1
|
Application of a fluorescent H 2S probe based on excited-state intramolecular proton transfer for detecting latent mechanism of H 2S-induced MCF-7 apoptosis. Future Med Chem 2022; 14:647-663. [PMID: 35383482 DOI: 10.4155/fmc-2021-0309] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: H2S is the third gas transmitter affecting the growth, reproduction and survival of cancer cells. However, the H2S anticancer and antitumor mechanism still needs to be further studied. Methods: Here, FHS-1 was synthesized utilizing excited-state intramolecular proton transfer to detect H2S in MCF-7 cells, and investigated the effects of varying concentrations NaHS on apoptosis. Results: The study found that FHS-1 detects H2S levels with high selectivity and pH stability and that H2S may regulate apoptosis in MCF-7 cells through the p53/mTOR/STAT3 pathway. Conclusion: Researching the influence of H2S on apoptosis can serve as a theoretical foundation for future research into H2S-related anticancer medicines, and the H2S probe can be used as an effective cancer screening tool.
Collapse
|
2
|
Torabi Dalivandan S, Plummer J, Gayther SA. Risks and Function of Breast Cancer Susceptibility Alleles. Cancers (Basel) 2021; 13:3953. [PMID: 34439109 PMCID: PMC8393346 DOI: 10.3390/cancers13163953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/30/2021] [Accepted: 07/31/2021] [Indexed: 12/22/2022] Open
Abstract
Family history remains one of the strongest risk factors for breast cancer. It is well established that women with a first-degree relative affected by breast cancer are twice as likely to develop the disease themselves. Twins studies indicate that this is most likely due to shared genetics rather than shared epidemiological/lifestyle risk factors. Linkage and targeted sequencing studies have shown that rare high- and moderate-penetrance germline variants in genes involved in the DNA damage response (DDR) including BRCA1, BRCA2, PALB2, ATM, and TP53 are responsible for a proportion of breast cancer cases. However, breast cancer is a heterogeneous disease, and there is now strong evidence that different risk alleles can predispose to different subtypes of breast cancer. Here, we review the associations between the different genes and subtype-specificity of breast cancer based on the most comprehensive genetic studies published. Genome-wide association studies (GWAS) have also been used to identify an additional hereditary component of breast cancer, and have identified hundreds of common, low-penetrance susceptibility alleles. The combination of these low penetrance risk variants, summed as a polygenic risk score (PRS), can identify individuals across the spectrum of disease risk. However, there remains a substantial bottleneck between the discovery of GWAS-risk variants and their contribution to tumorigenesis mainly because the majority of these variants map to the non-protein coding genome. A range of functional genomic approaches are needed to identify the causal risk variants and target susceptibility genes and establish their underlying role in disease biology. We discuss how the application of these multidisciplinary approaches to understand genetic risk for breast cancer can be used to identify individuals in the population that may benefit from clinical interventions including screening for early detection and prevention, and treatment strategies to reduce breast cancer-related mortalities.
Collapse
Affiliation(s)
| | | | - Simon A. Gayther
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; (S.T.D.); (J.P.)
| |
Collapse
|
3
|
Ana G, Kelly PM, Malebari AM, Noorani S, Nathwani SM, Twamley B, Fayne D, O’Boyle NM, Zisterer DM, Pimentel EF, Endringer DC, Meegan MJ. Synthesis and Biological Evaluation of 1-(Diarylmethyl)-1 H-1,2,4-triazoles and 1-(Diarylmethyl)-1 H-imidazoles as a Novel Class of Anti-Mitotic Agent for Activity in Breast Cancer. Pharmaceuticals (Basel) 2021; 14:169. [PMID: 33671674 PMCID: PMC7926793 DOI: 10.3390/ph14020169] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/21/2022] Open
Abstract
We report the synthesis and biochemical evaluation of compounds that are designed as hybrids of the microtubule targeting benzophenone phenstatin and the aromatase inhibitor letrozole. A preliminary screening in estrogen receptor (ER)-positive MCF-7 breast cancer cells identified 5-((2H-1,2,3-triazol-1-yl)(3,4,5-trimethoxyphenyl)methyl)-2-methoxyphenol 24 as a potent antiproliferative compound with an IC50 value of 52 nM in MCF-7 breast cancer cells (ER+/PR+) and 74 nM in triple-negative MDA-MB-231 breast cancer cells. The compounds demonstrated significant G2/M phase cell cycle arrest and induction of apoptosis in the MCF-7 cell line, inhibited tubulin polymerisation, and were selective for cancer cells when evaluated in non-tumorigenic MCF-10A breast cells. The immunofluorescence staining of MCF-7 cells confirmed that the compounds targeted tubulin and induced multinucleation, which is a recognised sign of mitotic catastrophe. Computational docking studies of compounds 19e, 21l, and 24 in the colchicine binding site of tubulin indicated potential binding conformations for the compounds. Compounds 19e and 21l were also shown to selectively inhibit aromatase. These compounds are promising candidates for development as antiproliferative, aromatase inhibitory, and microtubule-disrupting agents for breast cancer.
Collapse
Affiliation(s)
- Gloria Ana
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| | - Patrick M. Kelly
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| | - Azizah M. Malebari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Sara Noorani
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| | - Seema M. Nathwani
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.M.N.); (D.F.); (D.M.Z.)
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, Dublin 2, DO2R590 Dublin, Ireland;
| | - Darren Fayne
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.M.N.); (D.F.); (D.M.Z.)
| | - Niamh M. O’Boyle
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| | - Daniela M. Zisterer
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.M.N.); (D.F.); (D.M.Z.)
| | - Elisangela Flavia Pimentel
- Department of Pharmaceutical Sciences, University Vila Velha, Av. Comissário José Dantas de Melo, n°21, Boa Vista Vila Velha—Espírito Santo, Vila Velha 29102-920, Brazil; (E.F.P.); (D.C.E.)
| | - Denise Coutinho Endringer
- Department of Pharmaceutical Sciences, University Vila Velha, Av. Comissário José Dantas de Melo, n°21, Boa Vista Vila Velha—Espírito Santo, Vila Velha 29102-920, Brazil; (E.F.P.); (D.C.E.)
| | - Mary J. Meegan
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| |
Collapse
|
4
|
Kim HA, Lee D, Lee H, Lee J. Lysimachia christinae Hance as an anticancer agent against breast cancer cells. Food Sci Nutr 2020; 8:5717-5728. [PMID: 33133573 PMCID: PMC7590289 DOI: 10.1002/fsn3.1875] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the most common cancer in women, and metastasis is the leading cause of death in breast cancer patients. Although chemoprevention is widely employed to treat breast cancer, anticancer drugs can cause significant adverse effects. Lysimachia christinae Hance (LH) is a traditional Chinese medicinal plant with diverse therapeutic effects. However, its potential anticancer activity has not been fully investigated in breast cancers to date. Using high-performance liquid chromatography-mass spectrometry, we found that the main constituent of LH extract (LHE) was rutin. Our results indicated that LHE or rutin markedly decreased the proliferation and viability of estrogen receptor (ER)-positive MCF-7 and ER-negative HCC38 human breast cancer cells. LHE treatment induced morphological changes in apoptotic nuclei using 4',6-diamidino-2-phenylindole (DAPI) staining. Annexin V-fluorescein isothiocyanate (FITC) propidium iodide (PI) staining assay revealed that apoptosis significantly increased in both breast cancer cell types after LHE treatment. Additionally, the expression of poly (ADP-ribose) polymerase (PARP), Bcl-2, and phospho-Akt decreased, while that of cleaved PARP and p53 increased, in both cell types. Furthermore, LHE treatment inhibited epithelial-mesenchymal transition (EMT). LHE treatment significantly upregulated E-cadherin level in MCF-7 and HCC38 cells, while vimentin level was downregulated in HCC38 cells. In addition, transwell and wound-healing assays revealed that LHE or rutin inhibited breast cancer cell migration. Overall, these findings demonstrate that LHE is a promising therapeutic agent that acts by promoting apoptosis and reducing cell proliferation, EMT, and cell migration in ER-positive and ER-negative breast cancer cells.
Collapse
Affiliation(s)
- Hyun A. Kim
- Department of Food and NutritionChosun UniversityGwangjuKorea
| | | | - Hwan Lee
- College of PharmacyChosun UniversityGwangjuKorea
| | - Joomin Lee
- Department of Food and NutritionChosun UniversityGwangjuKorea
| |
Collapse
|
5
|
Jin Q, Liu G, Wang B, Li S, Ni K, Wang C, Ren J, Zhang S, Dai Y. High methionyl-tRNA synthetase expression predicts poor prognosis in patients with breast cancer. J Clin Pathol 2020; 73:803-812. [PMID: 32404475 PMCID: PMC7691814 DOI: 10.1136/jclinpath-2019-206175] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 04/07/2020] [Accepted: 04/10/2020] [Indexed: 01/03/2023]
Abstract
Aims Methionyl–tRNA synthetase (MARS) is known to play a critical role in initiating translation and protection against cellular damages in vivo. The aim of this study was to clarify the role of MARS in breast cancer (BC) progression. Methods The expressions of MARS messenger RNA (mRNA) and protein in human BC tissues and adjacent non-cancerous tissues were detected by quantitative real-time PCR, western blot and immunohistochemistry. The prognostic potential of MARS in patients with BC was assessed by univariate and multivariate survival analyses. The association between the MARS expression and BC progression was further evaluated by the bioinformatics database of UALCAN, Gene Expression Profiling Interactive Analysis (GEPIA) and Gene Expression Database of Normal and Tumor Tissues (GENT). The role of MARS in the proliferation, migration and epithelial-to-mesenchymal transition (EMT) of human breast cancer cell line (MCF-7 cells) was investigated after siRNA transfection. Results The expression level of MARS mRNA in the fresh BC tissues was significantly higher than that in the adjacent tissues. Immunohistochemistry showed that the expression level of MARS was closely associated with the clinicopathologial parameters of patients with BC, including the HER-2 status, Ki-67 status, molecular classification, tumour grade, N stage and tumour, node, metastasis (TNM) stage, and this finding was further confirmed by UALCAN database. The Kaplan-Meier analysis showed that high MARS expression and TNM stage were predictors of poor prognosis of patients with BC. The proliferation, migration and EMT capabilities of MCF-7 cells were significantly suppressed after MARS knockdown. An overview of UALCAN, GEPIA and GENT results suggested that MARS may be an oncogene of BC, as well as a potential therapeutic target of this malignant tumour. Conclusions High expression level of MARS in the human BC tissues was significantly associated with the unfavourable prognosis of patients with BC, suggesting that MARS may serve as a potential prognostic marker for the clinical diagnosis and prognostic prediction of BC.
Collapse
Affiliation(s)
- Qin Jin
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Gang Liu
- College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia, China.,Clinical Medicine Research Center of the Affiliated Hospital, Inner Mongolia Medical University, Hohhot, China
| | - Biao Wang
- College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Shubin Li
- College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Kan Ni
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Chunyu Wang
- College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Jingyu Ren
- College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Shu Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yanfeng Dai
- College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia, China
| |
Collapse
|
6
|
Recalde-Gil AM, Klein-Júnior L, Salton J, Bordignon S, Cechinel-Filho V, Matté C, Henriques A. Aromatase (CYP19) inhibition by biflavonoids obtained from the branches of Garcinia gardneriana (Clusiaceae). ACTA ACUST UNITED AC 2019; 74:279-282. [DOI: 10.1515/znc-2019-0036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 06/13/2019] [Indexed: 01/29/2023]
Abstract
Abstract
Overexpression of aromatase in breast cancer cells may substantially influence its progression and maintenance. In this sense, the inhibition of aromatase is a key target for the treatment of breast cancer in postmenopausal women. Although several flavonoids had already demonstrated the capacity of inhibiting aromatase activity, the role of biflavonoids as aromatase inhibitors is poorly studied. In this work, the biflavonoids isolated from Garcinia gardneriana, morelloflavone (1), Gb-2a (2) and Gb-2a-7-O-glucose (3) were submitted to in vitro assay to evaluate the aromatase modulatory effect. As results, it was demonstrated that all biflavonoids were able to inhibit the enzyme, with IC50 values ranging from 1.35 to 7.67 μM. This demonstrates that biflavonoids are an important source of scaffolds for the development of new aromatase inhibitors, focusing on the development of new anticancer agents.
Collapse
Affiliation(s)
- Angelica Maria Recalde-Gil
- Laboratório de Farmacognosia e Controle de Qualidade de Fitoterápicos, Faculdade de Farmácia , Universidade Federal do Rio Grande do Sul , Porto Alegre 90610-000 , Brazil
| | - Luiz Klein-Júnior
- Laboratório de Farmacognosia e Controle de Qualidade de Fitoterápicos, Faculdade de Farmácia , Universidade Federal do Rio Grande do Sul , Porto Alegre 90610-000 , Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas , Universidade do Vale do Itajaí – UNIVALI , Rua Uruguai, 458 – 88302-202 , Itajaí/SC , Brazil
| | - Juliana Salton
- Laboratório de Farmacognosia e Controle de Qualidade de Fitoterápicos, Faculdade de Farmácia , Universidade Federal do Rio Grande do Sul , Porto Alegre 90610-000 , Brazil
| | - Sérgio Bordignon
- Laboratório de Conservação da Biodiversidade e Manejo , Universidade La Salle , Canoas 92010-000 , Brazil
| | - Valdir Cechinel-Filho
- Programa de Pós-Graduação em Ciências Farmacêuticas , Universidade do Vale do Itajaí , Itajaí 88302-901 , Brazil
| | - Cristiane Matté
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica , ICBS, Universidade Federal do Rio Grande do Sul , Porto Alegre 90610-000 , Brazil
| | - Amélia Henriques
- Laboratório de Farmacognosia e Controle de Qualidade de Fitoterápicos, Faculdade de Farmácia , Universidade Federal do Rio Grande do Sul , Porto Alegre 90610-000 , Brazil
| |
Collapse
|
7
|
Barros-Oliveira MDC, Costa-Silva DR, Andrade DBD, Borges US, Silva VC, Borges RS, Lopes-Costa PV, Alencar AP, da Silva BB. Ki-67 antigen expression in the mammary epithelium of female rats in persistent estrus treated with anastrozole. Gynecol Endocrinol 2017; 33:359-362. [PMID: 28277134 DOI: 10.1080/09513590.2017.1290065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVES To evaluate Ki-67 antigen expression in mammary epithelium of female rats in persistent estrus treated with anastrozole. MATERIALS AND METHODS Twenty-eight Wistar-Hanover female rats in persistent estrus induced by subcutaneous injection of 1.25 mg of testosterone propionate in the second day of life were randomly divided into two groups, control and experimental, with 14 animals each. The animals of control group received only the vehicle (propyleneglycol) and the animals of group experimental received 0.125 mg daily of anastrozole by gavage during 28 days. After 28 days of treatment, all animals were sacrificed and the first pair of abdominal-inguinal mammary glands was removed and fixed in 10% buffered formalin to investigate Ki-67 antigen expression by immunohistochemistry. RESULTS The mean percentage of Ki-67-stained nuclei per 500 cells in the mammary epithelium was 76.97 ± 0.76 and 14.44 ± 2.02 [mean ± standard error of the mean (SEM)] in the control and experimental groups, respectively (p < 0.0001). CONCLUSIONS Anastrozole treatment significantly reduced Ki-67 expression in the mammary epithelium of rats in persistent estrus.
Collapse
Affiliation(s)
| | | | | | - Umbelina Soares Borges
- a Posgraduate Program of Sciences and Health, Federal University of Piauí , Teresina , Brazil
| | - Vladimir Costa Silva
- a Posgraduate Program of Sciences and Health, Federal University of Piauí , Teresina , Brazil
| | - Rafael Soares Borges
- a Posgraduate Program of Sciences and Health, Federal University of Piauí , Teresina , Brazil
| | - Pedro Vitor Lopes-Costa
- a Posgraduate Program of Sciences and Health, Federal University of Piauí , Teresina , Brazil
- b Mastology Unit of Getulio Vargas Hospital, Federal University of Piauí , Teresina , Brazil , and
| | | | - Benedito Borges da Silva
- a Posgraduate Program of Sciences and Health, Federal University of Piauí , Teresina , Brazil
- b Mastology Unit of Getulio Vargas Hospital, Federal University of Piauí , Teresina , Brazil , and
| |
Collapse
|
8
|
Barros-Oliveira MDC, Costa-Silva DR, Andrade DBD, Borges US, Tavares CB, Borges RS, Silva JDM, Silva BBD. Use of anastrozole in the chemoprevention and treatment of breast cancer: A literature review. Rev Assoc Med Bras (1992) 2017; 63:371-378. [DOI: 10.1590/1806-9282.63.04.371] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/19/2016] [Indexed: 11/22/2022] Open
Abstract
Summary Aromatase inhibitors have emerged as an alternative endocrine therapy for the treatment of hormone sensitive breast cancer in postmenopausal women. The use of third-generation inhibitors represented by exemestane, letrozol and anastrozole is currently indicated. Anastrozole is a nonsteroidal compound and a potent selective inhibitor of the aromatase enzyme. Although a few studies have shown that its pharmacodynamic and pharmacokinetic properties may be affected by interindividual variability, this drug has been recently used in all configurations of breast cancer treatment. In metastatic disease, it is currently considered the first-line treatment for postmenopausal women with estrogen receptor-positive breast tumors. Anastrozole has shown promising results in the adjuvant treatment of early-stage breast cancer in postmenopausal women. It has also achieved interesting results in the chemoprevention of the disease. Therefore, due to the importance of anastrozole both for endocrine treatment and chemoprevention of hormone-sensitive breast cancer in postmenopausal women, we proposed the current literature review in the SciELO and PubMed database of articles published in the last 10 years.
Collapse
|
9
|
Tamoxifen Resistance: Emerging Molecular Targets. Int J Mol Sci 2016; 17:ijms17081357. [PMID: 27548161 PMCID: PMC5000752 DOI: 10.3390/ijms17081357] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/10/2016] [Accepted: 08/16/2016] [Indexed: 12/12/2022] Open
Abstract
17β-Estradiol (E2) plays a pivotal role in the development and progression of breast cancer. As a result, blockade of the E2 signal through either tamoxifen (TAM) or aromatase inhibitors is an important therapeutic strategy to treat or prevent estrogen receptor (ER) positive breast cancer. However, resistance to TAM is the major obstacle in endocrine therapy. This resistance occurs either de novo or is acquired after an initial beneficial response. The underlying mechanisms for TAM resistance are probably multifactorial and remain largely unknown. Considering that breast cancer is a very heterogeneous disease and patients respond differently to treatment, the molecular analysis of TAM’s biological activity could provide the necessary framework to understand the complex effects of this drug in target cells. Moreover, this could explain, at least in part, the development of resistance and indicate an optimal therapeutic option. This review highlights the implications of TAM in breast cancer as well as the role of receptors/signal pathways recently suggested to be involved in the development of TAM resistance. G protein—coupled estrogen receptor, Androgen Receptor and Hedgehog signaling pathways are emerging as novel therapeutic targets and prognostic indicators for breast cancer, based on their ability to mediate estrogenic signaling in ERα-positive or -negative breast cancer.
Collapse
|
10
|
Glushkov A, Polenok E, Kostyanko M, Antonov A, Verzhbitskaya N, Vafin I, Ragozhina S. Postmenopausal Breast Cancer Risk in Relation to Antibodies Specific to Benzo[a]Pyrene, Estradiol and Progesterone. IRANIAN JOURNAL OF CANCER PREVENTION 2016; 9:e4212. [PMID: 27482331 PMCID: PMC4951760 DOI: 10.17795/ijcp-4212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 11/10/2015] [Accepted: 03/14/2016] [Indexed: 11/25/2022]
Abstract
BACKGROUND Antibodies might protect against low doses of environmental carcinogens by decreasing systemic uptake, activation of metabolic pathways, and redistribution of carcinogens within the organism. The features of antibody formation in relation to environmental carcinogens and sex steroids under natural conditions should be determined to identify breast cancer risk, then to develop cancer immune prevention strategies. OBJECTIVES The purpose of this study was to investigate antibodies specifications to benzo(a)pyrene, estradiol and progesterone in postmenopausal women with invasive breast cancer. PATIENTS AND METHODS A semi-quantitative non-competitive immunoassay of IgG antibodies to benzo(a)pyrene (IgG-Bp), estradiol (IgG-Es), and progesterone (IgG-Pg) has conducted. The assay has performed on 322 serum samples from patients with breast cancer and 179 serum samples from healthy postmenopausal women by using low-molecular-weight Bp, Es, and Pg conjugated with bovine serum albumin. ROC analysis has also conducted to determine the odds ratio (OR). RESULTS Combination of the high levels of IgG-Bp and IgG-Es without IpG-Pg was more frequent in breast cancer patients than that in healthy women, and the OR has increased to 3.8. Combination of the high levels of IgG-Pg with high levels of both IgG-Bp and IgG-Es were significantly more frequent in breast cancer patients (36.9%) than that in healthy women (5.6%), and the OR increased to 11.7. These differences have peculiarly expressed in breast cancer patients with hormone status ER+/PR- (OR = 26.7). The minimum OR (0.4) has obtained at low levels of the three antibodies. CONCLUSIONS Immunoassay of antibodies against environmental carcinogens and sex steroid hormones could use to detect breast cancer risk. Induction of antibodies against Bp for cancer immunoprevention could lead to antibody formation against steroid hormones, thereby increasing breast cancer risk.
Collapse
Affiliation(s)
- Andrew Glushkov
- Department of Molecular Human Ecology, Institute of Human Ecology Siberian Branch of Russian, Academy of Sciences, Kemerovo, Russia; Faculty of Biology, Kemerovo State University, Kemerovo, Russia
| | - Elena Polenok
- Department of Molecular Human Ecology, Institute of Human Ecology Siberian Branch of Russian, Academy of Sciences, Kemerovo, Russia
| | - Mikhail Kostyanko
- Department of Molecular Human Ecology, Institute of Human Ecology Siberian Branch of Russian, Academy of Sciences, Kemerovo, Russia; Faculty of Biology, Kemerovo State University, Kemerovo, Russia
| | | | | | | | | |
Collapse
|
11
|
Harvie M, Howell A, Evans DG. Can diet and lifestyle prevent breast cancer: what is the evidence? Am Soc Clin Oncol Educ Book 2016:e66-73. [PMID: 25993238 DOI: 10.14694/edbook_am.2015.35.e66] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Breast cancer is the most common cancer among women in both developed and less-developed countries. Rates of breast cancer are increasing worldwide, with a particular increase in postmenopausal and estrogen receptor-positive cases. The World Cancer Research Fund (WCRF) and American Cancer Society (ACS) cancer prevention guidelines recommend maintaining a healthy weight, undertaking at least 150 minutes of moderate intensity exercise per week, limiting alcohol consumption, and eating a plant-based diet. Observational data link adherence to physical activity and alcohol guidelines throughout life to a reduced risk of developing pre- and postmenopausal breast cancer. Weight control throughout life appears to prevent cases after menopause. Adherence to a healthy dietary pattern does not have specific effects on breast cancer risk but remains important as it reduces the risk for other common diseases, such as cardiovascular disease (CVD), diabetes, and dementia. Emerging data suggest that smoking during adolescence or early adulthood increases later risk of breast cancer. Lifestyle factors appear to modify risk among high-risk women with a family history and those with typical risk of the general population, although their effects among carriers of BRCA mutations are not well defined. Recent expert reports estimate that successful lifestyle changes could prevent 25% to 30% of cases of breast cancer. These reductions will only be achieved if we can implement targeted prevention programs for high-risk women and women in population-based breast screening programs during childhood, adolescence, and early adulthood when the rapidly developing breast is particularly susceptible to carcinogenesis.
Collapse
Affiliation(s)
- Michelle Harvie
- From the Genesis Breast Cancer Prevention Centre, University Hospital of South Manchester NHS Foundation Trust, Manchester, United Kingdom
| | - Anthony Howell
- From the Genesis Breast Cancer Prevention Centre, University Hospital of South Manchester NHS Foundation Trust, Manchester, United Kingdom
| | - D Gareth Evans
- From the Genesis Breast Cancer Prevention Centre, University Hospital of South Manchester NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|