6,807
Views
48
CrossRef citations to date
0
Altmetric
Focus on Bio-inspired nanomaterials

Functional biomimetic nanoparticles for drug delivery and theranostic applications in cancer treatment

, , , &
Pages 771-790 | Received 01 Jun 2018, Accepted 24 Sep 2018, Published online: 26 Oct 2018

References

  • Neal JW, Sledge GW. Decade in review-targeted therapy: successes, toxicities and challenges in solid tumours. Nat Rev Clin Oncol. 2014;11:627–628.
  • Metzger-Filho O, Moulin C, Awada A. Molecular targeted therapy in prevalent tumors: learning from the past and future perspectives. Curr Clin Pharmacol. 2010;5:166–177.
  • DeVita VT Jr., Chu E. A history of cancer chemotherapy. Cancer Res. 2008;68:8643–8653.
  • Cho K, Wang X, Nie S, et al. Therapeutic nanoparticles for drug delivery in cancer. Clin Cancer Res. 2008;14:1310–1316.
  • Parveen S, Sahoo SK. Polymeric nanoparticles for cancer therapy. J Drug Target. 2008;16:108–123.
  • Szakacs G, Paterson JK, Ludwig JA, et al. Targeting multidrug resistance in cancer. Nat Rev Drug Discov. 2006;5:219–234.
  • Rawat M, Singh D, Saraf S, et al. Nanocarriers: promising vehicle for bioactive drugs. Biol Pharm Bull. 2006;29:1790–1798.
  • Lipinski CA, Lombardo F, Dominy BW, et al. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev. 2001;46:3–26.
  • Zhang X, Huang Y, Li S. Nanomicellar carriers for targeted delivery of anticancer agents. Ther Deliv. 2014;5:53–68.
  • Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002;2:48–58.
  • Borst P, Elferink RO. Mammalian ABC transporters in health and disease. Annu Rev Biochem. 2002;71:537–592.
  • Fojo T, Bates S. Strategies for reversing drug resistance. Oncogene. 2003;22:7512–7523.
  • Hu CMJ, Zhang LF. Therapeutic nanoparticles to combat cancer drug resistance. Curr Drug Metab. 2009;10:836–841.
  • Haag R, Kratz F. Polymer therapeutics: concepts and applications. Angew Chem Int Ed. 2006;45:1198–1215.
  • Moghimi SM, Hunter AC, Murray JC. Nanomedicine: current status and future prospects. FASEB J. 2005;19:311–330.
  • Torchilin VP. Multifunctional nanocarriers. Adv Drug Deliv Rev. 2006;58:1532–1555.
  • Wang S, Huang P, Chen X. Hierarchical targeting strategy for enhanced tumor tissue accumulation/retention and cellular internalization. Adv Mater. 2016;28:7340–7364.
  • Wang S, Huang P, Chen X. Stimuli-responsive programmed specific targeting in nanomedicine. ACS Nano. 2016;10:2991–2994.
  • Calderon M, Quadir MA, Strumia M, et al. Functional dendritic polymer architectures as stimuli-responsive nanocarriers. Biochimie. 2010;92:1242–1251.
  • Aluri S, Janib SM, Mackay JA. Environmentally responsive peptides as anticancer drug carriers. Adv Drug Deliv Rev. 2009;61:940–952.
  • Vaupel P, Kallinowski F, Okunieff P. Blood-flow, oxygen and nutrient supply, and metabolic microenvironment of human-tumors – a review. Cancer Res. 1989;49:6449–6465.
  • Shenoy D, Little S, Langer R, et al. Poly(ethylene oxide)-modified poly(beta-amino ester) nanoparticles as a pH-sensitive system for tumor-targeted delivery of hydrophobic drugs: part 2. In vivo distribution and tumor localization studies. Pharm Res. 2005;22:2107–2114.
  • Shenoy D, Little S, Langer R, et al. Poly(ethylene oxide)-modified poly(beta-amino ester) nanoparticles as a pH-sensitive system for tumor-targeted delivery of hydrophobic drugs. 1. In vitro evaluations. Mol Pharm. 2005;2:357–366.
  • Shenoy DB, Amiji MM. Poly(ethylene oxide)-modified poly(epsilon-caprolactone) nanoparticles for targeted delivery of tamoxifen in breast cancer. Int J Pharm. 2005;293:261–270.
  • Kommareddy S, Amiji M. Preparation and evaluation of thiol-modified gelatin nanoparticles for intracellular DNA delivery in response to glutathione. Bioconjug Chem. 2005;16:1423–1432.
  • Jhaveri A, Pranali Deshpande TV. Stimuli-sensitive nanopreparations for combination cancer therapy. J Control Release. 2014;190:352–370.
  • Liu Y, Qu Z, Cao H, et al. pH switchable nanoassembly for imaging a broad range of malignant tumors. ACS Nano. 2017;11:12446–12452.
  • Schmaljohann D. Thermo- and pH-responsive polymers in drug delivery. Adv Drug Deliv Rev. 2006;58:1655–1670.
  • Gerweck LE, Seetharaman K. Cellular pH gradient in tumor versus normal tissue: potential exploitation for the treatment of cancer. Cancer Res. 1996;56:1194–1198.
  • Hunt CA, Macgregor RD, Siegel RA. Engineering targeted in vivo drug delivery. I. The physiological and physicochemical principles governing opportunities and limitations. Pharm Res. 1986;3:333–344.
  • Bolot G, David MJ, Taki T, et al. Analysis of glycosphingolipids of human head and neck carcinomas with comparison to normal tissue. Biochem Mol Biol Int. 1998;46:125–135.
  • Wike-Hooley JL, Haveman J, Reinhold HS. The relevance of tumour pH to the treatment of malignant disease. Radiother Oncol. 1984;2:343–366.
  • Wu H, Zhu L, Torchilin VP. pH-sensitive poly(histidine)-PEG/DSPE-PEG co-polymer micelles for cytosolic drug delivery. Biomaterials. 2013;34:1213–1222.
  • Kost J, Langer R. Responsive polymeric delivery systems. Adv Drug Deliver Rev. 2001;46:125–148.
  • Zhang HJ, Wang CL, Chen BA, et al. Daunorubicin-TiO2 nanocomposites as a “smart” pH-responsive drug delivery system. Int J Nanomed. 2012;7:235–242.
  • Ganta S, Devalapally H, Shahiwala A, et al. A review of stimuli-responsive nanocarriers for drug and gene delivery. J Control Release. 2008;126:187–204.
  • Gil ES, Hudson SM. Stimuli-responsive polymers and their bioconjugates. Prog Polym Sci. 2004;29:1173–1222.
  • Lee ES, Gao ZG, Bae YH. Recent progress in tumor pH targeting nanotechnology. J Control Release. 2008;132:164–170.
  • Chang GT, Yu L, Yang ZG, et al. A delicate ionizable-group effect on self-assembly and thermogelling of amphiphilic block copolymers in water. Polymer. 2009;50:6111–6120.
  • Knorr V, Russ V, Allmendinger L, et al. Acetal linked oligoethylenimines for use as pH-sensitive gene carriers. Bioconjug Chem. 2008;19:1625–1634.
  • Murthy N, Campbell J, Fausto N, et al. Design and synthesis of pH-responsive polymeric carriers that target uptake and enhance the intracellular delivery of oligonucleotides. J Control Release. 2003;89:365–374.
  • Tomlinson R, Heller J, Brocchini S, et al. Polyacetal-doxorubicin conjugates designed for pH-dependent degradation. Bioconjug Chem. 2003;14:1096–1106.
  • Ke CJ, Su TY, Chen HL, et al. Smart multifunctional hollow microspheres for the quick release of drugs in Intracellular Lysosomal compartments. Angew Chem Int Edit. 2011;50:8086–8089.
  • Liu J, Ma HL, Wei T, et al. CO2 gas induced drug release from pH-sensitive liposome to circumvent doxorubicin resistant cells. Chem Commun. 2012;48:4869–4871.
  • Guo XD, Zhang LJ, Chen Y, et al. Core/shell pH-sensitive micelles self-assembled from cholesterol conjugated oligopeptides for anticancer drug delivery. Aiche J. 2010;56:1922–1931.
  • Chen W, Meng FH, Cheng R, et al. pH-sensitive degradable polymersomes for triggered release of anticancer drugs: a comparative study with micelles. J Control Release. 2010;142:40–46.
  • Makhof A, Tozuka Y, Takeuchi H. pH-Sensitive nanospheres for colon-specific drug delivery in experimentally induced colitis rat model. Eur J Pharm Biopharm. 2009;72:1–8.
  • Bhattarai N, Gunn J, Zhang MQ. Chitosan-based hydrogels for controlled, localized drug delivery. Adv Drug Deliv Rev. 2010;62:83–99.
  • Yuba E, Kojima C, Sakaguchi N, et al. Gene delivery to dendritic cells mediated by complexes of lipoplexes and pH-sensitive fusogenic polymer-modified liposomes. J Control Release. 2008;130:77–83.
  • Shen M, Huang YZ, Han LM, et al. Multifunctional drug delivery system for targeting tumor and its acidic microenvironment. J Control Release. 2012;161:884–892.
  • Tomita S, Sato K, Anzai JI. Layer-by-layer assembled thin films composed of carboxyl-terminated poly(amidoamine) dendrimer as a pH-sensitive nano-device. J Colloid Interface Sci. 2008;326:35–40.
  • Hruby M, Konak C, Ulbrich K. Polymeric micellar pH-sensitive drug delivery system for doxorubicin. J Control Release. 2005;103:137–148.
  • Bae Y, Alani AW, Rockich NC, et al. Mixed pH-sensitive polymeric micelles for combination drug delivery. Pharm Res. 2010;27:2421–2432.
  • Liu X, Chen B, Li X, et al. Self-assembly of BODIPY based pH-sensitive near-infrared polymeric micelles for drug controlled delivery and fluorescence imaging applications. Nanoscale. 2015;7:16399–16416.
  • Liu Y, Feng L, Liu T, et al. Multifunctional pH-sensitive polymeric nanoparticles for theranostics evaluated experimentally in cancer. Nanoscale. 2014;6:3231–3242.
  • Gupta P, Vermani K, Garg S. Hydrogels: from controlled release to pH-responsive drug delivery. Drug Discov Today. 2002;7:569–579.
  • Bajaj I, Singhal R. Poly (glutamic acid) – an emerging biopolymer of commercial interest. Bioresour Technol. 2011;102:5551–5561.
  • Yue YM, Sheng X, Wang PX. Fabrication and characterization of microstructured and pH sensitive interpenetrating networks hydrogel films and application in drug delivery field. Eur Polym J. 2009;45:309–315.
  • Zignani M, Drummond DC, Meyer O, et al. In vitro characterization of a novel polymeric-based pH-sensitive liposome system. Biochim Biophys Acta. 2000;1463:383–394.
  • Mishra RK, Ramasamy K, Ahmad NA, et al. pH dependent poly[2-(methacryloyloxyethyl)trimetylammonium chloride-co-methacrylic acid]hydrogels for enhanced targeted delivery of 5-fluorouracil in colon cancer cells. J Mater Sci Mater Med. 2014;25:999–1012.
  • Li L, Gu J, Zhang J, et al. Injectable and biodegradable pH-responsive hydrogels for localized and sustained treatment of human fibrosarcoma. ACS Appl Mater Interfaces. 2015;7:8033–8040.
  • Torchilin VP. Recent advances with liposomes as pharmaceutical carriers. Nat Rev Drug Discov. 2005;4:145–160.
  • Allen TM, Cullis PR. Drug delivery systems: entering the mainstream. Science. 2004;303:1818–1822.
  • Drummond DC, Meyer O, Hong KL, et al. Optimizing liposomes for delivery of chemotherapeutic agents to solid tumors. Pharmacol Rev. 1999;51:691–743.
  • Zhou F, Huang L. Liposome-mediated cytoplasmic delivery of proteins: an effective means of accessing the MHC class I-restricted antigen presentation pathway. Immunomethods. 1994;4:229–235.
  • Reddy R, Zhou F, Huang L, et al. pH sensitive liposomes provide an efficient means of sensitizing target cells to class I restricted CTL recognition of a soluble protein. J Immunol Methods. 1991;141:157–163.
  • Yuba E, Kojima C, Harada A, et al. pH-Sensitive fusogenic polymer-modified liposomes as a carrier of antigenic proteins for activation of cellular immunity. Biomaterials. 2010;31:943–951.
  • Tachibana R, Harashima H, Shono M, et al. Intracellular regulation of macromolecules using pH-sensitive liposomes and nuclear localization signal: qualitative and quantitative evaluation of intracellular trafficking. Biochem Biophys Res Commun. 1998;251:538–544.
  • Chiang YT, Lo CL. pH-Responsive polymer-liposomes for intracellular drug delivery and tumor extracellular matrix switched-on targeted cancer therapy. Biomaterials. 2014;35:5414–5424.
  • Yuba E, Harada A, Sakanishi Y, et al. A liposome-based antigen delivery system using pH-sensitive fusogenic polymers for cancer immunotherapy. Biomaterials. 2013;34:3042–3052.
  • Yuba E, Tajima N, Yoshizaki Y, et al. Dextran derivative-based pH-sensitive liposomes for cancer immunotherapy. Biomaterials. 2014;35:3091–3101.
  • Lu J, Yoshimura K, Goto K, et al. Nanoformulation of geranylgeranyltransferase-I inhibitors for cancer therapy: liposomal encapsulation and pH-dependent delivery to cancer cells. PLoS One. 2015;10:e0137595.
  • Yuba E, Kanda Y, Yoshizaki Y, et al. pH-sensitive polymer-liposome-based antigen delivery systems potentiated with interferon-gamma gene lipoplex for efficient cancer immunotherapy. Biomaterials. 2015;67:214–224.
  • Chen Y, Sun J, Lu Y, et al. Complexes containing cationic and anionic pH-sensitive liposomes: comparative study of factors influencing plasmid DNA gene delivery to tumors. Int J Nanomed. 2013;8:1573–1593.
  • Turner C, Weir N, Catterall C, et al. The transfection of Jurkat T-leukemic cells by use of pH-sensitive immunoliposomes. J Liposome Res. 2002;12:45–50.
  • Ishida T, Okada Y, Kobayashi T, et al. Development of pH-sensitive liposomes that efficiently retain encapsulated doxorubicin (DXR) in blood. Int J Pharm. 2006;309:94–100.
  • Alb DB, LdG M, Soares DCF, et al. Long-circulating, pH-sensitive liposomes versus long-circulating, non-pH-sensitive liposomes as a delivery system for tumor identification. J Biomed Nanotechnol. 2013;9:1636–1643.
  • Hui H, Fan XD, Cao ZL. Thermo- and pH-sensitive dendrimer derivatives with a shell of poly (N,N-dimethylaminoethyl methacrylate) and study of their controlled drug release behavior. Polymer. 2005;46:9514–9522.
  • Lai PS, Lou PJ, Peng CL, et al. Doxorubicin delivery by polyamidoamine dendrimer conjugation and photochemical internalization for cancer therapy. J Control Release. 2007;122:39–46.
  • Iikuni N, Hahn BH, La Cava A. Potential for anti-DNA immunoglobulin peptide therapy in systemic lupus erythematosus. Expert Opin Biol Ther. 2009;9:201–206.
  • Xiao YF, Jie MM, Li BS, et al. Peptide-based treatment: a promising cancer therapy. J Immunol Res. 2015;2015:761820.
  • Li WJ, Nicol F, Szoka FC. GALA: a designed synthetic pH-responsive amphipathic peptide with applications in drug and gene delivery. Adv Drug Deliver Rev. 2004;56:967–985.
  • Subbarao NK, Parente RA, Szoka FC, et al. The pH-dependent bilayer destabilization by an amphipathic peptide. Biochemistry. 1987;26:2964–2972.
  • Parente RA, Nir S, Szoka FC. pH-dependent fusion of phosphatidylcholine small vesicles – induction by a synthetic amphipathic peptide. J Biol Chem. 1988;263:4724–4730.
  • Parente RA, Nir S, Szoka FC. Mechanism of leakage of phospholipid vesicle contents induced by the peptide gala. Biochemistry. 1990;29:8720–8728.
  • Goormaghtigh E, Demeutter J, Szoka F, et al. Secondary structure and orientation of the amphipathic peptide gala in lipid structures – an infrared-spectroscopic approach. Eur J Biochem. 1991;195:421–429.
  • Yoshizak Y, Yuba E, Komatsu T, et al. Improvement of peptide-based tumor immunotherapy using pH-sensitive fusogenic polymer-modified liposomes. Molecules. 2016;21:1–13.
  • Yamada Y, Shinohara Y, Kakudo T, et al. Mitochondrial delivery of mastoparan with transferrin liposomes equipped with a pH-sensitive fusogenic peptide for selective cancer therapy. Int J Pharm. 2005;303:1–7.
  • Morishita M, Takahashi Y, Nishikawa M, et al. Enhanced class I tumor antigen presentation via cytosolic delivery of exosomal cargos by tumor-cell-derived exosomes displaying a pH-sensitive fusogenic peptide. Mol Pharm. 2017;14:4079–4086.
  • Wang Q, Zhang X, Liao H, et al. Multifunctional shell–core nanoparticles for treatment of multidrug resistance hepatocellular carcinoma. Adv Funct Mater. 2018;28:1706124.
  • Norberg NS, Gamelin DR. Influence of surface modification on the luminescence of colloidal ZnO nanocrystals. J Phys Chem B. 2005;109:20810–20816.
  • Bang J, Yang H, Holloway PH. Enhanced and stable green emission of ZnO nanoparticles by surface segregation of Mg. Nanotechnology. 2006;17:973–978.
  • Rakshit S, Vasudevan S. Trap-state dynamics in visible-light-emitting ZnO:MgO nanocrystals. J Phys Chem C. 2008;112:4531–4537.
  • Muhammad F, Guo M, Guo Y, et al. Acid degradable ZnO quantum dots as a platform for targeted delivery of an anticancer drug. J Mater Chem. 2011;21:13406.
  • Zhang J, Wu D, Li MF, et al. Multifunctional mesoporous silica nanoparticles based on charge-reversal plug-gate nanovalves and acid-decomposable ZnO quantum dots for intracellular drug delivery. ACS Appl Mater Interfaces. 2015;7:26666–26673.
  • Kester M, Heakal Y, Fox T, et al. Calcium phosphate nanocomposite particles for in vitro imaging and encapsulated chemotherapeutic drug delivery to cancer cells. Nano Lett. 2008;8:4116–4121.
  • Ling D, Park W, Park SJ, et al. Multifunctional tumor pH-sensitive self-assembled nanoparticles for bimodal imaging and treatment of resistant heterogeneous tumors. J Am Chem Soc. 2014;136:5647–5655.
  • Wang J, Chen B, Zhao D, et al. Peptide decorated calcium phosphate/carboxymethyl chitosan hybrid nanoparticles with improved drug delivery efficiency. Int J Pharm. 2013;446:205–210.
  • Jelezova I, Drakalska E, Momekova D, et al. Curcumin loaded pH-sensitive hybrid lipid/block copolymer nanosized drug delivery systems. Eur J Pharm Sci. 2015;78:67–78.
  • Lu L, Unsworth LD. pH-triggered release of hydrophobic molecules from self-assembling hybrid nanoscaffolds. Biomacromolecules. 2016;17:1425–1436.
  • Li SX, Zheng JZ, Chen DJ, et al. Yolk-shell hybrid nanoparticles with magnetic and pH-sensitive properties for controlled anticancer drug delivery. Nanoscale. 2013;5:11718–11724.
  • Yang XQ, Grailer JJ, Rowland IJ, et al. Multifunctional stable and pH-responsive polymer vesicles formed by heterofunctional triblock copolymer for targeted anticancer drug delivery and ultrasensitive MR imaging. Acs Nano. 2010;4:6805–6817.
  • Chen DQ, Yang DZ, Dougherty CA, et al. In vivo targeting and positron emission tomography imaging of tumor with intrinsically radioactive metal-organic frameworks nanomaterials. ACS Nano. 2017;11:4315–4327.
  • Li ZY, Hu JJ, Xu Q, et al. A redox-responsive drug delivery system based on RGD containing peptide-capped mesoporous silica nanoparticles. J Mater Chem B. 2015;3:39–44.
  • Jin S, Wan JX, Meng LZ, et al. Biodegradation and toxicity of protease/redox/pH stimuli-responsive PEGlated PMAA nanohydrogels for targeting drug delivery. ACS Appl Mater Interfaces. 2015;7:19843–19852.
  • Noyhouzer T, L’Homme C, Beaulieu I, et al. Ferrocene-modified phospholipid: an innovative precursor for redox-triggered drug delivery vesicles selective to cancer cells. Langmuir. 2016;32:4169–4178.
  • Liu B, Chen YY, Li CX, et al. Poly(acrylic acid) modification of Nd3+-sensitized upconversion nanophosphors for highly efficient UCL imaging and pH-responsive drug delivery. Adv Funct Mater. 2015;25:4717–4729.
  • Ma N, Li Y, Xu HP, et al. Dual redox responsive assemblies formed from diselenide block copolymers. J Am Chem Soc. 2010;132:442.
  • Deng X, Yin Z, Lu J, et al. In situ monitoring of microrna replacement efficacy and accurate imaging-guided cancer therapy through light-up inter-polyelectrolyte nanocomplexes. Adv Sci. 2018;5:1700542.
  • Zhou J, Li MH, Lim WQ, et al. A transferrin-conjugated hollow nanoplatform for redox-controlled and targeted chemotherapy of tumor with reduced inflammatory reactions. Theranostics. 2018;8:518–532.
  • Kang Y, Lu L, Lan J, et al. Redox-responsive polymeric micelles formed by conjugating gambogic acid with bioreducible poly(amido amine)s for the co-delivery of docetaxel and MMP-9 shRNA. Acta Biomater. 2017;68:137–153.
  • Lux CD, Joshi-Barr S, Nguyen T, et al. Biocompatible polymeric nanoparticles degrade and release cargo in response to biologically relevant levels of hydrogen peroxide. J Am Chem Soc. 2012;134:15758–15764.
  • Shim MS, Xia YN. A reactive oxygen species (ROS)-responsive polymer for safe, efficient, and targeted gene delivery in cancer cells. Angew Chem Int Ed. 2013;52:6926–6929.
  • Hu QY, Katti PS, Gu Z. Enzyme-responsive nanomaterials for controlled drug delivery. Nanoscale. 2014;6:12273–12286.
  • Huang Z, Yao Q, Wei S, et al. Enzyme-Instructed Self-assembly in Biological Milieu for Theranostics Purpose. Curr Med Chem. 2017;25:1–13.
  • Ghadiali JE, Stevens MM. Enzyme-responsive nanoparticle systems. Adv Mater. 2008;20:4359–4363.
  • Andresen TL, Thompson DH, Kaasgaard T. Enzyme-triggered nanomedicine: drug release strategies in cancer therapy (invited review). Mol Membr Biol. 2010;27:353–363.
  • Ulijn RV. Enzyme-responsive materials: a new class of smart biomaterials. J Mater Chem. 2006;16:2217–2225.
  • Hu J, Zhang G, Liu S. Enzyme-responsive polymeric assemblies, nanoparticles and hydrogels. Chem Soc Rev. 2012;41:5933–5949.
  • de la Rica R, Aili D, Stevens MM. Enzyme-responsive nanoparticles for drug release and diagnostics. Adv Drug Deliv Rev. 2012;64:967–978.
  • Weissleder R, Tung CH, Mahmood U, et al. In vivo imaging of tumors with protease-activated near-infrared fluorescent probes. Nat Biotechnol. 1999;17:375–378.
  • Butterfield DA, Hardas SS, Lange ML. Oxidatively modified glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and Alzheimer’s disease: many pathways to neurodegeneration. J Alzheimers Dis. 2010;20:369–393.
  • Kundu JK, Surh YJ. Nrf2-Keap1 signaling as a potential target for chemoprevention of inflammation-associated carcinogenesis. Pharm Res. 2010;27:999–1013.
  • Gupta S, Andresen H, Ghadiali JE, et al. Kinase-actuated immunoaggregation of Peptide-conjugated gold nanoparticles. Small. 2010;6:1509–1513.
  • Gupta S, Andresen H, Stevens MM. Single-step kinase inhibitor screening using a peptide-modified gold nanoparticle platform. Chem Commun. 2011;47:2249–2251.
  • Deka SR, Yadav S, Mahato M, et al. Azobenzene-aminoglycoside: self-assembled smart amphiphilic nanostructures for drug delivery. Colloids Surf B Biointerfaces. 2015;135:150–157.
  • Welser K, Adsley R, Moore BM, et al. Protease sensing with nanoparticle based platforms. Analyst. 2011;136:29–41.
  • Anderson CF, Cui HG. Protease-Sensitive nanomaterials for cancer therapeutics and imaging. Ind Eng Chem Res. 2017;56:5761–5777.
  • Basel MT, Shrestha TB, Troyer DL, et al. Protease-sensitive, polymer-caged liposomes: a method for making highly targeted liposomes using triggered release. ACS Nano. 2011;5:2162–2175.
  • Kulkarni PS, Haldar MK, Nahire RR, et al. MMP-9 responsive PEG cleavable nanovesicles for efficient delivery of chemotherapeutics to pancreatic cancer. Mol Pharm. 2014;11:2390–2399.
  • Hou WX, Xia FF, Alves CS, et al. MMP2-targeting and redox-responsive PEGylated chlorine 6 nanoparticles for cancer near-infrared imaging and photodynamic therapy. ACS Appl Mater Inter. 2016;8:1447–1457.
  • Yamashita S, Yamashita J, Sakamoto K, et al. Increased expression of membrane-associated phospholipase-a2 shows malignant potential of human breast-cancer cells. Cancer. 1993;71:3058–3064.
  • Abe T, Sakamoto K, Kamohara H, et al. Group II phospholipase A2 is increased in peritoneal and pleural effusions in patients with various types of cancer. Int J Cancer. 1997;74:245–250.
  • Jensen SS, Andresen TL, Davidsen J, et al. Secretory phospholipase A2 as a tumor-specific trigger for targeted delivery of a novel class of liposomal prodrug anticancer ether lipids. Mol Cancer Ther. 2004;3:1451–1458.
  • Jin Y, Yang F, Du L. Nanoassemblies containing a fluorouracil/zidovudine glyceryl prodrug with phospholipase A2-triggered drug release for cancer treatment. Colloids Surf B Biointerfaces. 2013;112:421–428.
  • Weitzel JN, Pooler PA, Mohammed R, et al. A unique case of breast carcinoma producing pancreatic-type isoamylase. Gastroenterology. 1988;94:519–520.
  • Inaji H, Koyama H, Higashiyama M, et al. Immunohistochemical, ultrastructural and biochemical studies of an amylase-producing breast carcinoma. Virchows Arch A Pathol Anat Histopathol. 1991;419:29–33.
  • Cura JE, Blanzaco DP, Brisson C, et al. Phase I and pharmacokinetics study of crotoxin (cytotoxic PLA(2), NSC-624244) in patients with advanced cancer. Clin Cancer Res. 2002;8:1033–1041.
  • Bernardos A, Mondragon L, Aznar E, et al. Enzyme-responsive intracellular controlled release using nanometric silica mesoporous supports capped with “saccharides”. ACS Nano. 2010;4:6353–6368.
  • Butterfield DA, Hardas SS, Lange MLB. Oxidatively modified glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and Alzheimer’s disease: many pathways to neurodegeneration. J Alzheimers Dis. 2010;20:369–393.
  • Kundu JK, Surh YJ. Nrf2-keap1 signaling as a potential target for chemoprevention of inflammation-associated carcinogenesis. Pharm Res. 2010;27:999–1013.
  • Park C, Youn H, Kim H, et al. Cyclodextrin-covered gold nanoparticles for targeted delivery of an anti-cancer drug. J Mater Chem. 2009;19:2310–2315.