1,621
Views
107
CrossRef citations to date
0
Altmetric
Review

Carbon Nanotubes: Smart Drug/Gene Delivery Carriers

ORCID Icon, , , , ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 1681-1706 | Published online: 01 Mar 2021

References

  • Rahim M, Rizvi SMD, Iram S, Khan S, Bagga PS, Khan MS. Chapter 5 - Interaction of green nanoparticles with cells and organs. In: Grumezescu AM, editor. Inorganic Frameworks as Smart Nanomedicines. William Andrew Publishing; 2018:185–237.
  • Gao H, Jiang X. Chapter 1 - the medical applications of nanomaterials in the central nervous system. In: Jiang X, Gao H, editors. Neurotoxicity of Nanomaterials and Nanomedicine. Academic Press; 2017:1–31.
  • Wei M, Li S, Le W. Nanomaterials modulate stem cell differentiation: biological interaction and underlying mechanisms. J Nanobiotechnology. 2017;15:75. doi:10.1186/s12951-017-0310-5
  • Higuchi M, Takagi H, Owada Y, et al. Efficacy and tolerability of nanoparticle albumin-bound paclitaxel in combination with carboplatin as a late-phase chemotherapy for recurrent and advanced non-small-cell lung cancer: a multi-center study of the Fukushima lung cancer association group of surgeons. Oncol Lett. 2017;13:4315+. doi:10.3892/ol.2017.5998
  • Karimi M, Zare H, Bakhshian Nik A, et al. Nanotechnology in diagnosis and treatment of coronary artery disease. Nanomedicine. 2016;11:513–530. doi:10.2217/nnm.16.3
  • Shi J, Kantoff PW, Wooster R, Farokhzad OC. Cancer nanomedicine: progress, challenges and opportunities. Nat Rev Cancer. 2017;17:20–37.
  • Ruenraroengsak P, Chen S, Hu S, et al. Translocation of functionalized multi-walled carbon nanotubes across human pulmonary alveolar epithelium: dominant role of epithelial type 1 cells. ACS Nano. 2016;10:5070–5085. doi:10.1021/acsnano.5b08218
  • You DG, Deepagan VG, Um W, et al. ROS-generating TiO2 nanoparticles for non-invasive sonodynamic therapy of cancer. Sci Rep. 2016;6:23200. doi:10.1038/srep23200
  • Simeonidis K, Martinez-Boubeta C, Rivera-Gil P, et al. Regeneration of arsenic spent adsorbents by Fe/MgO nanoparticles. J Chem Technol Biotechnol. 2017;92:1876–1883. doi:10.1002/jctb.5187
  • Zhang J, Tang H, Liu Z, Chen B. Effects of major parameters of nanoparticles on their physical and chemical properties and recent application of nanodrug delivery system in targeted chemotherapy. Int J Nanomed. 2017;12:8483–8493. doi:10.2147/IJN.S148359
  • Kumar S, Rani R, Dilbaghi N, Tankeshwar K, Kim K-H. Carbon nanotubes: a novel material for multifaceted applications in human healthcare. Chem Soc Rev. 2017;46:158–196.
  • Cai Z, Zhang H, Wei Y, Cong F. Hyaluronan-inorganic nanohybrid materials for biomedical applications. Biomacromolecules. 2017;18:1677–1696. doi:10.1021/acs.biomac.7b00424
  • Cabrera I, Abasolo I, Corchero JL, et al. α-galactosidase-a loaded-nanoliposomes with enhanced enzymatic activity and intracellular penetration. Adv Healthc Mater. 2016;5:829–840. doi:10.1002/adhm.201500746
  • Karimi M, Ghasemi A, Zangabad PS, et al. Smart micro/nanoparticles in stimulus-responsive drug/gene delivery systems. Chem Soc Rev. 2016;45:1457–1501.
  • Costa PM, Bourgognon M, Wang JTW, Al-Jamal KT. Functionalised carbon nanotubes: from intracellular uptake and cell-related toxicity to systemic brain delivery. J Control Release. 2016;241:200–219. doi:10.1016/j.jconrel.2016.09.033
  • Alshehri R, Ilyas AM, Hasan A, Arnaout A, Ahmed F, Memic A. Carbon nanotubes in biomedical applications: factors, mechanisms, and remedies of toxicity. J Med Chem. 2016;59:8149–8167. doi:10.1021/acs.jmedchem.5b01770
  • Foo ME, Gopinath SCB. Feasibility of graphene in biomedical applications. Biomed Pharmacother. 2017;94:354–361. doi:10.1016/j.biopha.2017.07.122
  • Sajid MI, Jamshaid U, Jamshaid T, Zafar N, Fessi H, Elaissari A. Carbon nanotubes from synthesis to in vivo biomedical applications. Int J Pharm. 2016;501:278–299.
  • Skwarecki AS, Milewski S, Schielmann M, Milewska MJ. Antimicrobial molecular nanocarrier–drug conjugates. Nanomedicine. 2016;12:2215–2240. doi:10.1016/j.nano.2016.06.002
  • Singh B, Lohan S, Sandhu PS, Jain A, Mehta SK. Chapter 15 - functionalized carbon nanotubes and their promising applications in therapeutics and diagnostics. In: Grumezescu AM, editor. Nanobiomaterials in Medical Imaging. William Andrew Publishing; 2016:455–478.
  • Azqhandi MHA, Farahani BV, Dehghani N. Encapsulation of methotrexate and cyclophosphamide in interpolymer complexes formed between poly acrylic acid and poly ethylene glycol on multi-walled carbon nanotubes as drug delivery systems. Mater Sci Eng C. 2017;79:841–847. doi:10.1016/j.msec.2017.05.089
  • Ilbasmis-Tamer S, Unsal H, Tugcu-Demiroz F, Kalaycioglu GD, Degim IT, Aydogan N. Stimuli-responsive lipid nanotubes in gel formulations for the delivery of doxorubicin. Colloids Surf B. 2016;143:406–414. doi:10.1016/j.colsurfb.2016.03.070
  • Sharma P, Jain K, Jain NK, Mehra NK. Ex vivo and in vivo performance of anti-cancer drug loaded carbon nanotubes. J Drug Deliv Sci Tech. 2017;41:134–143. doi:10.1016/j.jddst.2017.07.011
  • Srivastava I, Misra SK, Ostadhossein F, Daza E, Singh J, Pan D. Surface chemistry of carbon nanoparticles functionally select their uptake in various stages of cancer cells. Nano Res. 2017;10:3269–3284. doi:10.1007/s12274-017-1518-2
  • Fedeli S, Brandi A, Venturini L, et al. The “click-on-tube” approach for the production of efficient drug carriers based on oxidized multi-walled carbon nanotubes. J Mater Chem B. 2016;4:3823–3831. doi:10.1039/C6TB00304D
  • Masotti A, Miller MR, Celluzzi A, et al. Regulation of angiogenesis through the efficient delivery of microRNAs into endothelial cells using polyamine-coated carbon nanotubes. Nanomedicine. 2016;12:1511–1522. doi:10.1016/j.nano.2016.02.017
  • Sun H, Ren J, Qu X. Carbon nanomaterials and DNA: from molecular recognition to applications. Acc Chem Res. 2016;49:461–470. doi:10.1021/acs.accounts.5b00515
  • Kong F, Liu F, Li W, et al. Smart carbon nanotubes with laser-controlled behavior in gene delivery and therapy through a non-digestive trafficking pathway. Small. 2016;12:6753–6766. doi:10.1002/smll.201601092
  • Cifuentes-Rius A, Boase NRB, Font I, et al. In vivo fate of carbon nanotubes with different physicochemical properties for gene delivery applications. ACS Appl Mater Interfaces. 2017;9:11461–11471. doi:10.1021/acsami.7b00677
  • Spinato C, Giust D, Vacchi IA, Ménard-Moyon C, Kostarelos K, Bianco A. Different chemical strategies to aminate oxidised multi-walled carbon nanotubes for siRNA complexation and delivery. J Mater Chem B. 2016;4:431–441. doi:10.1039/C5TB02088C
  • Taghavi S, HashemNia A, Mosaffa F, Askarian S, Abnous K, Ramezani M. Preparation and evaluation of polyethylenimine-functionalized carbon nanotubes tagged with 5TR1 aptamer for targeted delivery of Bcl-xL shRNA into breast cancer cells. Colloids Surf B. 2016;140:28–39. doi:10.1016/j.colsurfb.2015.12.021
  • Hernández-Rivera M, Zaibaq NG, Wilson LJ. Toward carbon nanotube-based imaging agents for the clinic. Biomaterials. 2016;101:229–240. doi:10.1016/j.biomaterials.2016.05.045
  • Wang Y, Liu J, Cui L, Losic D. Cytotoxicity, drug delivery, and photothermal therapy of functionalized carbon nanomaterials. In: Zhang M, Naik RR, Dai L, editors. Carbon Nanomaterials for Biomedical Applications. Cham: Springer International Publishing; 2016:81–111.
  • Budhathoki-Uprety J, Harvey JD, Isaac E, et al. Polymer cloaking modulates the carbon nanotube protein corona and delivery into cancer cells. J Mater Chem B. 2017;5:6637–6644. doi:10.1039/C7TB00695K
  • Hassan HAFM, Smyth L, Rubio N, et al. Carbon nanotubes’ surface chemistry determines their potency as vaccine nanocarriers in vitro and in vivo. J Control Release. 2016;225:205–216. doi:10.1016/j.jconrel.2016.01.030
  • Ema M, Gamo M, Honda K. Regul. Toxicol Pharmacol. 2016;74:42–63.
  • Karimi M, Solati N, Amiri M, et al. Carbon nanotubes part I: preparation of a novel and versatile drug-delivery vehicle. Expert Opin Drug Deliv. 2015;12:1071–1087.
  • Son KH, Hong JH, Lee JW. Carbon nanotubes as cancer therapeutic carriers and mediators. Int J Nanomed. 2016;11:5163–5185. doi:10.2147/IJN.S112660
  • Laura M, Franco T, Camillo La M, Adalberto B, Alberto B, Gianfranco R. Interactions and effects of BSA-functionalized single-walled carbon nanotubes on different cell lines. Nanotechnology. 2016;27:155704. doi:10.1088/0957-4484/27/15/155704
  • Al-Qattan MN, Deb PK, Tekade RK. Molecular dynamics simulation strategies for designing carbon-nanotube-based targeted drug delivery. Drug Discov Today. 2018;23:235–250. doi:10.1016/j.drudis.2017.10.002
  • Karimi M, Ghasemi A, Mirkiani S, Moosavi Basri SM, Hamblin MR. Carbon Nanotubes in Drug and Gene Delivery. Morgan & Claypool Publishers; 2017:5–6.
  • Zhu S, Zhu B, Huang A, et al. Application of virus targeting nanocarrier drug delivery system in virus-induced central nervous system disease treatment. ACS Appl Mater Interfaces. 2016;318:650–662.
  • Caoduro C, Hervouet E, Girard-Thernier C, et al. Carbon nanotubes as gene carriers: focus on internalization pathways related to functionalization and properties. Acta Biomater. 2017;49:36–44. doi:10.1016/j.actbio.2016.11.013
  • Cui X, Xu S, Wang X, Chen C. The nano-bio interaction and biomedical applications of carbon nanomaterials. Carbon. 2018;138:436–450. doi:10.1016/j.carbon.2018.07.069
  • Parton RG, Collins BM. Unraveling the architecture of caveolae. Proc Natl Acad Sci. 2016;113:14170. doi:10.1073/pnas.1617954113
  • Li Z, de Barros ALB, Soares DCF, Moss SN, Alisaraie L. Functionalized single-walled carbon nanotubes: cellular uptake, biodistribution and applications in drug delivery. Int J Pharm. 2017;524:41–54. doi:10.1016/j.ijpharm.2017.03.017
  • Eldawud R, Wagner A, Dong C, Stueckle TA, Rojanasakul Y, Dinu CZ. Toxicity screening of two prevalent metal organic frameworks for therapeutic use in human lung epithelial cells. Nano Impact. 2018;9:72–84.
  • Xie L, Wang G, Zhou H, et al. Functional long circulating single walled carbon nanotubes for fluorescent/photoacoustic imaging-guided enhanced phototherapy. Biomaterials. 2016;103:219–228. doi:10.1016/j.biomaterials.2016.06.058
  • Eldridge BN, Xing F, Fahrenholtz CD, Singh RN. Evaluation of multiwalled carbon nanotube cytotoxicity in cultures of human brain microvascular endothelial cells grown on plastic or basement membrane. Toxicol in Vitro. 2017;41:223–231. doi:10.1016/j.tiv.2017.03.002
  • Lacerda L, Russier J, Pastorin G, et al. Translocation mechanisms of chemically functionalised carbon nanotubes across plasma membranes. Biomaterials. 2012;33:3334–3343. doi:10.1016/j.biomaterials.2012.01.024
  • Bai W, Wu Z, Mitra S, Brown JM. Effects of multiwalled carbon nanotube surface modification and purification on bovine serum albumin binding and biological responses. J Nanomater. 2016;2016:2159537. doi:10.1155/2016/2159537
  • Ursini CL, Maiello R, Ciervo A, et al. Evaluation of uptake, cytotoxicity and inflammatory effects in respiratory cells exposed to pristine and -OH and -COOH functionalized multi-wall carbon nanotubes. J Appl Toxicol. 2016;36:394–403. doi:10.1002/jat.3228
  • Chatterjee N, Yang J, Yoon D, Kim S, Joo S-W, Choi J. Differential crosstalk between global DNA methylation and metabolomics associated with cell type specific stress response by pristine and functionalized MWCNT. Biomaterials. 2017;115:167–180. doi:10.1016/j.biomaterials.2016.11.005
  • Kafa H, Wang JT, Rubio N, et al. The interaction of carbon nanotubes with an in vitro blood-brain barrier model and mouse brain in vivo. Biomaterials. 2015;53:437–452. doi:10.1016/j.biomaterials.2015.02.083
  • Ren J, Shen S, Wang D, et al. The targeted delivery of anticancer drugs to brain glioma by PEGylated oxidized multi-walled carbon nanotubes modified with angiopep-2. Biomaterials. 2012;33:3324–3333. doi:10.1016/j.biomaterials.2012.01.025
  • Al-Jamal KT, Gherardini L, Bardi G, et al. Functional motor recovery from brain ischemic insult by carbon nanotube-mediated siRNA silencing. Proc Natl Acad Sci U S A. 2011;108:10952–10957. doi:10.1073/pnas.1100930108
  • Sciortino N, Fedeli S, Paoli P, et al. Multiwalled carbon nanotubes for drug delivery: efficiency related to length and incubation time. Int J Pharm. 2017;521:69–72. doi:10.1016/j.ijpharm.2017.02.023
  • Jin S, Wijesekara P, Boyer PD, Dahl KN, Islam MF. Length-dependent intracellular bundling of single-walled carbon nanotubes influences retention. J Mater Chem B. 2017;5:6657–6665. doi:10.1039/C7TB00735C
  • Shinde A, Tsai CSJ. Toxicity mechanism in fetal lung fibroblast cells for multi-walled carbon nanotubes defined by chemical impurities and dispersibility. Toxicol Res. 2016;5:248–258. doi:10.1039/C5TX00211G
  • Cui X, Wan B, Yang Y, Ren X, Guo L-H. Length effects on the dynamic process of cellular uptake and exocytosis of single-walled carbon nanotubes in murine macrophage cells. Sci Rep. 2017;7:1518. doi:10.1038/s41598-017-01746-9
  • Chatterjee N, Yang J, Kim S, Joo SW, Choi J. Diameter size and aspect ratio as critical determinants of uptake, stress response, global metabolomics and epigenetic alterations in multi-wall carbon nanotubes. Carbon. 2016;108:529–540. doi:10.1016/j.carbon.2016.07.031
  • Zhang M, Yang M, Morimoto T, et al. Size-dependent cell uptake of carbon nanotubes by macrophages: a comparative and quantitative study. Carbon. 2018;127:93–101. doi:10.1016/j.carbon.2017.10.085
  • Zhang M, Yang M, Okazaki T, Yudasaka M. Quantification of carbon nanotubes taken up by macrophage cells using optical absorption method. Surf Sci Nanotechnol. 2018;16:93–96. doi:10.1380/ejssnt.2018.93
  • Song Z-M, Wang L, Chen N, Cao A, Liu Y, Wang H. Biological effects of agglomerated multi-walled carbon nanotubes. Colloids Surf B. 2016;142:65–73. doi:10.1016/j.colsurfb.2016.02.032
  • Kuroda C, Ueda K, Haniu H, et al. Different aggregation and shape characteristics of carbon materials affect biological responses in RAW264 cells. Int J Nanomed. 2018;13:6079–6088. doi:10.2147/IJN.S172493
  • Roxbury D, Jena PV, Shamay Y, Horoszko CP, Heller DA. Cell membrane proteins modulate the carbon nanotube optical bandgap via surface charge accumulation. ACS Nano. 2016;10:499–506. doi:10.1021/acsnano.5b05438
  • Taghavi S, Nia AH, Abnous K, Ramezani M. Polyethylenimine-functionalized carbon nanotubes tagged with AS1411 aptamer for combination gene and drug delivery into human gastric cancer cells. Int J Pharm. 2017;516:301–312. doi:10.1016/j.ijpharm.2016.11.027
  • Summers HD, Rees P, Wang JTW, Al-Jamal KT. Spatially-resolved profiling of carbon nanotube uptake across cell lines. Nanoscale. 2017;9:6800–6807. doi:10.1039/C7NR01561E
  • Jiang W, Wang Q, Qu X, et al. Effects of charge and surface defects of multi-walled carbon nanotubes on the disruption of model cell membranes. Sci Total Environ. 2017;574:771–780. doi:10.1016/j.scitotenv.2016.09.150
  • Cai X, Ramalingam R, Wong HS, et al. Characterization of carbon nanotube protein corona by using quantitative proteomics. Nanomedicine. 2013;9:583–593. doi:10.1016/j.nano.2012.09.004
  • Corbo C, Molinaro R, Parodi A, Toledano Furman NE, Salvatore F, Tasciotti E. The impact of nanoparticle protein corona on cytotoxicity, immunotoxicity and target drug delivery. Nanomedicine. 2015;11:81–100. doi:10.2217/nnm.15.188
  • Ge C, Du J, Zhao L, et al. Binding of blood proteins to carbon nanotubes reduces cytotoxicity. Proc Natl Acad Sci U S A. 2011;108:16968–16973. doi:10.1073/pnas.1105270108
  • Elçin AE, Elçin AE, Elçin YM, Elçin YM. A comparative study on the in vitro cytotoxic responses of two mammalian cell types to fullerenes, carbon nanotubes and iron oxide nanoparticles. Drug Chem Toxicol. 2017;40:215–227. doi:10.1080/01480545.2016.1199563
  • Stan MS, Strugari AFG, Balas M, Nica IC. Chapter 2 - biomedical applications of carbon nanotubes with improved properties. In: Grumezescu AM, editor. Fullerenes, Graphenes and Nanotubes. William Andrew Publishing; 2018:31–65.
  • Karimi M, Solati N, Ghasemi A, et al. Temperature-responsive smart nanocarriers for delivery of therapeutic agents: applications and recent advances. Avci. 2015;12:1089–1105.
  • de Carvalho Lima EN, Piqueira JRC, Maria DA. Advances in carbon nanotubes for malignant melanoma: a chance for treatment. Mol Diag Ther. 2018;22:703–715. doi:10.1007/s40291-018-0363-7
  • García‐Hevia L, Villegas JC, Fernández F, et al. Multiwalled carbon nanotubes inhibit tumor progression in a mouse model. Adv Healthc Mater. 2016;5:1080–1087. doi:10.1002/adhm.201500753
  • Hopkins S, Gottipati MK, Montana V, Bekyarova E, Haddon RC, Parpura V. Leveraging science to advance health equity: a regional health policy research center’s approach. Neurol. 2018;1:327–338.
  • Judge A, McClintock K, Phelps JR, Maclachlan I. Hypersensitivity and loss of disease site targeting caused by antibody responses to PEGylated liposomes. Mol Ther. 2006;13:328–337. doi:10.1016/j.ymthe.2005.09.014
  • Sroda K, Rydlewski J, Langner M, Kozubek A, Grzybek M, Sikorski AF. Repeated injections of PEG-PE liposomes generate anti-PEG antibodies.. Cell Mol Biol Lett. 2005;10:37–47.
  • Dams ET, Laverman P, Oyen WJ, et al. Accelerated blood clearance and altered biodistribution of repeated injections of sterically stabilized liposomes. J Pharmacol Exp Ther. 2000;292:1071–1079.
  • Macdougall IC, Provenzano R, Sharma A, et al. Peginesatide for anemia in patients with chronic kidney disease not receiving dialysis. N Engl J Med. 2013;368:320–332. doi:10.1056/NEJMoa1203166
  • Eckardt KU. The safety and efficacy of peginesatide in patients with CKD. Nat Rev Nephrol. 2013;9:192–193. doi:10.1038/nrneph.2013.42
  • Elsadek NE, Abu Lila AS, Ishida T. 5 - immunological responses to PEGylated proteins: anti-PEG antibodies. In: Pasut G, Zalipsky S, editors. Polymer-Protein Conjugates. Elsevier; 2020:103–123.
  • Eldridge BN, Bernish BW, Fahrenholtz CD, Singh R. Photothermal therapy of glioblastoma multiforme using multiwalled carbon nanotubes optimized for diffusion in extracellular space. ACS Biomater Sci Eng. 2016;2:963–976. doi:10.1021/acsbiomaterials.6b00052
  • Li B, Zhang XX, Huang HY, et al. Effective deactivation of A549 tumor cells in vitro and in vivo by RGD-decorated chitosan-functionalized single-walled carbon nanotube loading docetaxel. Int J Pharm. 2018;543:8–20. doi:10.1016/j.ijpharm.2018.03.017
  • Kim SW, Kyung Lee Y, Yeon Lee J, Hee Hong J, Khang D. PEGylated anticancer-carbon nanotubes complex targeting mitochondria of lung cancer cells. Nanotechnology. 2017;28:465102. doi:10.1088/1361-6528/aa8c31
  • Weis SM, Cheresh DA. Integrins in angiogenesis and cancer. Cold Spring Harb Perspect Med. 2011;1:a006478. doi:10.1101/cshperspect.a006478
  • Koh B, Park SB, Yoon E, et al. αVβ3-targeted delivery of camptothecin-encapsulated carbon nanotube-cyclic RGD in 2D and 3D cancer cell culture. J Pharm Sci. 2019;108:3704–3712. doi:10.1016/j.xphs.2019.07.011
  • Liu X, Xu D, Liao C, Fang Y, Guo B. Development of a promising drug delivery for formononetin: cyclodextrin-modified single-walled carbon nanotubes. J Drug Deliv Sci Tech. 2018;43:461–468. doi:10.1016/j.jddst.2017.11.018
  • Wauthoz N, Balde A, Balde ES, Van Damme M, Duez P. Ethnopharmacology of Mangifera indica L. bark and pharmacological studies of its main C-glucosylxanthone, mangiferin. Int J Biomed Pharm Sci. 2007;1:112–119.
  • Harsha P, Thotakura N, Kumar M, et al. A novel PEGylated carbon nanotube conjugated mangiferin: an explorative nanomedicine for brain cancer cells. J Drug Deliv Sci Technol. 2019;53:101186. doi:10.1016/j.jddst.2019.101186
  • Raza K, Kumar D, Kiran C, et al. Conjugation of docetaxel with multiwalled carbon nanotubes and codelivery with piperine: implications on pharmacokinetic profile and anticancer activity. Mol Pharm. 2016;13:2423–2432. doi:10.1021/acs.molpharmaceut.6b00183
  • Razzazan A, Atyabi F, Kazemi B, Dinarvand R. In vivo drug delivery of gemcitabine with PEGylated single-walled carbon nanotubes. Mater Sci Eng C Mater. 2016;62:614–625. doi:10.1016/j.msec.2016.01.076
  • Pugazhendhi A, Edison T, Velmurugan BK, Jacob JA, Karuppusamy I. Toxicity of doxorubicin (Dox) to different experimental organ systems. Life Sci. 2018;200:26–30. doi:10.1016/j.lfs.2018.03.023
  • He H, Pham-Huy LA, Dramou P, Xiao D, Zuo P, Pham-Huy C. Carbon nanotubes: applications in pharmacy and medicine. Biomed Res Int. 2013;2013:578290. doi:10.1155/2013/578290
  • Liu D, Zhang Q, Wang J, Fan L, Zhu W, Cai D. Hyaluronic acid-coated single-walled carbon nanotubes loaded with doxorubicin for the treatment of breast cancer. Pharmazie. 2019;74:83–90. doi:10.1691/ph.2019.8152
  • Liu D, Zhang Q, Wang J, Fan L, Zhu W, Cai D. Hyaluronic acid-coated single-walled carbon nanotubes loaded with doxorubicin for the treatment of breast cancer. Die Pharmazie. 2019;74:83–90.
  • Yan Y, Wang R, Hu Y, et al. Stacking of doxorubicin on folic acid-targeted multiwalled carbon nanotubes for in vivo chemotherapy of tumors. Drug Deliv. 2018;25:1607–1616. doi:10.1080/10717544.2018.1501120
  • Uttekar PS, Lakade SH, Beldar VK, Harde MT. Facile synthesis of multi-walled carbon nanotube via folic acid grafted nanoparticle for precise delivery of doxorubicin. IET Nanobiotechnol. 2019;13:688–696. doi:10.1049/iet-nbt.2018.5421
  • Yu B, Tan L, Zheng R, Tan H, Zheng L. Targeted delivery and controlled release of Paclitaxel for the treatment of lung cancer using single-walled carbon nanotubes. Mater Sci Eng C Mater Biol Appl. 2016;68:579–584. doi:10.1016/j.msec.2016.06.025
  • Karnati KR, Wang Y. Understanding the co-loading and releasing of doxorubicin and paclitaxel using chitosan functionalized single-walled carbon nanotubes by molecular dynamics simulations. Phys Chem. 2018;20:9389–9400.
  • Saeednia L, Yao L, Cluff K, Asmatulu R. Sustained releasing of methotrexate from injectable and thermosensitive chitosan–carbon nanotube hybrid hydrogels effectively controls tumor cell growth. ACS Omega. 2019;4:4040–4048. doi:10.1021/acsomega.8b03212
  • Karimi M, Solati N, Amiri M, et al. Carbon nanotubes part I: preparation of a novel and versatile drug-delivery vehicle. Expert Opin Drug Deliv. 2015;12:1071–1087.
  • Behnam MA, Emami F, Sobhani Z, et al. Novel combination of silver nanoparticles and carbon nanotubes for plasmonic photo thermal therapy in melanoma cancer model. Adv Pharm Bull. 2018;8:49–55. doi:10.15171/apb.2018.006
  • Sobhani Z, Behnam MA, Emami F, Dehghanian A, Jamhiri I. Photothermal therapy of melanoma tumor using multiwalled carbon nanotubes. Int J Nanomed. 2017;12:4509–4517. doi:10.2147/IJN.S134661
  • Wang R, Xiao R, Zeng Z, Xu L, Wang J. Application of poly (ethylene glycol)–distearoylphosphatidylethanolamine (PEG-DSPE) block copolymers and their derivatives as nanomaterials in drug delivery. Int J Nanomed. 2012;7:4185–4198.
  • Kitiyanan B, Alvarez W, Harwell J, Resasco D. Controlled production of single-wall carbon nanotubes by catalytic decomposition of CO on bimetallic Co–Mo catalysts. Chem Phys Lett. 2000;317:497–503. doi:10.1016/S0009-2614(99)01379-2
  • Li Y, Li X, Doughty A, et al. Phototherapy using immunologically modified carbon nanotubes to potentiate checkpoint blockade for metastatic breast cancer. Nanomed. 2019;18:44–53. doi:10.1016/j.nano.2019.02.009
  • Virani NA, Davis C, McKernan P, et al. Phosphatidylserine targeted single-walled carbon nanotubes for photothermal ablation of bladder cancer. Nanotechnol. 2018;29:035101. doi:10.1088/1361-6528/aa9c0c
  • Han S, Kwon T, Um JE, Haam S, Kim WJ. Highly selective photothermal therapy by a phenoxylated-dextran-functionalized smart carbon nanotube platform. Adv Healthc Mater. 2016;5:1147–1156. doi:10.1002/adhm.201600015
  • Castano AP, Mroz P, Hamblin MR. Photodynamic therapy and anti-tumour immunity. Nat Rev Cancer. 2006;6:535–545. doi:10.1038/nrc1894
  • Zhang M, Wang W, Cui Y, et al. Magnetofluorescent Fe3O4/carbon quantum dots coated single-walled carbon nanotubes as dual-modal targeted imaging and chemo/photodynamic/photothermal triple-modal therapeutic agents. Chem Eng J. 2018;338:526–538. doi:10.1016/j.cej.2018.01.081
  • Hou L, Yang X, Ren J, et al. A novel redox-sensitive system based on single-walled carbon nanotubes for chemo-photothermal therapy and magnetic resonance imaging. Int J Nanomed. 2016;11:607–624.
  • Hasan M, Campbell E, Sizova O, et al. Multi-drug/gene NASH therapy delivery and selective hyperspectral NIR imaging using chirality-sorted single-walled carbon nanotubes. Cancers. 2019;11:1175. doi:10.3390/cancers11081175
  • Arosio P, Comito G, Orsini F, et al. Conjugation of a GM3 lactone mimetic on carbon nanotubes enhances the related inhibition of melanoma-associated metastatic events. Org Biomol. 2018;16:6086–6095. doi:10.1039/C8OB01817K
  • Dong Z, Wang Q, Huo M, et al. Mannose-modified multi-walled carbon nanotubes as a delivery nanovector optimizing the antigen presentation of dendritic cells. Chem Open. 2019;8:915–921. doi:10.1002/open.201900126
  • Zhang B, Wang H, Shen S, et al. Fibrin-targeting peptide CREKA-conjugated multi-walled carbon nanotubes for self-amplified photothermal therapy of tumor. Biomaterials. 2016;79:46–55. doi:10.1016/j.biomaterials.2015.11.061
  • Da Ros T, Ostric A, Andreola F, et al. Carbon nanotubes as nanovectors for intracellular delivery of laronidase in Mucopolysaccharidosis type I. Nanoscale. 2018;10:657–665. doi:10.1039/C7NR07393C
  • Golshadi M, Wright LK, Dickerson IM, Schrlau MG. High-efficiency gene transfection of cells through carbon nanotube arrays. Small. 2016;12:3014–3020. doi:10.1002/smll.201503878
  • Cao Y, Huang H-Y, Chen L-Q, et al. Enhanced lysosomal escape of pH-responsive polyethylenimine–betaine functionalized carbon nanotube for the codelivery of survivin small interfering RNA and doxorubicin. ACS Appl Mater Interfaces. 2019;11:9763–9776. doi:10.1021/acsami.8b20810
  • Demirer GS, Zhang H, Goh NS, Pinals RL, Chang R, Landry MP. Nanobiolistics: an emerging genetic transformation approach. bioRxiv. 2019;564427.
  • Kwak S-Y, Lew TTS, Sweeney CJ, et al. Chloroplast-selective gene delivery and expression in planta using chitosan-complexed single-walled carbon nanotube carriers. Nat Nanotechnol. 2019;14:447–455. doi:10.1038/s41565-019-0375-4
  • Ren X, Lin J, Wang X, et al. Photoactivatable RNAi for cancer gene therapy triggered by near-infrared-irradiated single-walled carbon nanotubes. Int J Nanomed. 2017;12:7885–7896. doi:10.2147/IJN.S141882
  • Pantarotto D, Singh R, McCarthy D, et al. Functionalized carbon nanotubes for plasmid DNA gene delivery. Angew Chem Int Ed Engl. 2004;43:5242–5246. doi:10.1002/anie.200460437
  • Hu F, Li Y, Wang Q, et al. Carbon nanotube-based DNA vaccine against koi herpesvirus given by intramuscular injection. Fish Shellfish Immunol. 2020;98:810–818. doi:10.1016/j.fsi.2019.11.035
  • Hashem Nia A, Behnam B, Taghavi S, et al. Evaluation of chemical modification effects on DNA plasmid transfection efficiency of single-walled carbon nanotube–succinate– polyethylenimine conjugates as non-viral gene carriers. Med Chem Comm. 2016;8:364–375. doi:10.1039/C6MD00481D
  • Munk M, de Souza Salomão R, Zanette LS, et al. Using carbon nanotubes to deliver genes to hard-to-transfect mammalian primary fibroblast cells. Biomed Phys Eng Express. 2017;3:045002. doi:10.1088/2057-1976/aa7927
  • Munk M, Ladeira LO, Carvalho BC, et al. Efficient delivery of DNA into bovine preimplantation embryos by multiwall carbon nanotubes. Sci Rep. 2016;6:33588. doi:10.1038/srep33588
  • Ghasemi A, Amiri H, Zare H, et al. Carbon nanotubes in microfluidic lab-on-a-chip technology: current trends and future perspectives. Microfluid Nanofluid. 2017;21.
  • Sukhodub LB, Sukhodub LF, Prylutskyy YI, et al. Composite material based on hydroxyapatite and multi-walled carbon nanotubes filled by iron: preparation, properties and drug release ability. Mater Sci Eng C. 2018;93:606–614. doi:10.1016/j.msec.2018.08.019
  • Havaldar R, Pilli SC, Putti BB. Insights into the effects of tensile and compressive loadings on human femur bone. Adv Biomed Res. 2014;3:101. doi:10.4103/2277-9175.129375
  • Moon SU, Kim J, Bokara KK, et al. Carbon nanotubes impregnated with subventricular zone neural progenitor cells promotes recovery from stroke. Int J Nanomed. 2012;7:2751–2765.
  • Sirivisoot S, Pareta R, Webster TJ. Electrically controlled drug release from nanostructured polypyrrole coated on titanium. Nanotechnol. 2011;22:085101. doi:10.1088/0957-4484/22/8/085101
  • Serra M, Arenal R, Tenne R. An overview of the recent advances in inorganic nanotubes. Nanoscale. 2019;11:8073–8090. doi:10.1039/C9NR01880H
  • Arbulu RC, Jiang YB, Peterson EJ, Qin Y. Metal-Organic Framework (MOF) nanorods, nanotubes, and nanowires. Angew Chem Int. 2018;57:5813–5817. doi:10.1002/anie.201802694
  • Wei Y-S, Zhang M, Kitta M, Liu Z, Horike S, Xu Q. A single-crystal open-capsule Metal–Organic Framework. J Am Chem Soc. 2019;141:7906–7916. doi:10.1021/jacs.9b02417
  • Huang J, Zhang X, Yan W, et al. Nanotubular topography enhances the bioactivity of titanium implants. Nanomed. 2017;13:1913–1923. doi:10.1016/j.nano.2017.03.017
  • Tharmavaram M, Pandey G, Rawtani D. Surface modified halloysite nanotubes: a flexible interface for biological, environmental and catalytic applications. Adv Colloid Interface Sci. 2018;261:82–101. doi:10.1016/j.cis.2018.09.001
  • Barman M, Mahmood S, Augustine R, Hasan A, Thomas S, Ghosal K. Natural halloysite nanotubes/chitosan based bio-nanocomposite for delivering norfloxacin, an anti-microbial agent in sustained release manner. Int J Biol Macromol. 2020;162:1849–1861. doi:10.1016/j.ijbiomac.2020.08.060
  • Tîlmaciu C-M, Morris MC. Carbon nanotube biosensors. Front Chem. 2015;3:59. doi:10.3389/fchem.2015.00059
  • Palmer BC, Phelan-Dickenson SJ, DeLouise LA. Multi-walled carbon nanotube oxidation dependent keratinocyte cytotoxicity and skin inflammation. Part Fibre Toxicol. 2019;16:3. doi:10.1186/s12989-018-0285-x
  • Dong J, Ma Q. Type 2 immune mechanisms in carbon nanotube-induced lung fibrosis. Front Immunol. 2018;9.
  • Bai Y, Zhang Y, Zhang J, et al. Repeated administrations of carbon nanotubes in male mice cause reversible testis damage without affecting fertility. Nat Nanotechnol. 2010;5:683–689. doi:10.1038/nnano.2010.153
  • Costa PM, Wang JT-W, Morfin J-F, et al. Functionalised carbon nanotubes enhance brain delivery of amyloid-targeting Pittsburgh compound B (PiB)-derived ligands. Nanotheranostics. 2018;2:168–183. doi:10.7150/ntno.23125
  • Long J, Xiao Y, Liu L, Cao Y. The adverse vascular effects of multi-walled carbon nanotubes (MWCNTs) to human vein endothelial cells (HUVECs) in vitro: role of length of MWCNTs. J Nanobiotechnology. 2017;15:80. doi:10.1186/s12951-017-0318-x
  • de Eguileor M, de Eguileor M, de Eguileor M, et al. Systemic distribution of single-walled carbon nanotubes in a novel model: alteration of biochemical parameters, metabolic functions, liver accumulation, and inflammation in vivo. Int J Nanomed. 2016;11:4299–4316. doi:10.2147/IJN.S109950
  • Sun X, Shao H, Xiang K, et al. Poly(dopamine)-modified carbon nanotube multilayered film and its effects on macrophages. Carbon. 2017;113:176–191. doi:10.1016/j.carbon.2016.11.040
  • Perepelytsina OM, Ugnivenko AP, Dobrydnev AV, Bakalinska ON, Marynin AI, Sydorenko MV. Influence of carbon nanotubes and its derivatives on tumor cells in vitro and biochemical parameters, cellular blood composition in vivo. Nanoscale Res Lett. 2018;13:286. doi:10.1186/s11671-018-2689-9
  • Dayani Y, Malmstadt N. Lipid bilayers covalently anchored to carbon nanotubes. Langmuir. 2012;28:8174–8182. doi:10.1021/la301094h
  • Hong G, Diao S, Antaris AL, Dai H. Carbon nanomaterials for biological imaging and nanomedicinal therapy. Chem Rev. 2015;115:10816–10906.
  • Singh R, Torti SV. Carbon nanotubes in hyperthermia therapy. Adv Drug Deliv Rev. 2013;65:2045–2060. doi:10.1016/j.addr.2013.08.001
  • Harrison BS, Atala A. Carbon nanotube applications for tissue engineering. Biomaterials. 2007;28:344–353. doi:10.1016/j.biomaterials.2006.07.044
  • Bianco A, Kostarelos K, Prato M. Applications of carbon nanotubes in drug delivery. Curr Opin Chem Biol. 2005;9:674–679. doi:10.1016/j.cbpa.2005.10.005
  • Ghasemi A, Amiri H, Zare H, et al. Carbon nanotubes in microfluidic lab-on-a-chip technology: current trends and future perspectives. Microfluid Nanofluidics. 2017;21:151.
  • Cherukuri P, Bachilo SM, Litovsky SH, Weisman RB, Am J. Near-infrared fluorescence microscopy of single-walled carbon nanotubes in phagocytic cells. Chem Soc. 2004;126:15638–15639. doi:10.1021/ja0466311
  • Zhang X, Meng L, Wang X, Lu Q. Preparation and cellular uptake of pH-dependent fluorescent single-wall carbon nanotubes. Chemistry. 2010;16:556–561. doi:10.1002/chem.200901168
  • Haniu H, Saito N, Matsuda Y, et al. Culture medium type affects endocytosis of multi-walled carbon nanotubes in BEAS-2B cells and subsequent biological response. Toxicol in Vitro. 2013;27:1679–1685. doi:10.1016/j.tiv.2013.04.012
  • Kam NWS, Liu Z, Dai H. Carbon nanotubes as intracellular transporters for proteins and DNA: an investigation of the uptake mechanism and pathway. Angew Chem Int Ed. 2006;45:577–581. doi:10.1002/anie.200503389
  • Song S, Fu H, He B, et al. Rho GTPases in A549 and Caco-2 cells dominating the endocytic pathways of nanocarbons with different morphologies. Int J Nanomed. 2018;13:4391–4404. doi:10.2147/IJN.S164866
  • Kang B, Yu DC, Chang SQ, Chen D, Dai YD, Ding Y. Intracellular uptake, trafficking and subcellular distribution of folate conjugated single walled carbon nanotubes within living cells. Nanotechnology. 2008;19:375103. doi:10.1088/0957-4484/19/37/375103
  • Kang B, Yu D, Dai Y, Chang S, Chen D, Ding Y. Cancer-cell targeting and photoacoustic therapy using carbon nanotubes as “bomb” agents. Small. 2009;5:1292–1301. doi:10.1002/smll.200801820
  • Zhang LW, Monteiro-Riviere NA. Lectins modulate multi-walled carbon nanotubes cellular uptake in human epidermal keratinocytes. Toxicol in Vitro. 2010;24:546–551. doi:10.1016/j.tiv.2009.11.007
  • Kang B, Chang S, Dai Y, Yu D, Chen D. Cell response to carbon nanotubes: size-dependent intracellular uptake mechanism and subcellular fate. Small. 2010;6:2362–2366. doi:10.1002/smll.201001260
  • Serag MF, Kaji N, Gaillard C, et al. Trafficking and subcellular localization of multiwalled carbon nanotubes in plant cells. ACS Nano. 2011;5:493–499. doi:10.1021/nn102344t
  • Zhou F, Xing D, Wu B, Wu S, Ou Z, Chen WR. New insights of transmembranal mechanism and subcellular localization of noncovalently modified single-walled carbon nanotubes. Nano Lett. 2010;10:1677–1681. doi:10.1021/nl100004m
  • Manzanares D, Ceña V. Endocytosis: the nanoparticle and submicron nanocompounds gateway into the cell. Pharmaceutics. 2020;12:371. doi:10.3390/pharmaceutics12040371
  • Rejman J, Oberle V, Zuhorn IS, Hoekstra D. Size-dependent internalization of particles via the pathways of clathrin- and caveolae-mediated endocytosis. Biochem J. 2004;377:159–169. doi:10.1042/bj20031253
  • Rappaport J, Manthe RL, Solomon M, Garnacho C, Muro S. A comparative study on the alterations of endocytic pathways in multiple lysosomal storage disorders. Mol Pharm. 2016;13:357–368. doi:10.1021/acs.molpharmaceut.5b00542
  • Zhang H, Li HX, Cheng HM. Water-soluble multiwalled carbon nanotubes functionalized with sulfonated polyaniline. J Phys Chem B. 2006;110:9095–9099. doi:10.1021/jp060193y
  • Hadidi N, Hosseini Shirazi SF, Kobarfard F, Nafissi-Varchehd N, Aboofazeli R. Evaluation of the effect of PEGylated single-walled carbon nanotubes on viability and proliferation of jurkat cells. Iran J Pharm Res. 2012;11:27–37.
  • Ketabi S, Rahmani L. Carbon nanotube as a carrier in drug delivery system for carnosine dipeptide: a computer simulation study. Mater Sci Eng. 2017;73:173–181. doi:10.1016/j.msec.2016.12.058
  • Farvadi F, Tamaddon A, Sobhani Z, Abolmaali SS. Polyionic complex of single-walled carbon nanotubes and PEG-grafted-hyperbranched polyethyleneimine (PEG-PEI-SWNT) for an improved doxorubicin loading and delivery: development and in vitro characterization. Artif Cells Nanomed Biotechnol. 2017;45(5):855–863. doi:10.1080/21691401.2016.1181642
  • Dong X, Sun Z, Wang X, Zhu D, Liu L, Leng X. Simultaneous monitoring of the drug release and antitumor effect of a novel drug delivery system-MWCNTs/DOX/TC. Drug Deliv. 2017;24:143–151. doi:10.1080/10717544.2016.1233592
  • Hossein Panahi F, Peighambardoust SJ, Davaran S, Salehi R. Development and characterization of PLA-mPEG copolymer containing iron nanoparticle-coated carbon nanotubes for controlled delivery of Docetaxel. Polymer. 2017;117:117–131. doi:10.1016/j.polymer.2017.03.084
  • Li H, Fan X, Chen X. Near-infrared light activation of proteins inside living cells enabled by carbon nanotube-mediated intracellular delivery. ACS Appl Mater Interfaces. 2016;8:4500–4507. doi:10.1021/acsami.6b00323
  • Mallakpour S, Khodadadzadeh L. Ultrasonic-assisted fabrication of starch/MWCNT-glucose nanocomposites for drug delivery. Ultrason Sonochem. 2018;40:402–409. doi:10.1016/j.ultsonch.2017.07.033
  • Oh Y, Jin JO, Oh J. Photothermal-triggered control of sub-cellular drug accumulation using doxorubicin-loaded single-walled carbon nanotubes for the effective killing of human breast cancer cells. Nanotechnology. 2017;28:1361–6528. doi:10.1088/1361-6528/aa5d7d
  • Singh RP, Sharma G, Koch B, Pandey BL, Koch B, Muthu MS. Chitosan-folate decorated carbon nanotubes for site specific lung cancer delivery. Mater Sci Eng C Mater. 2017;77:446–458. doi:10.1016/j.msec.2017.03.225
  • Xing J, Liu Z, Huang Y, et al. Lentinan-modified carbon nanotubes as an antigen delivery system modulate immune response in vitro and in vivo. ACS Appl Mater Interfaces. 2016;8:19276–19283. doi:10.1021/acsami.6b04591
  • Chowdhry A, Kaur J, Khatri M, Puri V, Tuli R, Puri S. Characterization of functionalized multiwalled carbon nanotubes and comparison of their cellular toxicity between HEK 293 cells and zebra fish in vivo. Heliyon. 2019;5:e02605. doi:10.1016/j.heliyon.2019.e02605
  • Sato Y, Yokoyama A, Nodasaka Y, et al. Long-term biopersistence of tangled oxidized carbon nanotubes inside and outside macrophages in rat subcutaneous tissue. Sci Rep. 2013;3:2516. doi:10.1038/srep02516
  • Schipper ML, Nakayama-Ratchford N, Davis CR, et al. A pilot toxicology study of single-walled carbon nanotubes in a small sample of mice. Nat Nanotechnol. 2008;3:216–221. doi:10.1038/nnano.2008.68