1
|
Parisian AD, Barratt SA, Hodges-Gallagher L, Ortega FE, Peña G, Sapugay J, Robello B, Sun R, Kulp D, Palanisamy GS, Myles DC, Kushner PJ, Harmon CL. Palazestrant (OP-1250), A Complete Estrogen Receptor Antagonist, Inhibits Wild-type and Mutant ER-positive Breast Cancer Models as Monotherapy and in Combination. Mol Cancer Ther 2024; 23:285-300. [PMID: 38102750 PMCID: PMC10911704 DOI: 10.1158/1535-7163.mct-23-0351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/01/2023] [Accepted: 12/13/2023] [Indexed: 12/17/2023]
Abstract
The estrogen receptor (ER) is a well-established target for the treatment of breast cancer, with the majority of patients presenting as ER-positive (ER+). Endocrine therapy is a mainstay of breast cancer treatment but the development of resistance mutations in response to aromatase inhibitors, poor pharmacokinetic properties of fulvestrant, agonist activity of tamoxifen, and limited benefit for elacestrant leave unmet needs for patients with or without resistance mutations in ESR1, the gene that encodes the ER protein. Here we describe palazestrant (OP-1250), a novel, orally bioavailable complete ER antagonist and selective ER degrader. OP-1250, like fulvestrant, has no agonist activity on the ER and completely blocks estrogen-induced transcriptional activity. In addition, OP-1250 demonstrates favorable biochemical binding affinity, ER degradation, and antiproliferative activity in ER+ breast cancer models that is comparable or superior to other agents of interest. OP-1250 has superior pharmacokinetic properties relative to fulvestrant, including oral bioavailability and brain penetrance, as well as superior performance in wild-type and ESR1-mutant breast cancer xenograft studies. OP-1250 combines well with cyclin-dependent kinase 4 and 6 inhibitors in xenograft studies of ER+ breast cancer models and effectively shrinks intracranially implanted tumors, resulting in prolonged animal survival. With demonstrated preclinical efficacy exceeding fulvestrant in wild-type models, elacestrant in ESR1-mutant models, and tamoxifen in intracranial xenografts, OP-1250 has the potential to benefit patients with ER+ breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Richard Sun
- Olema Pharmaceuticals, San Francisco, California
| | - David Kulp
- Olema Pharmaceuticals, San Francisco, California
| | | | | | | | | |
Collapse
|
2
|
Bailey S, Ezratty C, Mhango G, Lin JJ. Clinical and sociodemographic risk factors associated with the development of second primary cancers among postmenopausal breast cancer survivors. Breast Cancer 2023; 30:215-225. [PMID: 36316601 PMCID: PMC9974531 DOI: 10.1007/s12282-022-01411-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/18/2022] [Indexed: 02/24/2023]
Abstract
BACKGROUND Advancement in breast cancer (BC) diagnosis and treatment have increased the number of long-term survivors. Consequently, primary BC survivors are at a greater risk of developing second primary cancers (SPCs). The risk factors for SPCs among BC survivors including sociodemographic characteristics, cancer treatment, comorbidities, and concurrent medications have not been comprehensively examined. The purpose of this study is to assess the incidence and clinicopathologic factors associated with risk of SPCs in BC survivors. METHODS We analyzed 171, 311 women with early-stage primary BC diagnosed between January 2000 and December 2015 from the Medicare-linked Surveillance Epidemiology and End Results (SEER-Medicare) database. SPC was defined as any diagnosis of malignancy occurring within the study period and at least 6 months after primary BC diagnosis. Univariate analyses compared baseline characteristics between those who developed a SPC and those who did not. We evaluated the cause-specific hazard of developing a SPC in the presence of death as a competing risk. RESULTS Of the study cohort, 21,510 (13%) of BC survivors developed a SPC and BC was the most common SPC type (28%). The median time to SPC was 44 months. Women who were white, older, and with fewer comorbidities were more likely to develop a SPC. While statins [hazard ratio (HR) 1.066 (1.023-1.110)] and anti-hypertensives [HR 1.569 (1.512-1.627)] increased the hazard of developing a SPC, aromatase inhibitor therapy [HR 0.620 (0.573-0.671)] and bisphosphonates [HR 0.905 (0.857-0.956)] were associated with a decreased hazard of developing any SPC, including non-breast SPCs. CONCLUSION Our study shows that specific clinical factors including type of cancer treatment, medications, and comorbidities are associated with increased risk of developing SPCs among older BC survivors. These results can increase patient and clinician awareness, target cancer screening among BC survivors, as well as developing risk-adapted management strategies.
Collapse
Affiliation(s)
- Stacyann Bailey
- Department of Biomedical Engineering, Institute for Applied Life Sciences, University of Massachusetts Amherst, 240 Thatcher Road, Amherst, MA, 01003, USA.
| | - Charlotte Ezratty
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Grace Mhango
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jenny J. Lin
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
3
|
Miranda C, Galleguillos M, Torres R, Tardón K, Cáceres DD, Lee K, Redal MA, Varela NM, Quiñones LA. Preliminary Pharmacogenomic-Based Predictive Models of Tamoxifen Response in Hormone-dependent Chilean Breast Cancer Patients. Front Pharmacol 2021; 12:661443. [PMID: 34899282 PMCID: PMC8656167 DOI: 10.3389/fphar.2021.661443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022] Open
Abstract
Tamoxifen (TAM), a selective oestrogen receptor modulator, is one of the most used treatments in oestrogen receptor-positive (ER+) early and metastatic breast cancer (BC) patients. The response to TAM has a high degree of inter-individual variability. This is mainly due to genetic variants in CYP2D6 gene, as well as other genes encoding proteins involved in the TAM pharmacokinetic and/or pharmacodynamic. Therefore, prediction of the TAM response using these genetic factors together with other non-genetic variables may be relevant to improve breast cancer treatment. Thus, in this work, we used genetic polymorphisms and clinical variables for TAM response modelling. One hundred sixty-two ER + BC patients with 2 years of TAM treatment were retrospectively recruited, and the genetic polymorphisms CYP2D6*4, CYP3A4*1B (CYP3A4*1.001), CYP3A5*3, UGT2B7*2, UGT2B15*2, SULT1A1*2, and ESRA V364E were analyzed by PCR-RFLP. Concomitantly, the therapeutic response was obtained from clinical records for association with genotypes using univariate and multivariate biostatistical models. Our results show that UGT2B15*1/*2 genotype protects against relapse (OR = 0.09; p = 0.02), CYP3A5*3/*3 genotype avoids endometrial hyperplasia (OR = 0.07; p = 0.01), SULT1A1*1/*2 genotype avoids vaginal bleeding (OR = 0.09; p = 0.03) and ESRA 364E/364E genotype increases the probability of vaginal bleeding (OR = 5.68; p = 0.02). Logistic regression models, including genomic and non-genomic variables, allowed us to obtain preliminary predictive models to explain relapse (p = 0.010), endometrial hyperplasia (p = 0.002) and vaginal bleeding (p = 0.014). Our results suggest that the response to TAM treatment in ER + BC patients might be associated with the presence of the studied genetic variants in UGT2B15, CYP3A5, SULT1A1 and ESRA genes. After clinical validation protocols, these models might be used to help to predict a percentage of BC relapse and adverse reactions, improving the individual response to TAM-based treatment.
Collapse
Affiliation(s)
- Carla Miranda
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Macarena Galleguillos
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago, Chile
| | | | | | - Dante D Cáceres
- Institute of Population Health, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Kuen Lee
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago, Chile.,Faculty of Medicine, University of Chile, Santiago, Chile
| | - María A Redal
- Genetic Division, Department of Medicine, Hospital de Clínicas José de San Martín, Buenos Aires, Argentina.,Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | - Nelson M Varela
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago, Chile.,Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | - Luis A Quiñones
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago, Chile.,Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| |
Collapse
|
4
|
Segovia-Mendoza M, García-Quiroz J, Díaz L, García-Becerra R. Combinations of Calcitriol with Anticancer Treatments for Breast Cancer: An Update. Int J Mol Sci 2021; 22:12741. [PMID: 34884550 PMCID: PMC8657847 DOI: 10.3390/ijms222312741] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 12/12/2022] Open
Abstract
Preclinical, clinical, and epidemiological studies indicate that vitamin D3 (VD) deficiency is a risk factor for the development of breast cancer. Underlying mechanisms include the ability of calcitriol to induce cell differentiation, inhibit oncogenes expression, and modify different signaling pathways involved in the control of cell proliferation. In addition, calcitriol combined with different kinds of antineoplastic drugs has been demonstrated to enhance their beneficial effects in an additive or synergistic fashion. However, a recognized adjuvant regimen based on calcitriol for treating patients with breast cancer has not yet been fully established. Accordingly, in the present work, we review and discuss the preclinical and clinical studies about the combination of calcitriol with different oncological drugs, aiming to emphasize its main therapeutic benefits and opportunities for the treatment of this pathology.
Collapse
Affiliation(s)
- Mariana Segovia-Mendoza
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Janice García-Quiroz
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico;
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico;
| | - Rocío García-Becerra
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| |
Collapse
|
5
|
D'Amico P, Cristofanilli M. Standard of Care in Hormone Receptor-Positive Metastatic Breast Cancer: Can We Improve the Current Regimens or Develop Better Selection Tools? JCO Oncol Pract 2021; 18:331-334. [PMID: 34780297 DOI: 10.1200/op.21.00707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Paolo D'Amico
- Department of Medicine-Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Massimo Cristofanilli
- Department of Medicine-Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
6
|
Endocrine Therapy With or Without CDK4/6 Inhibitors in Women With Hormone-receptor Positive Breast Cancer: What do we Know About the Effects on Cognition? Clin Breast Cancer 2021; 22:191-199. [PMID: 34556423 DOI: 10.1016/j.clbc.2021.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/01/2021] [Accepted: 08/10/2021] [Indexed: 12/17/2022]
Abstract
Adjuvant endocrine therapy (ET) is the cornerstone of treatment for hormone-receptor positive breast cancer. Recently, ET is increasingly combined with "cyclin-dependent kinases 4 and 6'' (CDK4/6) inhibitors. Given the importance of estrogens in neural processes and the role of cyclin D in hippocampal cell proliferation, it is plausible that these therapies affect cognition, but studies on these potential cognitive effects are sparse. In this review, we summarize existing knowledge on the cognitive effects of ET and CDK4/6 inhibitors in pre-, peri- and postmenopausal patients with breast cancer. We show that several clinical studies support adverse cognitive effects, especially on verbal memory, after ET-induced decrease of estrogen-levels or inactivation of estrogen-receptors. Clinical studies on the cognitive effects of CDK4/6 inhibitors are virtually non-existent and no conclusions can yet be drawn. Longitudinal studies on the cognitive effects of the combined ET-CDK4/6 inhibitors are highly needed to properly inform patients about potential short-term and long-term cognitive side effects. These studies should preferably include cognitive assessments (including a measurement prior to ET), and be designed in such a way that they can account for variables such as type and duration of ET, CDK4/6 inhibition, menopausal status, and other disease- and treatment-related symptoms that can impact cognition, such as fatigue and distress.
Collapse
|
7
|
Schuler LA, Murdoch FE. Endogenous and Therapeutic Estrogens: Maestro Conductors of the Microenvironment of ER+ Breast Cancers. Cancers (Basel) 2021; 13:3725. [PMID: 34359625 PMCID: PMC8345134 DOI: 10.3390/cancers13153725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/25/2022] Open
Abstract
Estrogen receptor alpha (ERα) marks heterogeneous breast cancers which display a repertoire of somatic genomic mutations and an immune environment that differs from other breast cancer subtypes. These cancers also exhibit distinct biological behaviors; despite an overall better prognosis than HER2+ or triple negative breast cancers, disseminated dormant cells can lead to disease recurrence decades after the initial diagnosis and treatment. Estrogen is the best studied driver of these cancers, and antagonism or reduction of estrogen activity is the cornerstone of therapeutic approaches. In addition to reducing proliferation of ERα+ cancer cells, these treatments also alter signals to multiple other target cells in the environment, including immune cell subpopulations, cancer-associated fibroblasts, and endothelial cells via several distinct estrogen receptors. In this review, we update progress in our understanding of the stromal cells populating the microenvironments of primary and metastatic ER+ tumors, the effects of estrogen on tumor and stromal cells to modulate immune activity and the extracellular matrix, and net outcomes in experimental and clinical studies. We highlight new approaches that will illuminate the unique biology of these cancers, provide the foundation for developing new treatment and prevention strategies, and reduce mortality of this disease.
Collapse
Affiliation(s)
- Linda A. Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | | |
Collapse
|
8
|
Anurag M, Zhu M, Huang C, Vasaikar S, Wang J, Hoog J, Burugu S, Gao D, Suman V, Zhang XH, Zhang B, Nielsen T, Ellis MJ. Immune Checkpoint Profiles in Luminal B Breast Cancer (Alliance). J Natl Cancer Inst 2021; 112:737-746. [PMID: 31665365 DOI: 10.1093/jnci/djz213] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 09/12/2019] [Accepted: 10/25/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Unlike estrogen receptor (ER)-negative breast cancer, ER-positive breast cancer outcome is less influenced by lymphocyte content, indicating the presence of immune tolerance mechanisms that may be specific to this disease subset. METHODS A supervised analysis of microarray data from the ACOSOG Z1031 (Alliance) neoadjuvant aromatase inhibitor (AI) trial identified upregulated genes in Luminal (Lum) B breast cancers that correlated with AI-resistant tumor proliferation (percentage of Ki67-positive cancer nuclei, Pearson r > 0.4) (33 cases Ki67 > 10% on AI) vs LumB breast cancers that were more AI sensitive (33 cases Ki67 < 10% on AI). Overrepresentation analysis was performed using WebGestalt. All statistical tests were two-sided. RESULTS Thirty candidate genes positively correlated (r ≥ 0.4) with AI-resistant proliferation in LumB and were upregulated greater than twofold. Gene ontologies identified that the targetable immune checkpoint (IC) components IDO1, LAG3, and PD1 were overrepresented resistance candidates (P ≤ .001). High IDO1 mRNA was associated with poor prognosis in LumB disease (Molecular Taxonomy of Breast Cancer International Consortium, hazard ratio = 1.43, 95% confidence interval = 1.04 to 1.98, P = .03). IDO1 also statistically significantly correlated with STAT1 at protein level in LumB disease (Pearson r = 0.74). As a composite immune tolerance signature, expression of IFN-γ/STAT1 pathway components was associated with higher baseline Ki67, lower estrogen, and progesterone receptor mRNA levels and worse disease-specific survival (P = .002). In a tissue microarray analysis, IDO1 was observed in stromal cells and tumor-associated macrophages, with a higher incidence in LumB cases. Furthermore, IDO1 expression was associated with a macrophage mRNA signature (M1 by CIBERSORT Pearson r = 0.62 ) and by tissue microarray analysis. CONCLUSIONS Targetable IC components are upregulated in the majority of endocrine therapy-resistant LumB cases. Our findings provide rationale for IC inhibition in poor-outcome ER-positive breast cancer.
Collapse
MESH Headings
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antineoplastic Agents, Hormonal/therapeutic use
- Aromatase Inhibitors/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/immunology
- Cell Proliferation/physiology
- Drug Resistance, Neoplasm
- Female
- Humans
- Immune Tolerance
- Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology
- Interferon-gamma/metabolism
- Letrozole/therapeutic use
- Prognosis
- Programmed Cell Death 1 Receptor/biosynthesis
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/immunology
- STAT1 Transcription Factor/metabolism
- Signal Transduction
- Tissue Array Analysis
- Transcriptome
- Up-Regulation
- Lymphocyte Activation Gene 3 Protein
Collapse
|
9
|
Golbaghi G, Haghdoost MM, Yancu D, Santos YLDL, Doucet N, Patten SA, Sanderson JT, Castonguay A. Organoruthenium(II) Complexes Bearing an Aromatase Inhibitor: Synthesis, Characterization, in Vitro Biological Activity and in Vivo Toxicity in Zebrafish Embryos. Organometallics 2019; 38:702-711. [PMID: 31762529 DOI: 10.1021/acs.organomet.8b00897] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Third-generation aromatase inhibitors such as anastrozole (ATZ) and letrozole (LTZ) are widely used to treat estrogen receptor-positive ER+ breast cancers in postmenopausal women. Investigating their ability to coordinate metals could lead to the emergence of a new category of anticancer drug candidates with a broader spectrum of pharmacological activities. In this study, a series of ruthenium (II) arene complexes bearing the aromatase inhibitor anastrozole was synthesized and characterized. Among these complexes, [Ru(η 6 -C6H6)(PPh3)(η 1 -ATZ)Cl]BPh4 (3) was found to be the most stable in cell culture media, to lead to the highest cellular uptake and in vitro cytotoxicity in two ER+ human breast cancer cell lines (MCF7 and T47D), and to induce a decrease in aromatase activity in H295R cells. Exposure of zebrafish embryos to complex 3 (12.5 μM) did not lead to noticeable signs of toxicity over 96 h, making it a suitable candidate for further in vivo investigations.
Collapse
Affiliation(s)
- Golara Golbaghi
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Mohammad Mehdi Haghdoost
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Debbie Yancu
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Yossef López de Los Santos
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Nicolas Doucet
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Shunmoogum A Patten
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| | - J Thomas Sanderson
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Annie Castonguay
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| |
Collapse
|
10
|
Goel P, Alam O, Naim MJ, Nawaz F, Iqbal M, Alam MI. Recent advancement of piperidine moiety in treatment of cancer- A review. Eur J Med Chem 2018; 157:480-502. [DOI: 10.1016/j.ejmech.2018.08.017] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/26/2018] [Accepted: 08/04/2018] [Indexed: 12/23/2022]
|
11
|
Bjørklund G, Dadar M, Chirumbolo S, Lysiuk R. Flavonoids as detoxifying and pro-survival agents: What's new? Food Chem Toxicol 2017; 110:240-250. [PMID: 29079495 DOI: 10.1016/j.fct.2017.10.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/29/2017] [Accepted: 10/22/2017] [Indexed: 02/06/2023]
Abstract
The role of flavonoids in the survival machinery of cells has come in the spotlight due to the recent evidence of their effect on the relationship mitochondria-ER stress-proteasome, including the intracellular mechanisms of autophagy and apoptosis. Numerous experimental animal investigations and even human clinical studies have highlighted the major role of these natural compounds in the economy of life and their deep relationship with autotrophic organisms in the evolutionary space. Their role as anti-oxidant and oxidative stress preventive molecules has to date been investigated extensively in the literature. Despite this great amount of promising evidence, many concerns, however, remain, most of which dealing with biochemistry, bioavailability, pharmacokinetics, and interaction of flavonoids with gut microbiome, issues that make difficult any good attempt to introduce these molecules in the human healthcare systems as possible, encouraging therapeutic substances. This review tries to address and elucidate these items.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway.
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Salvatore Chirumbolo
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Roman Lysiuk
- Department of Pharmacognosy and Botany, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| |
Collapse
|
12
|
Di Matteo M, Ammazzalorso A, Andreoli F, Caffa I, De Filippis B, Fantacuzzi M, Giampietro L, Maccallini C, Nencioni A, Parenti MD, Soncini D, Del Rio A, Amoroso R. Synthesis and biological characterization of 3-(imidazol-1-ylmethyl)piperidine sulfonamides as aromatase inhibitors. Bioorg Med Chem Lett 2016; 26:3192-3194. [DOI: 10.1016/j.bmcl.2016.04.078] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 10/21/2022]
|
13
|
Turnbull AK, Arthur LM, Renshaw L, Larionov AA, Kay C, Dunbier AK, Thomas JS, Dowsett M, Sims AH, Dixon JM. Accurate Prediction and Validation of Response to Endocrine Therapy in Breast Cancer. J Clin Oncol 2015; 33:2270-8. [PMID: 26033813 DOI: 10.1200/jco.2014.57.8963] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Aromatase inhibitors (AIs) have an established role in the treatment of breast cancer. Response rates are only 50% to 70% in the neoadjuvant setting and lower in advanced disease. Accurate biomarkers are urgently needed to predict response in these settings and to determine which individuals will benefit from adjuvant AI therapy. PATIENTS AND METHODS Pretreatment and on-treatment (after 2 weeks and 3 months) biopsies were obtained from 89 postmenopausal women who had estrogen receptor-alpha positive breast cancer and were receiving neoadjuvant letrozole for transcript profiling. Dynamic clinical response was assessed with use of three-dimensional ultrasound measurements. RESULTS The molecular response to letrozole was characterized and a four-gene classifier of clinical response was established (accuracy of 96%) on the basis of the level of two genes before treatment (one gene [IL6ST] was associated with immune signaling, and the other [NGFRAP1] was associated with apoptosis) and the level of two proliferation genes (ASPM, MCM4) after 2 weeks of therapy. The four-gene signature was found to be 91% accurate in a blinded, completely independent validation data set of patients treated with anastrozole. Matched 2-week on-treatment biopsies were associated with improved predictive power as compared with pretreatment biopsies alone. This signature also significantly predicted recurrence-free survival (P = .029) and breast cancer -specific survival (P = .009). We demonstrate that the test can also be performed with use of quantitative polymerase chain reaction or immunohistochemistry. CONCLUSION A four-gene predictive model of clinical response to AIs by 2 weeks has been generated and validated. Deregulated immune and apoptotic responses before treatment and cell proliferation that is not reduced 2 weeks after initiation of treatment are functional characteristics of breast tumors that do not respond to AIs.
Collapse
Affiliation(s)
- Arran K Turnbull
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Laura M Arthur
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Lorna Renshaw
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Alexey A Larionov
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Charlene Kay
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Anita K Dunbier
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Jeremy S Thomas
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Mitch Dowsett
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| | - Andrew H Sims
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand.
| | - J Michael Dixon
- Arran K. Turnbull, Laura M. Arthur, Lorna Renshaw, Alexey A. Larionov, Charlene Kay, Jeremy S. Thomas, Andrew H. Sims, J. Michael Dixon, University of Edinburgh Cancer Research UK Centre, Edinburgh; Anita K. Dunbier, Mitch Dowsett, Institute of Cancer Research, London, United Kingdom; and Anita K. Dunbier, University of Otago, Dunedin, New Zealand
| |
Collapse
|
14
|
Abstract
Around 70% of all breast cancers are estrogen receptor alpha positive and hence their development is highly dependent on estradiol. While the invention of endocrine therapies has revolusioned the treatment of the disease, resistance to therapy eventually occurs in a large number of patients. This paper seeks to illustrate and discuss the complexity and heterogeneity of the mechanisms which underlie resistance and the approaches proposed to combat them. It will also focus on the use and development of methods for predicting which patients are likely to develop resistance.
Collapse
|
15
|
Miller WR, Larionov A. Changes in expression of oestrogen regulated and proliferation genes with neoadjuvant treatment highlight heterogeneity of clinical resistance to the aromatase inhibitor, letrozole. Breast Cancer Res 2010; 12:R52. [PMID: 20646288 PMCID: PMC2949641 DOI: 10.1186/bcr2611] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Revised: 05/25/2010] [Accepted: 07/20/2010] [Indexed: 12/14/2022] Open
Abstract
Introduction Clinical resistance is a major factor limiting benefits to endocrine therapy. Causes of resistance may be diverse and the mechanism of resistance in individual breast cancers is usually unknown. The present study illustrates how changes in the expression of proliferation and oestrogen-regulated genes occurring during neoadjuvant treatment with the aromatase inhibitor, letrozole, may define distinctive tumour subgroups and suggest different mechanisms of resistance in clinically endocrine resistant breast cancers. Methods Postmenopausal women with large primary oestrogen-receptor (ER)-rich breast cancers were treated neoadjuvantly with letrozole (2.5 mg daily) for three months. Clinical response was determined by ultrasound changes in tumour volume. Tumour ribonucleic acid (RNA) from biopsies taken before, after 14 days and after three months of treatment was hybridized on Affymetrix U133A chips. Changes in expression of KIAA0101, TFF3, SERPINA3, IRS-1 and TFF1 were taken as markers of oestrogen regulation and those in CDC2, CKS-2, Cyclin B1, Thymidine Synthetase and PCNA as markers of proliferation. Results Fifteen tumours with < 50% volume reduction over three months of treatment were classified as being clinically non-responsive. Gene expression changes after 14 days of treatment with letrozole revealed different patterns of change in oestrogen regulated and proliferation genes in individual resistant tumours. Tumours could be separated into three different subgroups as follows: i) nine cases in which both proliferation and oestrogen signalling signatures were generally reduced on treatment (ii) four cases in which both signatures were generally unaffected or increased with treatment and (iii) two cases in which expression of the majority of oestrogen-regulated genes decreased whereas proliferation genes remained unchanged or increased. In 14 out of 15 tumours, RNA profiles were also available after three months of treatment. Patterns of change observed after 14 days were maintained or accentuated at three months in nine tumours but changes in patterns were apparent in the remaining five cancers. Conclusions Different dynamic patterns of expression of oestrogen-regulated and proliferation genes were observed in tumours clinically resistant to neoadjuvant letrozole, thus illustrating heterogeneity of resistance and discriminating molecular sub-classes of resistant tumours. Molecular phenotyping might help to direct circumventing therapy suggesting the targeting of specific pathways in different tumour subtypes.
Collapse
Affiliation(s)
- William R Miller
- Breast Research Group, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK.
| | | |
Collapse
|
16
|
Gupta A, Saha P, Descôteaux C, Leblanc V, Asselin E, Bérubé G. Design, synthesis and biological evaluation of estradiol-chlorambucil hybrids as anticancer agents. Bioorg Med Chem Lett 2010; 20:1614-8. [PMID: 20137939 DOI: 10.1016/j.bmcl.2010.01.053] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 01/12/2010] [Accepted: 01/13/2010] [Indexed: 10/19/2022]
Abstract
A series of estradiol-chlorambucil hybrids was synthesized as anticancer drugs for site-directed chemotherapy of breast cancer. The novel compounds were synthesized in good yields through efficient modifications of estrone at position 16alpha of the steroid nucleus. The newly synthesized compounds were evaluated for their anticancer efficacy in different hormone-dependent and hormone-independent breast cancer cell lines. The novel hybrids showed significant in vitro anticancer activity when compared to chlorambucil. Structure-activity relationship (SAR) reveals the influence of the length of the spacer chain between carrier and drug molecule.
Collapse
Affiliation(s)
- Atul Gupta
- Département de Chimie-Biologie, Groupe de Recherche en Oncologie et Endocrinologie Moléculaires, Université du Québec à Trois-Rivières, C.P. 500, Trois-Rivières, Québec, Canada G9A 5H7
| | | | | | | | | | | |
Collapse
|
17
|
Miller WR, Larionov A, Renshaw L, Anderson TJ, Walker JR, Krause A, Sing T, Evans DB, Dixon JM. Gene expression profiles differentiating between breast cancers clinically responsive or resistant to letrozole. J Clin Oncol 2009; 27:1382-7. [PMID: 19224856 DOI: 10.1200/jco.2008.16.8849] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
PURPOSE Endocrine agents, such as letrozole, are established in the treatment of hormone-dependent breast cancer. However, response rates are only 50% to 70% in the neoadjuvant setting and lower in advanced disease. Thus there is a need to identify novel markers predicting for response and to understand molecular mechanisms of resistance. PATIENTS AND METHODS Sequential tumor biopsies were taken before and after 10 to 14 days of neoadjuvant treatment with letrozole in patients with estrogen receptor-rich breast cancer. Expression profiles on high-density microarray chips were then related to clinical responses as assessed from tumor volume measurements after 3 months of treatment. RESULTS Of 52 patients, 37 (71%) were classified as having a clinical response to letrozole and 15 being clinically resistant. Bioinformatic analysis identified 205 covariables (69 baseline expression, 45 day 14 expression, and 91 change in expression with treatment) which differentiated between clinical responders and nonresponders. Hierarchical clustering using the variables separated responders and nonresponders into two distinct groups. Ontological assessment indicated that discriminating genes were enriched toward cellular biosynthetic processes. In particular, functional gene assessment showed ribosomal protein probes to have higher baseline expression in tumors responsive to letrozole and increased expression with treatment in nonresponding cases. CONCLUSION To our knowledge, this is the first study to describe genetic covariables and molecular processes discriminating between tumors clinically responsive and resistant to an aromatase inhibitor. The understanding of such molecular phenotypes will be important in optimizing the clinical use of aromatase inhibitors, both in terms of identifying responsive breast cancers and developing new agents to target resistance pathways.
Collapse
Affiliation(s)
- William R Miller
- Breast Research Group, University of Edinburgh, Edinburgh, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Folkestad L, Bjarnason NH, Bjerregaard JK, Brixen K. The Effect of Aromatase Inhibitors on Bone Metabolism. Basic Clin Pharmacol Toxicol 2009; 104:3-10. [DOI: 10.1111/j.1742-7843.2008.00337.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
19
|
Bone loss in patients with breast cancer receiving aromatase inhibitors and associated treatment strategies. Cancer Treat Rev 2008; 34 Suppl 1:S31-42. [PMID: 18486346 DOI: 10.1016/j.ctrv.2008.03.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Hormone-receptor-positive breast cancer in postmenopausal women is treated increasingly with aromatase inhibitors because of increased efficacy and reduced incidence of endometrial cancer compared with tamoxifen. However, aromatase inhibitor therapy increases bone turnover as a result of nearly complete oestrogen depletion, leading to increases in bone loss and fragility fractures that erode patients' functional independence and quality of life. Management of patients with aromatase inhibitor-associated bone loss (AIBL) is currently evolving and intervention strategies are under investigation. Although no treatments are specifically approved for AIBL, bisphosphonates are currently the intervention of choice for patients with low bone mineral density or evidence of rapid bone turnover, along with adequate calcium and vitamin D supplementation and a healthy lifestyle. In this setting, the majority of information available regarding bisphosphonate efficacy is from studies of intravenous zoledronic acid (4 mg) every 6 months. Data from four large international studies (three of identical design in postmenopausal women and one in premenopausal women) indicate that zoledronic acid is effective in the management of AIBL. Treatment algorithms based on risk factors and bone mineral density are under development, and the results of ongoing studies should help define optimal bone health management for patients undergoing aromatase inhibitor treatment for early breast cancer.
Collapse
|
20
|
Miller WR, Larionov A, Anderson TJ, Walker JR, Krause A, Evans DB, Dixon JM. Predicting response and resistance to endocrine therapy. Cancer 2008; 112:689-694. [DOI: 10.1002/cncr.23187] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
21
|
Miller WR, Larionov AA, Renshaw L, Anderson TJ, White S, Murray J, Murray E, Hampton G, Walker JR, Ho S, Krause A, Evans DB, Dixon JM. Changes in breast cancer transcriptional profiles after treatment with the aromatase inhibitor, letrozole. Pharmacogenet Genomics 2007; 17:813-26. [PMID: 17885619 DOI: 10.1097/fpc.0b013e32820b853a] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The aim of the study was to identify changes in tumour expression profiling associated with short-term therapy of breast cancer patients with letrozole. EXPERIMENTAL DESIGN Microarray analysis was performed on RNA extracted from paired tumour core biopsies taken before and after 14 days of treatment with letrozole (2.5 mg/daily) in 58 patients. Changes in expression profile were identified by three different approaches on the basis of frequency of changes, magnitude of changes and significance analysis of microarray. RESULTS No single gene was consistently changed by therapy in all cases. Fifty-two genes, however, were downregulated and 36 upregulated in at least 45 of the 58 cases. In terms of quantitative change, 46 genes showed at least a median 1.5-fold change in expression. Significance analysis of microarray identified 62 genes that were significantly changed by therapy (P<0.0001, 56 downregulated and six upregulated). All three approaches showed that greater numbers of genes were downregulated rather than upregulated. Merging data produced a total of 143 genes, which were subject to gene ontology and cluster analysis. The ontology of the 91 downregulated genes showed that they were functionally associated with cell cycle progression, particularly mitosis. In contrast, upregulated genes were associated with organ development, connective tissue extracellular matrix regulation and inflammatory response. Cluster analysis segregated the patients into four groups differing in patterns of gene expression. CONCLUSION Genes have been identified which either change markedly or consistently in breast cancer after 14 days treatment with letrozole. These are new important data in understanding letrozole's molecular mechanism of action in breast cancers.
Collapse
|
22
|
Miller WR, Larionov A, Renshaw L, Anderson TJ, White S, Hampton G, Walker JR, Ho S, Krause A, Evans DB, Dixon JM. Aromatase inhibitors--gene discovery. J Steroid Biochem Mol Biol 2007; 106:130-42. [PMID: 17616392 DOI: 10.1016/j.jsbmb.2007.05.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Microarray analysis of tumour RNA is an extremely powerful tool which allows global gene expression to be measured. When used in combination with neoadjuvant treatment protocols in which therapy is given with the primary tumour within the breast, sequential biopsies may be analysed and results correlated with clinical and pathological response. In the present study, a neoadjuvant protocol has been used, administering the third generation inhibitor, letrozole, for 3 months and subjecting RNA extracted from biopsies taken before and after 10-14 days of treatment to microarray analysis. The objectives were to discover: (i) genes that change with estrogen deprivation (the only known biological effect of letrozole is to inhibit aromatase activity and reduce endogenous estrogens in postmenopausal women) and (ii) genes whose basal, on treatment or change in expression differ between tumours which are either responsive or resistant to treatment (so that predictive indices of response/resistance may be developed). Early changes in gene expression were identified by comparing paired tumour core biopsies taken before and after 14 days treatment in 58 patients using three different approaches based on frequency of changes, magnitude of changes and SAM analysis. All three approaches showed a greater number of genes were down-regulated than up-regulated. Merging of the data produced a total of 143 genes which were subject to gene ontology and cluster analysis. The ontology of the 91 down-regulated genes showed that they were functionally associated with cell cycle progression, particularly mitosis. In contrast, up-regulated genes were associated with organ development and extra-cellular matrix turnover and regulation. Clinical response was assessable in 52 patients; 37 (71%) tumours were classified as clinical responders (>50% reduction in volume at 3 months). Microarray analysis of pre- and 14-day biopsies identified 291 covariates (84 baselines, 72 14-day and 135 changes) highly predictive of response status. A similarity matrix using the covariates showed responding tumours have a similar genetic profile which was dissimilar to non-responding cancers whereas non-responsive cases were distinctive from each other. Changed genes predicting for response showed no concordance with those changed significantly by treatment in the overall group.
Collapse
Affiliation(s)
- William R Miller
- Breast Research Group, University of Edinburgh, Edinburgh, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Fujimura T, Takahashi S, Urano T, Kumagai J, Ogushi T, Horie-Inoue K, Ouchi Y, Kitamura T, Muramatsu M, Inoue S. Increased expression of estrogen-related receptor alpha (ERRalpha) is a negative prognostic predictor in human prostate cancer. Int J Cancer 2007; 120:2325-30. [PMID: 17294452 DOI: 10.1002/ijc.22363] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The nuclear receptor ERRalpha (estrogen-related receptor alpha) is known to modulate the estrogen-signaling pathway, but the biological significance of ERRalpha in the prostate remains unclear. We investigated the expression of ERRalpha in human prostate tissues and cancer cell lines to evaluate the potential roles of the receptor in prostate cancer (PC). Western blot analysis of ERRalpha was performed in three cell lines of human PC (LNCaP, DU145 and PC-3). The expressions of ERRalpha in cancerous lesions (n = 106) and benign foci (n = 99) of 106 surgically obtained prostate specimens were evaluated by immunohistochemistry. The relationships between the ERRalpha expression and clinicopathological features were evaluated. Western blot analysis using the polyclonal anti-ERRalpha antibody detected a 52 kD band in all three PC cell lines. Positive immunostaining of ERRalpha in the nuclei was found in 73 (69%) cancerous and 47 (47.5%) benign epithelium, whereas the stromal tissues were negative for ERRalpha. The mean immunoreactivity score (IR score) of the cancerous lesions (3.5 +/- 2.6) was significantly higher than that of the benign foci (1.8 +/- 2.1) (p < 0.0001). The IR score of the cancerous lesions significantly correlated with the Gleason score (p = 0.0135). Univariate and multivariate hazard analyses revealed significant correlations between elevated ERRalpha expression and poor cancer-specific survival (p = 0.0141 and 0.0367, respectively). The enhanced expression of ERRalpha might play a role in the development of human PC and serve as a significant prognostic factor for the disease.
Collapse
Affiliation(s)
- Tetsuya Fujimura
- Department of Urology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Breast cancer is the second most common cause of cancer-related death in women in the US and the UK, accounting for 15-17% of all female cancer deaths. Current treatment strategies include hormone therapy, such as anti-estrogens (tamoxifen) and aromatase inhibitors (exemastane, anastrozole, letrozole), as well as cytotoxics, such as the taxanes (paclitaxel, docetaxel). With multiple therapy choices, a method to prospectively screen patients prior to therapy selection is now needed. Pharmacogenetics seeks to develop screening mechanisms to optimise drug therapy. DNA variations in metabolism, transport and drug target genes may contribute to chemotherapy efficacy and toxicities. The status of the identification of genetic markers for breast cancer therapy selection is highlighted in this review.
Collapse
Affiliation(s)
- Sharon Marsh
- Washington University School of Medicine, Division of Oncology, St Louis, MO 63110, USA.
| | | |
Collapse
|
25
|
Cheng YSL, Mues G, Wood D, Ding J. Aromatase expression in normal human oral keratinocytes and oral squamous cell carcinoma. Arch Oral Biol 2006; 51:612-20. [PMID: 16510116 DOI: 10.1016/j.archoralbio.2006.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2005] [Revised: 12/28/2005] [Accepted: 01/11/2006] [Indexed: 10/24/2022]
Abstract
UNLABELLED Aromatase is the enzyme that catalyzes the conversion of androgen to oestrogen. Aromatase expression in extra-gonadal sites and local oestrogen synthesis play an important role in the physiological conditions and in the growth of certain neoplasms. OBJECTIVE The purpose of this study was to investigate aromatase expression in oral keratinocytes and oral squamous cell carcinoma (SCC). DESIGN Immunocytochemistry and RT-nested PCR were used to detect aromatase protein and mRNA expression in primary human oral epithelial cell culture and in an oral SCC cell line. Immunohistochemistry was used to detect aromatase protein expression in frozen and archival human tissue sections of normal oral epithelium and oral SCC. RESULTS Cytoplasmic immunostaining was found in normal oral keratinocytes and SCC cells in culture. The common coding region of aromatase mRNA was detected in the oral keratinocytes derived from five different normal individuals and in the SCC cell line. However, there were variations in aromatase exon 1 expression among normal oral keratinocyte samples. Cytoplasmic staining was found in normal oral epithelium and well-differentiated oral SCC but not in poorly differentiated oral SCC by immunohistochemistry. CONCLUSION Aromatase was expressed in normal oral keratinocytes and oral SCC both in cell culture and in tissues, indicating local oestrogen synthesis in normal and neoplastic conditions of oral epithelium.
Collapse
Affiliation(s)
- Yi-Shing Lisa Cheng
- Diagnostic Sciences, Baylor College of Dentistry-Texas A and M University Health Science Center, 3302 Gaston Ave., Dallas, TX 75246, USA.
| | | | | | | |
Collapse
|
26
|
Fisher B, Costantino JP, Wickerham DL, Cecchini RS, Cronin WM, Robidoux A, Bevers TB, Kavanah MT, Atkins JN, Margolese RG, Runowicz CD, James JM, Ford LG, Wolmark N. Tamoxifen for the prevention of breast cancer: current status of the National Surgical Adjuvant Breast and Bowel Project P-1 study. J Natl Cancer Inst 2005; 97:1652-62. [PMID: 16288118 DOI: 10.1093/jnci/dji372] [Citation(s) in RCA: 909] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Initial findings from the National Surgical Adjuvant Breast and Bowel Project Breast Cancer Prevention Trial (P-1) demonstrated that tamoxifen reduced the risk of estrogen receptor-positive tumors and osteoporotic fractures in women at increased risk for breast cancer. Side effects of varying clinical significance were observed. The trial was unblinded because of the positive results, and follow-up continued. This report updates our initial findings. METHODS Women (n = 13,388) were randomly assigned to receive placebo or tamoxifen for 5 years. Rates of breast cancer and other events were compared by the use of risk ratios (RRs) and 95% confidence intervals (CIs). Estimates of the net benefit from 5 years of tamoxifen therapy were compared by age, race, and categories of predicted breast cancer risk. Statistical tests were two-sided. RESULTS After 7 years of follow-up, the cumulative rate of invasive breast cancer was reduced from 42.5 per 1000 women in the placebo group to 24.8 per 1000 women in the tamoxifen group (RR = 0.57, 95% CI = 0.46 to 0.70) and the cumulative rate of noninvasive breast cancer was reduced from 15.8 per 1000 women in the placebo group to 10.2 per 1000 women in the tamoxifen group (RR = 0.63, 95% CI = 0.45 to 0.89). These reductions were similar to those seen in the initial report. Tamoxifen led to a 32% reduction in osteoporotic fractures (RR = 0.68, 95% CI = 0.51 to 0.92). Relative risks of stroke, deep-vein thrombosis, and cataracts (which increased with tamoxifen) and of ischemic heart disease and death (which were not changed with tamoxifen) were also similar to those initially reported. Risks of pulmonary embolism were approximately 11% lower than in the original report, and risks of endometrial cancer were about 29% higher, but these differences were not statistically significant. The net benefit achieved with tamoxifen varied according to age, race, and level of breast cancer risk. CONCLUSIONS Despite the potential bias caused by the unblinding of the P-1 trial, the magnitudes of all beneficial and undesirable treatment effects of tamoxifen were similar to those initially reported, with notable reductions in breast cancer and increased risks of thromboembolic events and endometrial cancer. Readily identifiable subsets of individuals comprising 2.5 million women could derive a net benefit from the drug.
Collapse
Affiliation(s)
- Bernard Fisher
- Operations Center, National Surgical Adjuvant Breast and Bowel Project, Pittsburgh, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|