1
|
Bertelsen BE, Almås B, Fjermeros K, Viste K, Geisler SB, Sauer T, Selsås K, Geisler J. Superior suppression of serum estrogens during neoadjuvant breast cancer treatment with letrozole compared to exemestane. Breast Cancer Res Treat 2024; 206:347-358. [PMID: 38649619 PMCID: PMC11182829 DOI: 10.1007/s10549-024-07313-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/22/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE The aromatase inhibitor letrozole and the aromatase inactivator exemestane are two of the most pivotal cancer drugs used for endocrine treatment of ER-positive breast cancer in all phases of the disease. Although both drugs inhibit CYP19 (aromatase) and have been used for decades, a direct head-to-head, intra-patient-cross-over comparison of their ability to decrease estrogen synthesis in vivo is still lacking. METHODS Postmenopausal breast cancer patients suitable for neoadjuvant endocrine therapy were randomized to receive either letrozole (2.5 mg o.d.) or exemestane (25 mg o.d.) for an initial treatment period, followed by a second treatment period on the alternative drug (intra-patient cross-over study design). Serum levels of estrone (E1), estradiol (E2), letrozole, exemestane, and 17-hydroxyexemestane were quantified simultaneously using a novel, ultrasensitive LC-MS/MS method established in our laboratory. RESULTS Complete sets of serum samples (baseline and during treatment with letrozole or exemestane) were available from 79 patients, including 40 patients starting with letrozole (cohort 1) and 39 with exemestane (cohort 2). Mean serum estrone and estradiol levels in cohort 1 were 174 pmol/L and 46.4 pmol/L at baseline, respectively. Treatment with letrozole suppressed serum E1 and E2 to a mean value of 0.2 pmol/L and 0.4 pmol/L (P < 0.001). After the cross-over to exemestane, mean serum levels of E1 and E2 increased to 1.4 pmol/L and 0.7 pmol/L, respectively. In cohort 2, baseline mean serum levels of E1 and E2 were 159 and 32.5 pmol/L, respectively. Treatment with exemestane decreased these values to 1.8 pmol/L for E1 and 0.6 pmol/L for E2 (P < 0.001). Following cross-over to letrozole, mean serum levels of E1 and E2 were significantly further reduced to 0.1 pmol/L and 0.4 pmol/L, respectively. Serum drug levels were monitored in all patients throughout the entire treatment and confirmed adherence to the protocol and drug concentrations within the therapeutic range for all patients. Additionally, Ki-67 values decreased significantly during treatment with both aromatase inhibitors, showing a trend toward a stronger suppression in obese women. CONCLUSION To the best of our knowledge, we present here for the first time a comprehensive and direct head-to-head, intra-patient-cross-over comparison of the aromatase inhibitor letrozole and the aromatase inactivator exemestane concerning their ability to suppress serum estrogen levels in vivo. All in all, our results clearly demonstrate that letrozole therapy results in a more profound suppression of serum E1 and E2 levels compared to exemestane.
Collapse
Affiliation(s)
- Bjørn-Erik Bertelsen
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland, University Hospital, Bergen, Norway
| | - Bjørg Almås
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland, University Hospital, Bergen, Norway
| | - Kamilla Fjermeros
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| | - Kristin Viste
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland, University Hospital, Bergen, Norway
| | | | - Torill Sauer
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Knut Selsås
- Department of Breast- and Endocrine Surgery, Akershus University Hospital, Lørenskog, Norway
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
2
|
Li C, Wang X, Nie M, Mao J, Wu X. Adding Letrozole to Growth Hormone and Gonadotropin-Releasing Hormone Analog Increases Height in Girls With Short Stature: A Hospital Record-Based Retrospective Study. Endocr Pract 2024; 30:639-646. [PMID: 38723894 DOI: 10.1016/j.eprac.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/28/2024] [Accepted: 04/19/2024] [Indexed: 05/23/2024]
Abstract
OBJECTIVE There have been rare data on letrozole for height improvement in girls. This study aimed to clarify the efficacy and safety of combination therapy with recombinant human growth hormone (rhGH), GnRHa, and letrozole in improving the height of girls with short stature and advanced bone age. METHODS This was a hospital record-based retrospective study. Follow-up was conducted on girls with short stature who received treatment with rhGH, GnRHa, and letrozole in our hospital. The treatment group included a total of 29 participants. Before treatment, the mean age of the patients was 11.17 years, and the mean treatment duration was 17.31 months. The control group consisted of 29 short-statured girls who received rhGH/GnRHa treatment, with the mean age and treatment duration of 12.43 years and 16.59 months, respectively. RESULTS The predicted adult heights (PAHs) before and after treatment were 155.38 and 161.32 cm (P < .001). The ΔPAH in the treatment group was 4 cm higher than that in the control group (5.85 vs 1.82 cm, P < .001). Significant differences were noted in the height standard deviation scores of bone age (P < .001) and chronological age (P = .003) before and after treatment. There was an increasing body mass index during therapy (P = .039). The height gain was 8.71 ± 4.46 cm, and the growth rate was 6.78 ± 3.84 cm per year. CONCLUSION Combined treatment with GH, GnRHa, and letrozole can enhance the adult height and PAH in short-statured girls, and no significant side effects have been reported.
Collapse
Affiliation(s)
- Chenyang Li
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xi Wang
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Nie
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangfeng Mao
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xueyan Wu
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
3
|
Akman T, Arendt LM, Geisler J, Kristensen VN, Frigessi A, Köhn-Luque A. Modeling of Mouse Experiments Suggests that Optimal Anti-Hormonal Treatment for Breast Cancer is Diet-Dependent. Bull Math Biol 2024; 86:42. [PMID: 38498130 PMCID: PMC11310292 DOI: 10.1007/s11538-023-01253-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/30/2023] [Indexed: 03/20/2024]
Abstract
Estrogen receptor positive breast cancer is frequently treated with anti-hormonal treatment such as aromatase inhibitors (AI). Interestingly, a high body mass index has been shown to have a negative impact on AI efficacy, most likely due to disturbances in steroid metabolism and adipokine production. Here, we propose a mathematical model based on a system of ordinary differential equations to investigate the effect of high-fat diet on tumor growth. We inform the model with data from mouse experiments, where the animals are fed with high-fat or control (normal) diet. By incorporating AI treatment with drug resistance into the model and by solving optimal control problems we found differential responses for control and high-fat diet. To the best of our knowledge, this is the first attempt to model optimal anti-hormonal treatment for breast cancer in the presence of drug resistance. Our results underline the importance of considering high-fat diet and obesity as factors influencing clinical outcomes during anti-hormonal therapies in breast cancer patients.
Collapse
Affiliation(s)
- Tuğba Akman
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, 0317, Oslo, Norway.
- Department of Computer Engineering, University of Turkish Aeronautical Association, 06790, Etimesgut, Ankara, Turkey.
| | - Lisa M Arendt
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Campus AHUS, Oslo, Norway
| | - Vessela N Kristensen
- Department of Medical Genetics, Institute of Clinical Medicine, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Arnoldo Frigessi
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, 0317, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Alvaro Köhn-Luque
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, 0317, Oslo, Norway.
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
4
|
Nolasco-Pérez TDJ, Cervantes-Candelas LA, Buendía-González FO, Aguilar-Castro J, Fernández-Rivera O, Salazar-Castañón VH, Legorreta-Herrera M. Immunomodulatory effects of testosterone and letrozole during Plasmodium berghei ANKA infection. Front Cell Infect Microbiol 2023; 13:1146356. [PMID: 37384220 PMCID: PMC10296187 DOI: 10.3389/fcimb.2023.1146356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/15/2023] [Indexed: 06/30/2023] Open
Abstract
Introduction Malaria is one of the leading health problems globally. Plasmodium infection causes pronounced sexual dimorphism, and the lethality and severity are more remarkable in males than in females. To study the role of testosterone in the susceptibility and mortality of males in malaria, it is common to increase its concentration. However, this strategy does not consider the enzyme CYP19A1 aromatase, which can transform it into oestrogens. Methods To avoid the interference of oestrogens, we inhibited in vivo CYP19A1 aromatase with letrozole and increased the testosterone level by exogen administration before infection with Plasmodium berghei ANKA. We measured the impact on free testosterone, 17β-oestradiol and dehydroepiandrosterone levels in plasma; additionally, we evaluated parasitaemia, body temperature, body mass, glucose levels and haemoglobin concentration. Furthermore, we evaluated the effects of testosterone on the immune response; we quantified the CD3+/CD4+, CD3+/CD8+, CD19+, Mac-3+ and NK cells in the spleen and the plasma concentrations of the cytokines IL-2, IL-4, IL-6, IFN-, IL-10, TNF-α and IL-17A. Finally, we quantified the levels of antibodies. Results We found that mice treated with the combination of letrozole and testosterone and infected with Plasmodium berghei ANKA had increased concentrations of free testosterone and DHEA but decreased levels of 17β-oestradiol. As a result, parasitaemia increased, leading to severe anaemia. Interestingly, testosterone increased temperature and decreased glucose concentration as a possible testosterone-mediated regulatory mechanism. The severity of symptomatology was related to critical immunomodulatory effects generated by free testosterone; it selectively increased CD3+CD8+ T and CD19+ cells but decreased Mac-3+. Remarkably, it reduced IL-17A concentration and increased IL-4 and TNF-α. Finally, it increased IgG1 levels and the IgG1/IgG2a ratio. In conclusion, free testosterone plays an essential role in pathogenesis in male mice by increasing CD8+ and decreasing Mac3+ cells and mainly reducing IL-17A levels, which is critical in the development of anaemia. Our results are important for understanding the mechanisms that regulate the exacerbated inflammatory response in infectious diseases and would be useful for the future development of alternative therapies to reduce the mortality generated by inflammatory processes.
Collapse
Affiliation(s)
- Teresita de Jesús Nolasco-Pérez
- Laboratorio de Inmunología Molecular, Unidad de Investigación Química Computacional, Síntesis y Farmacología en Moléculas de Interés Biológico, División de Estudios de Posgrado e Investigación, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Luis Antonio Cervantes-Candelas
- Laboratorio de Inmunología Molecular, Unidad de Investigación Química Computacional, Síntesis y Farmacología en Moléculas de Interés Biológico, División de Estudios de Posgrado e Investigación, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Fidel Orlando Buendía-González
- Laboratorio de Inmunología Molecular, Unidad de Investigación Química Computacional, Síntesis y Farmacología en Moléculas de Interés Biológico, División de Estudios de Posgrado e Investigación, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Jesús Aguilar-Castro
- Laboratorio de Inmunología Molecular, Unidad de Investigación Química Computacional, Síntesis y Farmacología en Moléculas de Interés Biológico, División de Estudios de Posgrado e Investigación, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Omar Fernández-Rivera
- Laboratorio de Inmunología Molecular, Unidad de Investigación Química Computacional, Síntesis y Farmacología en Moléculas de Interés Biológico, División de Estudios de Posgrado e Investigación, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Víctor Hugo Salazar-Castañón
- Laboratorio de Inmunología Molecular, Unidad de Investigación Química Computacional, Síntesis y Farmacología en Moléculas de Interés Biológico, División de Estudios de Posgrado e Investigación, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Martha Legorreta-Herrera
- Laboratorio de Inmunología Molecular, Unidad de Investigación Química Computacional, Síntesis y Farmacología en Moléculas de Interés Biológico, División de Estudios de Posgrado e Investigación, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| |
Collapse
|
5
|
Banjare L, Singh Y, Verma SK, Singh AK, Kumar P, Kumar S, Jain AK, Thareja S. Multifaceted 3D-QSAR analysis for the identification of pharmacophoric features of biphenyl analogues as aromatase inhibitors. J Biomol Struct Dyn 2023; 41:1322-1341. [PMID: 34963408 DOI: 10.1080/07391102.2021.2019122] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Aromatase, a cytochrome P450 enzyme, is responsible for the conversion of androgens to estrogens, which fuel the multiplication of cancerous cells. Inhibition of estrogen biosynthesis by aromatase inhibitors (AIs) is one of the highly advanced therapeutic approach available for the treatment of estrogen-positive breast cancer. Biphenyl moiety aids lipophilicity to the conjugated scaffold and enhances the accessibility of the ligand to the target. The present study is focused on the investigation of, the mode of binding of biphenyl with aromatase, prediction of ligand-target binding affinities, and pharmacophoric features essential for favorable for aromatase inhibition. A multifaceted 3D-QSAR (SOMFA, Field and Gaussian) along with molecular docking, molecular dynamic simulations and pharmacophore mapping were performed on a series of biphenyl bearing molecules (1-33) with a wide range of aromatase inhibitory activity (0.15-920 nM). Among the generated 3D-QSAR models, the Force field-based 3D-QSAR model (R2 = 0.9151) was best as compared to SOMFA and Gaussian Field (R2=0.7706, 0.9074, respectively). However, all the generated 3D-QSAR models were statistically fit, robust enough, and reliable to explain the variation in biological activity in relation to pharmacophoric features of dataset molecules. A four-point pharmacophoric features with three acceptor sites (A), one aromatic ring (R) features, AAAR_1, were obtained with the site and survival score values 0.890 and 4.613, respectively. The generated 3D-QSAR plots in the study insight into the structure-activity relationship of dataset molecules, which may help in the designing of potent biphenyl derivatives as newer inhibitors of aromatase.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Laxmi Banjare
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur, Chhattisgarh, India
| | - Yogesh Singh
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences Central, University of Punjab, Bathinda, Punjab, India
| | - Sant Kumar Verma
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Atul Kumar Singh
- Molecular Signaling and Drug Discovery Laboratory, Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences Central, University of Punjab, Bathinda, Punjab, India
| | - Shashank Kumar
- Molecular Signaling and Drug Discovery Laboratory, Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Akhlesh Kumar Jain
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur, Chhattisgarh, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences Central, University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
6
|
Ma Y, Jia R, Xia B, Tang B, Xu Z. Adult height in pubertal boys with short stature treated with GH/letrozole: a hospital record-based retrospective study. BMC Pediatr 2022; 22:371. [PMID: 35764954 PMCID: PMC9238122 DOI: 10.1186/s12887-022-03438-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 06/22/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The growth potential in pubertal boys with short stature is limited by the effect of estrogen on epiphyseal fusion. This study aims to identify the efficacy and safety of the combination of growth hormone (GH) and letrozole on adult height (AH) in pubertal boys with short stature.
Methods
This is a retrospective record based study. Pubertal boys with short stature who were treated with GH and letrozole were followed up at outpatient clinics in our hospital. Twenty subjects who reached AH are reported here.
Results
Baseline chronological age was 12.12 ± 1.14 yr and bone age was 13.00 ± 0.93 yr. The period of GH/letrozole treatment was 1.94 ± 0.67 yr. Height standard deviation score for bone age was increased from -1.46 ± 0.51 before treatment to -0.12 ± 0.57 after treatment (P < 0.001). The predicted AH before treatment, predicted AH after treatment, AH, and genetic target height were 161.02 ± 4.12 cm, 172.11 ± 4.20 cm, 172.67 ± 2.72 cm, and 167.67 ± 3.56 cm, respectively. There was a significant predicted AH difference before and after treatment (P < 0.001). There was a significant difference between predicted AH before treatment and genetic target height (P < 0.001). Predicted AH after therapy was higher than that of gene target height (P < 0.001), as well as AH and genetic target height (P < 0.001). There was no significant side effect.
Conclusions
GH and letrozole combination can enhance AH in pubertal boys with short stature.
Collapse
|
7
|
Bahrami N, Jabeen S, Tahiri A, Sauer T, Ødegård HP, Geisler SB, Gravdehaug B, Reitsma LC, Selsås K, Kristensen V, Geisler J. Lack of cross-resistance between non-steroidal and steroidal aromatase inhibitors in breast cancer patients: the potential role of the adipokine leptin. Breast Cancer Res Treat 2021; 190:435-449. [PMID: 34554372 PMCID: PMC8558290 DOI: 10.1007/s10549-021-06399-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 09/11/2021] [Indexed: 11/01/2022]
Abstract
PURPOSE The aromatase inactivator exemestane may cause clinical disease stabilization following progression on non-steroidal aromatase inhibitors like letrozole in patients with metastatic breast cancer, indicating that additional therapeutic effects, not necessarily related to estrogen-suppression, may be involved in this well-known "lack of cross-resistance". METHODS Postmenopausal women with ER positive, HER-2 negative, locally advanced breast cancer were enrolled in the NEOLETEXE-trial and randomized to sequential treatment starting with either letrozole (2.5 mg o.d.) or exemestane (25 mg o.d.) followed by the alternative aromatase inhibitor. Serum levels of 54 cytokines, including 12 adipokines were assessed using Luminex xMAP technology (multiple ELISA). RESULTS Serum levels of leptin were significantly decreased during treatment with exemestane (p < 0.001), regardless whether exemestane was given as first or second neoadjuvant therapy. In contrast, letrozole caused a non-significant increase in serum leptin levels in vivo. CONCLUSIONS Our findings suggest an additional and direct effect of exemestane on CYP-19 (aromatase) synthesis presumably due to effects on the CYP19 promoter use that is not present during therapy with the non-steroidal aromatase inhibitor letrozole. Our findings provide new insights into the influence of clinically important aromatase inhibitors on cytokine levels in vivo that contribute to the understanding of the clinically observed lack of cross-resistance between non-steroidal and steroidal aromatase inhibitors in breast cancer patients. TRIAL REGISTRATION Registered on March 23rd 2015 in the National trial database of Norway (Registration number: REK-SØ-84-2015).
Collapse
Affiliation(s)
- Nazli Bahrami
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway.,Department of Breast and Endocrine Surgery, Akershus University Hospital, Lørenskog, Norway
| | - Shakila Jabeen
- Department of Clinical Molecular Biology (EPIGEN), Akershus University Hospital, Lørenskog, Norway.,Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Andliena Tahiri
- Department of Clinical Molecular Biology (EPIGEN), Akershus University Hospital, Lørenskog, Norway.,Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Torill Sauer
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway.,Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, University of Oslo, Oslo, Norway
| | | | | | - Berit Gravdehaug
- Department of Breast and Endocrine Surgery, Akershus University Hospital, Lørenskog, Norway
| | | | - Knut Selsås
- Department of Breast and Endocrine Surgery, Akershus University Hospital, Lørenskog, Norway
| | - Vessela Kristensen
- Department of Clinical Molecular Biology (EPIGEN), Akershus University Hospital, Lørenskog, Norway.,Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway. .,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
8
|
Hyder T, Marino CC, Ahmad S, Nasrazadani A, Brufsky AM. Aromatase Inhibitor-Associated Musculoskeletal Syndrome: Understanding Mechanisms and Management. Front Endocrinol (Lausanne) 2021; 12:713700. [PMID: 34385978 PMCID: PMC8353230 DOI: 10.3389/fendo.2021.713700] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/12/2021] [Indexed: 12/31/2022] Open
Abstract
Aromatase inhibitors (AIs) are a key component in the chemoprevention and treatment of hormone receptor-positive (HR+) breast cancer. While the addition of AI therapy has improved cancer-related outcomes in the management of HR+ breast cancer, AIs are associated with musculoskeletal adverse effects known as the aromatase inhibitor-associated musculoskeletal syndrome (AIMSS) that limit its tolerability and use. AIMSS is mainly comprised of AI-associated bone loss and arthralgias that affect up to half of women on AI therapy and detrimentally impact patient quality of life and treatment adherence. The pathophysiology of AIMSS is not fully understood though has been proposed to be related to estrogen deprivation within the musculoskeletal and nervous systems. This review aims to characterize the prevalence, risk factors, and clinical features of AIMSS, and explore the syndrome's underlying mechanisms and management strategies.
Collapse
Affiliation(s)
- Tara Hyder
- University of Pittsburgh Physicians, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Christopher C Marino
- Mario Lemieux Center for Blood Cancers, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
| | - Sasha Ahmad
- Department of Sciences, Sewickley Academy, Pittsburgh, PA, United States
| | - Azadeh Nasrazadani
- UPMC Hillman Cancer Center, Magee Women's Hospital, Pittsburgh, PA, United States
| | - Adam M Brufsky
- UPMC Hillman Cancer Center, Magee Women's Hospital, Pittsburgh, PA, United States
| |
Collapse
|
9
|
Reis J, Thomas O, Lahooti M, Lyngra M, Schandiz H, Boavida J, Gjesdal KI, Sauer T, Geisler J, Geitung JT. Correlation between MRI morphological response patterns and histopathological tumor regression after neoadjuvant endocrine therapy in locally advanced breast cancer: a randomized phase II trial. Breast Cancer Res Treat 2021; 189:711-723. [PMID: 34357493 PMCID: PMC8505284 DOI: 10.1007/s10549-021-06343-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/28/2021] [Indexed: 11/09/2022]
Abstract
PURPOSE To correlate MRI morphological response patterns with histopathological tumor regression grading system based on tumor cellularity in locally advanced breast cancer (LABC)-treated neoadjuvant with third-generation aromatase inhibitors. METHODS Fifty postmenopausal patients with ER-positive/HER-2-negative LABC treated with neoadjuvant letrozole and exemestane given sequentially in an intra-patient cross-over regimen for at least 4 months with MRI response monitoring at baseline as well as after at least 2 and 4 months on treatment. The MRI morphological response pattern was classified into 6 categories: 0/complete imaging response; I/concentric shrinkage; II/fragmentation; III/diffuse; IV/stable; and V/progressive. Histopathological tumor regression was assessed based on the recommendations from The Royal College of Pathologists regarding tumor cellularity. RESULTS Following 2 and 4 months with therapy, the most common MRI pattern was pattern II (24/50 and 21/50, respectively). After 4 months on therapy, the most common histopathological tumor regression grade was grade 3 (21/50). After 4 months an increasing correlation is observed between MRI patterns and histopathology. The overall correlation, between the largest tumor diameter obtained from MRI and histopathology, was moderate and positive (r = 0.50, P-value = 2e-04). Among them, the correlation was highest in type IV (r = 0.53). CONCLUSION The type II MRI pattern "fragmentation" was more frequent in the histopathological responder group; and types I and IV in the non-responder group. Type II pattern showed the best endocrine responsiveness and a relatively moderate correlation between sizes obtained from MRI and histology, whereas type IV pattern indicated endocrine resistance but the strongest correlation between MRI and histology.
Collapse
Affiliation(s)
- Joana Reis
- Department of Diagnostic Imaging and Intervention, Akershus University Hospital (AHUS), Postboks 1000, 1478, Lørenskog, Norway. .,Institute of Clinical Medicine, Campus AHUS, University of Oslo, Postboks 1000, 1478, Lørenskog, Norway. .,Translational Cancer Research Group, Akershus University Hospital (AHUS), Postboks 1000, 1478, Lørenskog, Norway.
| | - Owen Thomas
- grid.411279.80000 0000 9637 455XHealth Services Research Department, Akershus University Hospital (AHUS), Postboks 1000, 1478 Lørenskog, Norway
| | - Maryam Lahooti
- grid.411279.80000 0000 9637 455XDepartment of Diagnostic Imaging and Intervention, Akershus University Hospital (AHUS), Postboks 1000, 1478 Lørenskog, Norway
| | - Marianne Lyngra
- grid.411279.80000 0000 9637 455XDepartment of Pathology, Akershus University Hospital (AHUS), Postboks 1000, 1478 Lørenskog, Norway
| | - Hossein Schandiz
- grid.411279.80000 0000 9637 455XDepartment of Pathology, Akershus University Hospital (AHUS), Postboks 1000, 1478 Lørenskog, Norway
| | - Joao Boavida
- grid.411279.80000 0000 9637 455XDepartment of Diagnostic Imaging and Intervention, Akershus University Hospital (AHUS), Postboks 1000, 1478 Lørenskog, Norway
| | - Kjell-Inge Gjesdal
- grid.411279.80000 0000 9637 455XDepartment of Diagnostic Imaging and Intervention, Akershus University Hospital (AHUS), Postboks 1000, 1478 Lørenskog, Norway ,Sunnmøre MR-Clinic, Agrinorbygget, Langelansveg 15, 6010 Ålesund, Norway
| | - Torill Sauer
- grid.5510.10000 0004 1936 8921Institute of Clinical Medicine, Campus AHUS, University of Oslo, Postboks 1000, 1478 Lørenskog, Norway ,grid.411279.80000 0000 9637 455XTranslational Cancer Research Group, Akershus University Hospital (AHUS), Postboks 1000, 1478 Lørenskog, Norway ,grid.411279.80000 0000 9637 455XDepartment of Pathology, Akershus University Hospital (AHUS), Postboks 1000, 1478 Lørenskog, Norway
| | - Jürgen Geisler
- grid.5510.10000 0004 1936 8921Institute of Clinical Medicine, Campus AHUS, University of Oslo, Postboks 1000, 1478 Lørenskog, Norway ,grid.411279.80000 0000 9637 455XTranslational Cancer Research Group, Akershus University Hospital (AHUS), Postboks 1000, 1478 Lørenskog, Norway ,grid.411279.80000 0000 9637 455XDepartment of Oncology, Akershus University Hospital (AHUS), Postboks 1000, 1478 Lørenskog, Norway
| | - Jonn Terje Geitung
- grid.411279.80000 0000 9637 455XDepartment of Diagnostic Imaging and Intervention, Akershus University Hospital (AHUS), Postboks 1000, 1478 Lørenskog, Norway ,grid.5510.10000 0004 1936 8921Institute of Clinical Medicine, Campus AHUS, University of Oslo, Postboks 1000, 1478 Lørenskog, Norway
| |
Collapse
|
10
|
Accuracy of breast MRI in patients receiving neoadjuvant endocrine therapy: comprehensive imaging analysis and correlation with clinical and pathological assessments. Breast Cancer Res Treat 2020; 184:407-420. [PMID: 32789592 PMCID: PMC7599143 DOI: 10.1007/s10549-020-05852-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/31/2020] [Indexed: 11/05/2022]
Abstract
Purpose To assess the accuracy of magnetic resonance imaging (MRI) measurements in locally advanced oestrogen receptor-positive and human epidermal growth factor receptor 2-negative breast tumours before, during and after neoadjuvant endocrine treatment (NET) for evaluation of tumour response in comparison with clinical and pathological assessments. Methods This prospective study enrolled postmenopausal patients treated neoadjuvant with letrozole and exemestane given sequentially in an intra-patient cross-over regimen. Fifty-four patients were initially recruited, but only 35 fulfilled the inclusion criteria and confirmed to participate with a median age of 77. Tumours were scanned with MRI prior to treatment, during the eighth week of treatment and prior to surgery. Additionally, changes in longest diameter on clinical examination (CE) and tumour size at pathology were determined. Pre- and post-operative measurements of tumour size were compared in order to evaluate tumour response. Results The correlation between post-treatment MRI size and pathology was moderate and higher with a correlation coefficient (r) 0.64 compared to the correlation between CE and pathology r = 0.25. Post-treatment MRI and clinical results had a negligible bias towards underestimation of lesion size. Tumour size on MRI and CE had 0.82 cm and 0.52 cm lower mean size than tumour size measured by pathology, respectively. Conclusions The higher correlation between measurements of residual disease obtained on MRI and those obtained with pathology validates the accuracy of imaging assessment during NET. MRI was found to be more accurate for estimating complete responses than clinical assessments and warrants further investigation in larger cohorts to validate this finding. Electronic supplementary material The online version of this article (10.1007/s10549-020-05852-7) contains supplementary material, which is available to authorized users.
Collapse
|
11
|
Bahrami N, Chang G, Kanaya N, Sauer T, Park D, Loeng M, Gravdehaug B, Chen S, Geisler J. Changes in serum estrogenic activity during neoadjuvant therapy with letrozole and exemestane. J Steroid Biochem Mol Biol 2020; 200:105641. [PMID: 32151708 DOI: 10.1016/j.jsbmb.2020.105641] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/21/2020] [Accepted: 02/22/2020] [Indexed: 12/29/2022]
Abstract
The aromatase inhibitors (AIs), letrozole (Femar®/Femara®) and exemestane (Aromasin®), are widely used to treat estrogen receptor (ER) positive breast cancer in postmenopausal patients. In the setting of metastatic breast cancer, these drugs may be used after another causing new responses in selected patients after progressing on the first choice. The precise explanation for this "lack of cross resistance" is still missing. NEOLETEXE is a neoadjuvant, randomized, open-label, cross-over trial. Postmenopausal patients with ER-positive, HER-2 negative, locally advanced breast cancer were enrolled. All patients were randomized to treatment starting with either letrozole or exemestane for at least 2 months followed by another 2 months on the alternative AI. The total estrogenic activities in blood samples were determined using the AroER tri-screen assay developed in the Chen laboratory. Using this highly sensitive assay, estrogenic activity was detected at three time points for all patients. Importantly, a significantly higher total estrogenic activity was found during therapy with exemestane compared to letrozole in 21 out of 26 patients. When letrozole was included in the AroER tri-screen assay, the estrogenic activities in most samples collected during exemestane treatment were further reduced, suggesting that low levels of androgens remained in specimens obtained after exemestane treatment. Our results suggest the AroER tri-screen to be a very sensitive method to estimate the overall estrogen-mediated activity in human samples even during therapy with highly potent aromatase inhibitors. In the present study, serum estrogen activity was significantly higher during exemestane therapy when compared to letrozole therapy.
Collapse
Affiliation(s)
- Nazli Bahrami
- Department of Oncology, Akershus University Hospital (AHUS), Lørenskog, Norway; Department of Breast and Endocrine Surgery, Akershus University Hospital, Lørenskog, Norway
| | - Gregory Chang
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Noriko Kanaya
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Torill Sauer
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Campus AHUS, Norway
| | - Daehoon Park
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | - Marie Loeng
- Department of Oncology, Akershus University Hospital (AHUS), Lørenskog, Norway
| | - Berit Gravdehaug
- Department of Breast and Endocrine Surgery, Akershus University Hospital, Lørenskog, Norway
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital (AHUS), Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Campus AHUS, Norway.
| |
Collapse
|
12
|
Eraso Y. Oestrogen receptors and breast cancer: are we prepared to move forward? A critical review. BIOSOCIETIES 2019. [DOI: 10.1057/s41292-019-00173-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
13
|
Bahrami N, Sauer T, Engebretsen S, Aljabri B, Bemanian V, Lindstrøm J, Lüders T, Kristensen V, Lorentzen A, Loeng M, Ødegård HP, Kvaløy JØ, Vestøl IB, Geisler SB, Gravdehaug B, Gundersen JM, Geisler J. The NEOLETEXE trial: a neoadjuvant cross-over study exploring the lack of cross resistance between aromatase inhibitors. Future Oncol 2019; 15:3675-3682. [DOI: 10.2217/fon-2019-0258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The aromatase inhibitor letrozole (Femar®/Femara®) and the aromatase inactivator exemestane (Aromasin®) differ in their biochemical effect on the aromatase enzyme. Letrozole is a competitive aromatase inhibitor while exemestane binds irreversibly to the aromatase enzyme. This pharmacological difference is of clinical interest since a lack of cross-resistance has been documented. It has been demonstrated in several clinical trials that exemestane may cause a disease regression following resistance to nonsteroidal aromatase inhibitors. The exact mechanism(s) behind this phenomenon is yet unknown. Here, we present the NEOLETEXE trial with the aim of exploring the individual mechanisms involved behind the observed lack of cross resistance. Clinical trial registration: The trial has been approved by the Regional Ethics Committee of South-East Norway (project number 2015/84).
Collapse
Affiliation(s)
- Nazli Bahrami
- Department of Oncology, Akershus University Hospital (AHUS), Lørenskog, Norway
- Department of Breast & Endocrine Surgery (AHUS), Akershus University Hospital, Lørenskog, Norway
- Translational Cancer Research Group, Akershus University Hospital, Lørenskog, Norway
| | - Torill Sauer
- Department of Pathology (AHUS), Akershus University Hospital, Lørenskog, Norway
- Translational Cancer Research Group, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Campus AHUS, University of Oslo, Norway
| | - Siri Engebretsen
- Department of Breast & Endocrine Surgery (AHUS), Akershus University Hospital, Lørenskog, Norway
- Translational Cancer Research Group, Akershus University Hospital, Lørenskog, Norway
| | - Belal Aljabri
- Department of Oncology, Akershus University Hospital (AHUS), Lørenskog, Norway
- Translational Cancer Research Group, Akershus University Hospital, Lørenskog, Norway
| | - Vahid Bemanian
- Department of Gene Technology (AHUS), Akershus University Hospital, Lørenskog, Norway
- Translational Cancer Research Group, Akershus University Hospital, Lørenskog, Norway
| | - Jonas Lindstrøm
- Health Services Research Center (AHUS), Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Campus AHUS, University of Oslo, Norway
| | - Torben Lüders
- Department of Clinical Molecular Biology (AHUS/EpiGen), Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Campus AHUS, University of Oslo, Norway
| | - Vessela Kristensen
- Department of Clinical Molecular Biology (AHUS/EpiGen), Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Campus AHUS, University of Oslo, Norway
| | - Annika Lorentzen
- Translational Cancer Research Group, Akershus University Hospital, Lørenskog, Norway
| | - Marie Loeng
- Translational Cancer Research Group, Akershus University Hospital, Lørenskog, Norway
| | - Hilde Presterud Ødegård
- Department of Oncology, Akershus University Hospital (AHUS), Lørenskog, Norway
- Translational Cancer Research Group, Akershus University Hospital, Lørenskog, Norway
| | - Jan Øyvind Kvaløy
- Department of Oncology, Akershus University Hospital (AHUS), Lørenskog, Norway
- Translational Cancer Research Group, Akershus University Hospital, Lørenskog, Norway
| | - Ingeborg Berge Vestøl
- Department of Oncology, Akershus University Hospital (AHUS), Lørenskog, Norway
- Translational Cancer Research Group, Akershus University Hospital, Lørenskog, Norway
| | - Stephanie Beate Geisler
- Department of Oncology, Akershus University Hospital (AHUS), Lørenskog, Norway
- Translational Cancer Research Group, Akershus University Hospital, Lørenskog, Norway
| | - Berit Gravdehaug
- Department of Breast & Endocrine Surgery (AHUS), Akershus University Hospital, Lørenskog, Norway
- Translational Cancer Research Group, Akershus University Hospital, Lørenskog, Norway
| | - Joanna Majak Gundersen
- Department of Breast & Endocrine Surgery (AHUS), Akershus University Hospital, Lørenskog, Norway
- Translational Cancer Research Group, Akershus University Hospital, Lørenskog, Norway
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital (AHUS), Lørenskog, Norway
- Translational Cancer Research Group, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Campus AHUS, University of Oslo, Norway
| |
Collapse
|
14
|
Lee JI, Yu JH, Anh SG, Lee HW, Jeong J, Lee KS. Aromatase Inhibitors and Newly Developed Nonalcoholic Fatty Liver Disease in Postmenopausal Patients with Early Breast Cancer: A Propensity Score-Matched Cohort Study. Oncologist 2019; 24:e653-e661. [PMID: 30679317 PMCID: PMC6693701 DOI: 10.1634/theoncologist.2018-0370] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 12/12/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Unlike tamoxifen, the relationship between aromatase inhibitor use in postmenopausal patients with breast cancer and nonalcoholic fatty liver disease (NAFLD) has not been delineated. MATERIALS AND METHODS A retrospective analysis of 253 patients with early breast cancer without baseline NAFLD and treated with nonsteroidal aromatase inhibitors was performed. Among them, 220 patients were matched for sex, age, and menstruation status with healthy patients, and the prevalence of NAFLD was compared. NAFLD was determined by hepatic steatosis index in the absence of other known liver diseases. The presence of significant liver fibrosis in patients with NAFLD was determined noninvasively by AST-platelet ratio index, FIB-4 score, and NAFLD fibrosis score (NFS). RESULTS Postmenopausal patients with breast cancer undergoing treatment with aromatase inhibitors had higher prevalence of NAFLD independent of body mass index (BMI) and underlying diabetes mellitus (DM). Although the aromatase inhibitor group showed higher fibrotic burden by NFS, independent of BMI and DM, the proportion of advanced fibrosis did not show statistically significant differences between AI-treated patients and the healthy patients. Those with abnormal baseline fasting glucose levels are suggested to have increased risk of NAFLD development after aromatase inhibitor treatment. In addition, patients with NAFLD developed after aromatase inhibitor use had significantly lower disease-free survival than those without NAFLD, although there was no significant difference in overall survival. CONCLUSION Results of this study suggest that inhibition of estrogen synthesis in postmenopausal women undergoing treatment with aromatase inhibitors could increase the risk of NAFLD, which might have some influence on the prognosis of patients with breast cancer. IMPLICATIONS FOR PRACTICE Unlike tamoxifen, the role of aromatase inhibitor treatment use in postmenopausal patients with breast cancer in development of fatty liver is not well known. In this propensity-matched cohort study, postmenopausal patients with breast cancer treated with aromatase inhibitors had increased risk of nonalcoholic fatty liver disease compared with healthy women after menopause, independent of obesity and diabetes mellitus. The results show possible adverse influence of the newly developed fatty liver on breast cancer disease-free survival and suggest a necessity for further validation. Fatty liver may need to be considered as an adverse event for aromatase inhibitor treatment.
Collapse
Affiliation(s)
- Jung Il Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung-Hwan Yu
- Department of Internal Medicine, Inha University College of Medicine, Incheon, Republic of Korea
| | - Sung Gwe Anh
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Woong Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joon Jeong
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kwan Sik Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
15
|
The beneficial androgenic action of steroidal aromatase inactivators in estrogen-dependent breast cancer after failure of nonsteroidal drugs. Cell Death Dis 2019; 10:494. [PMID: 31235695 PMCID: PMC6591174 DOI: 10.1038/s41419-019-1724-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/13/2019] [Accepted: 06/03/2019] [Indexed: 11/08/2022]
Abstract
Direct treatment of ER (+) breast cancer with Formestane diminishes the tumor within weeks. This is unlikely due to lack of estrogens alone. We proposed that it is the negative influence of androgens on the growth of ER(+) breast cancer. We investigated the influence of Formestane and Exemestane and of their major androgenic metabolites 4-hydroxytestosterone and 17-hydroexemestane on the proliferation of MCF-7 cells and ZR-75-1 cells. Inhibitory effects could be prevented by antiandrogens and siRNA. Activation of the AR in MCF-7 and U2-OS cells was tested by reporter gene assays. In vivo androgenicity was evaluated using the Hershberger assay. Influence on the cell cycle was demonstrated by flow-cytometry. Influence of androgens on the activity of CCND1 was demonstrated by Chip-qPCR. Antitumor activity was determined by topical treatment of DMBA tumors. We found that breast cancer cells can metabolize Formestane and Exemestane to androgenic compounds which inhibit proliferation. This can be explained by hindering the accessibility of CCND1 by histone modification. Androgenic metabolites can abolish the growth of DMBA-tumors and prevent the appearance of new tumors. The lack of cross-resistance between steroidal and nonsteroidal aromatase inhibitors is due to inhibitory effects of androgenic steroidal metabolites on the production of cyclin D1. These sterols not only inhibit proliferation of cancer cells but can also stop the growth of DMBA cancers upon direct absorption into the tumor. The quick and considerable effect on ER(+) tumors may open a new avenue for neodjuvant treatment.
Collapse
|
16
|
Choi MH. Mass spectrometry-based metabolic signatures of sex steroids in breast cancer. Mol Cell Endocrinol 2018; 466:81-85. [PMID: 28928086 DOI: 10.1016/j.mce.2017.09.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 09/15/2017] [Accepted: 09/15/2017] [Indexed: 11/16/2022]
Abstract
Owing to controversy over the effects of steroids on breast cancer pathophysiology, comprehensive quantification of steroid hormones has been extensively considered in both clinical practice and biomarker discovery studies. In contrast to the traditional immunoaffinity-based assays, which show cross-reactivity and have poor validity at low levels of sex steroids, mass spectrometry is becoming a promising tool for measuring steroid levels in complex biological specimens. The Endocrine Society has announced and continuously updated on technical advances to apply high-quality breakthroughs in the clinical sciences. To avoid incorrect estimation of the steroids of interest, however, further emphasis should be made on the efficient separation by chromatography, such as gas and liquid chromatography, prior to mass spectrometric (MS) detection. Recent advances in MS-based analysis of sex steroids associated with breast cancer enable accurate quantification of circulating as well as localized steroids from frozen tissue slices, allowing these assays to be more powerful in clinical practice.
Collapse
Affiliation(s)
- Man Ho Choi
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792, South Korea.
| |
Collapse
|
17
|
McNamara KM, Guestini F, Sauer T, Touma J, Bukholm IR, Lindstrøm JC, Sasano H, Geisler J. In breast cancer subtypes steroid sulfatase (STS) is associated with less aggressive tumour characteristics. Br J Cancer 2018; 118:1208-1216. [PMID: 29563635 PMCID: PMC5943586 DOI: 10.1038/s41416-018-0034-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 01/18/2018] [Accepted: 01/19/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The majority of breast cancer cases are steroid dependent neoplasms, with hormonal manipulation of either CYP19/aromatase or oestrogen receptor alpha axis being the most common therapy. Alternate pathways of steroid actions are documented, but their interconnections and correlations to BC subtypes and clinical outcome could be further explored. METHODS We evaluated selected steroid receptors (Androgen Receptor, Oestrogen Receptor alpha and Beta, Glucocorticoid Receptor) and oestrogen pathways (steroid sulfatase (STS), 17β-hydroxysteroid dehydrogenase 2 (17βHSD2) and aromatase) in a cohort of 139 BC cases from Norway. Using logistic and cox regression analysis, we examined interactions between these and clinical outcomes such as distant metastasis, local relapse and survival. RESULTS Our principal finding is an impact of STS expression on the risk for distant metastasis (p<0.001) and local relapses (p <0.001), HER2 subtype (p<0.015), and survival (p<0.001). The suggestion of a beneficial effect of alternative oestrogen synthesis pathways was strengthened by inverted, but non-significant findings for 17βHSD2. CONCLUSIONS Increased intratumoural metabolism of oestrogens through STS is associated with significantly lower incidence of relapse and/or distant metastasis and correspondingly improved prognosis. The enrichment of STS in the HER2 overexpressing subtype is intriguing, especially given the possible role of HER-2 over-expression in endocrine resistance.
Collapse
Affiliation(s)
- Keely M McNamara
- Department of Anatomic Pathology, School of Graduate Medicine, Tohoku University Japan, Sendai, Japan.
| | - Fouzia Guestini
- Department of Anatomic Pathology, School of Graduate Medicine, Tohoku University Japan, Sendai, Japan
| | - Torill Sauer
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Joel Touma
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Breast- and Endocrine Surgery, Akershus University Hospital, Lørenskog, Norway
| | - Ida Rashida Bukholm
- Department of Breast- and Endocrine Surgery, Akershus University Hospital, Lørenskog, Norway
| | - Jonas C Lindstrøm
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Helse Sør-Øst Health Services Research Centre, Akershus University Hospital, Lørenskog, Norway
| | - Hironobu Sasano
- Department of Anatomic Pathology, School of Graduate Medicine, Tohoku University Japan, Sendai, Japan
| | - Jürgen Geisler
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
18
|
Lane LC, Flowers J, Johnstone H, Cheetham T. Adult height in patients with familial male-limited precocious puberty and the role of an aromatase inhibitor in patient management. J Pediatr Endocrinol Metab 2018; 31:551-560. [PMID: 29654692 DOI: 10.1515/jpem-2017-0363] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 02/12/2018] [Indexed: 11/15/2022]
Abstract
BACKGROUND There is little adult height data in patients with familial male-limited precocious puberty (FMPP) and no management consensus. We assessed the treatment and adult height in local patients with FMPP and those reported in the literature. METHODS Growth data were obtained on four local patients with FMPP and a search performed to obtain management details and adult height data on cases in the literature. UK (90) population standards were used to calculate standard deviation scores (SDS). RESULTS Adult height data were available on 25 men with FMPP of whom 21 were treated. Median adult height SDS of patients was -1.5 SD with a mid-parental target of -0.6 SD (p=0.1). Eight patients (32%) had an adult height above the mid-parental target and seven patients (28%) had a height >2 SD below the mean. The median height SD was -0.03 in untreated patients and +0.5 SD in those receiving an aromatase inhibitor. There was no relationship between height and age at presentation. CONCLUSIONS Aromatase inhibitor therapy is associated with a positive height outcome in FMPP but the outcome with and without intervention is unpredictable. Clinicians need to be cautious when counselling families about the potential height outcome in FMPP.
Collapse
Affiliation(s)
- Laura C Lane
- Department of Paediatric Endocrinology, Great North Children's Hospital, Newcastle-Upon-Tyne, UK
| | - Josephine Flowers
- Department of Paediatrics, Sunderland Royal Hospital, Sunderland, UK
| | - Helen Johnstone
- Department of Paediatric Endocrinology, Great North Children's Hospital, Newcastle-Upon-Tyne, UK
| | - Tim Cheetham
- Department of Paediatric Endocrinology, Great North Children's Hospital, Newcastle-Upon-Tyne, UK.,Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
19
|
|
20
|
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: 0.9] [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
|
21
|
Wang Q, Gao S, Li H, Lv M, Lu C. Long noncoding RNAs (lncRNAs) in triple negative breast cancer. J Cell Physiol 2017; 232:3226-3233. [PMID: 28138992 DOI: 10.1002/jcp.25830] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 01/26/2017] [Indexed: 12/30/2022]
Abstract
Long noncoding RNAs (lncRNAs) are dysregulated in many cancer types, which are believed to play crucial roles in regulating several hallmarks of cancer biology. Triple Negative Breast Cancer (TNBC) is a very aggressive subtype of normal breast cancer, which has features of negativity for ER, PR, and HER2. Great efforts have been made to identify an association between lncRNAs expression profiles and TNBC, and to understand the functional role and molecular mechanism on aberrant-expressed lncRNAs. In this review, we summarized the existed knowledge on the systematics, biology, and function of lncRNAs. The advances from the most recent studies of lncRNAs in the predicament of breast cancer, TNBC, are highlighted, especially the functions of specifically selected lncRNAs. We also discussed the potential value of these lncRNAs in TNBC, providing clues for the diagnosis and treatments of TNBC.
Collapse
Affiliation(s)
- Qiuhong Wang
- Department of Clinical Laboratory, Nantong Maternal and Child Health Care Hospital Affiliated to Nantong University, Nantong, China
| | - Sheng Gao
- Department of Breast, Nanjing Maternal and Child Health Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Haibo Li
- Department of Clinical Laboratory, Nantong Maternal and Child Health Care Hospital Affiliated to Nantong University, Nantong, China
| | - Mingming Lv
- Department of Breast, Nanjing Maternal and Child Health Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China.,Nanjing Maternal and Child Health Medical Institute, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Cheng Lu
- Department of Breast, Nanjing Maternal and Child Health Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| |
Collapse
|
22
|
Jha T, Adhikari N, Halder AK, Saha A. Ligand- and Structure-Based Drug Design of Non-Steroidal Aromatase Inhibitors (NSAIs) in Breast Cancer. Oncology 2017. [DOI: 10.4018/978-1-5225-0549-5.ch004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aromatase is a multienzyme complex overexpressed in breast cancer and responsible for estrogen production. It is the potential target for designing anti-breast cancer drugs. Ligand and Structure-Based Drug Designing approaches (LBDD and SBDD) are involved in development of active and more specific Nonsteroidal Aromatase Inhibitors (NSAIs). Different LBDD and SBDD approaches are presented here to understand their utility in designing novel NSAIs. It is observed that molecules should possess a five or six membered heterocyclic nitrogen containing ring to coordinate with heme portion of aromatase for inhibition. Moreover, one or two hydrogen bond acceptor features, hydrophobicity, and steric factors may play crucial roles for anti-aromatase activity. Electrostatic, van der Waals, and p-p interactions are other important factors that determine binding affinity of inhibitors. HQSAR, LDA-QSAR, GQSAR, CoMFA, and CoMSIA approaches, pharmacophore mapping followed by virtual screening, docking, and dynamic simulation may be effective approaches for designing new potent anti-aromatase molecules.
Collapse
|
23
|
Rezvanpour A, Don-Wauchope AC. Clinical implications of estrone sulfate measurement in laboratory medicine. Crit Rev Clin Lab Sci 2016; 54:73-86. [DOI: 10.1080/10408363.2016.1252310] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Atoosa Rezvanpour
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario,Canada and
| | - Andrew C. Don-Wauchope
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario,Canada and
- Hamilton Regional Laboratory Medicine Program, Hamilton, Ontario, Canada
| |
Collapse
|
24
|
Cotargeting of CYP-19 (aromatase) and emerging, pivotal signalling pathways in metastatic breast cancer. Br J Cancer 2016; 116:10-20. [PMID: 27923036 PMCID: PMC5220158 DOI: 10.1038/bjc.2016.405] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/28/2016] [Accepted: 11/08/2016] [Indexed: 01/08/2023] Open
Abstract
Aromatase inhibition is one of the cornerstones of modern endocrine therapy of oestrogen receptor-positive (ER+) metastatic breast cancer (MBC). The nonsteroidal aromatase inhibitors anastrozole and letrozole, as well as the steroidal aromatase inactivator exemestane, are the preferred drugs and established worldwide in all clinical phases of the disease. However, although many patients suffering from MBC experience an initial stabilisation of their metastatic burden, drug resistance and disease progression occur frequently, following in general only a few months on treatment. Extensive translational research during the past two decades has elucidated the major pathways contributing to endocrine resistance and paved the way for clinical studies investigating the efficacy of novel drug combinations involving aromatase inhibitors and emerging drugable targets like mTOR, PI3K and CDK4/6. The present review summarises the basic research that provided the rationale for new drug combinations involving aromatase inhibitors and the main findings of pivotal clinical trials that have already started to change our way to treat hormone-sensitive MBC. The challenging situation of oestrogen receptor-positive and human epidermal growth factor receptor 2-positive (HER2+) MBC is also shortly reviewed to underline the complexity of the clinical scenario in the heterogeneous subgroups of hormone receptor-positive breast cancer patients and the increasing need for personalised medicine. Finally, we summarise some of the promising findings made with the combination of aromatase inhibitors with other potent endocrine treatment options like fulvestrant, a selective oestrogen receptor downregulator.
Collapse
|
25
|
Hudelist G, Czerwenka K, Keckstein J, Haas C, Fink-Retter A, Gschwantler-Kaulich D, Kubista E, Singer C. Expression of Aromatase and Estrogen Sulfotransferase in Eutopic and Ectopic Endometrium: Evidence for Unbalanced Estradiol Production in Endometriosis. Reprod Sci 2016; 14:798-805. [DOI: 10.1177/1933719107309120] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- G. Hudelist
- Department of Obstetrics and Gynaecology, Division of Special Gynecology, University of Vienna, Austria, Department of Obstetrics and Gynaecology, LKHVillach,Villach,Austria
| | - K. Czerwenka
- Department of Clinical Pathology, Division of Gynecopathology, University of Vienna, Austria
| | - J. Keckstein
- Department of Obstetrics and Gynaecology, LKHVillach,Villach,Austria
| | - C. Haas
- Department of Obstetrics and Gynaecology, Division of Special Gynecology, University of Vienna, Austria
| | - A. Fink-Retter
- Department of Obstetrics and Gynaecology, Division of Special Gynecology, University of Vienna, Austria
| | - D. Gschwantler-Kaulich
- Department of Obstetrics and Gynaecology, Division of Special Gynecology, University of Vienna, Austria
| | - E. Kubista
- Department of Obstetrics and Gynaecology, Division of Special Gynecology, University of Vienna, Austria
| | - C.F. Singer
- Department of Obstetrics and Gynaecology, Division of Special Gynecology, University of Vienna, Austria,
| |
Collapse
|
26
|
Mioranza E, Falci C, Dieci MV, Guarneri V, Conte P. The impact of adjuvant endocrine therapy in early breast cancer on quality-of-life: an overview of prospective trials. ACTA ACUST UNITED AC 2016. [DOI: 10.1080/23809000.2016.1157002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Eleonora Mioranza
- Division of Medical Oncology II, Veneto Istitute of Oncology IOV – IRCCS, Padua, Italy
| | - Cristina Falci
- Division of Medical Oncology II, Veneto Istitute of Oncology IOV – IRCCS, Padua, Italy
| | - Maria Vittoria Dieci
- Division of Medical Oncology II, Veneto Istitute of Oncology IOV – IRCCS, Padua, Italy
- Departement of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Valentina Guarneri
- Division of Medical Oncology II, Veneto Istitute of Oncology IOV – IRCCS, Padua, Italy
- Departement of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Pierfranco Conte
- Division of Medical Oncology II, Veneto Istitute of Oncology IOV – IRCCS, Padua, Italy
- Departement of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| |
Collapse
|
27
|
Thomsen KG, Lyng MB, Elias D, Vever H, Knoop AS, Lykkesfeldt AE, Lænkholm AV, Ditzel HJ. Gene expression alterations associated with outcome in aromatase inhibitor-treated ER+ early-stage breast cancer patients. Breast Cancer Res Treat 2015; 154:483-94. [PMID: 26585578 DOI: 10.1007/s10549-015-3644-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 11/13/2015] [Indexed: 01/05/2023]
Abstract
Aromatase inhibitors (AI), either alone or together with chemotherapy, have become the standard adjuvant treatment for postmenopausal, estrogen receptor-positive (ER+) breast cancer. Although AIs improve overall survival, resistance is still a major clinical problem, thus additional biomarkers predictive of outcome of ER+ breast cancer patients treated with AIs are needed. Global gene expression analysis was performed on ER+ primary breast cancers from patients treated with adjuvant AI monotherapy; half experienced recurrence (median follow-up 6.7 years). Gene expression alterations were validated by qRT-PCR, and functional studies evaluating the effect of siRNA-mediated gene knockdown on cell growth were performed. Twenty-six genes, including TFF3, DACH1, RGS5, and GHR, were shown to exhibit altered expression in tumors from patients with recurrence versus non-recurrent (fold change ≥1.5, p < 0.05), and the gene expression alterations were confirmed using qRT-PCR. Ten of these 26 genes could be linked in a network associated with cellular proliferation, growth, and development. TFF3, which encodes for trefoil factor 3 and is an estrogen-responsive oncogene shown to play a functional role in tamoxifen resistance and metastasis of ER+ breast cancer, was also shown to be upregulated in an AI-resistant cell line model, and reduction of TFF3 levels using TFF3-specific siRNAs decreased the growth of both the AI-resistant and -sensitive parental cell lines. Moreover, overexpression of TFF3 in parental AI-sensitive MCF-7/S0.5 cells resulted in reduced sensitivity to the AI exemestane, whereas TFF3 overexpression had no effect on growth in the absence of exemestane, indicating that TFF3 mediates growth and survival signals that abrogate the growth inhibitory effect of exemestane. We identified a panel of 26 genes exhibiting altered expression associated with disease recurrence in patients treated with adjuvant AI monotherapy, including TFF3, which was shown to exhibit a growth- and survival-promoting effect in the context of AI treatment.
Collapse
Affiliation(s)
- Karina G Thomsen
- Institute of Molecular Medicine, Department of Cancer and Inflammation Research, University of Southern Denmark, J. B. Winslowsvej 25, 5000, Odense, Denmark
| | - Maria B Lyng
- Institute of Molecular Medicine, Department of Cancer and Inflammation Research, University of Southern Denmark, J. B. Winslowsvej 25, 5000, Odense, Denmark
| | - Daniel Elias
- Institute of Molecular Medicine, Department of Cancer and Inflammation Research, University of Southern Denmark, J. B. Winslowsvej 25, 5000, Odense, Denmark
| | - Henriette Vever
- Institute of Molecular Medicine, Department of Cancer and Inflammation Research, University of Southern Denmark, J. B. Winslowsvej 25, 5000, Odense, Denmark
| | - Ann S Knoop
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, 2100, Copenhagen, Denmark
| | - Anne E Lykkesfeldt
- Breast Cancer Group, Cell Death and Metabolism, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | | | - Henrik J Ditzel
- Institute of Molecular Medicine, Department of Cancer and Inflammation Research, University of Southern Denmark, J. B. Winslowsvej 25, 5000, Odense, Denmark.
- Department of Oncology, Odense University Hospital, 5000, Odense, Denmark.
| |
Collapse
|
28
|
Egeland NG, Lunde S, Jonsdottir K, Lende TH, Cronin-Fenton D, Gilje B, Janssen EAM, Søiland H. The Role of MicroRNAs as Predictors of Response to Tamoxifen Treatment in Breast Cancer Patients. Int J Mol Sci 2015; 16:24243-75. [PMID: 26473850 PMCID: PMC4632748 DOI: 10.3390/ijms161024243] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 09/28/2015] [Accepted: 09/30/2015] [Indexed: 12/13/2022] Open
Abstract
Endocrine therapy is a key treatment strategy to control or eradicate hormone-responsive breast cancer. However, resistance to endocrine therapy leads to breast cancer relapse. The recent extension of adjuvant tamoxifen treatment up to 10 years actualizes the need for identifying biological markers that may be used to monitor predictors of treatment response. MicroRNAs are promising biomarkers that may fill the gap between preclinical knowledge and clinical observations regarding endocrine resistance. MicroRNAs regulate gene expression by posttranscriptional repression or degradation of mRNA, most often leading to gene silencing. MicroRNAs have been identified directly in the primary tumor, but also in the circulation of breast cancer patients. The few available studies investigating microRNA in patients suggest that seven microRNAs (miR-10a, miR-26, miR-30c, miR-126a, miR-210, miR-342 and miR-519a) play a role in tamoxifen resistance. Ingenuity Pathway Analysis (IPA) reveals that these seven microRNAs interact more readily with estrogen receptor (ER)-independent pathways than ER-related signaling pathways. Some of these pathways are targetable (e.g., PIK3CA), suggesting that microRNAs as biomarkers of endocrine resistance may have clinical value. Validation of the role of these candidate microRNAs in large prospective studies is warranted.
Collapse
Affiliation(s)
- Nina G Egeland
- Department of Pathology, Stavanger University Hospital, Gerd Ragna Bloch Thorsens Gate 8, 4011 Stavanger, Norway.
- Department of Mathematics and Natural Sciences, University of Stavanger, 4036 Stavanger, Norway.
| | - Siri Lunde
- Department of Breast and Endocrine Surgery, Stavanger University Hospital, 4011 Stavanger, Norway.
| | - Kristin Jonsdottir
- Department of Pathology, Stavanger University Hospital, Gerd Ragna Bloch Thorsens Gate 8, 4011 Stavanger, Norway.
| | - Tone H Lende
- Department of Breast and Endocrine Surgery, Stavanger University Hospital, 4011 Stavanger, Norway.
- Department of Clinical Science, University of Bergen, Postboks 7804, 5020 Bergen, Norway.
| | - Deirdre Cronin-Fenton
- Department of Clinical Epidemiology, Aarhus University, Science Center Skejby, Olof Palmes Allé 43, Aarhus N, 8200 Aarhus, Denmark.
| | - Bjørnar Gilje
- Department of Haematology and Oncology, Stavanger University Hospital, Gerd Ragna Bloch Thorsens Gate 8, 4011 Stavanger, Norway.
| | - Emiel A M Janssen
- Department of Pathology, Stavanger University Hospital, Gerd Ragna Bloch Thorsens Gate 8, 4011 Stavanger, Norway.
- Department of Mathematics and Natural Sciences, University of Stavanger, 4036 Stavanger, Norway.
| | - Håvard Søiland
- Department of Breast and Endocrine Surgery, Stavanger University Hospital, 4011 Stavanger, Norway.
- Department of Clinical Science, University of Bergen, Postboks 7804, 5020 Bergen, Norway.
| |
Collapse
|
29
|
Abstract
Estradiol quantitation is useful in the clinical assessment of diseases like hypogonadism, hirsutism, polycystic ovary syndrome (PCOS), amenorrhea, ovarian tumors and for monitoring response in women receiving aromatase inhibitor therapy. Physiologically relevant serum estradiol concentration in women can span across four orders of magnitude. For example, in women undergoing ovulation induction serum estradiol concentration can range between 250-2000 pg/mL whereas aromatase inhibitor therapy can decrease serum estradiol concentration to <5 pg/mL. While high-through-put automated un-extracted (direct) immunoassays accommodate the growing clinical need for estradiol quantitation, are amenable to implementation by most hospital clinical laboratories, they display a significant loss of specificity and accuracy at low concentrations. Most clinical scenarios (example: estradiol monitoring in fertility treatments) place a modest demand on accuracy and precision of the assay in use but accurate quantitation of estradiol in certain clinical scenarios (pediatric and male patients and for monitoring aromatase inhibitor therapy) can be challenging using currently available immunoassays since the direct immunoassays are prone to issues with sub-optimal accuracy and specificity due to cross reactivity with estradiol conjugates and metabolites. In this review we discuss the bases for the evolution of estradiol assays from extracted (indirect) radio-immunoassays to direct immunoassays to liquid-chromatography tandem mass spectrometry (LC-MS/MS) based assays, discuss technical factors relevant for development and optimization of a LC-MS/MS assay for estradiol and present the details and performance characteristics of an ultra-sensitive LC-MS/MS estradiol assay with a limit of quantitation of 0.2 pg/mL.
Collapse
Affiliation(s)
- Hemamalini Ketha
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, United States
| | - Adam Girtman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, United States
| | - Ravinder J Singh
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, United States.
| |
Collapse
|
30
|
Bayer J, Rune G, Schultz H, Tobia MJ, Mebes I, Katzler O, Sommer T. The effect of estrogen synthesis inhibition on hippocampal memory. Psychoneuroendocrinology 2015; 56:213-25. [PMID: 25863445 DOI: 10.1016/j.psyneuen.2015.03.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/05/2015] [Accepted: 03/05/2015] [Indexed: 11/12/2022]
Abstract
17-Beta-estradiol (E2) facilitates long term-potentiation (LTP) and increases spine synapse density in hippocampal neurons of ovariectomized rodents. Consistent with these beneficial effects on the cellular level, E2 improves hippocampus-dependent memory. A prominent approach to study E2 effects in rodents is the inhibition of its synthesis by letrozole, which reduces LTPs and spine synapse density. In the current longitudinal functional magnetic resonance imaging (fMRI) study, we translated this approach to humans and compared the impact of E2 synthesis inhibition on memory performance and hippocampal activity in post-menopausal women taking letrozole (n = 21) to controls (n = 24). In particular, we employed various behavioral memory paradigms that allow the disentanglement of hippocampus-dependent and -independent memory. Consistent with the literature on rodents, E2 synthesis inhibition specifically impaired hippocampus-dependent memory, however, this did not apply to the same degree to all of the employed paradigms. On the neuronal level, E2 depletion tended to decrease hippocampal activity during encoding, whereas it increased activity in the anterior cingulate and the dorsolateral prefrontal cortex. We thus infer that the inhibition of E2 synthesis specifically impairs hippocampal functioning in humans, whereas the increased prefrontal activity presumably reflects a compensatory mechanism, which is already known from studies on cognitive aging and Alzheimer's disease.
Collapse
Affiliation(s)
- Janine Bayer
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Gabriele Rune
- Department of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Heidrun Schultz
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Michael J Tobia
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Radiology, Pennsylvania State University College of Medicine, Hershey, PA, USA.
| | - Imke Mebes
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Olaf Katzler
- Mammazentrum am Jerusalem Krankenhaus, Hamburg, Germany.
| | - Tobias Sommer
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
31
|
Zucchini G, Geuna E, Milani A, Aversa C, Martinello R, Montemurro F. Clinical utility of exemestane in the treatment of breast cancer. Int J Womens Health 2015; 7:551-63. [PMID: 26064072 PMCID: PMC4455847 DOI: 10.2147/ijwh.s69475] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Breast cancer is the most prevalent cancer in women, causing a significant mortality worldwide. Different endocrine strategies are available for the treatment of hormone-sensitive breast cancer, including antiestrogen tamoxifen and fulvestrant, as well as third-generation aromatase inhibitors (AIs), such as letrozole, anastrozole, and exemestane. In this review, we will focus on exemestane, its clinical use, and its side effects. Exemestane is a steroidal third-generation AI now used in all treatment settings for breast cancer. In the metastatic disease, it has been extensively investigated as the first-, second-, and further-line treatment and it is now registered for the treatment of postmenopausal women with advanced estrogen-receptor-positive breast cancer whose disease has progressed following antiestrogen therapy. A potential lack of cross-resistance with nonsteroidal AIs has been described, giving additional therapeutic opportunities in sequences of endocrine agents. Exemestane is also approved for the adjuvant treatment of postmenopausal early breast cancer, either as upfront monotherapy for 5 years, as a switch following 2–3 years of tamoxifen, or as extended therapy beyond 5 years of adjuvant treatment. New promising data also showed a beneficial effect in young premenopausal early breast cancer patients, when administered together with ovarian suppression. Interesting results have also emerged when exemestane has been investigated as neodjuvant treatment as well as preventive agent in healthy women at high risk for breast cancer. Exemestane is generally well tolerated, with a side effect profile similar to that of other AIs, including menopausal symptoms, arthralgia, and bone loss. In conclusion, exemestane can be considered an effective and well-tolerated endocrine treatment option for all stages of breast cancer.
Collapse
Affiliation(s)
- Giorgia Zucchini
- Investigative Clinical Oncology, Fondazione del Piemonte per l'Oncologia-Candiolo Cancer Institute (IRCCs), Candiolo, Italy
| | - Elena Geuna
- Investigative Clinical Oncology, Fondazione del Piemonte per l'Oncologia-Candiolo Cancer Institute (IRCCs), Candiolo, Italy
| | - Andrea Milani
- Investigative Clinical Oncology, Fondazione del Piemonte per l'Oncologia-Candiolo Cancer Institute (IRCCs), Candiolo, Italy
| | | | | | - Filippo Montemurro
- Investigative Clinical Oncology, Fondazione del Piemonte per l'Oncologia-Candiolo Cancer Institute (IRCCs), Candiolo, Italy
| |
Collapse
|
32
|
Jha T, Adhikari N, Halder AK, Saha A. Ligand- and Structure-Based Drug Design of Non-Steroidal Aromatase Inhibitors (NSAIs) in Breast Cancer. QUANTITATIVE STRUCTURE-ACTIVITY RELATIONSHIPS IN DRUG DESIGN, PREDICTIVE TOXICOLOGY, AND RISK ASSESSMENT 2015. [DOI: 10.4018/978-1-4666-8136-1.ch011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Aromatase is a multienzyme complex overexpressed in breast cancer and responsible for estrogen production. It is the potential target for designing anti-breast cancer drugs. Ligand and Structure-Based Drug Designing approaches (LBDD and SBDD) are involved in development of active and more specific Nonsteroidal Aromatase Inhibitors (NSAIs). Different LBDD and SBDD approaches are presented here to understand their utility in designing novel NSAIs. It is observed that molecules should possess a five or six membered heterocyclic nitrogen containing ring to coordinate with heme portion of aromatase for inhibition. Moreover, one or two hydrogen bond acceptor features, hydrophobicity, and steric factors may play crucial roles for anti-aromatase activity. Electrostatic, van der Waals, and p-p interactions are other important factors that determine binding affinity of inhibitors. HQSAR, LDA-QSAR, GQSAR, CoMFA, and CoMSIA approaches, pharmacophore mapping followed by virtual screening, docking, and dynamic simulation may be effective approaches for designing new potent anti-aromatase molecules.
Collapse
|
33
|
Geissler K, Jäger E, Öhler L, Gisslinger H, Jäger U, Lechner K. Interleukin-10 inhibits autonomous myelopoiesis in patients with myelofibrosis. Eur J Haematol 2014; 95:239-43. [PMID: 25404526 DOI: 10.1111/ejh.12486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2014] [Indexed: 11/30/2022]
Abstract
The spontaneous formation of colony-forming units granulocyte/macrophage (CFU-GM) in semisolid cultures has been shown to be due to the endogenous release of cytokines and/or to the hypersensitivity of cells against growth factors. We have reported that increased autonomous CFU-GM growth is an in vitro characteristic of myelofibrosis (MF) which may reflect aberrant hematopoiesis in vivo. Because of its cytokine synthesis-inhibiting action, we speculated that interleukin-10 (IL-10) may inhibit pathological overproduction of myeloid cells in MF by suppression of autonomous myelopoiesis. In this study, IL-10 significantly inhibited autonomous CFU-GM formation in vitro from peripheral blood mononuclear cells (PB MNC) in 10 of 11 patients with MF tested. In all patients, there was a mean inhibition of 69% ranging from 35% to 100%. Suppression of autonomous CFU-GM formation by IL-10 was dose dependent and reversible by the addition of anti-IL-10 antibodies. Our results indicate that IL-10 is a potentially useful molecule to affect aberrant myelopoiesis in patients with MF.
Collapse
Affiliation(s)
- Klaus Geissler
- 5th Department of Internal Medicine - Oncology/Hematology, Hospital Hietzing, Vienna, Austria.,Ludwig Boltzmann Institute for Clinical Oncology, Vienna, Austria
| | - Eva Jäger
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Leopold Öhler
- Department of Internal Medicine I, St-Josef-Krankenhaus, Vienna, Austria
| | - Heinz Gisslinger
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Ulrich Jäger
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Klaus Lechner
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
34
|
Nantasenamat C, Worachartcheewan A, Prachayasittikul S, Isarankura-Na-Ayudhya C, Prachayasittikul V. QSAR modeling of aromatase inhibitory activity of 1-substituted 1,2,3-triazole analogs of letrozole. Eur J Med Chem 2013; 69:99-114. [DOI: 10.1016/j.ejmech.2013.08.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 07/28/2013] [Accepted: 08/07/2013] [Indexed: 10/26/2022]
|
35
|
Farhane S, Fournier MA, Poirier D. Chemical synthesis, characterisation and biological evaluation of lactonic-estradiol derivatives as inhibitors of 17β-hydroxysteroid dehydrogenase type 1. J Steroid Biochem Mol Biol 2013; 137:322-31. [PMID: 23685015 DOI: 10.1016/j.jsbmb.2013.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 04/28/2013] [Accepted: 05/01/2013] [Indexed: 11/17/2022]
Abstract
To control estradiol (E2) formation, we are interested in synthesizing inhibitors of 17β-hydroxyteroid dehydrogenase type 1 (17β-HSD1). Since the results of docking experiments have shown that E2-lactone derivatives substituted in position 19 or 20 (E-ring) could generate interactions with the active site of the enzyme, we carried out their chemical synthesis. After having prepared the 16β,17β-γ-lactone-E2 in four steps starting from estrone (E1), we introduced the molecular diversity by adding a hydroxymethyl, a methylcarboxylate, a carboxy or an allyl group. The allyl derivative was used as a key intermediate to generate a hydroxyethyl side chain in α or β position. Two lactols were also obtained from two hydroxyalkyl lactones. Enzymatic assays revealed that lactone and lactol derivatives weakly inhibited 17β-HSD1 in homogenized HEK-293 cells overexpressing 17β-HSD1 (34-60% at 1 μM) and in intact T-47D cells expressing 17β-HSD1 (10-40% at 10 μM). This article is part of a Special Issue entitled "Synthesis and biological testing of steroid derivatives as inhibitors".
Collapse
Affiliation(s)
- Siham Farhane
- Laboratory of Medicinal Chemistry, CHU de Québec (CHUL) - Research Center and Laval University, Québec (Québec), G1V 4G2, Canada
| | | | | |
Collapse
|
36
|
Abstract
Following their successful implementation for the treatment of metastatic breast cancer, the 'third-generation' aromatase inhibitors (anastrozole, letrozole, and exemestane) have now become standard adjuvant endocrine treatment for postmenopausal estrogen receptor-positive breast cancers. These drugs are characterized by potent aromatase inhibition, causing >98% inhibition of estrogen synthesis in vivo. A recent meta-analysis found no difference in anti-tumor efficacy between these three compounds. As of today, aromatase inhibitor monotherapy and sequential treatment using tamoxifen followed by an aromatase inhibitor for a total of 5 years are considered equipotent treatment options. However, current trials are addressing the potential benefit of extending treatment duration beyond 5 years. Regarding side effects, aromatase inhibitors are not found associated with enhanced risk of cardiovascular disease, and enhanced bone loss is prevented by adding bisphosphonates in concert for those at danger of developing osteoporosis. However, arthralgia and carpal tunnel syndrome preclude drug administration among a few patients. While recent findings have questioned the use of aromatase inhibitors among overweight and, in particular, obese patients, this problem seems to focus on premenopausal patients treated with an aromatase inhibitor and an LH-RH analog in concert, questioning the efficacy of LH-RH analogs rather than aromatase inhibitors among overweight patients. Finally, recent findings revealing a benefit from adding the mTOR inhibitor everolimus to endocrine treatment indicate targeted therapy against defined growth factor pathways to be a way forward, by reversing acquired resistance to endocrine therapy.
Collapse
Affiliation(s)
- Per Eystein Lønning
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.
| | | |
Collapse
|
37
|
Hu LX, Du YY, Zhang Y, Pan YY. Synergistic effects of exemestane and aspirin on MCF-7 human breast cancer cells. Asian Pac J Cancer Prev 2013; 13:5903-8. [PMID: 23317278 DOI: 10.7314/apjcp.2012.13.11.5903] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE The purpose of this study is to investigate the combined effects of exemestane and aspirin on MCF-7 human breast cancer cells. METHODS Antiproliferative effects of exemestane and aspirin, alone and in combination, on growth of MCF-7 human breast cancer cells were assessed using the MTT assay. Synergistic interaction between the two drugs was evaluated in vitro using the combination index (CI) method. The cell cycle distribution was analyzed by flow cytometry and Western blotting was used to investigate the expression of cyclooxygenase-1, cyclooxygenase-2 and Bcl-2. RESULTS MTT assays indicated that combination treatment obviously decreased the viability of MCF-7 human breast cancer cells compared to individual drug treatment (CI<1). In addition, the combination of exemestane and aspirin exhibited a synergistic inhibition of cell proliferation, significantly arrested the cell cycle in the G0/G1 phase and produced a stronger inhibitory effect on COX-1 and Bcl-2 expression than control or individual drug treatment. CONCLUSION These results indicate that the combination of exemestane and aspirin might become a useful method to the treatment of hormone- dependent breast cancer. The combination of the two inhibitors significantly increased the response as compared to single agent treatment, suggesting that combination treatment could become a highly effective approach for breast cancer.
Collapse
Affiliation(s)
- Li-Xia Hu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | | | | | | |
Collapse
|
38
|
Tilghman SL, Townley I, Zhong Q, Carriere PP, Zou J, Llopis SD, Preyan LC, Williams CC, Skripnikova E, Bratton MR, Zhang Q, Wang G. Proteomic signatures of acquired letrozole resistance in breast cancer: suppressed estrogen signaling and increased cell motility and invasiveness. Mol Cell Proteomics 2013; 12:2440-55. [PMID: 23704778 DOI: 10.1074/mcp.m112.023861] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Aromatase inhibitors, such as letrozole, have become the first-line treatment for postmenopausal women with estrogen-dependent breast cancer. However, acquired resistance remains a major clinical obstacle. Previous studies demonstrated constitutive activation of the MAPK signaling, overexpression of HER2, and down-regulation of aromatase and ERα in letrozole-resistant breast cancer cells. Given the complex signaling network involved in letrozole-refractory breast cancer and the lack of effective treatment for hormone resistance, further investigation of aromatase inhibitor resistance by a novel systems biology approach may reveal previously unconsidered molecular changes that could be utilized as therapeutic targets. This study was undertaken to characterize for the first time global proteomic alterations occurring in a letrozole-resistant cell line. A quantitative proteomic analysis of the whole cell lysates of LTLT-Ca (resistant) versus AC-1 cells (sensitive) was performed to identify significant protein expression changes. A total of 1743 proteins were identified and quantified, of which 411 were significantly up-regulated and 452 significantly down-regulated (p < 0.05, fold change > 1.20). Bioinformatics analysis revealed that acquired letrozole resistance is associated with a hormone-independent, more aggressive phenotype. LTLT-Ca cells exhibited 84% and 138% increase in migration and invasion compared with the control cells. The ROCK inhibitor partially abrogated the enhanced migration and invasion of the letrozole-resistant cells. Flow cytometric analyses also demonstrated an increase in vimentin and twist expression in letrozole-resistance cells, suggesting an onset of epithelial to mesenchymal transition (EMT). Moreover, targeted gene expression arrays confirmed a 28-fold and sixfold up-regulation of EGFR and HER2, respectively, whereas ERα and pS2 were dramatically reduced by 28-fold and 1100-fold, respectively. Taken together, our study revealed global proteomic signatures of a letrozole-resistant cell line associated with hormone independence, enhanced cell motility, EMT and the potential values of several altered proteins as novel prognostic markers or therapeutic targets for letrozole resistant breast cancer.
Collapse
Affiliation(s)
- Syreeta L Tilghman
- Department of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Basu S, Nachat-Kappes R, Caldefie-Chézet F, Vasson MP. Eicosanoids and adipokines in breast cancer: from molecular mechanisms to clinical considerations. Antioxid Redox Signal 2013; 18:323-60. [PMID: 22746381 DOI: 10.1089/ars.2011.4408] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Chronic inflammation is one of the foremost risk factors for different types of malignancies, including breast cancer. Additional risk factors of this pathology in postmenopausal women are weight gain, obesity, estrogen secretion, and an imbalance in the production of adipokines, such as leptin and adiponectin. Various signaling products of transcription factor, nuclear factor-kappaB, in particular inflammatory eicosanoids, reactive oxygen species (ROS), and cytokines, are thought to be involved in chronic inflammation-induced cancer. Together, these key components have an influence on inflammatory reactions in malignant tissue damage when their levels are deregulated endogenously. Prostaglandins (PGs) are well recognized in inflammation and cancer, and they are solely biosynthesized through cyclooxygenases (COXs) from arachidonic acid. Concurrently, ROS give rise to bioactive isoprostanes from arachidonic acid precursors that are also involved in acute and chronic inflammation, but their specific characteristics in breast cancer are less demonstrated. Higher aromatase activity, a cytochrome P-450 enzyme, is intimately connected to tumor growth in the breast through estrogen synthesis, and is interrelated to COXs that catalyze the formation of both inflammatory and anti-inflammatory PGs such as PGE(2), PGF(2α), PGD(2), and PGJ(2) synchronously under the influence of specific mediators and downstream enzymes. Some of the latter compounds upsurge the intracellular cyclic adenosine monophosphate concentration and appear to be associated with estrogen synthesis. This review discusses the role of COX- and ROS-catalyzed eicosanoids and adipokines in breast cancer, and therefore ranges from their molecular mechanisms to clinical aspects to understand the impact of inflammation.
Collapse
Affiliation(s)
- Samar Basu
- Biochemistry, Molecular Biology and Nutrition, University of Auvergne, Clermont-Ferrand, France.
| | | | | | | |
Collapse
|
40
|
Krishnan AV, Swami S, Feldman D. The potential therapeutic benefits of vitamin D in the treatment of estrogen receptor positive breast cancer. Steroids 2012; 77:1107-12. [PMID: 22801352 PMCID: PMC3429709 DOI: 10.1016/j.steroids.2012.06.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 06/11/2012] [Accepted: 06/26/2012] [Indexed: 01/08/2023]
Abstract
Calcitriol (1,25-dihydroxyvitamin D(3)), the hormonally active form of vitamin D, inhibits the growth of many malignant cells including breast cancer (BCa) cells. The mechanisms of calcitriol anticancer actions include cell cycle arrest, stimulation of apoptosis and inhibition of invasion, metastasis and angiogenesis. In addition we have discovered new pathways of calcitriol action that are especially relevant in inhibiting the growth of estrogen receptor positive (ER+) BCa cells. Calcitriol suppresses COX-2 expression and increases that of 15-PGDH thereby reducing the levels of inflammatory prostaglandins (PGs). Our in vitro and in vivo studies show that calcitriol decreases the expression of aromatase, the enzyme that catalyzes estrogen synthesis selectively in BCa cells and in the mammary adipose tissue surrounding BCa, by a direct repression of aromatase transcription via promoter II as well as an indirect effect due to the reduction in the levels of PGs, which are major stimulator of aromatase transcription through promoter II. Calcitriol down-regulates the expression of ERα and thereby attenuates estrogen signaling in BCa cells including the proliferative stimulus provided by estrogens. Thus the inhibition of estrogen synthesis and signaling by calcitriol and its anti-inflammatory actions will play an important role in inhibiting ER+BCa. We hypothesize that dietary vitamin D would exhibit similar anticancer activity due to the presence of the enzyme 25-hydroxyvitamin D-1α-hydroxylase (CYP27B1) in breast cells ensuring conversion of circulating 25-hydroxyvitamin D to calcitriol locally within the breast micro-environment where it can act in a paracrine manner to inhibit BCa growth. Cell culture and in vivo data in mice strongly suggest that calcitriol and dietary vitamin D would play a beneficial role in the prevention and/or treatment of ER+BCa in women.
Collapse
Affiliation(s)
| | | | - David Feldman
- Address correspondence and reprint requests to: David Feldman, MD, Stanford University School of Medicine, 300 Pasteur Drive, Room S025, Stanford CA 94305-5103, Fax: 650 725 7085,
| |
Collapse
|
41
|
Sherman MH, Downes M, Evans RM. Nuclear receptors as modulators of the tumor microenvironment. Cancer Prev Res (Phila) 2011; 5:3-10. [PMID: 22135047 DOI: 10.1158/1940-6207.capr-11-0528] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Over the past several decades of cancer research, the inherent complexity of tumors has become increasingly appreciated. In addition to acquired cell-intrinsic properties, tumor initiation and growth is supported by an abundance of parenchymal, inflammatory, and stromal cell types, which infiltrate and surround the tumor. Accumulating evidence shows that numerous components of this supportive milieu, referred to collectively as the tumor microenvironment, are indeed critical during the process of multistep tumorigenesis. These findings highlight the important interplay between neoplastic cells and tumor-associated cell types, and suggest that therapy should target both neoplastic cells and supportive stromal cells to effectively attenuate tumor growth. The nuclear receptor superfamily encompasses a druggable class of molecules expressed in numerous stromal and parenchymal cell types, whose established physiologic roles suggest their potential as therapeutic and preventive targets in the context of the reactive tumor microenvironment. In this minireview, we discuss recent evidence that tumor-associated inflammation, angiogenesis, and fibrosis can be modulated at the transcriptional level by nuclear receptors and their ligands. As these processes have been widely implicated in cancer initiation, progression, and resistance to current therapy, nuclear receptor ligands targeting the tumor microenvironment may be potent antitumor agents in combination therapies, including for preventing cancer development within high-risk populations.
Collapse
Affiliation(s)
- Mara H Sherman
- Gene Expression Laboratory, Howard Hughes Medical Institute, 10010 N Torrey Pines Road, La Jolla, CA, USA
| | | | | |
Collapse
|
42
|
Barnadas A, Estévez LG, Lluch-Hernández A, Rodriguez-Lescure A, Rodriguez-Sanchez C, Sanchez-Rovira P. An overview of letrozole in postmenopausal women with hormone-responsive breast cancer. Adv Ther 2011; 28:1045-58. [PMID: 22068628 DOI: 10.1007/s12325-011-0075-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Indexed: 01/10/2023]
Abstract
Third-generation aromatase inhibitors (AIs) have proven to be superior to tamoxifen in terms of time to disease progression in patients with hormone receptor (HR) positive (HR+) status and, nowadays, are used in the adjuvant and neoadjuvant settings, and first-line therapy for advanced breast cancer. Letrozole is a third generation AI, as are anastrozole and exemestane. In the past, clinical studies had demonstrated that letrozole was effective as a second-line treatment of metastatic breast cancer. In this paper, pharmacokinetic and pharmacodynamic properties of letrozole are reviewed along with its activity in preclinical and clinical settings. Additionally, the results of important clinical trials such as Breast International Group (BIG) 1-98, which tested the optimal initial adjuvant endocrine treatment and the sequential therapy with letrozole and tamoxifen, MA-17 that evaluates the benefits of extended adjuvant therapy, and other important studies in advanced and neoadjuvant disease, are reviewed. Safety comparisons of treatments are also addressed. Interestingly, about 50% of human epidermal growth factor receptor 2-positive (HER2+) breast cancers are HR+. However, HER2 positivity is a marker of antiestrogen treatment resistance. Because of this, a dual treatment is a logical approach when both receptors are overexpressed. The combination of lapatinib and letrozole leads to a significant improvement in the overall disease outcome. Also, the combination of everolimus plus letrozole has been tested in this setting. In fact, the coadministration of both agents seems to increase the efficacy of letrozole in newly-diagnosed HR+ patients. Once resistance to sequential trastuzumab and AI as monotherapy has been found, trastuzumab and letrozole combined in HR+ and HER2+ patients with advanced breast cancer can overcome resistance to both drugs administered as single agents, according to recently reported results.
Collapse
Affiliation(s)
- Agustí Barnadas
- Medical Oncology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|
43
|
Swami S, Krishnan AV, Wang JY, Jensen K, Peng L, Albertelli MA, Feldman D. Inhibitory effects of calcitriol on the growth of MCF-7 breast cancer xenografts in nude mice: selective modulation of aromatase expression in vivo. Discov Oncol 2011; 2:190-202. [PMID: 21686077 DOI: 10.1007/s12672-011-0073-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Calcitriol (1,25-dihydroxyvitamin D(3)), the hormonally active metabolite of vitamin D, exerts many anticancer effects in breast cancer (BCa) cells. We have previously shown using cell culture models that calcitriol acts as a selective aromatase modulator (SAM) and inhibits estrogen synthesis and signaling in BCa cells. We have now examined calcitriol effects in vivo on aromatase expression, estrogen signaling, and tumor growth when used alone and in combination with aromatase inhibitors (AIs). In immunocompromised mice bearing MCF-7 xenografts, increasing doses of calcitriol exhibited significant tumor inhibitory effects (~50% to 70% decrease in tumor volume). At the suboptimal doses tested, anastrozole and letrozole also caused significant tumor shrinkage when used individually. Although the combinations of calcitriol and the AIs caused a statistically significant increase in tumor inhibition in comparison to the single agents, the cooperative interaction between these agents appeared to be minimal at the doses tested. Calcitriol decreased aromatase expression in the xenograft tumors. Importantly, calcitriol also acted as a SAM in the mouse, decreasing aromatase expression in the mammary adipose tissue, while increasing it in bone marrow cells and not altering it in the ovaries and uteri. As a result, calcitriol significantly reduced estrogen levels in the xenograft tumors and surrounding breast adipose tissue. In addition, calcitriol inhibited estrogen signaling by decreasing tumor ERα levels. Changes in tumor gene expression revealed the suppressive effects of calcitriol on inflammatory and growth signaling pathways and demonstrated cooperative interactions between calcitriol and AIs to modulate gene expression. We hypothesize that cumulatively these calcitriol actions would contribute to a beneficial effect when calcitriol is combined with an AI in the treatment of BCa.
Collapse
Affiliation(s)
- Srilatha Swami
- Department of Medicine-Endocrinology, Stanford University School of Medicine, Room S025, 300 Pasteur Drive, Stanford, CA 94305-5103, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Vanoirbeek E, Krishnan AV, Eelen IG, Verlinden L, Bouillon R, Feldman D, Verstuyf A. The anti-cancer and anti-inflammatory actions of 1,25(OH)₂D₃. Best Pract Res Clin Endocrinol Metab 2011; 25:593-604. [PMID: 21872801 PMCID: PMC3164534 DOI: 10.1016/j.beem.2011.05.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Various epidemiological studies have shown an aetiological link between vitamin D deficiency and cancer incidence. The active metabolite of vitamin D, 1,25-dihydroxyvitamin D₃ [1,25(OH)₂D₃], has potent anti-cancer activities both in vitro and in vivo. These anti-cancer effects are attained by regulating the transcription of numerous genes that are involved in different pathways to reduce tumorigenesis and are dependent on the cancer cell type. Besides reducing cell growth and inducing apoptosis, 1,25(OH)₂D₃ also inhibits angiogenesis and metastasis. Moreover, its potency to inhibit inflammation also contributes to its anti-tumoral activity. Here, we report the different ways in which 1,25(OH)₂D₃ interferes with the malignant processes that are activated in cancer cells.
Collapse
Affiliation(s)
- Els Vanoirbeek
- Laboratory for Experimental Medicine and Endocrinology (LEGENDO), Katholieke Universiteit Leuven, Herestraat 49 bus 901 O&NI, Leuven, Belgium, tel. +32 16 347145, fax +32 16 345934
| | - Aruna V Krishnan
- Dept. of Medicine, Division of Endocrinology, Stanford University School of Medicine 300 Pasteur Drive, Stanford, CA 94305-5103, USA, tel: 650-725-2910, fax: 650-725-7085
| | - Ir Guy Eelen
- Laboratory for Experimental Medicine and Endocrinology (LEGENDO), Katholieke Universiteit Leuven, Herestraat 49 bus 901 O&NI, Leuven, Belgium, tel. +32 16 347145, fax +32 16 345934
| | - Lieve Verlinden
- Laboratory for Experimental Medicine and Endocrinology (LEGENDO), Katholieke Universiteit Leuven, Herestraat 49 bus 901 O&NI, Leuven, Belgium, tel. +32 16 347145, fax +32 16 345934
| | - Roger Bouillon
- Laboratory for Experimental Medicine and Endocrinology (LEGENDO), Katholieke Universiteit Leuven, Herestraat 49 bus 901 O&NI, Leuven, Belgium, tel. +32 16 345970, fax +32 16 345934
| | - David Feldman
- Dept. of Medicine, Division of Endocrinology, Stanford University School of Medicine 300 Pasteur Drive, Stanford, CA 94305-5103, USA, tel: 650-725-2910, fax: 650-725-7085
| | - Annemieke Verstuyf
- Laboratory for Experimental Medicine and Endocrinology (LEGENDO), Katholieke Universiteit Leuven, Herestraat 49 bus 901 O&NI, Leuven, Belgium, tel. +32 16 346209, fax +32 16 345934
| |
Collapse
|
45
|
Lønning PE, Haynes BP, Straume AH, Dunbier A, Helle H, Knappskog S, Dowsett M. Exploring breast cancer estrogen disposition: the basis for endocrine manipulation. Clin Cancer Res 2011; 17:4948-58. [PMID: 21791635 DOI: 10.1158/1078-0432.ccr-11-0043] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although normal breast tissue and breast cancer estrogens are known to be elevated compared with plasma estrogen levels, the mechanism behind this phenomenon has been an issue of debate for 2 decades. If local estrogen aromatization were to be confirmed as the main estrogen source in breast cancer tissue, tissue-specific inhibition of estrogen production, avoiding systemic side effects, would become a potentially attractive option for breast cancer treatment and prevention. Based on recent results from our groups exploring tissue estrogens, together with estrogen-synthesizing and estrogen-regulated gene expression levels, we propose a new model to explain elevated breast tissue estrogen levels. Although local estrogen production may be important, the local contribution is overruled by rapid plasma-to-tissue equilibration, including active uptake of circulating estrogens or enhanced tissue binding. As for breast cancer tissue levels, elevated levels of estradiol may be explained to a large extent by estrogen receptor binding and local conversion of estrone into estradiol. This model indicates that effective suppression of benign and malignant tissue estrogens as a treatment for ER+ breast cancer requires systemic suppression and will not be markedly affected by local enzyme targeting.
Collapse
Affiliation(s)
- Per E Lønning
- Section of Oncology, Institute of Medicine, University of Bergen, and Department of Oncology, Haukeland University Hospital, Bergen, Norway.
| | | | | | | | | | | | | |
Collapse
|
46
|
Lønning PE, Haynes BP, Straume AH, Dunbier A, Helle H, Knappskog S, Dowsett M. Recent data on intratumor estrogens in breast cancer. Steroids 2011; 76:786-91. [PMID: 21439992 DOI: 10.1016/j.steroids.2011.02.040] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The contribution of local synthesis versus circulatory delivery of normal breast as well as breast cancer tissue estrogens has remained a controversial area for decades. Novel data on tissue estrogen levels confirm a positive normal breast tissue to plasma concentration gradient for estrone, and to a smaller extent estradiol. Remarkably, this gradient is similar for pre- and post-menopausal women. Together with pharmacokinetic data on estrogen disposition, these findings suggest plasma and breast tissue estrogens to rapidly equilibrate, with circulating estrogens being a major contributor to breast tissue estrone levels. A likely explanation to the concentration gradient could be the fact that non-polar estrogens easily dissolve into tissue fat compartments as compared to plasma. While intratumor estrone levels are low as compared to benign tissue concentrations, intratumor estradiol is elevated in ER+ tumors. The correlation between intratumor estradiol levels and expression levels of dehydrogenases reducing estrone into estradiol but also intratumor ER concentrations are consistent with intratumor estrogen activation but also a scavenger effect of the ER.
Collapse
Affiliation(s)
- Per E Lønning
- Section of Oncology, Institute of Medicine, University of Bergen, Norway.
| | | | | | | | | | | | | |
Collapse
|
47
|
Geisler J, Sasano H, Chen S, Purohit A. Steroid sulfatase inhibitors: promising new tools for breast cancer therapy? J Steroid Biochem Mol Biol 2011; 125:39-45. [PMID: 21356310 DOI: 10.1016/j.jsbmb.2011.02.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 02/05/2011] [Accepted: 02/07/2011] [Indexed: 12/18/2022]
Abstract
Inhibition of aromatase is currently well-established as the major treatment option of hormone-dependent breast cancer in postmenopausal women. However, despite the effects of aromatase inhibitors in both early and metastatic breast cancer, endocrine resistance may cause relapses of the disease and progression of metastasis. Thus, driven by the success of manipulating the steroidogenic enzyme aromatase, several alternative enzymes involved in steroid synthesis and metabolism have recently been investigated as possible drug targets. One of the most promising targets is the steroid sulfatase (STS) which converts steroid sulfates like estrone sulfate (E1S) and dehydroepiandrosterone sulfate (DHEAS) to estrone (E1) and dehydroepiandrosterone (DHEA), respectively. Estrone and DHEA may thereafter be used for the synthesis of more potent estrogens and androgens that may eventually fuel hormone-sensitive breast cancer cells. The present review summarizes the biology behind steroid sulfatase and its inhibition, the currently available information derived from basic and early clinical trials in breast cancer patients, as well as ongoing research. Article from the Special Issue on Targeted Inhibitors.
Collapse
Affiliation(s)
- Jürgen Geisler
- Institute of Clinical Medicine, Division of Clinical Medicine and Laboratory Sciences, University of Oslo, Norway.
| | | | | | | |
Collapse
|
48
|
Krishnan AV, Feldman D. Mechanisms of the anti-cancer and anti-inflammatory actions of vitamin D. Annu Rev Pharmacol Toxicol 2011; 51:311-36. [PMID: 20936945 DOI: 10.1146/annurev-pharmtox-010510-100611] [Citation(s) in RCA: 322] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Calcitriol, the hormonally active form of vitamin D, is being evaluated in clinical trials as an anti-cancer agent. Calcitriol exerts multiple anti-proliferative, pro-apoptotic, and pro-differentiating actions on various malignant cells and retards tumor growth in animal models of cancer. Calcitriol also exhibits several anti-inflammatory effects including suppression of prostaglandin (PG) action, inhibition of p38 stress kinase signaling, and the subsequent production of pro-inflammatory cytokines and inhibition of NF-κB signaling. Calcitriol also decreases the expression of aromatase, the enzyme that catalyzes estrogen synthesis in breast cancer, both by a direct transcriptional repression and indirectly by reducing PGs, which are major stimulators of aromatase transcription. Other important effects include the suppression of tumor angiogenesis, invasion, and metastasis. These calcitriol actions provide a basis for its potential use in cancer therapy and chemoprevention. We summarize the status of trials involving calcitriol and its analogs, used alone or in combination with known anti-cancer agents.
Collapse
Affiliation(s)
- Aruna V Krishnan
- Department of Medicine, Stanford University School of Medicine, California 94305, USA
| | | |
Collapse
|
49
|
Shehu A, Albarracin C, Devi YS, Luther K, Halperin J, Le J, Mao J, Duan RW, Frasor J, Gibori G. The stimulation of HSD17B7 expression by estradiol provides a powerful feed-forward mechanism for estradiol biosynthesis in breast cancer cells. Mol Endocrinol 2011; 25:754-66. [PMID: 21372145 DOI: 10.1210/me.2010-0261] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Our laboratory has previously cloned and purified an ovarian protein found to be a novel 17β-hydroxysteroid dehydrogenase type 7 enzyme (HSD17B7) (formerly prolactin receptor-associated protein) that converts the weak estrogen, estrone, to the highly potent estradiol. The regulation of this enzyme has not yet been explored. In this report, we show high expression of HSD17B7 in human ductal carcinoma and breast cancer cell lines and present evidence for a strong up-regulation of this enzyme by estradiol at the level of mRNA, protein expression, and promoter activity in MCF-7 cells. The effect of estradiol is mediated by estrogen receptor (ER)α, whereas ERβ prevents this stimulation. ER antagonists, ICI 182,780 and 4-hydroxytamoxifen, prevent estradiol-induced stimulation of the endogenously expressed HSD17B7, suggesting that these inhibitors not only block estradiol action but also its production. We have identified a -185-bp region of the hsd17b7 promoter that is highly conserved among rat, mouse, and human and confers regulation by estradiol in MCF-7 cells. This region is devoid of a classical estradiol-response element but contains a nuclear factor 1 (NF1) site that is essential for estradiol action. We found that estradiol stimulates the recruitment and DNA binding of NF1 to this region of the hsd17b7 promoter. Furthermore, knockdown of NF1 family members, NF1B, NF1A, and NF1X, completely prevents induction of this gene by estradiol. In summary, our findings demonstrate that estradiol stimulates HSD17B7 transcriptional activity in breast cancer cells through a novel mechanism requiring NF1 and strongly suggest a positive feedback mechanism to increase local estradiol synthesis causing growth of estrogen-dependent breast cancers.
Collapse
Affiliation(s)
- Aurora Shehu
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Oster A, Klein T, Henn C, Werth R, Marchais‐Oberwinkler S, Frotscher M, Hartmann RW. Bicyclic Substituted Hydroxyphenylmethanone Type Inhibitors of 17 β‐Hydroxysteroid Dehydrogenase Type 1 (17 β‐HSD1): The Role of the Bicyclic Moiety. ChemMedChem 2011; 6:476-87. [DOI: 10.1002/cmdc.201000457] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Revised: 01/01/2011] [Indexed: 11/11/2022]
Affiliation(s)
- Alexander Oster
- Pharmaceutical and Medicinal Chemistry, Saarland University, & the Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2 3, P.O. Box 151150, 66123 Saarbrücken (Germany), Fax: (+49) 681‐302‐70308
| | - Tobias Klein
- Pharmaceutical and Medicinal Chemistry, Saarland University, & the Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2 3, P.O. Box 151150, 66123 Saarbrücken (Germany), Fax: (+49) 681‐302‐70308
| | - Claudia Henn
- Pharmaceutical and Medicinal Chemistry, Saarland University, & the Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2 3, P.O. Box 151150, 66123 Saarbrücken (Germany), Fax: (+49) 681‐302‐70308
| | - Ruth Werth
- Pharmaceutical and Medicinal Chemistry, Saarland University, & the Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2 3, P.O. Box 151150, 66123 Saarbrücken (Germany), Fax: (+49) 681‐302‐70308
| | - Sandrine Marchais‐Oberwinkler
- Pharmaceutical and Medicinal Chemistry, Saarland University, & the Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2 3, P.O. Box 151150, 66123 Saarbrücken (Germany), Fax: (+49) 681‐302‐70308
| | - Martin Frotscher
- Pharmaceutical and Medicinal Chemistry, Saarland University, & the Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2 3, P.O. Box 151150, 66123 Saarbrücken (Germany), Fax: (+49) 681‐302‐70308
| | - Rolf W. Hartmann
- Pharmaceutical and Medicinal Chemistry, Saarland University, & the Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus C2 3, P.O. Box 151150, 66123 Saarbrücken (Germany), Fax: (+49) 681‐302‐70308
| |
Collapse
|