;256:170–181.
  • Liu Y, Yao L, Cao W, et al. Dendritic cell targeting peptide-based nanovaccines for enhanced cancer immunotherapy. ACS Appl Bio Mater. 2019;2:1241–1254.
  • Duan H, Liu Y, Gao Z, et al. Recent advances in drug delivery systems for targeting cancer stem cells. Acta Pharm Sin B. 2021;11:55–70.
  • Choi KY, Yoon HY, Kim J-H, et al. Smart nanocarrier based on pegylated hyaluronic acid for cancer therapy. Acs Nano. 2011;5:8591–8599.
  • Li M, Sun J, Zhang W, et al. Drug delivery systems based on CD44-targeted glycosaminoglycans for cancer therapy. Carbohydr Polym. 2021;251:117103.
  • Yang Z, Sun N, Cheng R, et al. Hybrid nanoparticles coated with hyaluronic acid lipoid for targeted co-delivery of paclitaxel and curcumin to synergistically eliminate breast cancer stem cells. J Mater Chem B. 2017;5:6762–6775.
  • Zahiri M, Babaei M, Abnous K, et al. Hybrid nanoreservoirs based on dextran-capped dendritic mesoporous silica nanoparticles for CD133-targeted drug delivery. J Cell Physiol. 2020;235:1036–1050.
  • Ishiguro K, Yan IK, Lewis‐Tuffin L, et al. Targeting liver cancer stem cells using engineered biological nanoparticles for the treatment of hepatocellular cancer. Hepatol Commun. 2020;4(2):298–313.
  • Ou W, Thapa RK, Jiang L, et al. Regulatory T cell-targeted hybrid nanoparticles combined with immuno-checkpoint blockage for cancer immunotherapy. J Control Release. 2018;281:84–96.
  • Buabeid MA, Arafa EA, Murtaza G. Emerging prospects for nanoparticle-enabled cancer immunotherapy. J Immunol Res. 2020;2020:9624532.
  • Tanaka A, Sakaguchi S. Targeting Treg cells in cancer immunotherapy. Eur J Immunol. 2019;49:1140–1146.
  • Fang Y, Yu A, Ye L, et al. Research progress in tumor targeted immunotherapy. Expert Opin Drug Deliv. 2021;18:1067–1090.
  • Sacchetti C, Rapini N, Magrini A, et al. In vivo targeting of intratumor regulatory T cells using PEG-modified single-walled carbon nanotubes. Bioconjug Chem. 2013;24:852–858.
  • Phung CD, Pham TT, Nguyen HT, et al. 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.
  • Tang L, Zhang A, Zhang Z, et al. Multifunctional inorganic nanomaterials for cancer photoimmunotherapy. Cancer Commun (Lond). 2022;42:141–163.
  • Gao S, Yang D, Fang Y, et al. Engineering nanoparticles for targeted remodeling of the tumor microenvironment to improve cancer immunotherapy. Theranostics. 2019;9:126–151.
  • Musetti S, Huang L. Nanoparticle-mediated remodeling of the tumor microenvironment to enhance immunotherapy. ACS Nano. 2018;12:11740–11755.
  • Tang L, Mei Y, Shen Y, et al. Nanoparticle-mediated targeted drug delivery to remodel tumor microenvironment for cancer therapy. Int J Nanomedicine. 2021;16:5811–5829.
  • Cao Q, Wang W, Zhou M, et al. Induction of antitumor immunity in mice by the combination of nanoparticle-based photothermolysis and anti-PD-1 checkpoint inhibition. Nanomedicine. 2020;25:102169.
  • Pan D, Zheng X, Zhang L, et al. Synergistic disruption of metabolic homeostasis through hyperbranched poly(ethylene glycol) conjugates as nanotherapeutics to constrain cancer growth. Adv Mater. 2022: e2109036.
  • Zheng X, Pan D, Chen X, et al. Self-stabilized supramolecular assemblies constructed from pegylated dendritic peptide conjugate for augmenting tumor retention and therapy. Adv Sci (Weinh). 2021;8:e2102741.
  • Shang Y, Zheng N, Wang Z. Tetraphenylsilane-cored star-shaped polymer micelles with ph/redox dual response and active targeting function for drug-controlled release. Biomacromolecules. 2019;20:4602–4610.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.