DOI QR코드

DOI QR Code

Tumor microenvironment-responsive nanoparticles for cancer theragnostic applications

  • Uthaman, Saji (Department of Polymer Science and Engineering, Chungnam National University) ;
  • Huh, Kang Moo (Department of Polymer Science and Engineering, Chungnam National University) ;
  • Park, In-Kyu (Department of Biomedical Sciences, BK21 PLUS Centre for Creative Biomedical Scientists, Chonnam National University Medical School)
  • Received : 2018.06.16
  • Accepted : 2018.08.02
  • Published : 2018.09.30

Abstract

Background: Cancer is one of the deadliest threats to human health. Abnormal physiochemical conditions and dysregulated biosynthetic intermediates in the tumor microenvironment (TME) play a significant role in modulating cancer cells to evade or defend conventional anti-cancer therapy such as surgery, chemotherapy and radiotherapy. One of the most important challenges in the development of anti-tumor therapy is the successful delivery of therapeutic and imaging agents specifically to solid tumors. Main body: The recent progresses in development of TME responsive nanoparticles offers promising strategies for combating cancer by making use of the common attributes of tumor such as acidic and hypoxic microenvironments. In this review, we discussed the prominent strategies utilized in the development of tumor microenvironment-responsive nanoparticles and mode of release of therapeutic cargo. Conclusion: Tumor microenvironment-responsive nanoparticles offers a universal approach for anti-cancer therapy.

Keywords

Acknowledgement

Grant : Externally Actuatable Nanorobot System for Precise Targeting and Controlled Releasing of Drugs

References

  1. Xu L, et al. A reactive oxygen species-responsive prodrug micelle with efficient cellular uptake and excellent bioavailability. J Mater Chem B. 2018; 6:1076-84.
  2. Shi JJ, Kantoff PW, Wooster R, Farokhzad OC. Cancer nanomedicine: progress, challenges and opportunities. Nat Rev Cancer. 2017;17:20-37.
  3. Panieri E, Santoro MM. ROS homeostasis and metabolism: a dangerous liason in cancer cells. Cell Death Dis. 2016;7:e2253
  4. Jiang W, von Roemeling CA, Chen YX, Qie YQ, Liu XJ, Chen JZ, Kim BYS. Designing nanomedicine for immuno-oncology. Nat Biomed Eng. 2017;1 :0029.
  5. Singh R, et al. Dose-dependent therapeutic distinction between active and passive targeting revealed using transferrin-coated PGMA nanoparticles. Small. 2016;12:351-9.
  6. Du J, Lane LA, Nie S. Stimuli-responsive nanoparticles for targeting the tumor microenvironment. J Control Release. 2015;219:205-14.
  7. Rosenblum D, Joshi N, Tao W, Karp JM, Peer D. Progress and challenges towards targeted delivery of cancer therapeutics. Nat Commun. 2018;9:1410.
  8. Xia F, et al. pH-responsive gold nanoclusters-based nanoprobes for lung cancer targeted near-infrared fluorescence imaging and chemophotodynamic therapy. Acta biomaterialia. 2018;68:308-19.
  9. Hao YW, et al. Tumor acidity-activatable manganese phosphate nanoplatform for amplification of photodynamic cancer therapy and magnetic resonance imaging. Acta Biomater. 2017;62:293-305.
  10. Shi XX, et al. pH-responsive unimolecular micelles based on amphiphilic star-like copolymers with high drug loading for effective drug delivery and cellular imaging. J Mater Chem B. 2017;5:6847-59.
  11. Lin WJ, Yao N, Qian L, Zhang XF, Chen Q, Wang JF, Zhang LJ. pH-responsive unimolecular micelle-gold nanoparticles-drug nanohybrid system for cancer theranostics. Acta Biomater. 2017;58:455-65.
  12. Fang S, et al. Dual-stimuli responsive Nanotheranostics for multimodal imaging guided Trimodal synergistic therapy. Small. 2017;13.
  13. Chen Q, Feng LZ, Liu JJ, Zhu WW, Dong ZL, Wu YF, Liu Z. Intelligent albumin-MnO2 nanoparticles as pH-/H2O2-responsive dissociable Nanocarriers to modulate tumor hypoxia for effective combination therapy (vol 28, pg 7129, 2016). Adv Mater. 2018;30.
  14. Feng LL, et al. Controllable generation of free radicals from multifunctional heat-responsive Nanoplatform for targeted Cancer therapy. Chem Mater. 2018;30:526-39.
  15. Yang GB, et al. Manganese dioxide coated WS2@Fe3O4/sSiO(2) Nanocomposites for pH-responsive MR imaging and oxygen-elevated synergetic therapy. Small. 2018;14.
  16. Cho MH, Choi ES, Kim S, Goh SH, Choi Y. Redox-responsive manganese dioxide nanoparticles for enhanced MR imaging and radiotherapy of lung Cancer. Front Chem. 2017;5:109.
  17. Moreira AF, Dias DR, Correia IJ. Stimuli-responsive mesoporous silica nanoparticles for cancer therapy: a review. Micropor Mesopor Mat. 2016; 236:141-57.
  18. Li JM, Liu F, Shao Q, Min YZ, Costa M, Yeow EKL, Xing BG. Enzymeresponsive cell-penetrating peptide conjugated Mesoporous silica quantum dot Nanocarriers for controlled release of nucleus-targeted drug molecules and real-time intracellular fluorescence imaging of tumor cells. Adv Healthc Mater. 2014;3:1230-9.
  19. Hu QY, Katti PS, Gu Z. Enzyme-responsive nanomaterials for controlled drug delivery. Nanoscale. 2014;6:12273-86.
  20. de la Rica R, Aili D, Stevens MM. Enzyme-responsive nanoparticles for drug release and diagnostics. Adv Drug Deliver Rev. 2012;64:967-78.
  21. Bailey KM, Wojtkowiak JW, Cornnell HH, Ribeiro MC, Balagurunathan Y, Hashim AI, Gillies RJ. Mechanisms of buffer therapy resistance. Neoplasia. 2014;16:354-364 e351-353.
  22. Bogurcu N, Seidel S, Garvalov BK, Acker T. Analysis of hypoxia and the hypoxic response in tumor Xenografts. Methods Mol Biol. 2018;1742: 283-300.
  23. Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat Rev Cancer. 2011;11:393-410.
  24. Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science. 2005;307:58-62.
  25. Radisky ES, Raeeszadeh-Sarmazdeh M, Radisky DC. Therapeutic potential of matrix metalloproteinase inhibition in breast Cancer. J Cell Biochem. 2017;118:3531-48.
  26. Kessenbrock K, Plaks V, Werb Z. Matrix Metalloproteinases: regulators of the tumor microenvironment. Cell. 2010;141:52-67.
  27. Jiang T, Olson ES, Nguyen QT, Roy M, Jennings PA, Tsien RY. Tumor imaging by means of proteolytic activation of cell-penetrating peptides. P Natl Acad Sci USA. 2004;101:17867-72.
  28. Wang L, Huo M, Chen Y, Shi J. Tumor Microenvironment-Enabled Nanotherapy. Adv Healthc Mater. 2017;7:1704490.
  29. Mo R, Gu Z. Tumor microenvironment and intracellular signal-activated nanomaterials for anticancer drug delivery. Mater Today. 2016;19:274-83.
  30. John JV, Uthaman S, Augustine R, Chen HY, Park IK, Kim I. pH/redox dual stimuli-responsive sheddable nanodaisies for efficient intracellular tumourtriggered drug delivery. J Mater Chem B. 2017;5:5027-36.
  31. Johnson RP, et al. Glutathione and endosomal pH-responsive hybrid vesicles fabricated by zwitterionic polymer block poly(L-aspartic acid) as a smart anticancer delivery platform. React Funct Polym. 2017;119:47-56.
  32. John JV, Uthaman S, Augustine R, Lekshmi KM, Park IK, Kim I. Biomimetic pH/redox dual stimuli-responsive zwitterionic polymer block poly((L)-histidine) micelles for intracellular delivery of doxorubicin into tumor cells. J Polym Sci Pol Chem. 2017;55:2061-70.
  33. Johnson RP, et al. Poly(PEGA)-b-poly(L-lysine)-b-poly(L-histidine) hybrid vesicles for Tumoral pH-triggered intracellular delivery of doxorubicin hydrochloride. Acs Appl Mater Inter. 2015;7:21770-9.
  34. Kang EB, Lee JE, Mazrad ZAI, In I, Jeong JH, Park SY. pH-responsible fluorescent carbon nanoparticles for tumor selective theranostics via pHturn on/off fluorescence and photothermal effect in vivo and in vitro. Nanoscale. 2018;10:2512-23.
  35. Zhang M, et al. Ingenious pH-sensitive dextran/mesoporous silica nanoparticles based drug delivery systems for controlled intracellular drug release. Int J Biol Macromol. 2017;98:691-700.
  36. Zhu S, Lansakara-P DS, Li X, Cui Z. Lysosomal delivery of a lipophilic gemcitabine prodrug using novel acid-sensitive micelles improved its antitumor activity. Bioconjug Chem. 2012;23:966-80.
  37. Thambi T, Deepagan VG, Yoo CK, Park JH. Synthesis and physicochemical characterization of amphiphilic block copolymers bearing acid-sensitive orthoester linkage as the drug carrier. Polymer. 2011;52:4753-9.
  38. Zha Q, Wang X, Cheng X, Fu SX, Yang GQ, Yao WJ, Tang RP. Aciddegradable carboxymethyl chitosan nanogels via an ortho ester linkage mediated improved penetration and growth inhibition of 3-D tumor spheroids in vitro. Mat Sci Eng C-Mater. 2017;78:246-57.
  39. Belali S, Karimi AR, Hadizadeh M. Cell-specific and pH-sensitive nanostructure hydrogel based on chitosan as a photosensitizer carrier for selective photodynamic therapy. Int J Biol Macromol. 2018;110: 437-48.
  40. Tao YC, Liu SW, Zhang Y, Chi ZG, Xu JR. A pH-responsive polymer based on dynamic imine bonds as a drug delivery material with pseudo target release behavior. Polym Chem-Uk. 2018;9:878-84.
  41. Popat A, Liu J, Lu GQ, Qiao SZ. A pH-responsive drug delivery system based on chitosan coated mesoporous silica nanoparticles. J Mater Chem. 2012;22: 11173-8.
  42. Liao JH, et al. Tumor-targeting and pH-responsive nanoparticles from hyaluronic acid for the enhanced delivery of doxorubicin. Int J Biol Macromol. 2018;113:737-47.
  43. He H, et al. Selective cancer treatment via photodynamic sensitization of hypoxia-responsive drug delivery. Nanoscale. 2017;10:2856-65.
  44. Perche F, Biswas S, Wang T, Zhu L, Torchilin VP. Hypoxia- targeted siRNA delivery. Angew Chem Int Edit. 2014;53:3362-6.
  45. Xie ZQ, et al. Targeting tumor hypoxia with stimulus-responsive nanocarriers in overcoming drug resistance and monitoring anticancer efficacy. Acta Biomater. 2018;71:351-62.
  46. Yang GB, et al. Smart Nanoreactors for pH-responsive tumor homing, mitochondria-targeting, and enhanced photodynamic-immunotherapy of Cancer. Nano Lett. 2018;18:2475-84.
  47. Sun L, et al. Cell-permeable, MMP-2 Activatable, nickel ferrite and his-tagged fusion protein self-assembled fluorescent Nanoprobe for tumor magnetictargeting and imaging. Acs Appl Mater Inter. 2017;9:39209-22.
  48. Ma P, et al. Overcoming multidrug resistance through the GLUT1-mediated and enzyme-triggered mitochondrial targeting conjugate with redoxsensitive paclitaxel release. Acs Appl Mater Inter. 2018;15(10):12351-63.
  49. Ansari C, et al. Development of novel tumor-targeted Theranostic nanoparticles activated by membrane-type matrix Metalloproteinases for combined Cancer magnetic resonance imaging and therapy. Small. 2014;10:566-75.
  50. Sharma A, Kim EJ, Shi H, Lee JY, Chung BG, Kim JS. Development of a theranostic prodrug for colon cancer therapy by combining ligand-targeted delivery and enzyme-stimulated activation. Biomaterials. 2018;155:145-51.
  51. Luo C, et al. Facile Fabrication of Tumor Redox-Sensitive Nanoassemblies of Small-Molecule Oleate Prodrug as Potent Chemotherapeutic Nanomedicine. Small (Weinheim an der Bergstrasse, Germany). 2016;12(46):6353-62. https://doi.org/10.1002/smll.201601597
  52. Sun C, Li X, Du X, Wang T. Redox-responsive micelles for triggered drug delivery and effective laryngopharyngeal cancer therapy. Int J Biol Macromol. 2018;112:65-73.
  53. Zhou Y, Wang S, Ying X, Wang Y, Geng P, Deng A, Yu Z. Doxorubicinloaded redox-responsive micelles based on dextran and indomethacin for resistant breast cancer. Int J Nanomedicine. 2017;12:6153-68.
  54. Xia YC, et al. Polycarbonate-based core-crosslinked redox-responsive nanoparticles for targeted delivery of anticancer drug. J Mater Chem B. 2018;6:3348-57.
  55. Aubry S, et al. Cell-surface thiols affect cell entry of disulfide-conjugated peptides. FASEB J. 2009;23:2956-67.
  56. Lux CD, Joshi-Barr S, Nguyen T, Mahmoud E, Schopf E, Fomina N, Almutairi A. Biocompatible polymeric nanoparticles degrade and release cargo in response to biologically relevant levels of hydrogen peroxide. J Am Chem Soc. 2012;134:15758-64.
  57. Sun C, et al. A ROS-responsive polymeric micelle with a ${\pi}$-conjugated thioketal moiety for enhanced drug loading and efficient drug delivery. Org Biomol Chem. 2017;15:9176-85.
  58. Dai L, et al. Photosensitizer enhanced disassembly of amphiphilic micelle for ROS-response targeted tumor therapy in vivo. Biomaterials. 2016;104:1-17.
  59. Yu L, Yang Y, Du F-SS, Li Z-CC. ROS-responsive Chalcogen-containing polycarbonates for photodynamic therapy. Biomacromolecules. 2018;19:2182-93.
  60. Xiong D, Zhang XF, Peng SY, Gu HW, Zhang LJ. Smart pH-sensitive micelles based on redox degradable polymers as DOX/GNPs carriers for controlled drug release and CT imaging. Colloid Surface B. 2018;163:29-40.
  61. Chen Q, Feng L, Liu J, Zhu W, Dong Z, Wu Y, Liu Z. Intelligent Albumin-MnO2Nanoparticles as pH-/H2O2-Responsive Dissociable Nanocarriers to Modulate Tumor Hypoxia for Effective Combination Therapy. Advanced materials (Deerfield Beach, Fla). 2018;30:7129-36.
  62. Jia X, Zhang Y, Zou Y, Wang Y, Niu D, He Q, Huang Z, Zhu W, Tian H, Shi J, Li Y. Dual Intratumoral Redox/Enzyme-Responsive NO-Releasing Nanomedicine for the Specific, High-Efficacy, and Low-Toxic Cancer Therapy. Advanced Materials. 2018;30(30):1704490.
  63. Feng QS, et al. Self-assembly of gold nanoparticles shows microenvironment-mediated dynamic switching and enhanced brain tumor targeting. Theranostics. 2017;7:1875-89.
  64. Feng Q, et al. Hypoxia-specific therapeutic agents delivery nanotheranostics: a sequential strategy for ultrasound mediated on-demand tritherapies and imaging of cancer. J Control Release. 2018;275:192-200.
  65. He H, Zhu RY, Sun W, Cai KM, Chen YB, Yin LC. Selective cancer treatment via photodynamic sensitization of hypoxia-responsive drug delivery. Nanoscale. 2018;10:2856-65.

Cited by

  1. Integrating nanomedicine into clinical radiotherapy regimens vol.144, pp.None, 2018, https://doi.org/10.1016/j.addr.2019.07.002
  2. Causes, consequences, and therapy of tumors acidosis vol.38, pp.1, 2018, https://doi.org/10.1007/s10555-019-09792-7
  3. H2O2-responsive nano-prodrug for podophyllotoxin delivery vol.7, pp.6, 2019, https://doi.org/10.1039/c9bm00344d
  4. In Situ Synthesis of Amino Acid Functionalized Carbon Dots with Tunable Properties and Their Biological Applications vol.2, pp.8, 2018, https://doi.org/10.1021/acsabm.9b00374
  5. Nanotechnology and Immunotherapy in Ovarian Cancer: Tracing New Landscapes vol.370, pp.3, 2019, https://doi.org/10.1124/jpet.118.254979
  6. Hyaluronic Acid-Conjugated with Hyperbranched Chlorin e6 Using Disulfide Linkage and Its Nanophotosensitizer for Enhanced Photodynamic Therapy of Cancer Cells vol.12, pp.19, 2018, https://doi.org/10.3390/ma12193080
  7. A γ-cyclodextrin-based metal-organic framework embedded with graphene quantum dots and modified with PEGMA via SI-ATRP for anticancer drug delivery and therapy vol.11, pp.43, 2018, https://doi.org/10.1039/c9nr06195a
  8. Cancer nanomedicine: focus on recent developments and self-assembled peptide nanocarriers vol.7, pp.48, 2018, https://doi.org/10.1039/c9tb01842e
  9. Tumor microenvironment-responsive contrast agents for specific cancer imaging: a narrative review vol.3, pp.4, 2018, https://doi.org/10.1097/jbr.0000000000000075
  10. Synthetic immunity by remote control vol.10, pp.8, 2018, https://doi.org/10.7150/thno.41305
  11. Nano-Therapies for Glioblastoma Treatment vol.12, pp.1, 2018, https://doi.org/10.3390/cancers12010242
  12. Enhanced cytotoxicity of a redox-sensitive hyaluronic acid-based nanomedicine toward different oncocytes via various internalization mechanisms vol.27, pp.1, 2018, https://doi.org/10.1080/10717544.2019.1709919
  13. Extracellular Acidosis Promotes Metastatic Potency via Decrease of the BMAL1 Circadian Clock Gene in Breast Cancer vol.9, pp.4, 2018, https://doi.org/10.3390/cells9040989
  14. Factors Influencing the Delivery Efficiency of Cancer Nanomedicines vol.21, pp.4, 2018, https://doi.org/10.1208/s12249-020-01691-3
  15. Overview of tumor environment responsive nano-drug delivery systems in tumor immunotherapy vol.512, pp.None, 2018, https://doi.org/10.1088/1755-1315/512/1/012096
  16. Enhanced nanoparticle accumulation by tumor-acidity-activatable release of sildenafil to induce vasodilation vol.8, pp.11, 2018, https://doi.org/10.1039/d0bm00466a
  17. Affinity-Driven Design of Cargo-Switching Nanoparticles to Leverage a Cholesterol-Rich Microenvironment for Atherosclerosis Therapy vol.14, pp.6, 2018, https://doi.org/10.1021/acsnano.9b08216
  18. Evaluating the benefits of renin-angiotensin system inhibitors as cancer treatments vol.211, pp.None, 2018, https://doi.org/10.1016/j.pharmthera.2020.107527
  19. Smart stimuli-responsive biopolymeric nanomedicines for targeted therapy of solid tumors vol.15, pp.22, 2018, https://doi.org/10.2217/nnm-2020-0146
  20. Development of pH/Glutathione-Responsive Theranostic Agents Activated by Glutathione S-Transferase π for Human Colon Cancer vol.63, pp.17, 2018, https://doi.org/10.1021/acs.jmedchem.0c00354
  21. Electrospray-based synthesis of fluorescent poly(D,L-lactide-co-glycolide) nanoparticles for the efficient delivery of an anticancer drug and self-monitoring its effect i vol.1, pp.8, 2018, https://doi.org/10.1039/d0ma00646g
  22. An Overview of Bioactive Natural Products-Based Nano-Drug Delivery Systems in Antitumor Chemotherapy vol.271, pp.None, 2018, https://doi.org/10.1051/e3sconf/202127103042
  23. TME-Responsive Multistage Nanoplatform for siRNA Delivery and Effective Cancer Therapy vol.16, pp.None, 2018, https://doi.org/10.2147/ijn.s322901
  24. Drug Delivery of Natural Products Through Nanocarriers for Effective Breast Cancer Therapy: A Comprehensive Review of Literature vol.16, pp.None, 2021, https://doi.org/10.2147/ijn.s328135
  25. Underlying mechanisms and drug intervention strategies for the tumour microenvironment vol.40, pp.1, 2021, https://doi.org/10.1186/s13046-021-01893-y
  26. Tumor-Acidity Responsive Polymeric Nanoparticles for Targeting Delivery of Angiogenesis Inhibitor for Enhanced Antitumor Efficacy With Decreased Toxicity vol.9, pp.None, 2018, https://doi.org/10.3389/fbioe.2021.664051
  27. Noyes-Whitney Dissolution Model-Based pH-Sensitive Slow Release of Paclitaxel (Taxol) from Human Hair-Derived Keratin Microparticle Carriers vol.2021, pp.None, 2018, https://doi.org/10.1155/2021/6657482
  28. Nanotherapeutics and Nanotheragnostics for Cancers: Properties, Pharmacokinetics, Biopharmaceutics, and Biosafety vol.27, pp.None, 2021, https://doi.org/10.2174/1381612827666210804102645
  29. Reactive Oxygen Species‐Regulating Strategies Based on Nanomaterials for Disease Treatment vol.8, pp.3, 2018, https://doi.org/10.1002/advs.202002797
  30. Surface engineered amphiphilic carbon dots: solvatochromic behavior and applicability as a molecular probe vol.9, pp.5, 2021, https://doi.org/10.1039/d0tb02007a
  31. Multifunctional and stimuli-responsive nanocarriers for targeted therapeutic delivery vol.18, pp.2, 2018, https://doi.org/10.1080/17425247.2021.1828339
  32. Emerging Strategies in Anticancer Combination Therapy Employing Silica‐Based Nanosystems vol.16, pp.3, 2018, https://doi.org/10.1002/biot.201900438
  33. Nanovectorization of Prostate Cancer Treatment Strategies: A New Approach to Improved Outcomes vol.13, pp.5, 2018, https://doi.org/10.3390/pharmaceutics13050591
  34. Intelligent Nanoparticle-Based Dressings for Bacterial Wound Infections vol.4, pp.5, 2018, https://doi.org/10.1021/acsabm.0c01168
  35. Role of intravital imaging in nanomedicine-assisted anti-cancer therapy vol.69, pp.None, 2018, https://doi.org/10.1016/j.copbio.2020.12.024
  36. Evaluation of the Anticancer Activity of Phytomolecules Conjugated Gold Nanoparticles Synthesized by Aqueous Extracts of Zingiber officinale (Ginger) and Nigella sativa L. Seeds (Black Cumin) vol.14, pp.12, 2018, https://doi.org/10.3390/ma14123368
  37. Anisamide-modified dual-responsive drug delivery system with MRI capacity for cancer targeting therapy vol.340, pp.None, 2021, https://doi.org/10.1016/j.molliq.2021.116889