Generic placeholder image

Current Cancer Therapy Reviews

Editor-in-Chief

ISSN (Print): 1573-3947
ISSN (Online): 1875-6301

Review Article

Breast Cancer: A Review of Risk Factors and New Insights into Treatment

Author(s): Kimia Karami and Khatereh Anbari*

Volume 17, Issue 3, 2021

Published on: 20 January, 2021

Page: [207 - 233] Pages: 27

DOI: 10.2174/1573394717999210120195208

Price: $65

Abstract

Today, despite significant advances in cancer treatment have been made, breast cancer remains one of the main health problems and considered a top biomedical investigation urgency. The present study reviewed the common conventional chemotherapy agents and also some alternative and complementary approaches such as oncolytic virotherapy, bacteriotherapy, nanotherapy, immunotherapy, and natural products, which are recommended for breast cancer treatment. In addition to current surgery approaches such as mastectomy, in recent years, a number of novel techniques such as robotic mastectomies, nipple-sparing mastectomy, skin-sparing mastectomy, daycase mastectomy were used in breast cancer surgery. In this review, we summarize new insights into risk factors, surgical and non-surgical treatments for breast cancer.

Keywords: Cancer therapy, surgical treatment, non-surgical treatment, chemotherapy, breast cancer, mastectomy.

Graphical Abstract
[1]
Benson JR, Jatoi I. The global breast cancer burden. Future Oncol 2012; 8(6): 697-702.
[http://dx.doi.org/10.2217/fon.12.61] [PMID: 22764767]
[2]
Dyrstad SW, Yan Y, Fowler AM, Colditz GA. Breast cancer risk associated with benign breast disease: systematic review and meta-analysis. Breast Cancer Res Treat 2015; 149(3): 569-75.
[http://dx.doi.org/10.1007/s10549-014-3254-6] [PMID: 25636589]
[3]
Ghoncheh M, Pournamdar Z, Salehiniya H. Incidence and mortality and epidemiology of breast cancer in the world. Asian Pac J Cancer Prev 2016; 17(S3): 43-6.
[http://dx.doi.org/10.7314/APJCP.2016.17.S3.43] [PMID: 27165206]
[4]
Anbari K, Sahraei N, Ahmadi SAY, Baharvand P. Barriers of breast cancer screening from the viewpoint of women in Khorramabad (West of Iran). Res J Pharm Biol Chem Sci 2016; 7(6): 2044-9.
[5]
Gomez SL, Von Behren J, McKinley M, et al. Breast cancer in Asian Americans in California, 1988-2013: increasing incidence trends and recent data on breast cancer subtypes. Breast Cancer Res Treat 2017; 164(1): 139-47.
[http://dx.doi.org/10.1007/s10549-017-4229-1] [PMID: 28365834]
[6]
Nafissi N, Khayamzadeh M, Zeinali Z, Pazooki D, Hosseini M, Akbari ME. Epidemiology and histopathology of breast cancer in Iran versus other middle eastern countries. Middle East J Cancer 2018; 9(3): 243-51.
[7]
Williams LJ, Fletcher E, Douglas A, et al. Retrospective cohort study of breast cancer incidence, health service use and outcomes in Europe: a study of feasibility. Eur J Public Health 2018; 28(2): 327-32.
[http://dx.doi.org/10.1093/eurpub/ckx127] [PMID: 29020283]
[8]
Abdulrahman GO Jr, Rahman GA. Epidemiology of breast cancer in europe and Africa. J Cancer Epidemiol 2012; 2012: 915610.
[http://dx.doi.org/10.1155/2012/915610] [PMID: 22693503]
[9]
Khakbazan Z, Taghipour A, Latifnejad Roudsari R, Mohammadi E. Help seeking behavior of women with self-discovered breast cancer symptoms: a meta-ethnographic synthesis of patient delay. PLoS One 2014; 9(12): e110262.
[http://dx.doi.org/10.1371/journal.pone.0110262] [PMID: 25470732]
[10]
Carioli G, Malvezzi M, Rodriguez T, Bertuccio P, Negri E, La Vecchia C. Trends and predictions to 2020 in breast cancer mortality: Americas and Australasia. Americas and Australasia Breast 2018; 37: 163-9.
[http://dx.doi.org/10.1016/j.breast.2017.12.004] [PMID: 29246526]
[11]
Colditz GA, Wei EK. Preventability of cancer: the relative contributions of biologic and social and physical environmental determinants of cancer mortality. Annu Rev Public Health 2012; 33: 137-56.
[http://dx.doi.org/10.1146/annurev-publhealth-031811-124627] [PMID: 22224878]
[12]
Rojas K, Stuckey A. Breast cancer epidemiology and risk factors. Clin Obstet Gynecol 2016; 59(4): 651-72.
[http://dx.doi.org/10.1097/GRF.0000000000000239] [PMID: 27681694]
[13]
Sayad S, Ahmadi SAY, Nekouian R, Panahi M, Anbari K. Epidemiological and pathological characteristics of post-surgical cases of invasive breast cancer among ethnicities of Iran in 2018: A single center cross-sectional study. Arch Oncol 2020; 26(1): 6-9.
[http://dx.doi.org/10.2298/AOO190626007S]
[14]
Gaitanidis A, Alevizakos M, Tsalikidis C, Tsaroucha A, Simopoulos C, Pitiakoudis M. Refusal of cancer-directed surgery by breast cancer patients: Risk factors and survival outcomes. Clin Breast Cancer 2018; 18(4): e469-76.
[http://dx.doi.org/10.1016/j.clbc.2017.07.010] [PMID: 28784267]
[15]
Ozsoy A, Barca N, Dolek BA, et al. The relationship between breast cancer and risk factors: A single-center study. Eur J Breast Health 2017; 13(3): 145-9.
[http://dx.doi.org/10.5152/tjbh.2017.3180] [PMID: 28894854]
[16]
Lee A, Mavaddat N, Wilcox AN, et al. BOADICEA: a comprehensive breast cancer risk prediction model incorporating genetic and nongenetic risk factors. Genet Med 2019; 21(8): 1708-18.
[http://dx.doi.org/10.1038/s41436-018-0406-9] [PMID: 30643217]
[17]
Sun Y-S, Zhao Z, Yang Z-N, et al. Risk factors and preventions of breast cancer. Int J Biol Sci 2017; 13(11): 1387-97.
[http://dx.doi.org/10.7150/ijbs.21635] [PMID: 29209143]
[18]
Christie EL, Pattnaik S, Beach J, et al. Multiple ABCB1 transcriptional fusions in drug resistant high-grade serous ovarian and breast cancer. Nat Commun 2019; 10(1): 1295.
[http://dx.doi.org/10.1038/s41467-019-09312-9] [PMID: 30894541]
[19]
Brewer HR, Jones ME, Schoemaker MJ, Ashworth A, Swerdlow AJ. Family history and risk of breast cancer: an analysis accounting for family structure. Breast Cancer Res Treat 2017; 165(1): 193-200.
[http://dx.doi.org/10.1007/s10549-017-4325-2] [PMID: 28578505]
[20]
Alsheh Ali M, Czene K, Hall P, Humphreys K. Association of microcalcification clusters with short-term invasive breast cancer risk and breast cancer risk factors. Sci Rep 2019; 9(1): 14604.
[http://dx.doi.org/10.1038/s41598-019-51186-w] [PMID: 31601987]
[21]
Rizzolo P, Zelli V, Silvestri V, et al. Insight into genetic susceptibility to male breast cancer by multigene panel testing: Results from a multicenter study in Italy. Int J Cancer 2019; 145(2): 390-400.
[http://dx.doi.org/10.1002/ijc.32106] [PMID: 30613976]
[22]
Mihalcea DJ, Florescu M, Vinereanu D. Mechanisms and genetic susceptibility of chemotherapy-induced cardiotoxicity in patients with breast cancer. Am J Ther 2017; 24(1): e3-e11.
[http://dx.doi.org/10.1097/MJT.0000000000000453] [PMID: 27145188]
[23]
Bodelon C, Oh H, Chatterjee N, et al. Association between breast cancer genetic susceptibility variants and terminal duct lobular unit involution of the breast. Int J Cancer 2017; 140(4): 825-32.
[http://dx.doi.org/10.1002/ijc.30512] [PMID: 27859137]
[24]
Hormozi M, Salehi Marzijerani A, Baharvand P. Effects of hydroxytyrosol on epression of aoptotic genes and activity of antioxidant enzymes in LS180 cells. Cancer Manag Res 2020; 12: 7913-9.
[http://dx.doi.org/10.2147/CMAR.S253591] [PMID: 32943925]
[25]
Alshammari FD. Breast cancer genetic susceptibility: With focus in Saudi Arabia. J Oncol Sci 2019; 5(1): 6-12.
[http://dx.doi.org/10.1016/j.jons.2019.02.001]
[26]
Engin A. Obesity-associated breast cancer: Analysis of risk factors. Advances in Experimental Medicine and Biology. Springer New York LLC 2017; pp. 571-606.
[27]
Garcia-Estevez L, Moreno-Bueno G. Updating the role of obesity and cholesterol in breast cancer. Breast Cancer Res 2019; 21(1): 35.
[http://dx.doi.org/10.1186/s13058-019-1124-1] [PMID: 30823902]
[28]
Guenancia C, Lefebvre A, Cardinale D, et al. Obesity as a risk factor for anthracyclines and trastuzumab cardiotoxicity in breast cancer: A systematic review and meta-analysis. J Clin Oncol 2016; 34(26): 3157-65.
[http://dx.doi.org/10.1200/JCO.2016.67.4846] [PMID: 27458291]
[29]
Ahmadipour S, Baharvand P, Rahmani P, Hasanvand A, Mohsenzadeh A. Effect of synbiotic on the treatment of jaundice in full term neonates: A randomized clinical trial. Pediatr Gastroenterol Hepatol Nutr 2019; 22(5): 453-9.
[http://dx.doi.org/10.5223/pghn.2019.22.5.453] [PMID: 31555570]
[30]
Agurs-Collins T, Ross SA, Dunn BK. The many faces of obesity and its influence on breast cancer risk. Front Oncol 2019; 9: 765.
[http://dx.doi.org/10.3389/fonc.2019.00765] [PMID: 31555578]
[31]
Atoum MF, Alzoughool F, Al-Hourani H. Linkage between obesity leptin and breast cancer. Breast Cancer (Auckl) 2020; 14: 1178223419898458.
[http://dx.doi.org/10.1177/1178223419898458] [PMID: 31975779]
[32]
Lee K, Kruper L, Dieli-Conwright CM, Mortimer JE. The impact of obesity on breast cancer diagnosis and treatment. Curr Oncol Rep 2019; 21(5): 41.
[http://dx.doi.org/10.1007/s11912-019-0787-1] [PMID: 30919143]
[33]
Nindrea RD, Aryandono T, Lazuardi L, Dwiprahasto I. Association of overweight and obesity with breast cancer during premenopausal period in Asia: A meta-analysis. Int J Prev Med 2019; 10(1): 192.
[http://dx.doi.org/10.4103/ijpvm.IJPVM_372_18] [PMID: 31772724]
[34]
Zych M, Stec K, Bąblewska A, Pilis K. Importance of diet in reducing cancer incidence in Poland: a review. Rocz Panstw Zakl Hig 2019; 70(4): 337-45.http://wydawnictwa.pzh.gov.pl/roczniki_pzh/
[PMID: 31960665]
[35]
Vahid F, Shivappa N, Hatami M, et al. Association between dietary inflammatory index (DII) and risk of breast cancer: A case-control study. Asian Pac J Cancer Prev 2018; 19(5): 1215-21.
[PMID: 29801404]
[36]
Alexander DD, Morimoto LM, Mink PJ, Lowe KA. Summary and meta-analysis of prospective studies of animal fat intake and breast cancer. Nutr Res Rev 2010; 23(1): 169-79.
[http://dx.doi.org/10.1017/S095442241000003X] [PMID: 20181297]
[37]
Linos E, Willett WC, Cho E, Frazier L. Adolescent diet in relation to breast cancer risk among premenopausal women 2010.
[http://dx.doi.org/10.1158/1055-9965.EPI-09-0802]
[38]
Rogers IS, Northstone K, Dunger DB, Cooper AR, Ness AR, Emmett PM. Diet throughout childhood and age at menarche in a contemporary cohort of British girls. Public Health Nutr 2010; 13(12): 2052-63.
[http://dx.doi.org/10.1017/S1368980010001461] [PMID: 20529402]
[39]
Parohan M, Sadeghi A, Khatibi SR, et al. Dietary total antioxidant capacity and risk of cancer: a systematic review and meta-analysis on observational studies. Crit Rev Oncol Hematol 2019; 138: 70-86.
[http://dx.doi.org/10.1016/j.critrevonc.2019.04.003] [PMID: 31092388]
[40]
Baharvand P, Hormozi M. Can parents’ educational level and occupation affect perceived parental support and metabolic control in adolescents with type 1 diabetes? J Educ Health Promot 2019; 8: 11.
[http://dx.doi.org/10.1093/eurpub/ckx231] [PMID: 30815482]
[41]
Sadeghi M, Vahid F, Rahmani D, Akbari ME, Davoodi SH. The association between dietary patterns and breast cancer pathobiological factors progesterone receptor (pr) and estrogen receptors (ER): New findings from iranian case-control study. Nutr Cancer 2019; 71(8): 1290-8.
[http://dx.doi.org/10.1080/01635581.2019.1602658] [PMID: 31007067]
[42]
Xiao Y, Xia J, Li L, et al. Associations between dietary patterns and the risk of breast cancer: a systematic review and meta-analysis of observational studies. Breast Cancer Res 2019; 21(1): 16.
[http://dx.doi.org/10.1186/s13058-019-1096-1] [PMID: 30696460]
[43]
Dandamudi A, Tommie J, Nommsen-Rivers L, Couch S. Dietary patterns and breast cancer risk: A systematic review. Int Institute Anticancer Res 2018; 38: 3209-22.http://ar.iiarjournals.org/content/38/6/3209.full
[http://dx.doi.org/10.21873/anticanres.12586]
[44]
Steck SE, Murphy EA. Dietary patterns and cancer risk. Nat Rev Cancer 2020; 20(2): 125-38.
[http://dx.doi.org/10.1038/s41568-019-0227-4] [PMID: 31848467]
[45]
Karimi Z, Jessri M, Houshiar-Rad A, Mirzaei HR, Rashidkhani B. Dietary patterns and breast cancer risk among women. Public Health Nutr 2014; 17(5): 1098-106.
[http://dx.doi.org/10.1017/S1368980013001018] [PMID: 23651876]
[46]
Niazi M, Galehdar N, Jamshidi M, Mohammadi R, Moayyedkazemi A. A review of the role of statins in heart failure treatment. Curr Clin Pharmacol 2020; 15(1): 30-7.
[http://dx.doi.org/10.2174/1574884714666190802125627] [PMID: 31376825]
[47]
Saremi A, Bonchenari MS. Physical activity and breast cancer prevalence: A case-control study in Arak, Iran (2017-2018). Iran Q J Breast Dis 2019; 12(1): 29-38.
[http://dx.doi.org/10.30699/acadpub.ijbd.12.01.29]
[48]
Eliassen AH, Hankinson SE, Rosner B, Holmes MD, Willett WC. Physical activity and risk of breast cancer among postmenopausal women. Arch Intern Med 2010; 170(19): 1758-64.
[http://dx.doi.org/10.1001/archinternmed.2010.363] [PMID: 20975025]
[49]
Rainey L, Eriksson M, Trinh T, et al. The impact of alcohol consumption and physical activity on breast cancer: The role of breast cancer risk. Int J Cancer 2020; 147(4): 931-9.
[http://dx.doi.org/10.1002/ijc.32846] [PMID: 31863475]
[50]
Carreras G, Lachi A, Boffi R, et al. Burden of disease from breast cancer attributable to smoking and second-hand smoke exposure in Europe. Int J Cancer 2020; 147(9): 2387-93.
[http://dx.doi.org/10.1002/ijc.33021] [PMID: 32356370]
[51]
Murithi M, Ogeto TK, Walekhwa M, et al. Oral contraceptives and intrauterine devices as risk factors for breast and cervical cancers: A systematic review. Int J Res Med Sci 2020; 8(6): 8082.
[http://dx.doi.org/10.18203/2320-6012.ijrms20202294]
[52]
Ji L-W, Jing C-X, Zhuang S-L, Pan W-C, Hu X-P. Effect of age at first use of oral contraceptives on breast cancer risk: An updated meta-analysis. Medicine (Baltimore) 2019; 98(36): e15719.
[http://dx.doi.org/10.1097/MD.0000000000015719] [PMID: 31490359]
[53]
Rojas-Lima E, Gamboa-Loira B, Cebrián ME, Rothenberg SJ, López-Carrillo L. A cumulative index of exposure to endogenous estrogens and breast cancer by molecular subtypes in northern Mexican women. Breast Cancer Res Treat 2020; 180(3): 791-800.
[http://dx.doi.org/10.1007/s10549-020-05562-0] [PMID: 32086656]
[54]
Baharvand P, Malekshahi F. Relationship between anger and drug addiction potential as factors affecting the health of medical students. J Educ Health Promot 2019; 8: 157.
[PMID: 31544122]
[55]
Jerry DJ, Shull JD, Hadsell DL, et al. Genetic variation in sensitivity to estrogens and breast cancer risk. Mamm Genome 2018; 29(1-2): 24-37.
[http://dx.doi.org/10.1007/s00335-018-9741-z] [PMID: 29487996]
[56]
Nishiyama K, Taira N, Mizoo T, et al. Influence of breast density on breast cancer risk: a case control study in Japanese women. Breast Cancer 2020; 27(2): 277-83.
[http://dx.doi.org/10.1007/s12282-019-01018-6] [PMID: 31650498]
[57]
Niazi M, Veiskaramian A, Mokhayeri Y. Toward nonalcoholic fatty liver treatment: A review on herbal medicine treatment. J Crit Rev 2020; 7(3): 554-64.
[58]
Key TJ. Endogenous oestrogens and breast cancer risk in premenopausal and postmenopausal women. Steroids 2011; 76(8): 812-5.
[http://dx.doi.org/10.1016/j.steroids.2011.02.029] [PMID: 21477610]
[59]
Travis RC, Key TJ. Oestrogen exposure and breast cancer risk. Breast Cancer Res 2003; 5(5): 239-47.
[http://dx.doi.org/10.1186/bcr628] [PMID: 12927032]
[60]
Ruiz R, Herrero C, Strasser-Weippl K, et al. Epidemiology and pathophysiology of pregnancy-associated breast cancer: A review. Breast 2017; 35: 136-41.
[http://dx.doi.org/10.1016/j.breast.2017.07.008] [PMID: 28732325]
[61]
Beral V, Bull D, Doll R, Peto R, Reeves G. Breast cancer and breastfeeding: collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women without the disease. Lancet 2002; 360(9328): 187-95.
[http://dx.doi.org/10.1016/S0140-6736(02)09454-0] [PMID: 12133652]
[62]
Sharma GN, Dave R, Sanadya J, Sharma P, Sharma KK. Various types and management of breast cancer: an overview. J Adv Pharm Technol Res 2010; 1(2): 109-26.
[PMID: 22247839]
[63]
Lee K-L, Kuo Y-C, Ho Y-S, Huang Y-H. Triple-negative breast cancer: Current understanding and future therapeutic breakthrough targeting cancer stemness. Cancers (Basel) 2019; 11(9): 1334.
[http://dx.doi.org/10.3390/cancers11091334] [PMID: 31505803]
[64]
Momenimovahed Z, Salehiniya H. Epidemiological characteristics of and risk factors for breast cancer in the world. Breast Cancer (Dove Med Press) 2019; 11: 151-64.
[http://dx.doi.org/10.2147/BCTT.S176070] [PMID: 31040712]
[65]
Rana HQ, Sacca R, Drogan C, et al. Prevalence of germline variants in inflammatory breast cancer. Cancer 2019; 125(13): 2194-202.
[http://dx.doi.org/10.1002/cncr.32062] [PMID: 30933323]
[66]
Bostanci Z, Kruper L. Paget’s disease Clinical algorithms in general surgery. Cham: Springer International Publishing 2019; pp. 95-6.
[http://dx.doi.org/10.1007/978-3-319-98497-1_25]
[67]
Chen S, Chen H, Yi Y, et al. Comparative study of breast cancer with or without concomitant Paget disease: An analysis of the SEER database. Cancer Med 2019; 8(8): 4043-54.
[http://dx.doi.org/10.1002/cam4.2242] [PMID: 31134761]
[68]
Fragomeni SM, Sciallis A, Jeruss JS. Molecular subtypes and local-regional control of breast cancer. In: Surgical Oncology Clinics of North America 2018.
[http://dx.doi.org/10.1016/j.soc.2017.08.005]
[69]
Yersal O, Barutca S. Biological subtypes of breast cancer: Prognostic and therapeutic implications. World J Clin Oncol 2014; 5(3): 412-24.
[http://dx.doi.org/10.5306/wjco.v5.i3.412] [PMID: 25114856]
[70]
Kondov B, Milenkovikj Z, Kondov G, et al. Presentation of the molecular subtypes of breast cancer detected by immunohistochemistry in surgically treated patients. Open Access Maced J Med Sci 2018; 6(6): 961-7.
[http://dx.doi.org/10.3889/oamjms.2018.231] [PMID: 29983785]
[71]
Mohit E, Hashemi A, Allahyari M. Breast cancer immunotherapy: monoclonal antibodies and peptide-based vaccines. Expert Rev Clin Immunol 2014; 10(7): 927-61.
[http://dx.doi.org/10.1586/1744666X.2014.916211] [PMID: 24867051]
[72]
García-Aranda M, Redondo M. Protein kinase targets in breast cancer. Int J Mol Sci 2017; 18(12): E2543.
[http://dx.doi.org/10.3390/ijms18122543] [PMID: 29186886]
[73]
Lodi M, Scheer L, Reix N, et al. Breast cancer in elderly women and altered clinico-pathological characteristics: a systematic review. Breast Cancer Res Treat 2017; 166(3): 657-68.
[http://dx.doi.org/10.1007/s10549-017-4448-5] [PMID: 28803352]
[74]
Raichand S, Dunn AG, Ong MS, Bourgeois FT, Coiera E, Mandl KD. Conclusions in systematic reviews of mammography for breast cancer screening and associations with review design and author characteristics. Syst Rev 2017; 6(1): 105.
[http://dx.doi.org/10.1186/s13643-017-0495-6] [PMID: 28532422]
[75]
Gupta S. Data mining classification techniques applied for breast cancer diagnosis and prognosis. Indian J Comput Sci Eng 2011; 2(2): 1-8.
[76]
Anbari K, Ahmadi SAY, Baharvand P, Sahraei N. Investigation of breast cancer screening among the women of Khorramabad (West of Iran): A cross-sectional study. Epidemiol Biostat Public Health 2017; 14(1): 1-8.
[77]
Sathian B, Asim M, Mekkodathil A, James S, Mancha A, Ghosh A. Knowledge regarding breast self-examination among the women in Nepal: A meta-analysis. Nepal J Epidemiol 2019; 9(2): 761-8.
[http://dx.doi.org/10.3126/nje.v9i2.24684] [PMID: 31285936]
[78]
Oh KM, Taylor KL, Jacobsen KH. Breast cancer screening among Korean Americans: A systematic review. J Community Health 2017; 42(2): 324-32.
[http://dx.doi.org/10.1007/s10900-016-0258-7] [PMID: 27678390]
[79]
Zhu X, Huang JM, Zhang K, et al. Diagnostic value of contrast-enhanced spectral mammography for screening breast cancer: Systematic review and Meta-analysis. Clin Breast Cancer 2018; 18(5): e985-95.
[http://dx.doi.org/10.1016/j.clbc.2018.06.003] [PMID: 29983379]
[80]
Zhang X, Liu Y, Luo H, Zhang J. PETCT and MRI for identifying axillary lymph node metastases in breast cancer patients: Systematic Review and Meta‐Analysis. J Magn Reson Imaging 2020; 52(6): 1840-51.
[http://dx.doi.org/10.1002/jmri.27246] [PMID: 32567090]
[81]
Sauder CAM, Bateni SB, Davidson AJ, Nishijima DK. Breast conserving surgery compared with mastectomy in male breast cancer: A brief systematic review. Clin Breast Cancer 2020; 20(3): e309-14.
[http://dx.doi.org/10.1016/j.clbc.2019.12.004] [PMID: 32171701]
[82]
De La Cruz LM, Thiruchelvam PTR, Shivani J, Trina J, Blankenship SA, Fisher CS. Saving the male breast: A systematic literature review of breast-conservation surgery for male breast cancer. Ann Surg Oncol 2019; 26(12): 3939-44.
[http://dx.doi.org/10.1245/s10434-019-07588-1] [PMID: 31250345]
[83]
Bellavance EC, Kesmodel SB. Decision-making in the surgical treatment of breast cancer: Factors influencing women’s choices for mastectomy and breast conserving surgery. Front Oncol 2016; 6: 74.
[http://dx.doi.org/10.3389/fonc.2016.00074] [PMID: 27066455]
[84]
Riedel F, Hennigs A, Hug S, et al. Is mastectomy oncologically safer than breast-conserving treatment in early breast cancer. Breast Care (Basel) 2017; 12(6): 385-90.
[http://dx.doi.org/10.1159/000485737] [PMID: 29456470]
[85]
Davey S, Roche A, Pegba-Otemolu I, Ibrahim A, Ainsworth R. Comparison of patient satisfaction on a day-case mastectomy pathway for breast cancer versus a traditional inpatient delivery model, using a validated questionnaire. Anticancer Res 2020; 40(4): 2179-83.
[http://dx.doi.org/10.21873/anticanres.14178] [PMID: 32234912]
[86]
Harrison C, Remoundos DD, Harvey K, et al. Day Case Mastectomy: A teaching hospital experience of introducing a change in practice. Eur J Surg Oncol 2017; 43(5): S41.
[http://dx.doi.org/10.1016/j.ejso.2017.01.160]
[87]
Knabben L, Kanagalingam G, Imboden S, Günthert AR. Acellular dermal matrix (permacol®) for heterologous immediate breast reconstruction after skin-sparing mastectomy in patients with breast cancer: A single-institution experience and a review of the literature. Front Med (Lausanne) 2017; 3: 72.
[http://dx.doi.org/10.3389/fmed.2016.00072] [PMID: 28105410]
[88]
Agha RA, Wellstead G, Sagoo H, et al. Nipple sparing versus skin sparing mastectomy: A systematic review protocol. BMJ Open 2016; 6(5): e010151.
[http://dx.doi.org/10.1136/bmjopen-2015-010151] [PMID: 27207622]
[89]
Agha RA, Al Omran Y, Wellstead G, et al. Systematic review of therapeutic nipple-sparing versus skin-sparing mastectomy. BJS Open 2018; 3(2): 135-45.
[http://dx.doi.org/10.1002/bjs5.50119] [PMID: 30957059]
[90]
Galimberti V, Vicini E, Corso G, et al. Nipple-sparing and skin-sparing mastectomy: Review of aims, oncological safety and contraindications. Breast 2017; 34(Suppl. 1): S82-4.
[http://dx.doi.org/10.1016/j.breast.2017.06.034] [PMID: 28673535]
[91]
Houvenaeghel G, Bannier M, Rua S, et al. Breast cancer robotic nipple sparing mastectomy: evaluation of several surgical procedures and learning curve. World J Surg Oncol 2019; 17(1): 27.
[http://dx.doi.org/10.1186/s12957-019-1567-y] [PMID: 30728011]
[92]
Park HS, Lee J, Lee DW, et al. Robot-assisted Nipple-sparing mastectomy with immediate breast reconstruction: An initial experience. Sci Rep 2019; 9(1): 15669.
[http://dx.doi.org/10.1038/s41598-019-51744-2] [PMID: 31666551]
[93]
Tong CWS, Wu M, Cho WCS, To KKW. Recent advances in the treatment of breast cancer. Front Oncol 2018; 8: 227.
[http://dx.doi.org/10.3389/fonc.2018.00227] [PMID: 29963498]
[94]
Barroso-Sousa R, Shapiro GI, Tolaney SM. Clinical development of the CDK4/6 inhibitors ribociclib and abemaciclib in breast cancer. Breast Care (Basel) 2016; 11(3): 167-73.
[http://dx.doi.org/10.1159/000447284] [PMID: 27493615]
[95]
Di Leo A, Seok Lee K, Ciruelos E, et al. Abstract S4-07: BELLE-3: A phase III study of buparlisib + fulvestrant in postmenopausal women with HR+, HER2–, aromatase inhibitor-treated, locally advanced or metastatic breast cancer, who progressed on or after mTOR inhibitor-based treatment. American Association for Cancer Research (AACR) 2017; 04-7.
[96]
Blackwell K, Burris H, Gomez P, et al. Phase I/II dose-escalation study of PI3K inhibitors pilaralisib or voxtalisib in combination with letrozole in patients with hormone-receptor-positive and HER2-negative metastatic breast cancer refractory to a non-steroidal aromatase inhibitor. Breast Cancer Res Treat 2015; 154(2): 287-97.
[http://dx.doi.org/10.1007/s10549-015-3615-9] [PMID: 26497877]
[97]
Schmid P, Pinder SE, Wheatley D, et al. Phase II randomized preoperative window-of-opportunity study of the PI3K inhibitor pictilisib plus anastrozole compared with anastrozole alone in patients with estrogen receptor-positive breast cancer. J Clin Oncol 2016; 34(17): 1987-94.
[http://dx.doi.org/10.1200/JCO.2015.63.9179] [PMID: 26976426]
[98]
Fleming GF, Ma CX, Huo D, et al. Phase II trial of temsirolimus in patients with metastatic breast cancer. Breast Cancer Res Treat 2012; 136(2): 355-63.
[http://dx.doi.org/10.1007/s10549-011-1910-7] [PMID: 22245973]
[99]
Munster PN, Thurn KT, Thomas S, et al. A phase II study of the histone deacetylase inhibitor vorinostat combined with tamoxifen for the treatment of patients with hormone therapy-resistant breast cancer. Br J Cancer 2011; 104(12): 1828-35.
[http://dx.doi.org/10.1038/bjc.2011.156] [PMID: 21559012]
[100]
Guerin M, Rezai K, Isambert N, et al. PIKHER2: A phase IB study evaluating buparlisib in combination with lapatinib in trastuzumab-resistant HER2-positive advanced breast cancer. Eur J Cancer 2017; 86: 28-36.
[http://dx.doi.org/10.1016/j.ejca.2017.08.025] [PMID: 28950146]
[101]
Milojkovic Kerklaan B, Diéras V, Le Tourneau C, et al. Phase I study of lonafarnib (SCH66336) in combination with trastuzumab plus paclitaxel in Her2/neu overexpressing breast cancer: EORTC study 16023. Cancer Chemother Pharmacol 2013; 71(1): 53-62.
[http://dx.doi.org/10.1007/s00280-012-1972-1] [PMID: 23053259]
[102]
Lianos GD, Vlachos K, Zoras O, Katsios C, Cho WC, Roukos DH. Potential of antibody-drug conjugates and novel therapeutics in breast cancer management. OncoTargets Ther 2014; 7: 491-500.
[PMID: 24711706]
[103]
Bang YJ, Giaccone G, Im SA, et al. First-in-human phase 1 study of margetuximab (MGAH22), an Fc-modified chimeric monoclonal antibody, in patients with HER2-positive advanced solid tumors. Ann Oncol 2017; 28(4): 855-61.
[http://dx.doi.org/10.1093/annonc/mdx002] [PMID: 28119295]
[104]
FDA approves imlygic talimogene laherparepvec as first oncolytic viral therapy in the US
[105]
Bauerschmitz GJ, Ranki T, Kangasniemi L, et al. Tissue-specific promoters active in CD44+CD24-/low breast cancer cells. Cancer Res 2008; 68(14): 5533-9.
[http://dx.doi.org/10.1158/0008-5472.CAN-07-5288] [PMID: 18632604]
[106]
Koski A, Kangasniemi L, Escutenaire S, et al. Treatment of cancer patients with a serotype 5/3 chimeric oncolytic adenovirus expressing GMCSF. Mol Ther 2010; 18(10): 1874-84.
[http://dx.doi.org/10.1038/mt.2010.161] [PMID: 20664527]
[107]
Gomes EM, Rodrigues MS, Phadke AP, et al. Antitumor activity of an oncolytic adenoviral-CD40 ligand (CD154) transgene construct in human breast cancer cells. Clin Cancer Res 2009; 15(4): 1317-25.
[http://dx.doi.org/10.1158/1078-0432.CCR-08-1360] [PMID: 19228733]
[108]
Shayestehpour M, Moghim S, Salimi V, et al. Targeting human breast cancer cells by an oncolytic adenovirus using microRNA-targeting strategy. Virus Res 2017; 240: 207-14.
[http://dx.doi.org/10.1016/j.virusres.2017.08.016] [PMID: 28867494]
[109]
Hu Z, Zhang Z, Guise T, Seth P. Systemic delivery of an oncolytic adenovirus expressing soluble transforming growth factor-β receptor II-Fc fusion protein can inhibit breast cancer bone metastasis in a mouse model. Hum Gene Ther 2010; 21(11): 1623-9.
[http://dx.doi.org/10.1089/hum.2010.018] [PMID: 20712434]
[110]
Hu Z, Gerseny H, Zhang Z, et al. Oncolytic adenovirus expressing soluble TGFβ receptor II-Fc-mediated inhibition of established bone metastases: a safe and effective systemic therapeutic approach for breast cancer. Mol Ther 2011; 19(9): 1609-18.
[http://dx.doi.org/10.1038/mt.2011.114] [PMID: 21712815]
[111]
Yang Y, Xu W, Neill T, et al. Systemic delivery of an oncolytic adenovirus expressing decorin for the treatment of breast cancer bone metastases. Hum Gene Ther 2015; 26(12): 813-25.
[http://dx.doi.org/10.1089/hum.2015.098] [PMID: 26467629]
[112]
Zhang Z, Krimmel J, Zhang Z, Hu Z, Seth P. Systemic delivery of a novel liver-detargeted oncolytic adenovirus causes reduced liver toxicity but maintains the antitumor response in a breast cancer bone metastasis model. Hum Gene Ther 2011; 22(9): 1137-42.
[http://dx.doi.org/10.1089/hum.2011.003] [PMID: 21480822]
[113]
MacLeod SH, Elgadi MM, Bossi G, et al. HER3 targeting of adenovirus by fiber modification increases infection of breast cancer cells in vitro, but not following intratumoral injection in mice. Cancer Gene Ther 2012; 19(12): 888-98.
[http://dx.doi.org/10.1038/cgt.2012.79] [PMID: 23099884]
[114]
Sakr HI, Coleman DT, Cardelli JA, Mathis JM. Characterization of an oncolytic adenovirus vector constructed to target the cMet receptor. Oncolytic Virother 2015; 4: 119-32.
[http://dx.doi.org/10.2147/OV.S87369] [PMID: 26866014]
[115]
Christensen JG, Burrows J, Salgia R. c-Met as a target for human cancer and characterization of inhibitors for therapeutic intervention. Cancer Letters Cancer Lett 2005; 225: 1-26.
[116]
Platonov ME, Borovjagin AV, Kaverina N, et al. KISS1 tumor suppressor restricts angiogenesis of breast cancer brain metastases and sensitizes them to oncolytic virotherapy in vitro. Cancer Lett 2018; 417: 75-88.
[http://dx.doi.org/10.1016/j.canlet.2017.12.024] [PMID: 29269086]
[117]
Garza-Morales R, Gonzalez-Ramos R, Chiba A, et al. Temozolomide enhances triple-negative breast cancer virotherapy in vitro. Cancers (Basel) 2018; 10(5): 144.
[http://dx.doi.org/10.3390/cancers10050144] [PMID: 29772755]
[118]
Zhu W, Wei L, Zhang H, Chen J, Qin X. Oncolytic adenovirus armed with IL-24 inhibits the growth of breast cancer in vitro and in vivo. J Exp Clin Cancer Res 2012; 31(1): 51.
[http://dx.doi.org/10.1186/1756-9966-31-51] [PMID: 22640485]
[119]
Zhu W, Zhang H, Shi Y, Song M, Zhu B, Wei L. Oncolytic adenovirus encoding tumor necrosis factor-related apoptosis inducing ligand (TRAIL) inhibits the growth and metastasis of triple-negative breast cancer. Cancer Biol Ther 2013; 14(11): 1016-23.
[http://dx.doi.org/10.4161/cbt.26043] [PMID: 24025362]
[120]
Rahman M, Davis SR, Pumphrey JG, et al. TRAIL induces apoptosis in triple-negative breast cancer cells with a mesenchymal phenotype. Breast Cancer Res Treat 2009; 113(2): 217-30.
[http://dx.doi.org/10.1007/s10549-008-9924-5] [PMID: 18266105]
[121]
Menotti L, Cerretani A, Hengel H, Campadelli-Fiume G. Construction of a fully retargeted herpes simplex virus 1 recombinant capable of entering cells solely via human epidermal growth factor receptor 2. J Virol 2008; 82(20): 10153-61.
[http://dx.doi.org/10.1128/JVI.01133-08] [PMID: 18684832]
[122]
Cheng L, Jiang H, Fan J, et al. A novel oncolytic herpes simplex virus armed with the carboxyl-terminus of murine MyD116 has enhanced anti-tumour efficacy against human breast cancer cells. Oncol Lett 2018; 15(5): 7046-52.
[http://dx.doi.org/10.3892/ol.2018.8247] [PMID: 29849789]
[123]
Walker JD, Sehgal I, Kousoulas KG. Oncolytic herpes simplex virus 1 encoding 15-prostaglandin dehydrogenase mitigates immune suppression and reduces ectopic primary and metastatic breast cancer in mice. J Virol 2011; 85(14): 7363-71.
[http://dx.doi.org/10.1128/JVI.00098-11] [PMID: 21543507]
[124]
Sahin TT, Kasuya H, Nomura N, et al. Impact of novel oncolytic virus HF10 on cellular components of the tumor microenviroment in patients with recurrent breast cancer. Cancer Gene Ther 2012; 19(4): 229-37.
[http://dx.doi.org/10.1038/cgt.2011.80] [PMID: 22193629]
[125]
Nagano S, Perentes JY, Jain RK, Boucher Y. Cancer cell death enhances the penetration and efficacy of oncolytic herpes simplex virus in tumors. Cancer Res 2008; 68(10): 3795-802.
[http://dx.doi.org/10.1158/0008-5472.CAN-07-6193] [PMID: 18483263]
[126]
Cody JJ, Markert JM, Hurst DR. Histone deacetylase inhibitors improve the replication of oncolytic herpes simplex virus in breast cancer cells. PLoS One 2014; 9(3): e92919.
[http://dx.doi.org/10.1371/journal.pone.0092919] [PMID: 24651853]
[127]
Zhuang X, Zhang W, Chen Y, et al. Doxorubicin-enriched, ALDH(br) mouse breast cancer stem cells are treatable to oncolytic herpes simplex virus type 1. BMC Cancer 2012; 12: 549.
[http://dx.doi.org/10.1186/1471-2407-12-549] [PMID: 23176143]
[128]
Chen X, Han J, Chu J, et al. A combinational therapy of EGFR-CAR NK cells and oncolytic herpes simplex virus 1 for breast cancer brain metastases. Oncotarget 2016; 7(19): 27764-77.
[http://dx.doi.org/10.18632/oncotarget.8526] [PMID: 27050072]
[129]
Gentschev I, Stritzker J, Hofmann E, et al. Use of an oncolytic vaccinia virus for the treatment of canine breast cancer in nude mice: preclinical development of a therapeutic agent. Cancer Gene Ther 2009; 16(4): 320-8.
[http://dx.doi.org/10.1038/cgt.2008.87] [PMID: 18949014]
[130]
Gholami S, Marano A, Chen NG, et al. A novel vaccinia virus with dual oncolytic and anti-angiogenic therapeutic effects against triple-negative breast cancer. Breast Cancer Res Treat 2014; 148(3): 489-99.
[http://dx.doi.org/10.1007/s10549-014-3180-7] [PMID: 25391896]
[131]
John LB, Howland LJ, Flynn JK, et al. Oncolytic virus and anti-4-1BB combination therapy elicits strong antitumor immunity against established cancer. Cancer Res 2012; 72(7): 1651-60.
[http://dx.doi.org/10.1158/0008-5472.CAN-11-2788] [PMID: 22315352]
[132]
Mostafa AA, Meyers DE, Thirukkumaran CM, et al. Oncolytic reovirus and immune checkpoint inhibition as a novel immunotherapeutic strategy for breast cancer. Cancers (Basel) 2018; 10(6): 1-18.
[http://dx.doi.org/10.3390/cancers10060205] [PMID: 29914097]
[133]
Skelding KA, Barry RD, Shafren DR. Systemic targeting of metastatic human breast tumor xenografts by Coxsackievirus A21. Breast Cancer Res Treat 2009; 113(1): 21-30.
[http://dx.doi.org/10.1007/s10549-008-9899-2] [PMID: 18256929]
[134]
Miyamoto S, Inoue H, Nakamura T, et al. Coxsackievirus B3 is an oncolytic virus with immunostimulatory properties that is active against lung adenocarcinoma. Cancer Res 2012; 72(10): 2609-21.
[http://dx.doi.org/10.1158/0008-5472.CAN-11-3185] [PMID: 22461509]
[135]
Holl EK, Brown MC, Boczkowski D, et al. Recombinant oncolytic poliovirus, PVSRIPO, has potent cytotoxic and innate inflammatory effects, mediating therapy in human breast and prostate cancer xenograft models. Oncotarget 2016; 7(48): 79828-41.
[http://dx.doi.org/10.18632/oncotarget.12975] [PMID: 27806313]
[136]
Kaur S, Kaur S. Bacteriocins as potential anticancer agents. Front Pharmacol 2015; 6: 272.
[http://dx.doi.org/10.3389/fphar.2015.00272] [PMID: 26617524]
[137]
Paiva AD, de Oliveira MD, de Paula SO, Baracat-Pereira MC, Breukink E, Mantovani HC. Toxicity of bovicin HC5 against mammalian cell lines and the role of cholesterol in bacteriocin activity. 2012.
[http://dx.doi.org/10.1099/mic.0.062190-0]
[138]
Chumchalová J, Šmarda J. Human tumor cells are selectively inhibited by colicins. Folia Microbiol (Praha) 2003; 48(1): 111-5.
[http://dx.doi.org/10.1007/BF02931286] [PMID: 12744087]
[139]
Baindara P, Gautam A, Raghava GPS, Korpole S. Anticancer properties of a defensin like class IId bacteriocin Laterosporulin10. Sci Rep 2017; 7(1): 46541.
[http://dx.doi.org/10.1038/srep46541] [PMID: 28422156]
[140]
Begde D, Bundale S, Mashitha P, Rudra J, Nashikkar N, Upadhyay A. Immunomodulatory efficacy of nisin--a bacterial lantibiotic peptide. J Pept Sci 2011; 17(6): 438-44.
[http://dx.doi.org/10.1002/psc.1341] [PMID: 21294231]
[141]
Um S, Choi TJ, Kim H, et al. Ohmyungsamycins A and B: cytotoxic and antimicrobial cyclic peptides produced by Streptomyces sp. from a volcanic island. J Org Chem 2013; 78(24): 12321-9.
[http://dx.doi.org/10.1021/jo401974g] [PMID: 24266328]
[142]
Mohamed MS, Fattah SA, Mostafa HM. azurin as antitumor protein and its effect on the cancer cell lines. Curr Res J Biol Sci 2010; 2(6): 396-401.
[143]
Gao M, Zhou J, Su Z, Huang Y. Bacterial cupredoxin azurin hijacks cellular signaling networks: Protein-protein interactions and cancer therapy. Protein Sci 2017; 26(12): 2334-41.
[http://dx.doi.org/10.1002/pro.3310] [PMID: 28960574]
[144]
Sung WS, Park Y, Choi C-H, Hahm K-S, Lee DG. Mode of antibacterial action of a signal peptide, Pep27 from Streptococcus pneumoniae. Biochem Biophys Res Commun 2007; 363(3): 806-10.
[http://dx.doi.org/10.1016/j.bbrc.2007.09.041] [PMID: 17900534]
[145]
Karpiński TM, Adamczak A. Anticancer activity of bacterial proteins and peptides. Pharmaceutics 2018; 10(2): 54.
[http://dx.doi.org/10.3390/pharmaceutics10020054] [PMID: 29710857]
[146]
Fiedler HP, Bruntner C, Riedlinger J, et al. Proximicin A, B and C, novel aminofuran antibiotic and anticancer compounds isolated from marine strains of the actinomycete Verrucosispora. J Antibiot (Tokyo) 2008; 61(3): 158-63.
[http://dx.doi.org/10.1038/ja.2008.125] [PMID: 18503194]
[147]
Matsuo Y, Kanoh K, Yamori T, et al. Urukthapelstatin A, a novel cytotoxic substance from marine-derived Mechercharimyces asporophorigenens YM11-542. I. Fermentation, isolation and biological activities. J Antibiot (Tokyo) 2007; 60(4): 251-5.
[http://dx.doi.org/10.1038/ja.2007.30] [PMID: 17456975]
[148]
Lewis DJ, Dao H Jr, Nagarajan P, Duvic M. Primary cutaneous anaplastic large-cell lymphoma: Complete remission for 13 years after denileukin diftitox. JAAD Case Rep 2017; 3(6): 501-4.
[http://dx.doi.org/10.1016/j.jdcr.2017.06.031] [PMID: 29296637]
[149]
Lutz MB, Baur AS, Schuler-Thurner B, Schuler G. Immunogenic and tolerogenic effects of the chimeric IL-2-diphtheria toxin cytocidal agent Ontak® on CD25+ cells. OncoImmunology 2014; 3(3): e28223.
[http://dx.doi.org/10.4161/onci.28223] [PMID: 25050193]
[150]
Bandala C, Cortés-Algara AL, Mejía-Barradas CM, et al. Botulinum neurotoxin type A inhibits synaptic vesicle 2 expression in breast cancer cell lines. Int J Clin Exp Pathol 2015; 8(7): 8411-8.
[PMID: 26339411]
[151]
Hemmati M, Tarighi P, Amoozadeh S, Farajollahi MM. Expression and purification of the recombinant pseudomonas exotoxin a conjugated to herceptin and its anti-proliferation effects on Sk-Br-3 2017.
[http://dx.doi.org/10.21859/mci-supp-33]
[152]
Goldufsky J, Wood S, Hajihossainlou B, et al. Pseudomonas aeruginosa exotoxin T induces potent cytotoxicity against a variety of murine and human cancer cell lines. J Med Microbiol 2015; 64(Pt 2): 164-73.
[http://dx.doi.org/10.1099/jmm.0.000003] [PMID: 25627204]
[153]
Lee JH, Moore LD, Kumar S, Pritchard DG, Ponnazhagan S, Deivanayagam C. Bacteriophage hyaluronidase effectively inhibits growth, migration and invasion by disrupting hyaluronan-mediated Erk1/2 activation and RhoA expression in human breast carcinoma cells. Cancer Lett 2010; 298(2): 238-49.
[http://dx.doi.org/10.1016/j.canlet.2010.07.011] [PMID: 20688428]
[154]
Zheng JH, Min J-J. Targeted cancer therapy using engineered Salmonella typhimurium. Chonnam Med J 2016; 52(3): 173-84.
[http://dx.doi.org/10.4068/cmj.2016.52.3.173] [PMID: 27689027]
[155]
Gunn GR, Zubair A, Peters C, Pan Z-K, Wu T-C, Paterson Y. Two Listeria monocytogenes vaccine vectors that express different molecular forms of human papilloma virus-16 (HPV-16) E7 induce qualitatively different T cell immunity that correlates with their ability to induce regression of established tumors immortalized by HPV-16. J Immunol 2001; 167(11): 6471-9.
[http://dx.doi.org/10.4049/jimmunol.167.11.6471] [PMID: 11714814]
[156]
Shahabi V, Seavey MM, Maciag PC, Rivera S, Wallecha A. Development of a live and highly attenuated Listeria monocytogenes-based vaccine for the treatment of Her2/neu-overexpressing cancers in human. Cancer Gene Ther 2011; 18(1): 53-62.
[http://dx.doi.org/10.1038/cgt.2010.48] [PMID: 20725099]
[157]
Asoudeh-Fard A, Barzegari A, Dehnad A, Bastani S, Golchin A, Omidi Y. Lactobacillus plantarum induces apoptosis in oral cancer KB cells through upregulation of PTEN and downregulation of MAPK signalling pathways. Bioimpacts 2017; 7(3): 193-8.
[http://dx.doi.org/10.15171/bi.2017.22] [PMID: 29159146]
[158]
Albalawi AE, Alanazi AD, Baharvand P, Sepahvand M, Mahmoudvand H. High potency of organic and inorganic nanoparticles to treat cystic Echinococcosis: An evidence-based review. Nanomaterials (Basel) 2020; 10(12): 2538.
[http://dx.doi.org/10.3390/nano10122538] [PMID: 33348662]
[159]
Hawkins MJ, Soon-Shiong P, Desai N. Protein nanoparticles as drug carriers in clinical medicine. Adv Drug Deliv Rev 2008; 60(8): 876-85.
[http://dx.doi.org/10.1016/j.addr.2007.08.044] [PMID: 18423779]
[160]
Clinical trials | The Perth Blood Institute Limited.
[161]
Caraglia M, De Rosa G, Salzano G, et al. Nanotech revolution for the anti-cancer drug delivery through blood-brain barrier. Curr Cancer Drug Targets 2012; 12(3): 186-96.
[http://dx.doi.org/10.2174/156800912799277421] [PMID: 22268384]
[162]
Lin Z, Liu Y, Ma X, et al. Photothermal ablation of bone metastasis of breast cancer using PEGylated multi-walled carbon nanotubes. Sci Rep 2015; 5: 11709.
[http://dx.doi.org/10.1038/srep11709] [PMID: 26122018]
[163]
Judge AD, Sood V, Shaw JR, Fang D, McClintock K, MacLachlan I. Sequence-dependent stimulation of the mammalian innate immune response by synthetic siRNA. Nat Biotechnol 2005; 23(4): 457-62.
[http://dx.doi.org/10.1038/nbt1081] [PMID: 15778705]
[164]
Alam S, Bowser BS, Israr M, Conway MJ, Meyers C. Adeno-associated virus type 2 infection of nude mouse human breast cancer xenograft induces necrotic death and inhibits tumor growth. Cancer Biol Ther 2014; 15(8): 1013-28.
[http://dx.doi.org/10.4161/cbt.29172] [PMID: 24834917]
[165]
Lei N, Gong C, Qian Z, et al. Therapeutic application of injectable thermosensitive hydrogel in preventing local breast cancer recurrence and improving incision wound healing in a mouse model. Nanoscale 2012; 4(18): 5686-93.
[http://dx.doi.org/10.1039/c2nr30731f] [PMID: 22875402]
[166]
Lin Z, Xu S, Gao W, et al. A comparative investigation between paclitaxel nanoparticle- and nanocrystal-loaded thermosensitive PECT hydrogels for peri-tumoural administration. Nanoscale 2016; 8(44): 18782-91.
[http://dx.doi.org/10.1039/C6NR05498F] [PMID: 27801924]
[167]
Fathi M, Alami-Milani M, Geranmayeh MH, Barar J, Erfan-Niya H, Omidi Y. Dual thermo-and pH-sensitive injectable hydrogels of chitosan/(poly(N-isopropylacrylamide-co-itaconic acid)) for doxorubicin delivery in breast cancer. Int J Biol Macromol 2019; 128: 957-64.
[http://dx.doi.org/10.1016/j.ijbiomac.2019.01.122] [PMID: 30685304]
[168]
Liu Z, Xu G, Wang C, Li C, Yao P. Shear-responsive injectable supramolecular hydrogel releasing doxorubicin loaded micelles with pH-sensitivity for local tumor chemotherapy. Int J Pharm 2017; 530(1-2): 53-62.
[http://dx.doi.org/10.1016/j.ijpharm.2017.07.063] [PMID: 28739501]
[169]
Johnson R, Sabnis N, McConathy WJ, Lacko AG. The potential role of nanotechnology in therapeutic approaches for triple negative breast cancer. Pharmaceutics 2013; 5(2): 353-70.
[http://dx.doi.org/10.3390/pharmaceutics5020353] [PMID: 24244833]
[170]
Senkus E, Kyriakides S, Ohno S, et al. Primary breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2015; 26(Suppl. 5): v8-v30.
[http://dx.doi.org/10.1093/annonc/mdv298] [PMID: 26314782]
[171]
Talluri SV, Kuppusamy G, Karri VVSR, Tummala S, Madhunapantula SV. Lipid-based nanocarriers for breast cancer treatment - comprehensive review. Drug Deliv 2016; 23(4): 1291-305.
[http://dx.doi.org/10.3109/10717544.2015.1092183] [PMID: 26430913]
[172]
Regina F, Rocha G, Molon RS, et al. Use of salivary biomarkers for diagnosis of periodontal disease activity: A literature review. World J Dent 2013; 4(4): 250-5.
[http://dx.doi.org/10.5005/jp-journals-10015-1240]
[173]
Vogel CL, Cobleigh MA, Tripathy D, et al. Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J Clin Oncol 2002; 20(3): 719-26.
[http://dx.doi.org/10.1200/JCO.2002.20.3.719] [PMID: 11821453]
[174]
Tolaney SM, Krop IE. Mechanisms of trastuzumab resistance in breast cancer. Anticancer Agents Med Chem 2009; 9(3): 348-55.
[http://dx.doi.org/10.2174/1871520610909030348] [PMID: 19275526]
[175]
Cortés J, Fumoleau P, Bianchi GV, et al. Pertuzumab monotherapy after trastuzumab-based treatment and subsequent reintroduction of trastuzumab: activity and tolerability in patients with advanced human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol 2012; 30(14): 1594-600.
[http://dx.doi.org/10.1200/JCO.2011.37.4207] [PMID: 22393084]
[176]
Baselga J, Gelmon KA, Verma S, et al. Phase II trial of pertuzumab and trastuzumab in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer that progressed during prior trastuzumab therapy. J Clin Oncol 2010; 28(7): 1138-44.
[http://dx.doi.org/10.1200/JCO.2009.24.2024] [PMID: 20124182]
[177]
Peddi PF, Hurvitz SA. Trastuzumab emtansine: the first targeted chemotherapy for treatment of breast cancer. Future Oncol 2013; 9(3): 319-26.
[http://dx.doi.org/10.2217/fon.13.7] [PMID: 23469968]
[178]
Kaufman B, Trudeau M, Awada A, et al. Lapatinib monotherapy in patients with HER2-overexpressing relapsed or refractory inflammatory breast cancer: final results and survival of the expanded HER2+ cohort in EGF103009, a phase II study. Lancet Oncol 2009; 10(6): 581-8.
[http://dx.doi.org/10.1016/S1470-2045(09)70087-7] [PMID: 19394894]
[179]
Tao Z, Li SX, Shen K, Zhao Y, Zeng H, Ma X. Safety and efficacy profile of Neratinib: A systematic review and meta-analysis of 23 prospective clinical trials. Clin Drug Investig 2019; 39(1): 27-43.
[http://dx.doi.org/10.1007/s40261-018-0719-0] [PMID: 30370488]
[180]
Baselga J, Albanell J, Ruiz A, et al. Phase II and tumor pharmacodynamic study of gefitinib in patients with advanced breast cancer. J Clin Oncol 2005; 23(23): 5323-33.
[http://dx.doi.org/10.1200/JCO.2005.08.326] [PMID: 15939921]
[181]
Lin NU, Winer EP, Wheatley D, et al. A phase II study of afatinib (BIBW 2992), an irreversible ErbB family blocker, in patients with HER2-positive metastatic breast cancer progressing after trastuzumab. Breast Cancer Res Treat 2012; 133(3): 1057-65.
[http://dx.doi.org/10.1007/s10549-012-2003-y] [PMID: 22418700]
[182]
Mahaddalkar T, Lopus M. From natural products to designer drugs: Development and molecular mechanisms action of novel anti-microtubule breast cancer therapeutics. Curr Top Med Chem 2017; 17(22): 2559-68.
[http://dx.doi.org/10.2174/1568026617666170104144240] [PMID: 28056739]
[183]
Bishayee A, Sethi G. Bioactive natural products in cancer prevention and therapy: Progress and promise. Semin Cancer Biol 2016; 40-41: 1-3.
[http://dx.doi.org/10.1016/j.semcancer.2016.08.006] [PMID: 27565447]
[184]
Degner SC, Papoutsis AJ, Selmin O, Romagnolo DF. Targeting of aryl hydrocarbon receptor-mediated activation of cyclooxygenase-2 expression by the indole-3-carbinol metabolite 3,3′-diindolylmethane in breast cancer cells. J Nutr 2009; 139(1): 26-32.
[http://dx.doi.org/10.3945/jn.108.099259] [PMID: 19056653]
[185]
Riby JE, Firestone GL, Bjeldanes LF. 3,3′-diindolylmethane reduces levels of HIF-1α and HIF-1 activity in hypoxic cultured human cancer cells. Biochem Pharmacol 2008; 75(9): 1858-67.
[http://dx.doi.org/10.1016/j.bcp.2008.01.017] [PMID: 18329003]
[186]
Licznerska BE, Szaefer H, Murias M, Bartoszek A, Baer-Dubowska W. Modulation of CYP19 expression by cabbage juices and their active components: indole-3-carbinol and 3,3′-diindolylmethene in human breast epithelial cell lines. Eur J Nutr 2013; 52(5): 1483-92.
[http://dx.doi.org/10.1007/s00394-012-0455-9] [PMID: 23090135]
[187]
Thomson CA, Chow HHS, Wertheim BC, et al. A randomized, placebo-controlled trial of diindolylmethane for breast cancer biomarker modulation in patients taking tamoxifen. Breast Cancer Res Treat 2017; 165(1): 97-107.
[http://dx.doi.org/10.1007/s10549-017-4292-7] [PMID: 28560655]
[188]
Chan HY, Wang H, Leung LK. The red clover (Trifolium pratense) isoflavone biochanin A modulates the biotransformation pathways of 7,12-dimethylbenz[a]anthracene. Br J Nutr 2003; 90(1): 87-92.
[http://dx.doi.org/10.1079/BJN2003868] [PMID: 12844379]
[189]
Sehdev V, Lai JC, Bhushan A, Sehdev V, Lai JCK. Biochanin A modulates cell viability, invasion, and growth promoting signaling pathways in HER-2-positive breast cancer cells. J Oncol 2009; 2009: 121458.
[http://dx.doi.org/10.1155/2009/121458] [PMID: 20169097]
[190]
Liu Q, Loo WTY, Sze SCW, Tong Y. Curcumin inhibits cell proliferation of MDA-MB-231 and BT-483 breast cancer cells mediated by down-regulation of NFkappaB, cyclinD and MMP-1 transcription. Phytomedicine 2009; 16(10): 916-22.
[http://dx.doi.org/10.1016/j.phymed.2009.04.008] [PMID: 19524420]
[191]
Kakarala M, Brenner DE, Korkaya H, et al. Targeting breast stem cells with the cancer preventive compounds curcumin and piperine. Breast Cancer Res Treat 2010; 122(3): 777-85.
[http://dx.doi.org/10.1007/s10549-009-0612-x] [PMID: 19898931]
[192]
Chen Y, Shu W, Chen W, Wu Q, Liu H, Cui G. Curcumin, both histone deacetylase and p300/CBP-specific inhibitor, represses the activity of nuclear factor kappa B and Notch 1 in Raji cells. Basic Clin Pharmacol Toxicol 2007; 101(6): 427-33.
[http://dx.doi.org/10.1111/j.1742-7843.2007.00142.x] [PMID: 17927689]
[193]
Yang J, Cao Y, Sun J, Zhang Y. Curcumin reduces the expression of Bcl-2 by upregulating miR-15a and miR-16 in MCF-7 cells. Med Oncol 2010; 27(4): 1114-8.
[http://dx.doi.org/10.1007/s12032-009-9344-3] [PMID: 19908170]
[194]
Limtrakul P, Chearwae W, Shukla S, Phisalphong C, Ambudkar SV. Modulation of function of three ABC drug transporters, P-glycoprotein (ABCB1), mitoxantrone resistance protein (ABCG2) and multidrug resistance protein 1 (ABCC1) by tetrahydrocurcumin, a major metabolite of curcumin. Mol Cell Biochem 2007; 296(1-2): 85-95.
[http://dx.doi.org/10.1007/s11010-006-9302-8] [PMID: 16960658]
[195]
Huang PH, Huang CY, Chen MC, et al. Emodin and Aloe-Emodin suppress breast cancer cell proliferation through ER α inhibition. Evid Based Complement Alternat Med 2013; 2013: 376123.
[http://dx.doi.org/10.1155/2013/376123] [PMID: 23864887]
[196]
Nandakumar V, Vaid M, Katiyar SK. (-)-Epigallocatechin-3-gallate reactivates silenced tumor suppressor genes, Cip1/p21 and p16INK4a, by reducing DNA methylation and increasing histones acetylation in human skin cancer cells. Carcinogenesis 2011; 32(4): 537-44.
[http://dx.doi.org/10.1093/carcin/bgq285] [PMID: 21209038]
[197]
Han SG, Han SS, Toborek M, Hennig B. EGCG protects endothelial cells against PCB 126-induced inflammation through inhibition of AhR and induction of Nrf2-regulated genes. Toxicol Appl Pharmacol 2012; 261(2): 181-8.https://miami.pure.elsevier.com/en/publications/egcg-protects-endothelial-cells-against-pcb-126-induced-inflammat
[http://dx.doi.org/10.1016/j.taap.2012.03.024] [PMID: 22521609]
[198]
Hong OY, Noh EM, Jang HY, et al. Epigallocatechin gallate inhibits the growth of MDA-MB-231 breast cancer cells via inactivation of the β-catenin signaling pathway. Oncol Lett 2017; 14(1): 441-6.https://europepmc.org/articles/PMC5494649
[http://dx.doi.org/10.3892/ol.2017.6108] [PMID: 28693189]
[199]
Farabegoli F, Govoni M, Spisni E, Papi A. EGFR inhibition by (-)-epigallocatechin-3-gallate and IIF treatments reduces breast cancer cell invasion. Biosci Rep 2017; 37(3): 20170168.https://portlandpress.com/bioscirep/article-pdf/37/3/BSR20170168/430362/bsr-2017-0168.pdf
[http://dx.doi.org/10.1042/BSR20170168] [PMID: 28465354]
[200]
Li MJ, Yin YC, Wang J, Jiang YF. Green tea compounds in breast cancer prevention and treatment. World J Clin Oncol 2014; 5(3): 520-8.
[http://dx.doi.org/10.5306/wjco.v5.i3.520] [PMID: 25114865]
[201]
Mitra S, Dash R. Natural products for the management and prevention of breast cancer. Evid Based Complement Alternat Med 2018; 2018: 8324696.
[http://dx.doi.org/10.1155/2018/8324696] [PMID: 29681985]
[202]
Qiao J, Gu C, Shang W, et al. Effect of green tea on pharmacokinetics of 5-fluorouracil in rats and pharmacodynamics in human cell lines in vitro. Food Chem Toxicol 2011; 49(6): 1410-5.
[http://dx.doi.org/10.1016/j.fct.2011.03.033] [PMID: 21440026]
[203]
Stearns ME, Amatangelo MD, Varma D, Sell C, Goodyear SM. Combination therapy with epigallocatechin-3-gallate and doxorubicin in human prostate tumor modeling studies: inhibition of metastatic tumor growth in severe combined immunodeficiency mice. Am J Pathol 2010; 177(6): 3169-79.
[http://dx.doi.org/10.2353/ajpath.2010.100330] [PMID: 20971741]
[204]
Dharmappa KK, Mohamed R, Shivaprasad HV, Vishwanath BS. Genistein, a potent inhibitor of secretory phospholipase A2: a new insight in down regulation of inflammation. Inflammopharmacology 2010; 18(1): 25-31.
[http://dx.doi.org/10.1007/s10787-009-0018-8] [PMID: 19894024]
[205]
Chung M-H, Kim D-H, Na H-K, et al. Genistein inhibits phorbol ester-induced NF-κB transcriptional activity and COX-2 expression by blocking the phosphorylation of p65/RelA in human mammary epithelial cells. Mutat Res Fundam Mol Mech Mutagen 2014; 768(C): 74-83.https://linkinghub.elsevier.com/retrieve/pii/S0027510714000694
[http://dx.doi.org/10.1016/j.mrfmmm.2014.04.003] [PMID: 24742714]
[206]
Pons DG, Nadal-Serrano M, Blanquer-Rossello MM, Sastre-Serra J, Oliver J, Roca P. Genistein modulates proliferation and mitochondrial functionality in breast cancer cells depending on ERalpha/ERbeta ratio. J Cell Biochem 2014; 115(5): 949-58.
[http://dx.doi.org/10.1002/jcb.24737] [PMID: 24375531]
[207]
Chen J, Duan Y, Zhang X, Ye Y, Ge B, Chen J. Genistein induces apoptosis by the inactivation of the IGF-1R/p-Akt signaling pathway in MCF-7 human breast cancer cells. Food Funct 2015; 6(3): 995-1000.
[http://dx.doi.org/10.1039/C4FO01141D] [PMID: 25675448]
[208]
Xie Q, Bai Q, Zou LY, et al. Genistein inhibits DNA methylation and increases expression of tumor suppressor genes in human breast cancer cells. Genes Chromosomes Cancer 2014; 53(5): 422-31.
[http://dx.doi.org/10.1002/gcc.22154] [PMID: 24532317]
[209]
Tominaga Y, Wang A, Wang RH, Wang X, Cao L, Deng CX. Genistein inhibits Brca1 mutant tumor growth through activation of DNA damage checkpoints, cell cycle arrest, and mitotic catastrophe. Cell Death Differ 2007; 14(3): 472-9.www.nature.com/cdd
[http://dx.doi.org/10.1038/sj.cdd.4402037] [PMID: 17024228]
[210]
Takeshima M, Ono M, Higuchi T, Chen C, Hara T, Nakano S. Anti-proliferative and apoptosis-inducing activity of lycopene against three subtypes of human breast cancer cell lines. Cancer Sci 2014; 105(3): 252-7.
[http://dx.doi.org/10.1111/cas.12349] [PMID: 24397737]
[211]
Peng SJ, Li J, Zhou Y, et al. In vitro effects and mechanisms of lycopene in MCF-7 human breast cancer cells. Genet Mol Res 2017; 16(2): 16029434.https://pubmed.ncbi.nlm.nih.gov/28407181/
[212]
Pledgie-Tracy A, Sobolewski MD, Davidson NE. Sulforaphane induces cell type-specific apoptosis in human breast cancer cell lines. Mol Cancer Ther 2007; 6(3): 1013-21.
[http://dx.doi.org/10.1158/1535-7163.MCT-06-0494] [PMID: 17339367]
[213]
Kim SH, Park HJ, Moon DO. Sulforaphane sensitizes human breast cancer cells to paclitaxel-induced apoptosis by downregulating the NF-κB signaling pathway. Oncol Lett 2017; 13(6): 4427-32.
[http://dx.doi.org/10.3892/ol.2017.5950] [PMID: 28599444]
[214]
Lewinska A, Bednarz D, Adamczyk-Grochala J, Wnuk M. Phytochemical-induced nucleolar stress results in the inhibition of breast cancer cell proliferation. Redox Biol 2017; 12: 469-82.
[http://dx.doi.org/10.1016/j.redox.2017.03.014] [PMID: 28334682]
[215]
Li Y, Meeran SM, Tollefsbol TO. Combinatorial bioactive botanicals re-sensitize tamoxifen treatment in ER-negative breast cancer via epigenetic reactivation of ERα expression. Sci Rep 2017; 7(1): 1-15.www.nature.com/scientificreports/
[http://dx.doi.org/10.1038/s41598-017-09764-3] [PMID: 28839265]
[216]
Kim HN, Kim DH, Kim EH, et al. Sulforaphane inhibits phorbol ester-stimulated IKK-NF-κB signaling and COX-2 expression in human mammary epithelial cells by targeting NF-κB activating kinase and ERK. Cancer Lett 2014; 351(1): 41-9.
[http://dx.doi.org/10.1016/j.canlet.2014.03.037] [PMID: 24747121]
[217]
Li Q, Yao Y, Eades G, Liu Z, Zhang Y, Zhou Q. Downregulation of miR-140 promotes cancer stem cell formation in basal-like early stage breast cancer. Oncogene 2014; 33(20): 2589-600.
[http://dx.doi.org/10.1038/onc.2013.226] [PMID: 23752191]
[218]
Li Y, Zhang T, Korkaya H, et al. Sulforaphane, a dietary component of broccoli/broccoli sprouts, inhibits breast cancer stem cells. Clin Cancer Res 2010; 16(9): 2580-90.
[http://dx.doi.org/10.1158/1078-0432.CCR-09-2937] [PMID: 20388854]
[219]
Zhang CH, Wang J, Zhang LX, et al. Sulforaphane, a dietary component of broccoli/broccoli sprouts, inhibits breast cancer stem cells. Clin Cancer Res 2017; 16(9): 2580-90.
[220]
Li W, Liu J, Jackson K, Shi R, Zhao Y. Sensitizing the therapeutic efficacy of taxol with shikonin in human breast cancer cells. PLoS One 2014; 9(4): e94079.
[http://dx.doi.org/10.1371/journal.pone.0094079] [PMID: 24710512]
[221]
Duru N, Gernapudi R, Zhou Q. Chemopreventive activities of shikonin in breast cancer. Biochem Pharmacol 2014; 3: 163.
[222]
Lu W, Lin C, King TD, Chen H, Reynolds RC, Li Y. Silibinin inhibits Wnt/β-catenin signaling by suppressing Wnt co-receptor LRP6 expression in human prostate and breast cancer cells. Cell Signal 2012; 24(12): 2291-6.
[http://dx.doi.org/10.1016/j.cellsig.2012.07.009] [PMID: 22820499]
[223]
Kim S, Kim SH, Hur SM, et al. Silibinin prevents TPA-induced MMP-9 expression by down-regulation of COX-2 in human breast cancer cells. J Ethnopharmacol 2009; 126(2): 252-7.
[http://dx.doi.org/10.1016/j.jep.2009.08.032] [PMID: 19715751]
[224]
Molavi O, Narimani F, Asiaee F, et al. Silibinin sensitizes chemo-resistant breast cancer cells to chemotherapy. Pharm Biol 2017; 55(1): 729-39.
[http://dx.doi.org/10.1080/13880209.2016.1270972] [PMID: 28027688]
[225]
Chavoshi H, Vahedian V, Saghaei S, Pirouzpanah MB, Raeisi M, Samadi N. Adjuvant therapy with silibinin improves the efficacy of paclitaxel and cisplatin in MCF-7 breast cancer cells. Asian Pac J Cancer Prev 2017; 18(8): 2243-7.
[PMID: 28843263]
[226]
Wang H, Khor TO, Shu L, et al. Plants vs. cancer: a review on natural phytochemicals in preventing and treating cancers and their druggability. Anticancer Agents Med Chem 2012; 12(10): 1281-305.https://www.researchwithrutgers.com/en/publications/plants-vs-cancer-a-review-on-natural-phytochemicals-in-preventing
[http://dx.doi.org/10.2174/187152012803833026] [PMID: 22583408]
[227]
Elsayed HE, Ebrahim HY, Mohyeldin MM, et al. Rutin as A novel c-Met inhibitory lead for the control of triple negative breast malignancies. Nutr Cancer 2017; 69(8): 1256-71.
[http://dx.doi.org/10.1080/01635581.2017.1367936] [PMID: 29083228]
[228]
Iriti M, Kubina R, Cochis A, et al. Rutin, a quercetin glycoside, restores chemosensitivity in human breast cancer cells. Phyther Res 2017; 31(10): 1529-38.
[http://dx.doi.org/10.1002/ptr.5878]
[229]
American Cancer Society. Breast cancer occurrence 3 breast cancer risk factors 12 what is the american cancer society doing about breast cancer. 2020.
[230]
Yin SY, Wei WC, Jian FY, Yang NS. Therapeutic applications of herbal medicines for cancer patients. Evid Based Complement Alternat Med 2013; 2013: 302426.
[http://dx.doi.org/10.1155/2013/302426] [PMID: 23956768]
[231]
Yamamoto S, Sobue T, Kobayashi M, Sasaki S, Tsugane S. Soy, isoflavones, and breast cancer risk in Japan. J Natl Cancer Inst 2003; 95(12): 906-13.
[http://dx.doi.org/10.1093/jnci/95.12.906] [PMID: 12813174]
[232]
Gerber B, Scholz C, Reimer T, Briese V, Janni W. Complementary and alternative therapeutic approaches in patients with early breast cancer: a systematic review. Breast Cancer Res Treat 2006; 95(3): 199-209.
[http://dx.doi.org/10.1007/s10549-005-9005-y] [PMID: 16254687]
[233]
Lesperance ML, Olivotto IA, Forde N, et al. Mega-dose vitamins and minerals in the treatment of non-metastatic breast cancer: an historical cohort study. Breast Cancer Res Treat 2002; 76(2): 137-43.
[http://dx.doi.org/10.1023/A:1020552501345] [PMID: 12452451]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy