Generic placeholder image

Pharmaceutical Nanotechnology

Editor-in-Chief

ISSN (Print): 2211-7385
ISSN (Online): 2211-7393

Review Article

Bioresponse Inspired Nanomaterials for Targeted Drug and Gene Delivery

Author(s): Shrestha Sharma, Md. Noushad Javed, Faheem Hyder Pottoo*, Syed Arman Rabbani, Md. Abul Barkat, Harshita, Md. Sarafroz and Mohd. Amir

Volume 7, Issue 3, 2019

Page: [220 - 233] Pages: 14

DOI: 10.2174/2211738507666190429103814

Abstract

The traditional drug delivery techniques are unresponsive to the altering metabolic states of the body and fail to achieve target specific drug delivery, which results in toxic plasma concentrations. In order to harmonize the drug release profiles, diverse biological and pathological pathways and factors involved have been studied and consequently, nanomaterials and nanostructures are engineered in a manner so that they respond and interact with the target cells and tissues in a controlled manner to induce promising pharmacological responses with least undesirable effects. The bioinspired nanoparticles such as carbon nanotubes, metallic nanoparticles, and quantum dots sense the localized host environment for diagnosis and treatment of pathological states. These biocompatible polymeric- based nanostructures bind drugs to the specific receptors, which renders them as ideal vehicles for the delivery of drugs and gene. The ultimate goal of bioinspired nanocomposites is to achieve personalized diagnostic and therapeutic outcomes. This review briefly discussed current trends; role, recent advancements as well as different approaches, which are being used for designing and fabrication of some bioinspired nanocarriers.

Keywords: Bioinspired nanocarriers, biomimetics, carbon nano tubes, nanomaterials, polymers, quantum dots.

Graphical Abstract
[1]
Alam MS, Garg A, Pottoo FH, et al. Gum ghatti mediated, one pot green synthesis of optimized gold nanoparticles: investigation of process-variables impact using Box-Behnken based statistical design. Int J Biol Macromol 2017; 104(Pt A): 758-67.
[2]
Sharma S, Sahni J, Ali J, Baboota S. Patent perspective for potential antioxidant compounds-rutin and quercetin. Recent Pat Nanomed 2013; 3(1): 62-8.
[3]
Pottoo FH, Tabassum N. Triple drug combination for treatment of status epilepticus and/or partial seizures and/or partial seizures with associated neurological disorders. WO2017130208A1, 2017.
[4]
Pottoo FH, Tabassum N, Javed MN, et al. The synergistic effect of raloxifene, fluoxetine, and bromocriptine protects against pilocarpine-induced status epilepticus and temporal lobe epilepsy. Mol Neurobiol 2019; 56(2): 1233-47.
[5]
Sharma S, Ali A, Ali J, Sahni JK, Baboota S. Rutin : therapeutic potential and recent advances in drug delivery. Expert Opin Investig Drugs 2013; 22(8): 1063-79.
[6]
Barkat A, Harshita H, Beg S, et al. Current progress in synthesis, characterization and applications of silver nanoparticles: precepts and prospects. Recent Pat Antiinfect Drug Discov 2018; 13(1): 53-69.
[7]
Wong IY, Bhatia SN, Toner M. Nanotechnology: emerging tools for biology and medicine. Genes Dev 2013; 27(22): 2397-408.
[8]
Whitesides GM. The “right” size in nanobiotechnology. Nat Biotechnol 2003; 21(10): 1161-5.
[9]
Barkat MA, Harshita H, Ahmad I, et al. Nanosuspension-based aloe vera gel of silver sulfadiazine with improved wound healing activity. AAPS PharmSciTech 2017; 18(8): 3274-85.
[10]
Sharma S, Narang JK, Ali J, Baboota S. Synergistic antioxidant action of vitamin E and rutin SNEDDS in ameliorating oxidative stress in a Parkinson’s disease model. Nanotechnology 2016; 27(37)375101
[11]
Sharma S, Sahni JK, Ali J, Baboota S. Effect of high-pressure homogenization on formulation of TPGS loaded nanoemulsion of rutin - pharmacodynamic and antioxidant studies. Drug Deliv 2015; 22(4): 541-51.
[12]
Galaev IY, Mattiasson B. ‘Smart’ polymers and what they could do in biotechnology and medicine. Trends Biotechnol 1999; 17(8): 335-40.
[13]
Kumar A, Srivastava A, Galaev IY, Mattiasson B. Smart polymers: physical forms and bioengineering applications. Prog Polym Sci 2007; 32(10): 1205-37.
[14]
Sahoo SK, Labhasetwar V. Nanotech approaches to drug delivery and imaging. Drug Discov Today 2003; 8(24): 1112-20.
[15]
Gilmore JL, Yi X, Quan L, Kabanov AV. Novel nanomaterials for clinical neuroscience. J Neuroimmune Pharmacol 2008; 3(2): 83-94.
[16]
Sharma S, Kumar A, Sahni JK, Ali J, Baboota S. Nanoemulsion based hydrogel containing omega 3 fatty acids as a surrogate of betamethasone dipropionate for topical delivery. Adv Sci Lett 2012; 6(1): 221-31.
[17]
Nigar S, Pottoo FH, Tabassum N, Verma SK, Javed MN. Molecular insights into the role of inflammation and oxidative stress in epilepsy. J Adv Med Pharma Sci 2016; 10(1): 1-9.
[18]
Pottoo FH, Tabassum N, Darzi MM. Bromocriptine mesylate protects against status epilepticus and temporal lobe epilepsy: neurobehavioral, histopathological and neurochemical evidences. Int Neuropsychiatr Dis Treat 2016; 6(4): 1-13.
[19]
Schmaljohann D. Thermo- and pH-responsive polymers in drug delivery. Adv Drug Deliv Rev 2006; 58(15): 1655-70.
[20]
Chen S, Singh J. Controlled delivery of testosterone from smart polymer solution based systems: in vitro evaluation. Int J Pharm 2005; 295(1-2): 183-90.
[21]
Amit K, Sonam R. Pulsatile drug delivery system: method and technology review. Int J Drug Dev Res 2012; 4(4): 95-107.
[22]
Bawa P, Pillay V, Choonara YE, du Toit LC. Stimuli-responsive polymers and their applications in drug delivery. Biomed Mater 2009; 4(2)022001
[23]
Lalwani1 A, Santani DD. Pulsatile drug delivery systems. Int J Pharma Sci 2007; 69(4): 489.
[24]
Al-Tahami K, Singh J. Smart polymer based delivery systems for peptides and proteins. Recent Pat Drug Deliv Formul 2007; 1(1): 65-71.
[25]
Kopeček J. Smart and genetically engineered biomaterials and drug delivery systems. Eur J Pharm Sci 2003; 20(1): 1-16.
[26]
Roy I, Gupta MN. Smart Polymeric materials: emerging biochemical applications. Chem Biol 2003; 10(12): 1161-71.
[27]
Mulens V, Morales MD, Barber DF. Development of magnetic nanoparticles for cancer gene therapy: a comprehensive review. ISRN Nanomater 2013; 2013: 1-14.
[28]
Hosseinkhani H, Chen Y-R, He W, Hong P-D, Yu D-S, Domb AJ. Engineering of magnetic DNA nanoparticles for tumor-targeted therapy. J Nanopart Res 2013; 15(1): 1345.
[29]
Pisanic TR, Blackwell JD, Shubayev VI, Fiñones RR, Jin S. Nanotoxicity of iron oxide nanoparticle internalization in growing neurons. Biomaterials 2007; 28(16): 2572-81.
[30]
Jain TK, Reddy MK, Morales MA, Leslie-Pelecky DL, Labhasetwar V. Biodistribution, clearance, and biocompatibility of iron oxide magnetic nanoparticles in rats. Mol Pharm 2008; 5(2): 316-27.
[31]
Berry CC. Progress in functionalization of magnetic nanoparticles for applications in biomedicine. J Phys D Appl Phys 2009; 42(22)224003
[32]
Giri J, Ray A, Dasgupta S, Datta D, Bahadur D. Investigation on Tc tuned nano particles of magnetic oxides for hyperthermia applications. Biomed Mater Eng 2003; 13(4): 387-99.
[33]
Shinkai M. Functional magnetic particles for medical application. J Biosci Bioeng 2002; 94(6): 606-13.
[34]
Jin H, Kang KA. Application of novel metal nanoparticles as optical/thermal agents in optical mammography and hyperthermic treatment for breast cancer. Adv Exp Med Biol 2007; 599: 45-52.
[35]
Johannsen M, Gneveckow U, Thiesen B, et al. Thermotherapy of prostate cancer using magnetic nanoparticles: feasibility, imaging, and three-dimensional temperature distribution. Eur Urol 2007; 52(6): 1653-61.
[36]
van Landeghem FKH, Maier-Hauff K, Jordan A, et al. Post-mortem studies in glioblastoma patients treated with thermotherapy using magnetic nanoparticles. Biomaterials 2009; 30(1): 52-7.
[37]
Walther W, Stein U, Schlag PM. Use of the human MDR1 promoter for heat-inducible expression of therapeutic genes. Int J Cancer 2002; 98(2): 291-6.
[38]
Ito A, Shinkai M, Honda H, Kobayashi T. Heat-inducible TNF-alpha gene therapy combined with hyperthermia using magnetic nanoparticles as a novel tumor-targeted therapy. Cancer Gene Ther 2001; 8(9): 649-54.
[39]
Barry SE. Challenges in the development of magnetic particles for therapeutic applications. Int J Hyperthermia 2008; 24(6): 451-66.
[40]
Gupta P, Vermani K, Garg S. Hydrogels: from controlled release to pH-responsive drug delivery. Drug Discov Today 2002; 7(10): 569-79.
[41]
Brigger I, Dubernet C, Couvreur P. Nanoparticles in cancer therapy and diagnosis. Adv Drug Deliv Rev 2002; 54(5): 631-51.
[42]
Shenoy D, Little S, Langer R, Amiji M. 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(5): 357-66.
[43]
Chawla JS, Amiji MM. Biodegradable poly(epsilon -caprolactone) nanoparticles for tumor-targeted delivery of tamoxifen. Int J Pharm 2002; 249(1-2): 127-38.
[44]
Harrison SC. Viral membrane fusion. Nat Struct Mol Biol 2008; 15(7): 690-8.
[45]
Procko E, Berguig GY, Shen BW, et al. A computationally designed inhibitor of an Epstein-Barr viral Bcl-2 protein induces apoptosis in infected cells. Cell 2014; 157(7): 1644-56.
[46]
López-Otín C, Hunter T. The regulatory crosstalk between kinases and proteases in cancer. Nat Rev Cancer 2010; 10(4): 278-92.
[47]
Turk B. Targeting proteases: successes, failures and future prospects. Nat Rev Drug Discov 2006; 5(9): 785-99.
[48]
Yu I-M, Zhang W, Holdaway HA, et al. Structure of the immature dengue virus at low pH primes proteolytic maturation. Science 2008; 319(5871): 1834-7.
[49]
Weissleder R, Tung CH, Mahmood U, Bogdanov A. In vivo imaging of tumors with protease-activated near-infrared fluorescent probes. Nat Biotechnol 1999; 17(4): 375-8.
[50]
Law B, Curino A, Bugge TH, Weissleder R, Tung C-H. Design, synthesis, and characterization of urokinase plasminogen-activator-sensitive near-infrared reporter. Chem Biol 2004; 11(1): 99-106.
[51]
Chen J, Tung C-H, Allport JR, Chen S, Weissleder R, Huang PL. Near-infrared fluorescent imaging of matrix metalloproteinase activity after myocardial infarction. Circulation 2005; 111(14): 1800-5.
[52]
Jaffer FA, Libby P, Weissleder R. Optical and multimodality molecular imaging: insights into atherosclerosis. Arterioscler Thromb Vasc Biol 2009; 29(7): 1017-24.
[53]
Chaterji S, Kwon IK, Park K. Smart polymeric gels: redefining the limits of biomedical devices. Prog Polym Sci 2007; 32(8-9): 1083-122.
[54]
Aronoff DM, Neilson EG. Antipyretics: mechanisms of action and clinical use in fever suppression. Am J Med 2001; 111(4): 304-15.
[55]
Gil ES, Hudson SM. Stimuli-reponsive polymers and their bioconjugates. Prog Polym Sci 2004; 29(12): 1173-222.
[56]
Bae YH, Okano T, Kim SW. A new thermo-sensitive hydrogel: Interpenetrating polymer networks from N-acryloylpyrrolidine and poly(oxyethylene). Die Makromolekulare Chemie, Rapid Communications 1988; 9(3): 185-9.
[57]
Okuyama Y, Yoshida R, Sakai K, Okano T, Sakurai Y. Swelling controlled zero order and sigmoidal drug release from thermo-responsive poly(N-isopropyl-acrylamide-co-butyl methacrylate) hydrogel. J Biomater Sci Polym Ed 1993; 4(5): 545-56.
[58]
Issels R. Hyperthermia Combined with chemotherapy - biological rationale, clinical application, and treatment results. ORT 1999; 22(5): 374-81.
[59]
Chung JE, Yokoyama M, Yamato M, Aoyagi T, Sakurai Y, Okano T. Thermo-responsive drug delivery from polymeric micelles constructed using block copolymers of poly(N-isopropylacrylamide) and poly (butylmethacrylate). J Control Release 1999; 62(1): 115-27.
[60]
Kohori F, Sakai K, Aoyagi T, et al. Control of adriamycin cytotoxic activity using thermally responsive polymeric micelles composed of poly(N-isopropyl-acrylamide-co-N,N-dimethylacrylamide)-b-poly(d,l-lactide). Colloids Surf B 1999; 16(1): 195-205.
[61]
Shiga T. Deformation and viscoelastic behavior of polymer gels in electric fields. In: neutron spin echospectroscopy viscoelasticity rheology [Internet]. Springer, Berlin, Heidelberg; 1997.
[62]
Gong JP, Nitta T, Osada Y. Electrokinetic modeling of the contractile phenomena of polyelectrolyte gels. one-dimensional capillary model. J Phys Chem 1994; 98(38): 9583-7.
[63]
Kwon EJ, Lo JH, Bhatia SN. Smart nanosystems: Bio-inspired technologies that interact with the host environment. Proc Natl Acad Sci USA 2015; 112(47): 14460-6.
[64]
Veiseh O, Tang BC, Whitehead KA, Anderson DG, Langer R. Managing diabetes with nanomedicine: challenges and opportunities. Nat Rev Drug Discov 2015; 14(1): 45-57.
[65]
Gu Z, Dang TT, Ma M, et al. Glucose-responsive microgels integrated with enzyme nanocapsules for closed-loop insulin delivery. ACS Nano 2013; 7(8): 6758-66.
[66]
Lin KY, Lo JH, Consul N, Kwong GA, Bhatia SN. Self-titrating anticoagulant nanocomplexes that restore homeostatic regulation of the coagulation cascade. ACS Nano 2014; 8(9): 8776-85.
[67]
Zhao MD, Yang JJ, Cheng JL, et al. Hyaluronic acid reagent functional chitosan-PEI conjugate with AQP2-siRNA suppressed endometriotic lesion formation. Int J Nanomedicine 2016; 11: 1323.
[68]
Das D, Patra P, Ghosh P, Rameshbabu AP, Dhara S, Pal S. Dextrin and poly(lactide)-based biocompatible and biodegradable nanogel for cancer targeted delivery of doxorubicin hydrochloride. Polym Chem 2016; 7(17): 2965-75.
[69]
Kommareddy S, Amiji M. Poly(Ethylene Glycol)-Modified Thiolated gelatin nanoparticles for glutathione-responsive intracellular dna delivery. Nanomedicine 2007; 3(1): 32-42.
[70]
Van S, Das SK, Wang X, et al. Synthesis, characterization, and biological evaluation of poly(L-γ-glutamyl-glutamine)- paclitaxel nanoconjugate. Int J Nanomedicine 2010; 5: 825-37.
[71]
Bai RG, Muthoosamy K, Shipton FN, et al. The biogenic synthesis of a reduced graphene oxide-silver (RGO-Ag) nanocomposite and its dual applications as an antibacterial agent and cancer biomarker sensor. RSC Advances 2016; 6(43): 36576-87.
[72]
Ma N, Zhang B, Liu J, Zhang P, Li Z, Luan Y. Green fabricated reduced graphene oxide: evaluation of its application as nano-carrier for pH-sensitive drug delivery. Int J Pharm 2015; 496(2): 984-92.
[73]
Li Z, Xu W, Wang Y, et al. Quantum dots loaded nanogels for low cytotoxicity, pH-sensitive fluorescence, cell imaging and drug delivery. Carbohydr Polym 2015; 121: 477-85.
[74]
Bwatanglang IB, Mohammad F, Yusof NA, et al. Folic acid targeted Mn: ZnS quantum dots for theranostic applications of cancer cell imaging and therapy. Int J Nanomedicine 2016; 11: 413.
[75]
Jeyamohan P, Hasumura T, Nagaoka Y, Yoshida Y, Maekawa T, Kumar DS. Accelerated killing of cancer cells using a multifunctional single-walled carbon nanotube-based system for targeted drug delivery in combination with photothermal therapy. Int J Nanomedicine 2013; 8: 2653-67.
[76]
Javed MN, Kohli K, Amin S. Risk assessment integrated qbd approach for development of optimized bicontinuous mucoadhesive limicubes for oral delivery of rosuvastatin. AAPS PharmSciTech 2018; 19(3): 1377-91.

© 2024 Bentham Science Publishers | Privacy Policy