Drug Res (Stuttg)
DOI: 10.1055/a-0945-1469
Review
© Georg Thieme Verlag KG Stuttgart · New York

Application of Carbon Nanotubes in Breast Cancer Therapy

Mahdis Tajabadi
Student of Research Committee, Islamic Azad University of Medical Science, Tehran, Iran
› Author Affiliations
Further Information

Correspondence

Mahdis Tajabadi
Tehran Medical Branch, Islamic Azad University,
Khaghani Street, Shariati Avenue,
Tehran, Iran
Phone: +93/865/65 639   

Publication History

received 17 December 2018

accepted 28 May 2019

Publication Date:
28 June 2019 (online)

 

Abstract

Conjugated single-walled carbon nanotubes (SWNT) have been shown to be promising in cancer-targeted accumulation and is biocompatible, easily excreted, and possesses little toxicity. The present study aims at reviewing the recent advancements in carbon nanotubes especially SWNT for improving the treatment of breast cancer. Nanotube drug delivery system is a potential high efficacy therapy with minimum side effects for future tumor therapy with low doses of drug.


#

Introduction

Cancer remain one of the greatest challenges faced across the world today. Deaths due to cancer accounted for approximately 8.2 million in 2012, and breast cancer leading to 521 000 deaths of all. According to American Cancer Society, the numbers of diagnosed and death by invasive breast cancers in 2014 was estimated to be respectively 232 670 and 40 000 [1].

Breast tumor remains the most common women malignant tumor throughout the world, and also a major cancer death cause [2]. According to American Cancer Society, it is the second main cause of cancer-related death in The United State of America. The basic hallmark of breast tumor is the progression and metastasis to several organs including liver, lung, bone and/or brain, which leads to a multiple organ dysfunction, thereby increasing the incidence of death in patients with breast cancer [3] [4]. Metastasis occur when cancer cells leaves the primary cancer site and travels to another organ through the blood vessels or lymphatic system [5] [6].

Nanomaterials possessing opto-thermal principles of transduction have been shown promising for cancer therapy. Nanotubes (NTs) are suitable in this regards, such that, they possess a high photo-thermal cell destruction and optical absorbance efficiencies [7] [8]. The quantity of light energy used is fairly small as compared to other procedures, since the optical absorbance of NTs is high in the 700–1 100 nm near-infrared (NIR) range. This has been used lately to design several kinds of optical actuators and sensors based on the photo-mechanical actuation of carbon nanotubes (CNTs) [9] [10] [11]. CNT [12] are regarded as a flawless materials for various uses [13], this ranges from field emission display [14] to ultrastrong fibers [15]. Lately, CNT have received great attention in the field of molecular biology [16], where appropriately functionalized CNT can serve as vaccine and drug delivery systems (DDS) [17] or protein transporters [18]. Due to the extensive π-π stacking reaction between associated ligand and CNTs side walls, conjugation of ligand and functionalization is possible [19] [20], multi-component targeting using photo-thermal cell destruction might lead to the improvement of micro-surgery, cancer therapies, and cell repair [21].

The present study aims at reviewing the recent advances of carbon nanotubes especially Single-walled carbon nanotube (SWNT) for improving targeted breast cancer therapy.


#

Carbon Nanotubes

CNTs are cylindrical carbons made of benzene rings, they finds applications in biology as protein and DNA detecting sensors, protein or vaccine carrier and diagnostic tool for several kinds of serum samples for proteins discrimination [22]. These structure can also be produced from graphite sheets where carbon atoms are located in the apexes of a hexagonal net. They are chemically stable, possess high thermal and electrical conductance, mechanical strength and surface area [23]. CNTs are completely insoluble in all solvents, which gives them toxicity and health-related concerns. Nevertheless, introduction of chemical modification to CNTs improves their solubility, and interaction with active molecules including proteins, peptides, nucleic acids, proteins, and other therapeutic agents [24]. Anticancer drugs (methotrexate) or antifungal agents (amphotericin B) have covalently been attached to CNTs with a fluorescent agent (FITC). Drugs attached to CNTs have been shown efficient internalized into the cells in comparison with drugs without CNTs [25] [26]. The multiple CNTs surface modification or tips of CNTs permits them to carry several molecules, and thus providing a fundamental merit for cancer therapy. CNTs come in two principal forms; these are the SWCNT and Multi-walled carbon nanotube (MWNTs) ([Fig. 1]). Targeted drug delivery using CNTs have shown improvements by integrating molecules specific of the biological receptors with these nanotubes [27]. In a study, glycol-MWCNT were designed where glucosamine was covalently attached to the carbon nanotube. These tubes exhibited sugar-like interaction with breast cancer cells, hence assisting the biological molecules into the cells [28].

Zoom Image
Fig. 1 Single-walled carbon nanotube (SWCNT) and multiwalled carbon nanotube (MWCNT) [rerif] [50].

Single-walled carbon nanotube

SWCNT is a newer type of nanomaterial possessing unique mechanical, electrical, structural, and optical features which are promising for several biomedical applications, including biosensors [29], drug delivery transporters [30] and novel biomaterials [31]. In the quest of achieving a specific tumor cells targeting for photothermal ablation, SWNTs was attached to folate for targeting the receptors of folate in folate positive tumor cells [32] [33] or noncovalently conjugated (through adsorption) or indirectly through streptavidin-biotin conjugation [34] to antibodies targeting a specific receptors on the tumor cells. The direct covalent conjugation for specific cancer by targeting antibodies of SWNTs has also been studied [35], nevertheless, the application of such antibody-SWNT conjugates for specific photothermal ablation of tumor cells with NIR light has not been reported. Studies have also revealed that CNTs can enhance the pharmacokinetic properties of drugs like Docetaxel, which are insoluble and have low tissue permeability [36]. Needle-like structure of carbon nanotube ease internalization of the biomolecules into the target cells. CNTs have been reported effective for the administration of several antitumor drugs like topoisomerase I inhibitors, platinum and anti-microtubule drugs, genes, siRNA, immunogenic compounds and aptamers for anticancer treatments [37].


#

Multi-walled carbon nanotube

A MWNTs encompass an exclusive nanomaterial type that is remarkably durable and electrically conductive owing to the high aspect ratio and chemistry of the particles [38] [39]. MWNTs are comparatively long, with a cylindrical shaped-nanoparticles whose honeycomb structure produces high surface area relative to their volume, and high porosity. The size of these pores ranges from 4 and 30 nm in diameter [40]. The particles are flexible and durable [41], comparable to the extracellular matrix (ECM). Because of the importance of cells’ physical microenvironment, the ability of MWNT to mimic the normal ECM might be able to modify tumor cell behavior in a way that is hypothetically clinically applicable.


#
#

Characteristics of Carbon Nanotubes as a Drug Delivery System

Large surface area and thermal and electrical stability of carbon nanotubes allow the attachment of various biological molecules to its surface, making them suitable candidate to form drug delivery system. For example, Cationic fullerenes have tendency to bind to anionic tumor cells hence providing affinity for targeted delivery of the drugs [23] The conjugation scheme of antibodies (Ab) is shown in [Fig. 2]. This strategy works fine for the attachment of specific and non-specific Abs on the surface of the NT. TEM of the Ab conjugated SWCNT shows a direct confirmation that the Ab were functionalized on the SWCNT on several sites, as depicted in [Fig. 3]. This has been confirmed in previous research using atomic force microscopy (AFM) and confocal microscopy [42]. Since the NTs conjugated with Abs were washed three times prior to imaging, it is predictable that all the Abs aggregates are covalently LINKED to the CNTs. This depicted a greater number of Abs molecules in a specific site, capable of attachment to SWCNT without any activity loss for drug delivery.

Zoom Image
Fig. 2 Conjugation schematic of SWNTs with mAbs. 1-pyrenebutanoyl succinimide was adsorbed on the NT via π–π stacking. The succinimidyl ester can react with lysine side chain amines available on Abs to form amide bonds. PEG serves the purpose of insulator to hinder possible bio-fouling (Image adapted from [32] with permission).
Zoom Image
Fig. 3 TEM of the functionalization of mAbs aggregates specific to IGF1 receptor in breast cancer cells. Each site comprising 10–25 Ab molecules [rerif] [32].

#

Carbon Nanotubes Improves Breast Cancer Therapy

The improvement of efficient and novel DDS is critical for developing the pharmacological profiles of several therapeutic molecules classes. Several DDS are presently in use. Amongst nanomaterials, CNT have materialized as an effective tool and new substitute for translocating and conveying therapeutic molecules. They can act as a surface modifier using proteins, bioactive peptides, drugs and nucleic acids for carrying the drugs they contain into the target organs and cells. Since surface modified CNT are not immunogenic, and possesses a lower toxicity, such a design is of a great potential in nanomedicine and bionanotechnology [43].

Singh et al. [44] synthesized and studied paclitaxel loaded thiamine and riboflavin conjugated multi walled CNT (PTX-Tm-MWCNTs and PTX-Rf-MWCNTs) for targeted delivery to MCF-7 breast cancer cell lines. They found that PTX-Tm-MWCNTs and PTX-Rf-MWCNTs showed a great potential in breast cancer treatment, however, more thorough findings are required in future.

Al Faraj et al. [45] fabricated a new diagnostic and therapeutic method through doxorubicin-loaded SWCNTs, targeting treatment of metastatic regions, they followed preferential improved and homing therapeutic effects through noninvasive free-breathing bioluminescence imaging and magnetic resonance imaging (MRI). The noninvasive MRI combination to localize sensitively of cancer sites, through specific magnetic positioning that improves nanocarriers magnetic targeting permitted increasing efficacy of treatment.

Molecular targeting and photodynamic therapy is promising for selective treatment of tumor. Monoclonal antibodies that are specific to the HER2 and IGF1 cell surface antigens can bind to SWCNT for targeting SWCNT on breast cancer cells for specific near-infrared phototherapy. When this nanotube is conjugated with both the receptors, there is a selective attachment to the breast cancer cells as compared to those conjugated with non-specific antibodies.

Mashal et al. showed that SWCNTs is a probable dielectric contrast enhancer between normal and malignant breast tumor microwave detection and enhances selective malignant tissue heating for microwave hyperthermia treatment of breast cancer. This study was the first step in the direction of fabricating functionalized, cancer-targeting SWCNTs as theranostic (integrated therapeutic and diagnostic) agents for microwave breast tumor diagnosis and treatment. It was observed that in microwave heating studies, there was a significantly higher temperature increase in SWCNTs containing mixtures. This increase in temperature was linearly scaled with the efficient mixtures conductivity [46].

Metastasized cancerous cells lack contact inhibition where past studies have proposed that restoration of these contact inhibition may restore the normal cells properties. Dineshkumar et al. aimed at restoring the contact inhibition of cancer cells with multi-walled CNTs. They analyzed the adhesion, proliferation, western blots, autophagy assays, and immunochemical staining to evaluate E-cadherin expression and adhesion. Breast cancer cells seeded on a MWCNT-collagen coated surface showed an increased cell adhesion and decreased cell migration indicating an upregulation of E-cadherin. This study provided an alternative approach to cancer therapy [47].

Xiao et al., [48] investigated two unique optical characteristics of SWNTs - very strong NIR absorbance and a very strong Raman signals. HER2 IgY-SWNT complex was synthesized through covalently functionalizing SWNTs with anti-HER2 IgY antibody to produce SWNTs possessing high sensitivity and specificity for the IgY antibody. The complex formed was effectively applied in vitro for the selective destruction and detection of HER2-expressing breast cancer cells. Raman signal arising from tumor cells was identified at the unicellular level. The uniqueness of this dual-function agent lies in the fact that it needs no internalization by the tumor cells for achieving selective photothermal ablation, and therefore offering the merit of being easily extended to other tumor types. Conversely, more research is required for this investigation to be applied in the clinical trials. In a recent study, polyethylene glycol (for its physiochemical characteristics and biocompatibility) and estradiol-E2 (estradiol receptors are over expressed in breast cancer cells) were used for CNT modification and lobaplatin (third generation, platinum anticancer drug) was loaded for the treatment of breast cancer. It was seen that incubating cells after 72 h at the dose of 200 µg/ml of PEG-E2-CNT-lobaplatin significantly inhibited the tumorigenic activity of the cells, without any toxic effects [49]


#

Conclusion

This review reveals the understanding of the relationship between cells and their microenvironment. It has been shown that CNTs specifically SWNTs, can imitate the natural ECM to facilitate the adhesion of cell and regulates the expression of gene. This area of research offers a novel method of diagnosing and treating breast cancer and should be considered for additional investigation to develop it towards clinical use.


#

Contributors’ Statement

Dr. Mahdis Tajabadi: conceptualized and designed the study, drafted the initial manuscript, and reviewed and revised the manuscript. Designed the data collection instruments, collected data, carried out the initial analyses, and reviewed and revised the manuscript. Coordinated and supervised data collection, and critically reviewed the manuscript for important intellectual content.


#
#

Conflict of Interest

The authors deny any conflict of interest in any terms or by anymeans during the study. All the fees provided by research centerfund and deployed accordingly.

  • References

  • 1 Mourouti N, Panagiotakos DB, Kotteas EA. et al. Optimizing diet and nutrition for cancer survivors: A review. Maturitas 2017; 105: 33-36
  • 2 DeSantis C, Siegel R, Bandi P. et al. Breast cancer statistics, 2011. CA: A Cancer Journal for Clinicians 2011; 61: 408-418
  • 3 Valastyan S, Weinberg RA. Tumor metastasis: Molecular insights and evolving paradigms. Cell 2011; 147: 275-292
  • 4 Lorzadeh N, Kazemirad S, Lorzadeh M. et al. Comparison of the effect of oral and intravenous fluid therapy on women with oligohydramnios. Res J Obstet Gynecol 2008; 1: 25-29
  • 5 Fidler IJ. The pathogenesis of cancer metastasis: The ‘seed and soil’ hypothesis revisited. Nature Reviews Cancer 2003; 3: 453
  • 6 Nguyen DX, Bos PD, Massagué J. Metastasis: From dissemination to organ-specific colonization. Nature Reviews Cancer 2009; 9: 274
  • 7 Kam NWS, O'Connell M, Wisdom JA. et al. Carbon nanotubes as multifunctional biological transporters and near-infrared agents for selective cancer cell destruction. Proceedings of the National Academy of Sciences 2005; 102: 11600-11605
  • 8 Akbariasbagh F, Lorzadeh N, Azmoodeh A. et al. Association among diameter and volume of follicles, oocyte maturity, and competence in intracytoplasmic sperm injection cycles. Minerva Ginecologica 2015; 67: 397-403
  • 9 Lu S, Panchapakesan B. Optically driven nanotube actuators. Nanotechnology 2005; 16: 2548
  • 10 Lu S, Panchapakesan B. Nanotube micro-optomechanical actuators. Applied Physics Letters 2006; 88: 253107
  • 11 Lu S, Panchapakesan B. Photomechanical responses of carbon nanotube/polymer actuators. Nanotechnology 2007; 18: 305502
  • 12 Haddon RC. Carbon nanotubes. ACS Publications; 2002
  • 13 Milne W, Teo K, Amaratunga G. et al. Carbon nanotubes as field emission sources. Journal of Materials Chemistry 2004; 14: 933-943
  • 14 Berger J, Reist M, Mayer JM. et al. Structure and interactions in covalently and ionically crosslinked chitosan hydrogels for biomedical applications. European Journal of Pharmaceutics and Biopharmaceutics 2004; 57: 19-34
  • 15 Ericson LM, Fan H, Peng H. et al. Macroscopic, neat, single-walled carbon nanotube fibers. Science 2004; 305: 1447-1450
  • 16 Martin CR, Kohli P. The emerging field of nanotube biotechnology. Nature Reviews Drug Discovery 2003; 2: 29
  • 17 Bianco A, Prato M. Can carbon nanotubes be considered useful tools for biological applications?. Advanced Materials 2003; 15: 1765-1768
  • 18 Kam NWS, Dai H. Carbon nanotubes as intracellular protein transporters: Generality and biological functionality. Journal of the American Chemical Society 2005; 127: 6021-6026
  • 19 Mehra NK, Mishra V, Jain N. A review of ligand tethered surface engineered carbon nanotubes. Biomaterials 2014; 35: 1267-1283
  • 20 Mehra NK, Jain NK. Multifunctional hybrid-carbon nanotubes: New horizon in drug delivery and targeting. Journal of Drug Targeting 2016; 24: 294-308
  • 21 Lorzadeh N, Ghasem Nejad A, Mohmad Pour J. The Effect of metformin on outcome of Intrauterine insemination (IUI) in insulin non-resistant infertile women with polycystic ovarian syndrome. The Iranian. Journal of Obstetrics, Gynecology and Infertility 2014; 17: 1-11
  • 22 Roldo M, Fatouros DG. Biomedical applications of carbon nanotubes. Annual Reports Section" C"(Physical Chemistry) 2013; 109: 10-35
  • 23 Casais-Molina ML, Cab C, Canto G. et al. Carbon Nanomaterials for Breast Cancer Treatment %J Journal of Nanomaterials. 2018; 2018: 9, doi:10.1155/2018/2058613 [published Online First: Epub Date]
  • 24 Bianco A, Kostarelos K, Prato M. Applications of carbon nanotubes in drug delivery. Current Opinion in Chemical Biology 2005; 9: 674-679
  • 25 Wu W, Wieckowski S, Pastorin G. et al. Targeted delivery of amphotericin B to cells by using functionalized carbon nanotubes. Angewandte Chemie International Edition 2005; 44: 6358-6362
  • 26 Pastorin G, Wu W, Wieckowski S. et al. Double functionalisation of carbon nanotubes for multimodal drug delivery. Chemical Communications 2006; 11: 1182-1184
  • 27 Sanginario A, Miccoli B, Demarchi D. Carbon Nanotubes as an Effective Opportunity for Cancer Diagnosis and Treatment. Biosensors 2017; 7 9 DOI: 10.3390/bios7010009 [published. Online First: Epub Date]
  • 28 Fahrenholtz CD, Hadimani M, King SB. et al. Targeting breast cancer with sugar-coated carbon nanotubes. Nanomedicine (London, England) 2015; 10: 2481-2497, doi:10.2217/NNM.15.90 [published Online First: Epub Date]
  • 29 Chen RJ, Bangsaruntip S, Drouvalakis KA. et al. Noncovalent functionalization of carbon nanotubes for highly specific electronic biosensors. Proceedings of the National Academy of Sciences 2003; 100: 4984-4989
  • 30 Radomski A, Jurasz P, Alonso-Escolano D. et al. Nanoparticle-induced platelet aggregation and vascular thrombosis. British Journal of Pharmacology 2005; 146: 882-893
  • 31 Narayan RJ, Jin C, Menegazzo N. et al. Nanoporous hard carbon membranes for medical applications. Journal of Nanoscience and Nanotechnology 2007; 7: 1486-1493
  • 32 Shao N, Lu S, Wickstrom E. et al. Integrated molecular targeting of IGF1R and HER2 surface receptors and destruction of breast cancer cells using single wall carbon nanotubes. Nanotechnology 2007; 18: 315101
  • 33 Browne BC, Eustace AJ, Kennedy S. et al. Evaluation of IGF1R and phosphorylated IGF1R as targets in HER2-positive breast cancer cell lines and tumours. Breast Cancer Research and Treatment 2012; 136: 717-727
  • 34 Chakravarty P, Marches R, Zimmerman NS. et al. Thermal ablation of tumor cells with antibody-functionalized single-walled carbon nanotubes. Proceedings of the National Academy of Sciences 2008; 105: 8697-8702
  • 35 McDevitt MR, Chattopadhyay D, Kappel BJ. et al. Tumor targeting with antibody-functionalized, radiolabeled carbon nanotubes. Journal of Nuclear Medicine 2007; 48: 1180-1189
  • 36 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. Molecular Pharmaceutics 2016; 13: 2423-2432. doi:10.1021/acs.molpharmaceut.6b00183 [published Online First: Epub Date]
  • 37 Son KH, Hong JH, Lee JW. Carbon nanotubes as cancer therapeutic carriers and mediators. International Journal of Nanomedicine 2016; 11: 5163
  • 38 Liu J, Appaix F, Bibari O. et al. Control of neuronal network organization by chemical surface functionalization of multi-walled carbon nanotube arrays. Nanotechnology 2011; 22: 195101
  • 39 MacDonald RA, Laurenzi BF, Viswanathan G. et al. Collagen–carbon nanotube composite materials as scaffolds in tissue engineering. Journal of Biomedical Materials Research Part A: An Official Journal of The Society for Biomaterials, The Japanese Society for Biomaterials, and The Australian Society for Biomaterials and the Korean Society for Biomaterials 2005; 74: 489-496
  • 40 Tonelli FM, Santos AK, Gomes KN. et al. Carbon nanotube interaction with extracellular matrix proteins producing scaffolds for tissue engineering. International Journal of Nanomedicine 2012; 7: 4511
  • 41 Riaz M, Fulati A, Amin G. et al. Buckling and elastic stability of vertical ZnO nanotubes and nanorods. Journal of Applied Physics 2009; 106: 034309
  • 42 Teker K, Wickstrom E, Panchapakesan B. Biomolecular tuning of electronic transport properties of carbon nanotubes via antibody functionalization. IEEE Sensors Journal 2006; 6: 1422-1428
  • 43 Tajallaie-Asl F, Mardani M, Shahsavari S. et al. Menstruation phytotherapy according to iran ethnobotanical sources. J. Pharm. Sci. & Res 2017; 9: 986-990
  • 44 Singh S, Mehra NK, Jain N. Development and characterization of the paclitaxel loaded riboflavin and thiamine conjugated carbon nanotubes for cancer treatment. Pharmaceutical Research 2016; 33: 1769-1781
  • 45 Al Faraj A, Shaik AS, Halwani R. et al. Magnetic targeting and delivery of drug-loaded SWCNTs theranostic nanoprobes to lung metastasis in breast cancer animal model: Noninvasive monitoring using magnetic resonance imaging. Molecular Imaging and Biology 2016; 18: 315-324
  • 46 Mashal A, Sitharaman B, Li X. et al. Toward carbon-nanotube-based theranostic agents for microwave detection and treatment of breast cancer: Enhanced dielectric and heating response of tissue-mimicking materials. IEEE Transactions on Biomedical Engineering 2010; 57: 1831-1834
  • 47 Dineshkumar B, Krishnakumar K, Bhatt A. et al. Single-walled and multi-walled carbon nanotubes based drug delivery system: Cancer therapy: A review. Indian Journal of Cancer 2015; 52: 262
  • 48 Xiao Y, Gao X, Taratula O. et al. Anti-HER2 IgY antibody-functionalized single-walled carbon nanotubes for detection and selective destruction of breast cancer cells. BMC cancer 2009; 9: 351
  • 49 Yu S, Zhang Y, Chen L. et al. Antitumor effects of carbon nanotube-drug complex against human breast cancer cells. Experimental and therapeutic medicine 2018; 16: 1103-1110. doi:10.3892/etm.2018.6334 [published Online First: Epub Date]|
  • 50 Gajewicz A, Rasulev B, Dinadayalane TC. et al. Advancing risk assessment of engineered nanomaterials: Application of computational approaches. Advanced Drug Delivery Reviews 2012; 64: 1663-1693

Correspondence

Mahdis Tajabadi
Tehran Medical Branch, Islamic Azad University,
Khaghani Street, Shariati Avenue,
Tehran, Iran
Phone: +93/865/65 639   

  • References

  • 1 Mourouti N, Panagiotakos DB, Kotteas EA. et al. Optimizing diet and nutrition for cancer survivors: A review. Maturitas 2017; 105: 33-36
  • 2 DeSantis C, Siegel R, Bandi P. et al. Breast cancer statistics, 2011. CA: A Cancer Journal for Clinicians 2011; 61: 408-418
  • 3 Valastyan S, Weinberg RA. Tumor metastasis: Molecular insights and evolving paradigms. Cell 2011; 147: 275-292
  • 4 Lorzadeh N, Kazemirad S, Lorzadeh M. et al. Comparison of the effect of oral and intravenous fluid therapy on women with oligohydramnios. Res J Obstet Gynecol 2008; 1: 25-29
  • 5 Fidler IJ. The pathogenesis of cancer metastasis: The ‘seed and soil’ hypothesis revisited. Nature Reviews Cancer 2003; 3: 453
  • 6 Nguyen DX, Bos PD, Massagué J. Metastasis: From dissemination to organ-specific colonization. Nature Reviews Cancer 2009; 9: 274
  • 7 Kam NWS, O'Connell M, Wisdom JA. et al. Carbon nanotubes as multifunctional biological transporters and near-infrared agents for selective cancer cell destruction. Proceedings of the National Academy of Sciences 2005; 102: 11600-11605
  • 8 Akbariasbagh F, Lorzadeh N, Azmoodeh A. et al. Association among diameter and volume of follicles, oocyte maturity, and competence in intracytoplasmic sperm injection cycles. Minerva Ginecologica 2015; 67: 397-403
  • 9 Lu S, Panchapakesan B. Optically driven nanotube actuators. Nanotechnology 2005; 16: 2548
  • 10 Lu S, Panchapakesan B. Nanotube micro-optomechanical actuators. Applied Physics Letters 2006; 88: 253107
  • 11 Lu S, Panchapakesan B. Photomechanical responses of carbon nanotube/polymer actuators. Nanotechnology 2007; 18: 305502
  • 12 Haddon RC. Carbon nanotubes. ACS Publications; 2002
  • 13 Milne W, Teo K, Amaratunga G. et al. Carbon nanotubes as field emission sources. Journal of Materials Chemistry 2004; 14: 933-943
  • 14 Berger J, Reist M, Mayer JM. et al. Structure and interactions in covalently and ionically crosslinked chitosan hydrogels for biomedical applications. European Journal of Pharmaceutics and Biopharmaceutics 2004; 57: 19-34
  • 15 Ericson LM, Fan H, Peng H. et al. Macroscopic, neat, single-walled carbon nanotube fibers. Science 2004; 305: 1447-1450
  • 16 Martin CR, Kohli P. The emerging field of nanotube biotechnology. Nature Reviews Drug Discovery 2003; 2: 29
  • 17 Bianco A, Prato M. Can carbon nanotubes be considered useful tools for biological applications?. Advanced Materials 2003; 15: 1765-1768
  • 18 Kam NWS, Dai H. Carbon nanotubes as intracellular protein transporters: Generality and biological functionality. Journal of the American Chemical Society 2005; 127: 6021-6026
  • 19 Mehra NK, Mishra V, Jain N. A review of ligand tethered surface engineered carbon nanotubes. Biomaterials 2014; 35: 1267-1283
  • 20 Mehra NK, Jain NK. Multifunctional hybrid-carbon nanotubes: New horizon in drug delivery and targeting. Journal of Drug Targeting 2016; 24: 294-308
  • 21 Lorzadeh N, Ghasem Nejad A, Mohmad Pour J. The Effect of metformin on outcome of Intrauterine insemination (IUI) in insulin non-resistant infertile women with polycystic ovarian syndrome. The Iranian. Journal of Obstetrics, Gynecology and Infertility 2014; 17: 1-11
  • 22 Roldo M, Fatouros DG. Biomedical applications of carbon nanotubes. Annual Reports Section" C"(Physical Chemistry) 2013; 109: 10-35
  • 23 Casais-Molina ML, Cab C, Canto G. et al. Carbon Nanomaterials for Breast Cancer Treatment %J Journal of Nanomaterials. 2018; 2018: 9, doi:10.1155/2018/2058613 [published Online First: Epub Date]
  • 24 Bianco A, Kostarelos K, Prato M. Applications of carbon nanotubes in drug delivery. Current Opinion in Chemical Biology 2005; 9: 674-679
  • 25 Wu W, Wieckowski S, Pastorin G. et al. Targeted delivery of amphotericin B to cells by using functionalized carbon nanotubes. Angewandte Chemie International Edition 2005; 44: 6358-6362
  • 26 Pastorin G, Wu W, Wieckowski S. et al. Double functionalisation of carbon nanotubes for multimodal drug delivery. Chemical Communications 2006; 11: 1182-1184
  • 27 Sanginario A, Miccoli B, Demarchi D. Carbon Nanotubes as an Effective Opportunity for Cancer Diagnosis and Treatment. Biosensors 2017; 7 9 DOI: 10.3390/bios7010009 [published. Online First: Epub Date]
  • 28 Fahrenholtz CD, Hadimani M, King SB. et al. Targeting breast cancer with sugar-coated carbon nanotubes. Nanomedicine (London, England) 2015; 10: 2481-2497, doi:10.2217/NNM.15.90 [published Online First: Epub Date]
  • 29 Chen RJ, Bangsaruntip S, Drouvalakis KA. et al. Noncovalent functionalization of carbon nanotubes for highly specific electronic biosensors. Proceedings of the National Academy of Sciences 2003; 100: 4984-4989
  • 30 Radomski A, Jurasz P, Alonso-Escolano D. et al. Nanoparticle-induced platelet aggregation and vascular thrombosis. British Journal of Pharmacology 2005; 146: 882-893
  • 31 Narayan RJ, Jin C, Menegazzo N. et al. Nanoporous hard carbon membranes for medical applications. Journal of Nanoscience and Nanotechnology 2007; 7: 1486-1493
  • 32 Shao N, Lu S, Wickstrom E. et al. Integrated molecular targeting of IGF1R and HER2 surface receptors and destruction of breast cancer cells using single wall carbon nanotubes. Nanotechnology 2007; 18: 315101
  • 33 Browne BC, Eustace AJ, Kennedy S. et al. Evaluation of IGF1R and phosphorylated IGF1R as targets in HER2-positive breast cancer cell lines and tumours. Breast Cancer Research and Treatment 2012; 136: 717-727
  • 34 Chakravarty P, Marches R, Zimmerman NS. et al. Thermal ablation of tumor cells with antibody-functionalized single-walled carbon nanotubes. Proceedings of the National Academy of Sciences 2008; 105: 8697-8702
  • 35 McDevitt MR, Chattopadhyay D, Kappel BJ. et al. Tumor targeting with antibody-functionalized, radiolabeled carbon nanotubes. Journal of Nuclear Medicine 2007; 48: 1180-1189
  • 36 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. Molecular Pharmaceutics 2016; 13: 2423-2432. doi:10.1021/acs.molpharmaceut.6b00183 [published Online First: Epub Date]
  • 37 Son KH, Hong JH, Lee JW. Carbon nanotubes as cancer therapeutic carriers and mediators. International Journal of Nanomedicine 2016; 11: 5163
  • 38 Liu J, Appaix F, Bibari O. et al. Control of neuronal network organization by chemical surface functionalization of multi-walled carbon nanotube arrays. Nanotechnology 2011; 22: 195101
  • 39 MacDonald RA, Laurenzi BF, Viswanathan G. et al. Collagen–carbon nanotube composite materials as scaffolds in tissue engineering. Journal of Biomedical Materials Research Part A: An Official Journal of The Society for Biomaterials, The Japanese Society for Biomaterials, and The Australian Society for Biomaterials and the Korean Society for Biomaterials 2005; 74: 489-496
  • 40 Tonelli FM, Santos AK, Gomes KN. et al. Carbon nanotube interaction with extracellular matrix proteins producing scaffolds for tissue engineering. International Journal of Nanomedicine 2012; 7: 4511
  • 41 Riaz M, Fulati A, Amin G. et al. Buckling and elastic stability of vertical ZnO nanotubes and nanorods. Journal of Applied Physics 2009; 106: 034309
  • 42 Teker K, Wickstrom E, Panchapakesan B. Biomolecular tuning of electronic transport properties of carbon nanotubes via antibody functionalization. IEEE Sensors Journal 2006; 6: 1422-1428
  • 43 Tajallaie-Asl F, Mardani M, Shahsavari S. et al. Menstruation phytotherapy according to iran ethnobotanical sources. J. Pharm. Sci. & Res 2017; 9: 986-990
  • 44 Singh S, Mehra NK, Jain N. Development and characterization of the paclitaxel loaded riboflavin and thiamine conjugated carbon nanotubes for cancer treatment. Pharmaceutical Research 2016; 33: 1769-1781
  • 45 Al Faraj A, Shaik AS, Halwani R. et al. Magnetic targeting and delivery of drug-loaded SWCNTs theranostic nanoprobes to lung metastasis in breast cancer animal model: Noninvasive monitoring using magnetic resonance imaging. Molecular Imaging and Biology 2016; 18: 315-324
  • 46 Mashal A, Sitharaman B, Li X. et al. Toward carbon-nanotube-based theranostic agents for microwave detection and treatment of breast cancer: Enhanced dielectric and heating response of tissue-mimicking materials. IEEE Transactions on Biomedical Engineering 2010; 57: 1831-1834
  • 47 Dineshkumar B, Krishnakumar K, Bhatt A. et al. Single-walled and multi-walled carbon nanotubes based drug delivery system: Cancer therapy: A review. Indian Journal of Cancer 2015; 52: 262
  • 48 Xiao Y, Gao X, Taratula O. et al. Anti-HER2 IgY antibody-functionalized single-walled carbon nanotubes for detection and selective destruction of breast cancer cells. BMC cancer 2009; 9: 351
  • 49 Yu S, Zhang Y, Chen L. et al. Antitumor effects of carbon nanotube-drug complex against human breast cancer cells. Experimental and therapeutic medicine 2018; 16: 1103-1110. doi:10.3892/etm.2018.6334 [published Online First: Epub Date]|
  • 50 Gajewicz A, Rasulev B, Dinadayalane TC. et al. Advancing risk assessment of engineered nanomaterials: Application of computational approaches. Advanced Drug Delivery Reviews 2012; 64: 1663-1693

Zoom Image
Fig. 1 Single-walled carbon nanotube (SWCNT) and multiwalled carbon nanotube (MWCNT) [rerif] [50].
Zoom Image
Fig. 2 Conjugation schematic of SWNTs with mAbs. 1-pyrenebutanoyl succinimide was adsorbed on the NT via π–π stacking. The succinimidyl ester can react with lysine side chain amines available on Abs to form amide bonds. PEG serves the purpose of insulator to hinder possible bio-fouling (Image adapted from [32] with permission).
Zoom Image
Fig. 3 TEM of the functionalization of mAbs aggregates specific to IGF1 receptor in breast cancer cells. Each site comprising 10–25 Ab molecules [rerif] [32].