We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

HER-2/neu and MYC gene silencing in breast cancer: therapeutic potential and advancement in nonviral nanocarrier systems

    Jananee Padayachee

    Nano-Gene & Drug Delivery Laboratory, Discipline of Biochemistry, School of Life Sciences, College of Agriculture, Engineering & Science, University of KwaZulu-Natal (Westville Campus), Private Bag X54001, Durban 4000, Kwa-Zulu Natal, South Africa

    ,
    Aliscia Daniels

    Nano-Gene & Drug Delivery Laboratory, Discipline of Biochemistry, School of Life Sciences, College of Agriculture, Engineering & Science, University of KwaZulu-Natal (Westville Campus), Private Bag X54001, Durban 4000, Kwa-Zulu Natal, South Africa

    ,
    Adhika Balgobind

    Nano-Gene & Drug Delivery Laboratory, Discipline of Biochemistry, School of Life Sciences, College of Agriculture, Engineering & Science, University of KwaZulu-Natal (Westville Campus), Private Bag X54001, Durban 4000, Kwa-Zulu Natal, South Africa

    ,
    Mario Ariatti

    Nano-Gene & Drug Delivery Laboratory, Discipline of Biochemistry, School of Life Sciences, College of Agriculture, Engineering & Science, University of KwaZulu-Natal (Westville Campus), Private Bag X54001, Durban 4000, Kwa-Zulu Natal, South Africa

    &
    Moganavelli Singh

    *Author for correspondence: Tel.: +27 ƒ31 2601 7170;

    E-mail Address: singhm1@ukzn.ac.za

    Nano-Gene & Drug Delivery Laboratory, Discipline of Biochemistry, School of Life Sciences, College of Agriculture, Engineering & Science, University of KwaZulu-Natal (Westville Campus), Private Bag X54001, Durban 4000, Kwa-Zulu Natal, South Africa

    Published Online:https://doi.org/10.2217/nnm-2019-0459

    Globally, breast cancer is the second leading cause of cancer-related mortality among women, with approximately 1.4 million new cases diagnosed annually. Associated genetic perturbations are emerging in the face of intense scientific enquiry, facilitating its classification, prognostication and treatment. RNAi, utilizing siRNA, is a powerful treatment strategy to silence disease-causing genes. However, therapeutic siRNA instability and poor cellular uptake have limited its clinical application, necessitating the use of nanocarriers. In this review, we highlight the RNAi mechanism, HER-2/neu and MYC as breast cancer gene targets, and nonviral nanocarriers as potentially safe and efficient delivery systems.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Huang C, Li M, Chen C, Yao Q. Small interfering RNA therapy in cancer: mechanism, potential targets, and clinical applications. Expert Opin. Ther. Tar. 12, 637–645 (2008).
    • 2. Oh YK, Park TG. siRNA delivery systems for cancer treatment. Adv. Drug Deliv. Rev. 61, 850–862 (2009).
    • 3. Maiyo F, Singh M. Selenium nanoparticles: potential in cancer gene and drug delivery. Nanomedicine 12(9), 1075–1089 (2017).
    • 4. Benson JR, Jatoi I. The global breast cancer burden. Future Oncol. 8, 697–702 (2012).
    • 5. World Health Organization. www.who.int/cancer/detection/breastcancer/en/index1
    • 6. American Cancer Society. Breast cancer facts and figures 2017/2018 (2018). www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/breast-cancer-facts-and-figures-2017-2018.pdf
    • 7. Napoli C, Lemieux C, Jorgensen R. Introduction of a chimeric chalcone synthase gene into petunia results in reversible co-suppression of homologous genes in trans. Plant Cell 2, 279–289 (1990).
    • 8. Singh A, Trivedi P, Jain NK. Advances in siRNA delivery in cancer therapy. Artif. Cells. Nanomed. Biotech. 46(2), 274–283 (2018).
    • 9. Dana H, Chalbatani GM, Mahmoodzadeh H et al. Molecular mechanisms and biological functions of siRNA. Int. J. Biomed. Sci. 13(2), 48–57 (2017).
    • 10. Zhang H, Kolb FA, Jaskiewicz L, Westhof E, Filipowicz W. Single processing centre models for human Dicer and bacterial RNase III. Cell 118, 57–68 (2004).
    • 11. Huynh A, Madu CO, Lu Y. siRNA: a promising new tool for future breast cancer therapy. Oncomedicine 3, 74–81 (2018).
    • 12. Mansoori B, Shotorbani SS, Baradaran B. RNA interference and its role in cancer therapy. Adv. Pharm. Bull. 4(4), 313–321 (2014).
    • 13. Kim VN, Han J, Siomi MC. Biogenesis of small RNAs in animals. Nat. Rev. Mol. Cell. Biol. 10, 126–139 (2009).
    • 14. Matranga C, Tomari Y, Shin C, Bartel DP, Zamore PD. Passenger-strand cleavage facilitates assembly of siRNA into AgO2-containing RNAi enzyme complexes. Cell 123, 607–620 (2005).
    • 15. Bartlett DW, Davis ME. Insights into the kinetics of siRNA-mediated gene silencing from live-cell and live-animal bioluminescent imaging. Nucleic Acids Res. 34, 322–333 (2006).
    • 16. Naab TJ, Gautam A, Ricks-Santi L et al. MYC amplification in subtypes of breast cancers in African American women. BMC Cancer 18, 274–280 (2018).
    • 17. Lodish A, Berk A, Zipursky SL et al. Proto-oncogenes and tumour suppressor genes. In: Molecular Cell Biology (4th Edition). WH Freeman (Ed.). MacMillan, NY, USA (2000).
    • 18. Haschek WM, Rousseaux CG, Wallig MA. Manifestaions of toxic cell injury: cell injury/death and chemical carcinogenisis. In: Fundamentals of Toxicologic Pathology (2nd Edition). Academic Press, MA, USA 9–42 (2010).
    • 19. Igbal N, Igbal N. Human epidermal growth factor receptor 2 (HER2) in cancers: overexpression and therapeutics implications. Mol. Biol. Int. 2014, 852748 (2014). • Describes the role of HER2 in various cancers and treatment strategies that target HER2.
    • 20. Mitri Z, Constantine T, O'Regan R. The HER2 receptor in breast cancer: pathophysiology, clinical use and new advances in therapy. Chemother. Res. Pract. 2012, 743193 (2012).
    • 21. Harbeck N, Pegram MD, Rüschoff J, Mobus V. Targeted therapy in metastatic breast cancer: the HER2/neu oncogene. Breast Care 5(Suppl. 1), 3–7 (2010).
    • 22. Fernández Val JF, Losada J, Arregui Murua MA, Sarría R. Cell proliferation, nuclear ploidy, and EGFR and HER2/neu tyrosine kinase oncoproteins in infiltrating ductal breast carcinoma. Cancer Genet. Cytogenet. 138, 69–72 (2002).
    • 23. Warren CM, Landgraf R. Signalling through ERBB receptors: multiple layers of diversity and control. Cell. Signal. 18, 923–933 (2006).
    • 24. Engel RH, Kaklamani VG. HER2-positive breast cancer: current and future treatment strategies. Drugs 67, 1329–1341 (2007).
    • 25. Ngamcherdtrakul W, Morry J, Gu S et al. Cationic polymer modified mesoporous silica nanoparticles for targeted siRNA delivery to HER2+ breast cancer. Adv. Funct. Mater. 25(18), 2646–2659 (2015).
    • 26. Tan WB, Jiang S, Zhang Y. Quantum-dot based nanoparticles for targeted silencing of HER2/neu gene via RNA interference. Biomaterials 28, 1565–1571 (2007).
    • 27. Wang Y, Morrison G, Gillihan R et al. Different mechanisms for resistance to trastuzumab versus lapatinib in HER2-positive breast cancers – role of estrogen receptor and HER2 reactivation. Breast Cancer Res. 13(6), R121 (2011).
    • 28. Wong LL, Zhang D, Chang CF, Koay ESC. Silencing of the PP2A catalytic subunit causes HER-2/neu positive breast cancer cells to undergo apoptosis. Exp. Cell Res. 16, 3387–3396 (2010).
    • 29. Tai W, Qin B, Cheng K. Inhibition of breast cancer cell growth and invasiveness by dual silencing of HER-2 and VEGF. Mol. Pharm. 7, 543–556 (2010).
    • 30. Klauber-DeMore N, Schulte BA, Wang GY. Targeting MYC for triple-negative breast cancer treatment. Oncoscience 5, 5–6 (2018).
    • 31. Horiuchi D, Camardr R, Zhou AY et al. PIM1 kinase inhibition as a targeted therapy against triple-negative breast tumours with elevated MYC expression. Nat. Med. 22(11), 1321–1329 (2016).
    • 32. Chen H, Liu H, Quig G. Targeting oncogenic Myc as a strategy for cancer treatment. Signal Transduct. Tar. 3, 5 (2018).
    • 33. Fallah Y, Brundage J, Allegakoen P, Shajahan-Haq AN. MYC-driven pathways in breast cancer sub-types. Biomolecules 7, 53 (2017). • Outlines the role of MYC in different breast cancer subtypes.
    • 34. Perou CM. Molecular stratification of triple-negative breast cancers. Oncologist 16(Suppl. 1), 61–70 (2011).
    • 35. Lee K, Giltane JM, Balko JM et al. MYC and MCL1 cooperatively promote chemotherapy-resistant breast cancer stem cells through regulation of mitochondrial oxidative phosphorylation. Cell Metab. 26(4), 633–647 (2017).
    • 36. Carey JPW, Karakas C, Bui T et al. Synthetic lethality of PARP inhibitors in combination with MYC blockade is independent of BRCA status in triple-negative breast cancer. Cancer Res. 78(3), 742–757 (2017).
    • 37. Stine ZE, Walton ZE, Altman BJ, Hsieh AL, Dang CV. MYC, metabolism and cancer. Cancer Discov. 5(10), 1024–1039 (2015).
    • 38. Meyer N, Penn LZ. Reflecting on 25 years with MYC. Nat. Rev. Cancer 8, 976–990 (2008).
    • 39. Xu J, Cen Y, Olopade OI. MYC and breast cancer. Genes Cancer 1(6), 629–640 (2010). •• Describes MYC protein and biological function, MYC upregulation in the breast cancer subtypes and therapeutic strategies for breast cancer.
    • 40. Conacci-Sorrel M, McFerrin L, Eisenman RN. An overview of MYC and its interactome. CSH Perspect. Med. 4, a014357 (2014).
    • 41. Tian T, Xiaoyi L, Zhang J. mTOR signalling in cancer and mTOR inhibitor in solid tumour targeting therapy. Int. J. Mol. Sci. 20, 755 (2019).
    • 42. Muthulagu N, Junttila MR, Wiese KE et al. BIM is the primary mediator of MYC-induced apoptosis in multiple solid tumours. Cell. Rep. 8(5), 1347–1353 (2014).
    • 43. Liu Y, Zhu YH, Mao CQ et al. Triple-negative breast cancer therapy and CDK1 siRNA delivered by cationic lipid assisted PEG-PLA nanoparticles. J. Control. Rel. 192, 114–121 (2014).
    • 44. Yang A, Qin S, Schulte BA, Ethier SP, Kenneth DT, Wang GY. MYC inhibition depletes cancer stem-like cells in triple negative breast cancer. Cancer Res. 77(23), 6641–6650 (2017).
    • 45. Jones SK, Merkel OM. Tackling breast cancer chemoresistance with nano-formulated siRNA. Gene Ther. 23(12), 821–828 (2016).
    • 46. Mbatha LS, Maiyo FC, Singh M. Dendrimer functionalized folate-targeted gold nanoparticles for luciferase gene silencing in vitro: a proof of principle study. Acta Pharm. 69, 49–61 (2019).
    • 47. Deng Y, Wang CC, Choy KW et al. Therapeutic potentials of gene silencing by RNA interference: principles, challenges, and new strategies. Gene 538, 217–227 (2014).
    • 48. Thanki K, Zeng X, Justesen S et al. Engineering of small interfering RNA-loaded lipidoid-poly(DL-lactic-co-glycidic acid) hybrid nanoparticles for highly efficient and safe gene silencing. a quality by design-based approach. Eur. J. Pharma. Biopharm. 120, 22–33 (2017).
    • 49. Mbatha LS, Singh M. Starburst poly(amidoamine) dendrimer grafted gold nanoparticles as a scaffold for folic acid-targeted plasmid DNA delivery in vitro. J. Nanosci. Nanotechnol. 19(4), 1959–1970 (2019).
    • 50. Draz MS, Fang BA, Zhang P et al. Nanoparticle mediated systemic delivery of siRNA for treatment of cancers and viral infections. Theranostics 4(9), 872–892 (2014).
    • 51. Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv. Drug Deliv. Rev. 99(Pt A), 28–55 (2016).
    • 52. Shen Z, Fisher A, Liu NK, Li Y. PEGylated “stealth” nanoparticles and liposomes: engineering of biomaterials for drug delivery systems. In: Beyond Polyethylene Glycol. Woodhead Publishing Series in Biomaterials, 1–26 (2018).
    • 53. Çağdaş M, Sezer AD, Bucak S. Liposomes as potential drug carrier systems for drug delivery. In: Application of Nanotechnology in Drug Delivery. InTechOpen Ltd, London, UK (2014).
    • 54. Kesharwani P, Gajbhiye V, Jain NK. A review of nanocarriers for the delivery of small interfering RNA. Biomaterials 33, 7138–7150 (2012).
    • 55. Garćia-Pinel B, Porras-Alcalá C, Ortega-Rodŕiguez A et al. Lipid-based nanoparticles: applications and recent advances in cancer treatment. Nanomaterials 9(4), 638 (2019). •• Outlines lipid-based nanoparticles and their applications in cancer treatment.
    • 56. Liu Y, Huang L. Designer lipids advance systemic siRNA delivery. Mol. Ther. 18, 669–670 (2010).
    • 57. Talluri SV, Kuppusamy G, Karri VVSR, Tummala S, Madhunapantula SV. Lipid-based nanocarriers for breast cancer treatment-comprehensive review. Drug Deliv. 23(4), 1291–1305 (2016).
    • 58. Hoekstra D, Rejman J, Wasungu L, Shi F, Zuhorn I. Gene delivery by cationic lipids: in and out of an endosome. Biochem. Soc. Trans. 35, 68–71 (2007).
    • 59. Nourbakhsh M, Jaafari MR, Lage H, Abnous K. Nanolipoparticles-mediated MDR1 siRNA delivery reduces doxorubicin resistance in breast cancer cells and silences MDR1 expression in xenograft model of human breast cancer. Iran. J. Basic Med. Sci. 18(4), 385–392 (2015).
    • 60. Zhang Y, Li H, Sun J et al. DC-Chol/DOPE cationic liposomes: a comparative study of the influence factors on plasmid pDNA and siRNA gene delivery. Int. J. Pharm. 390, 198–207 (2010).
    • 61. Pirollo KF, Chang EH. Targeted delivery of small interfering RNA: approaching effective cancer therapies. Cancer Res. 68(5), 1247–1250 (2008).
    • 62. Brown S, Khan DR. The treatment of breast cancer using liposome technology. J. Drug Deliv. 2012, 212965 (2012).
    • 63. Gao J, Zhang J, He J et al. Tumour-targeted PE38KDEL delivery via PEGylated anti-HER2 immunoliposomes. Int. J. Pharm. 374(1–2), 145–152 (2009).
    • 64. Beltrán-Grazia E, López-Camacho A, Higura-Ciapara I, Velázquez-Fernandez J, Vallejo-Cardon A. Nanomedicine review: clinical developments in liposomal applcations. Cancer Nano. 10, 11 (2019).
    • 65. Pandey H, Rani R, Agarwal V. Liposomes and their applications in cancer therapy. Braz. Arch. Biol.Technol. 59, e16150477 (2016).
    • 66. Barenholz Y. Doxil (R) the first FDA-approved nano-drug lessons learned. J. Control. Rel. 160, 117–134 (2012).
    • 67. Batist G, Gelman KA, Chi LN et al. Safety, pharmacokinetics and efficacy of CPX-1 liposome injected in patients with solid tumours. Clin. Cancer Res. 15, 692–700 (2009).
    • 68. Boeckle S, von Gersdorff K, van der Piepen S, Culmsee C, Wagner E, Ogris M. Purification of polyethylenimine polyplexes highlights the role of free polycations in gene transfer. J. Gene Med. 6, 1102–1111 (2004).
    • 69. Xin Y, Huang M, Guo WW, Huang Q, Zhang L, Jiang G. Nano-based delivery of RNAi in cancer therapy. Mol. Cancer 16, 134 (2017).
    • 70. Ragelle H, Riva R, Vandermeulen G et al. Chitosan nanoparticles for siRNA delivery: optimizing formulation to increase the stability and efficiency. J. Control. Rel. 176, 54–63 (2014).
    • 71. Mao S, Sun W, Kissel T. Chitosan-based formulations for delivery of DNA and siRNA. Adv. Drug Deliv. Rev. 62, 12–27 (2010).
    • 72. Grzelinski M, Urban-Klein B, Martens T et al. RNA interference-mediated gene silencing of pleiotrophin through polyethylenimine-complexed small interfering RNAs in vivo exerts antitumoral effects in glioblastoma xenografts. Human Gene Ther. 17, 751–766 (2006).
    • 73. Garcia-Fuentes M, Alonso MJ. Chitosan based nanocarriers: where do we stand? J. Control. Rel. 161, 496–504 (2012).
    • 74. Yang S, Chen Y, Ahmadie R, Ho EA. Advancements in the field of intravaginal siRNA delivery. J. Control. Rel. 167, 29–39 (2013).
    • 75. Alinej V, Somi MH, Baradarem B et al. Co-delivery of IL178RB siRNA and doxorubicin by chitosan-based nanoparticles for enhanced anticancer efficacy in breast cancer cells. Biomed. Pharmacother. 83, 229–240 (2016).
    • 76. Yu S, Chen Y, Li X, Gao Z, Liu G. Chitosan nanoparticle-delivered siRNA induces CXCR4 expression and sensitizes breast cancer cells to cisplatin. Biosci. Rep. 37(3), BSR20170122 (2017).
    • 77. Safari J, Zarnegar Z. Advanced drug delivery systems: nanotechnology of health design: A review. J. Saudi Chem. Soc. 18, 85–99 (2014).
    • 78. Dzmitruk V, Apartsin E, Ihnatsyeu-Kachan A, Abashkin V, Shcharbin D, Bryszewska M. Dendrimers show promise for siRNA and microRNA therapeutics. Pharmaceutics 10, 126 (2018).
    • 79. Abbasi E, Aval S, Akbarzadeh A et al. Dendrimers: synthesis, applications, and properties. Nanoscale Res. Lett. 9, 247 (2014).
    • 80. Khan OF, Zaia EW, Yin H et al. Ionizable amphiphilic dendrimer-based nanomaterials with alkyl-chain-substituted amines for tunable siRNA delivery to the liver endothelium in vivo. Angew. Chem. Int. Ed. 53, 14397–14401 (2014).
    • 81. Liu X, Liu C, Zhou J et al. Promoting siRNA delivery via enhanced cellular uptake using arginine-decorated amphiphilic dendrimer. Nanoscale 7, 3867–3875 (2015).
    • 82. Deshahri A, Sadeghpour H. Surface decorations of poly(amido amine) dendrimer by various pendant moieties for improve delivery of nucleic acid materials. Colloids Surf. B 132, 85–102 (2015).
    • 83. Finlay J, Roberts CM, Lowe G, Loeza J, Rossi JJ, Glackin CA. RNA based TWIST1 inhibition via dendrimer complex to reduce breast cancer cell metastasis. BioMed Res. Int. 2015, 382745 (2015).
    • 84. Li J, Liu J, Guo N, Zhang Z. Reversal of multidrug resistance in breast cancer MCF-7/ADR cells by h-R3-siMDR1-PAMAM complexes. Int. J. Pharm. 511(1), 436–445 (2016).
    • 85. Ngamcherdtrakul W, Castro DJ, Gu S et al. Current development of targeted oligonucleotide-based cancer therapies: perspective on HER2-positive breast cancer treatment. Cancer Treat. Rev. 45, 19–29 (2016).
    • 86. Herizchi R, Abbasi E, Milani M, Akbarzadeh A. Current methods for synthesis of gold nanoparticles. Artif. Cells Nanomed. Biotechnol. 44(2), 596–602 (2016).
    • 87. Moon HJ, Ku M, Lee H, Yoon N, Yang J, Bong KW. Implantable photothermal agents based on gold nanorods-encapsulated microcube. Sci. Rep. 8(1), 13683 (2018).
    • 88. Shahbazi R, Asik E, Kahraman N, Turk M, Ozpolat B, Ulubayram K. Modified gold-based siRNA nanotherapeutics for targeted therapy of triple-negative breast cancer. Nanomedicine 12(16), 1961–1973 (2017).
    • 89. Daniels AN, Singh M. Sterically stabilized siRNA:gold nanocomplexes enhance c-MYC silencing in a breast cancer cell model. Nanomedicine 14(11), 1387–1401 (2019).
    • 90. Ahwazi RP, Kiani M, Dinarvand M et al. Immobilization of HIV-1 TAT peptide on gold nanoparticles: a feasible approach for siRNA delivery. J. Cell. Phys. 235(3), 2049–2059 (2020).
    • 91. Shen J, Kim H-C, Mu C et al. Multifunctional gold nanorods for siRNA gene silencing and photothermal therapy. Adv. Healthcare Mater. 3(10), 1629–1637 (2014a).
    • 92. Yang Z, Liu T, Xie Y et al. Chitosan layered gold nanorods as synergistic therapeutics for photothermal ablation and gene silencing in triple-negative breast cancer. Acta Biomater. 25, 194–204 (2015).
    • 93. Wang S, Tian Y, Tian W et al. Selectively sensitizing malignant cells to photothermal therapy using a CD44-targeting heat shock protein 72 depletion nanosystem. ACS Nano. 10(9), 8578–8590 (2016).
    • 94. Moodley T, Singh M. Polymeric mesoporous silica nanoparticles for enhanced delivery of 5-fluorouracil in vitro. Pharmaceutics 11(6), 288 (2019).
    • 95. Xu C, Lei C, Yu C. Mesoporous silica nanoparticles for protein protection and delivery. Front. Chem. 7, 290 (2019).
    • 96. Shen J, Kim H-C, Su H et al. Cyclodextrin and polyethylenimine functionalized mesoporous silica nanoparticles for delivery of siRNA cancer therapeutics. Theranostics 4(5), 487–497 (2014b).
    • 97. Na H-K, Kim M-H, Park K et al. Efficient functional delivery of siRNA using mesoporous silica nanoparticles with ultralarge pores. Small 8(11), 1752–1761 (2012).
    • 98. Meng H, Mai WX, Zhang H et al. Codelivery of an optimal drug/siRNA combination using mesoporous silica nanoparticles to overcome drug resistance in breast cancer in vitro and in vivo. ACS Nano 7(2), 994–1005 (2013).
    • 99. Matea CT, Mocan T, Tabaran F et al. Quantum dots in imaging, drug delivery and sensor applications. Int. J. Nanom. 12, 5421–5431 (2017).
    • 100. Zhao J, Qiu X, Wang Z, Pan J, Chen J, Han J. Application of quantum dots as vectors in targeted survivin gene siRNA delivery. OncoTargets Ther. 6, 303–309 (2013).
    • 101. Yoo D, Park Y, Cheon B, Park M-H. Carbon dots as an effective fluorescent sensing platform for metal ion detection. Nanoscale Res. Lett. 14(1), 272 (2019).
    • 102. Fernández-Delgado N, Herrera M, Tavabi AH et al. Structural and chemical characterization of CdSe-ZnS core–shell quantum dots. Appl. Surf. Sci. 457, 93–97 (2018).
    • 103. Kim MW, Jeong HY, Kang SJ et al. Anti-EGF receptor aptamer-guided co-delivery of anti-cancer siRNAs and quantum dots for theranostics of triple-negative breast cancer. Theranostics 9(3), 837–852 (2019).
    • 104. Majidi S, Zeinali Sehrig F, Samiei M et al. Magnetic nanoparticles: applications in gene delivery and gene therapy. Artif. Cells Nanomed. Biotechnol. 44(4), 1186–1193 (2015).
    • 105. Ali A, Zafar H, Zia M et al. Synthesis, characterization, applications, and challenges of iron oxide nanoparticles. Nanotechnol. Sci. Appl. 9, 49–67 (2016).
    • 106. Ben Djemaa S, David S, Hervé-Aubert K et al. Formulation and in vitro evaluation of a siRNA delivery nanosystem decorated with gH625 peptide for triple negative breast cancer theranosis. Eur. J. Pharm. Biopharm. 131, 99–108 (2018).
    • 107. Ansari MO, Ahmad MF, Shadab GGHA, Siddique HR. Superparamagnetic iron oxide nanoparticles based cancer theranostics: a double edge sword to fight against cancer. J. Drug Deliv. Sci. Technol. 45, 177–183 (2018).
    • 108. Bruniaux J, Allard-Vannier E, Aubrey N et al. Magnetic nanocarriers for the specific delivery of siRNA: contribution of breast cancer cells active targeting for down-regulation efficiency. Int. J. Pharm. 569, 118572 (2019).
    • 109. Dalmina M, Pittella F, Sierra JA et al. Magnetically responsive hybrid nanoparticles for in vitro siRNA delivery to breast cancer cells. Mater. Sci. Eng. C 99, 1182–1190 (2019).
    • 110. Guruprasath P, Kim J, Gunassekaran GR et al. Interleukin-4 receptor-targeted delivery of Bcl-xL siRNA sensitizes tumors to chemotherapy and inhibits tumor growth. Biomaterials 142, 101–111 (2017).
    • 111. Sanginario A, Miccoli B, Demarchi D. Carbon nanotubes as an effective opportunity for cancer diagnosis and treatment. Biosensors 7(1), 9 (2017).
    • 112. Wong BS, Yoong SL, Jagusiak A et al. Carbon nanotubes for delivery of small molecule drugs. Adv. Drug Deliv. Rev. 65(15), 1964–2015 (2013).
    • 113. Mohammadi M, Salmasi Z, Hashemi M, Mosaffa F, Abnous K, Ramezani M. Single-walled carbon nanotubes functionalized with aptamer and piperazine–polyethylenimine derivative for targeted siRNA delivery into breast cancer cells. Int. J. Pharm. 485(1–2), 50–60 (2015).
    • 114. Alhaji SY, Chowdhury EH, Rosli R, Hassan F, Abdullah S. Gene delivery potential of biofunctional carbonate apatite nanoparticles in lungs. BioMed Res. Int. 2014, 646787 (2014).
    • 115. Chowdhury EH, Akaike T. High performance DNA nano-carriers of carbonate apatite: multiple factors in regulation of particle synthesis and transfection efficiency. Int. J. Nanomed. 2(1), 101–106 (2007).
    • 116. Hossain SM, Shetty J, Tha KK, Chowdhury EH. α-Ketoglutaric acid-modified carbonate apatite enhances cellular uptake and cytotoxicity of a Raf-kinase inhibitor in breast cancer cells through inhibition of MAPK and PI-3 kinase pathways. Biomedicines 7(1), 4 (2019).
    • 117. Fatemian T, Moghimi HR, Chowdhury EH. Intracellular delivery of siRNAs targeting AKT and ERBB2 genes enhances chemosensitization of breast cancer cells in a culture and animal model. Pharmaceutics 11(9), 458 (2019).
    • 118. Tiash S, Chua MJ, Chowdhury EH. Knockdown of ROS1 gene sensitizes breast tumor growth to doxorubicin in a syngeneic mouse model. Int. J. Oncol. 48(6), 2359–2366 (2016).
    • 119. Uddin M, Balaravi Pillai B, Tha K, Ashaie M, Karim M, Chowdhury E. Carbonate apatite nanoparticles-facilitated intracellular delivery of siRNA(s) targeting calcium ion channels efficiently kills breast cancer cells. Toxics 6(3), 34 (2018).