1
|
Hutten SJ, Jonkers J. MIND the translational gap: Preclinical models of ductal carcinoma in situ. Clin Transl Med 2023; 13:e1376. [PMID: 37620984 PMCID: PMC10449811 DOI: 10.1002/ctm2.1376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023] Open
Affiliation(s)
- Stefan J. Hutten
- Division of Molecular PathologyOncode Institute, Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jos Jonkers
- Division of Molecular PathologyOncode Institute, Netherlands Cancer InstituteAmsterdamThe Netherlands
| |
Collapse
|
2
|
Tong JH, Elmore S, Huang SS, Tachachartvanich P, Manz K, Pennell K, Wilson MD, Borowsky A, La Merrill MA. Chronic Exposure to Low Levels of Parabens Increases Mammary Cancer Growth and Metastasis in Mice. Endocrinology 2023; 164:bqad007. [PMID: 36683225 PMCID: PMC10205179 DOI: 10.1210/endocr/bqad007] [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: 09/09/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/24/2023]
Abstract
Methylparaben (MP) and propylparaben (PP) are commonly used as food, cosmetic, and drug preservatives. These parabens are detected in the majority of US women and children, bind and activate estrogen receptors (ER), and stimulate mammary tumor cell growth and invasion in vitro. Hemizygous B6.FVB-Tg (MMTV-PyVT)634Mul/LellJ female mice (n = 20/treatment) were exposed to MP or PP at levels within the US Food and Drug Administration's "human acceptable daily intake." These paraben-exposed mice had increased mammary tumor volume compared with control mice (P < 0.001) and a 28% and 91% increase in the number of pulmonary metastases per week compared with the control mice, respectively (P < 0.0001). MP and PP caused differential expression of 288 and 412 mammary tumor genes, respectively (false discovery rate < 0.05), a subset of which has been associated with human breast cancer metastasis. Molecular docking and luciferase reporter studies affirmed that MP and PP bound and activated human ER, and RNA-sequencing revealed increased ER expression in mammary tumors among paraben-exposed mice. However, ER signaling was not enriched in mammary tumors. Instead, both parabens strongly impaired tumor RNA metabolism (eg, ribosome, spliceosome), as evident from enriched KEGG pathway analysis of differential mammary tumor gene expression common to both paraben treatments (MP, P < 0.001; PP, P < 0.01). Indeed, mammary tumors from PP-exposed mice had an increased retention of introns (P < 0.05). Our data suggest that parabens cause substantial mammary cancer metastasis in mice as a function of their increasing alkyl chain length and highlight the emerging role of aberrant spliceosome activity in breast cancer metastasis.
Collapse
Affiliation(s)
- Jason H Tong
- Department of Environmental Toxicology, University of California at Davis, Davis, CA 95616, USA
| | - Sarah Elmore
- Department of Environmental Toxicology, University of California at Davis, Davis, CA 95616, USA
| | - Shenq-Shyang Huang
- Department of Environmental Toxicology, University of California at Davis, Davis, CA 95616, USA
| | - Phum Tachachartvanich
- Department of Environmental Toxicology, University of California at Davis, Davis, CA 95616, USA
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | - Katherine Manz
- School of Engineering, Brown University, Providence, RI 02912, USA
| | - Kurt Pennell
- School of Engineering, Brown University, Providence, RI 02912, USA
| | - Machelle D Wilson
- Department of Public Health Sciences, University of California at Davis, Davis, CA 95616, USA
| | - Alexander Borowsky
- Department of Pathology and Laboratory Medicine, University of California at Davis, Sacramento, CA 95817, USA
| | - Michele A La Merrill
- Department of Environmental Toxicology, University of California at Davis, Davis, CA 95616, USA
| |
Collapse
|
3
|
Jacobson M, Mills K, Graves G, Wolfman W, Fortier M. Directive clinique n o 422f : Ménopause et cancer du sein. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2021; 43:1457-1465.e1. [PMID: 34895584 DOI: 10.1016/j.jogc.2021.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
OBJECTIF Proposer des stratégies pour améliorer les soins aux femmes en périménopause ou ménopausées d'après les plus récentes données probantes publiées. POPULATION CIBLE Femmes en périménopause ou ménopausées. BéNéFICES, RISQUES ET COûTS: La population cible bénéficiera des plus récentes données scientifiques publiées que leur communiqueront les fournisseurs de soins de santé. Aucun coût ni préjudice ne sont associés à cette information, car les femmes seront libres de choisir parmi les différentes options thérapeutiques, y compris le statu quo, pour la prise en charge des symptômes et morbidités associés à la ménopause. DONNéES PROBANTES: Les auteurs ont interrogé les bases de données PubMed, Medline et Cochrane Library pour extraire des articles publiés entre 2002 et 2020 en utilisant des termes MeSH spécifiques à chacun des sujets abordés dans les 7 chapitres. MéTHODES DE VALIDATION: Les auteurs ont évalué la qualité des données probantes et la force des recommandations en utilisant le cadre méthodologique d'évaluation, de développement et d'évaluation (GRADE). Voir l'annexe A en ligne (tableau A1 pour les définitions et tableau A2 pour l'interprétation des recommandations fortes et faibles). PROFESSIONNELS CONCERNéS: médecins, y compris gynécologues, obstétriciens, médecins de famille, internistes, urgentologues; infirmières, y compris infirmières autorisées et infirmières praticiennes; pharmaciens; stagiaires, y compris étudiants en médecine, résidents, moniteurs cliniques; et autres fournisseurs de soins auprès de la population cible. RéSUMé POUR TWITTER: Prise en charge de la ménopause chez les survivantes et « présurvivantes » du cancer du sein et les femmes ayant un risque élevé de cancer du sein : mise à jour sur les données récentes. DÉCLARATIONS SOMMAIRES: RECOMMANDATIONS.
Collapse
|
4
|
Jacobson M, Mills K, Graves G, Wolfman W, Fortier M. Guideline No. 422f: Menopause and Breast Cancer. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2021; 43:1450-1456.e1. [PMID: 34895583 DOI: 10.1016/j.jogc.2021.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Provide strategies for improving the care of perimenopausal and postmenopausal women based on the most recent published evidence. TARGET POPULATION Perimenopausal and postmenopausal women. BENEFITS, HARMS, AND COSTS Target population will benefit from the most recent published scientific evidence provided via the information from their health care provider. No harms or costs are involved with this information since women will have the opportunity to choose among the different therapeutic options for the management of the symptoms and morbidities associated with menopause, including the option to choose no treatment. EVIDENCE Databases consulted were PubMed, MEDLINE, and the Cochrane Library for the years 2002-2020, and MeSH search terms were specific for each topic developed through the 7 chapters. VALIDATION METHODS The authors rated the quality of evidence and strength of recommendations using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach. See online Appendix A (Tables A1 for definitions and A2 for interpretations of strong and weak recommendations). INTENDED AUDIENCE: physicians, including gynaecologists, obstetricians, family physicians, internists, emergency medicine specialists; nurses, including registered nurses and nurse practitioners; pharmacists; medical trainees, including medical students, residents, fellows; and other providers of health care for the target population. SUMMARY STATEMENTS RECOMMENDATIONS.
Collapse
|
5
|
Salvatore S, Nappi RE, Casiraghi A, Ruffolo AF, Degliuomini R, Parma M, Leone Roberti Maggiore U, Athanasiou S, Candiani M. Microablative Fractional CO 2 Laser for Vulvovaginal Atrophy in Women With a History of Breast Cancer: A Pilot Study at 4-week Follow-up. Clin Breast Cancer 2021; 21:e539-e546. [PMID: 33745867 DOI: 10.1016/j.clbc.2021.01.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 11/24/2020] [Accepted: 01/05/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Breast cancer (BC) is the most common female cancer worldwide. Menopausal symptoms are a well-known side effect in women with BC and have a significant negative impact on quality of life (QoL) and sexuality. Nowadays, hormonal replacement therapy and local estrogens are the most common prescriptions to treat vulvovaginal (VVA) symptoms. However, in women with a history of BC, proper therapy for such conditions remains an often inadequately addressed clinical problem. A treatment with microablative fractional CO2 laser (MLT) can produce a remodeling of the vaginal connective tissue without causing damage to the surrounding tissue. The aim of this pilot study is to assess the efficacy and safety of MLT for treating VVA symptoms in women with a history of BC at 20-week follow-up since the first laser treatment. PATIENTS AND METHODS Women with BC and VVA symptoms were enrolled in the study and treated with 5 laser applications (one every 4 weeks). The rate of satisfied patients at 20 weeks of follow-up was evaluated with a 5-point Likert scale. Changes of the Vaginal Health Index (VHI) after treatment was compared with baseline. Effects of the laser treatment on VVA symptoms was measured using a 10-cm visual analog scale (VAS). Changes in overall QoL were assessed with a generic QoL questionnaire: the Short Form 12 (SF-12) that we analyzed considering its physical (PCS12) and mental (MCS12) domains. Sexual function was evaluated by the Female Sexual Function Index (FSFI). RESULTS In this prospective cohort study, we enrolled 40 women with a history of BC and who currently were or (Group 2) who had been (Group 1) on treatment with endocrine therapy for their condition. Six (15.0%) women were very satisfied, 25 (62.5%) were satisfied, 6 (15.0%) were uncertain, and 3 (7.5%) were dissatisfied with the MLT. VVA symptoms and VHI improved significantly at 20 weeks from baseline (P < .05) with no differences between the 2 groups (P > .05). In terms of QoL measured by the SF-12, the PCS12 and the MCS12 significantly improved at the 20-week follow-up. A significant improvement in total FSFI and in all domains was reported in both study groups (P < .05) with no differences between groups (P > .05). CONCLUSION MLT was safe and effective in treating VVA symptoms in women with a history of BC, irrespective of being previously or currently on endocrine therapies.
Collapse
Affiliation(s)
- Stefano Salvatore
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; Division of Gynecology and Obstetrics, Urogynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rossella E Nappi
- Research Centre for Reproductive Medicine, Gynecological Endocrinology and Menopause, IRCCS Policlinico S. Matteo Foundation and Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Arianna Casiraghi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; Division of Gynecology and Obstetrics, Urogynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Alessandro F Ruffolo
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; Division of Gynecology and Obstetrics, Urogynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rebecca Degliuomini
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; Division of Gynecology and Obstetrics, Urogynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Parma
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; Division of Gynecology and Obstetrics, Urogynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Stavros Athanasiou
- First Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Athens, Greece
| | - Massimo Candiani
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy; Division of Gynecology and Obstetrics, Urogynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
6
|
Fractional microablative CO2 laser in breast cancer survivors affected by iatrogenic vulvovaginal atrophy after failure of nonestrogenic local treatments: a retrospective study. Menopause 2019; 25:657-662. [PMID: 29286986 DOI: 10.1097/gme.0000000000001053] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Vulvovaginal atrophy (VVA) is a condition frequently observed in menopause. Its symptoms can significantly affect the quality of life of patients. Since VVA is related to estrogen deficiency, chemotherapy and hormone therapy for breast cancer (BC) might cause VVA by inducing menopause. Given the lack of effective treatment for VVA in BC survivors, we retrospectively evaluated the efficacy and tolerability of fractional microablative CO2 laser therapy in these patients. METHODS We treated 82 BC survivors with three cycles of CO2 laser after failure of topical nonestrogenic therapy. The severity of symptoms was assessed with a visual analog scale (VAS) at baseline and after completion of laser therapy. Differences in mean VAS scores of each symptom before and after treatment were assessed with multiple t tests for pairwise comparisons. Multivariate analyses were used to adjust the final mean scores for the main confounding factors. RESULTS Pre versus post-treatment differences in mean VAS scores were significant for sensitivity during sexual intercourse, vaginal dryness, itching/stinging, dyspareunia and dysuria (P < 0.001 for all), bleeding (P = 0.001), probe insertion (P = 0.001), and movement-related pain (P = 0.011). Multivariate analyses confirmed that results were significant, irrespective of patients' age and type of adjuvant therapy. CONCLUSION This study shows that CO2 laser treatment is effective and safe in BC patients with iatrogenic menopause. However, the optimal number of cycles to administer and the need for retreatment remain to be defined. Prospective trials are needed to compare CO2 laser therapy with therapeutic alternatives.
Collapse
|
7
|
Behbod F, Gomes AM, Machado HL. Modeling Human Ductal Carcinoma In Situ in the Mouse. J Mammary Gland Biol Neoplasia 2018; 23:269-278. [PMID: 30145750 PMCID: PMC6244883 DOI: 10.1007/s10911-018-9408-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/22/2018] [Indexed: 12/13/2022] Open
Abstract
Breast cancer development is a multi-step process in which genetic and molecular heterogeneity occurs at multiple stages. Ductal carcinoma arises from pre-invasive lesions such as atypical ductal hyperplasia (ADH) and ductal carcinoma in situ (DCIS), which progress to invasive and metastatic cancer. The feasibility of obtaining tissue samples from all stages of progression from the same patient is low, and thus molecular studies dissecting the mechanisms that mediate the transition from pre-invasive DCIS to invasive carcinoma have been hampered. In the past 25 years, numerous mouse models have been developed that partly recapitulate the histological and biological properties of early stage lesions. In this review, we discuss in vivo model systems of breast cancer progression from syngeneic mouse models to human xenografts, with particular focus on how accurately these models mimic human disease.
Collapse
Affiliation(s)
- Fariba Behbod
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Angelica M Gomes
- Department of Biochemistry and Molecular Biology, Tulane Cancer Center, Tulane University School of Medicine, 1430 Tulane Ave, #8543, New Orleans, LA, USA
| | - Heather L Machado
- Department of Biochemistry and Molecular Biology, Tulane Cancer Center, Tulane University School of Medicine, 1430 Tulane Ave, #8543, New Orleans, LA, USA.
| |
Collapse
|
8
|
Mori H, Cardiff RD, Borowsky AD. Aging Mouse Models Reveal Complex Tumor-Microenvironment Interactions in Cancer Progression. Front Cell Dev Biol 2018; 6:35. [PMID: 29651417 PMCID: PMC5884881 DOI: 10.3389/fcell.2018.00035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/15/2018] [Indexed: 12/15/2022] Open
Abstract
Mouse models and genetically engineered mouse models (GEMM) are essential experimental tools for the understanding molecular mechanisms within complex biological systems. GEMM are especially useful for inferencing phenocopy information to genetic human diseases such as breast cancer. Human breast cancer modeling in mice most commonly employs mammary epithelial-specific promoters to investigate gene function(s) and, in particular, putative oncogenes. Models are specifically useful in the mammary epithelial cell in the context of the complete mammary gland environment. Gene targeted knockout mice including conditional targeting to specific mammary cells can reveal developmental defects in mammary organogenesis and demonstrate the importance of putative tumor suppressor genes. Some of these models demonstrate a non-traditional type of tumor suppression which involves interplay between the tumor susceptible cell and its host/environment. These GEMM help to reveal the processes of cancer progression beyond those intrinsic to cancer cells. Furthermore, the, analysis of mouse models requires appropriate consideration of mouse strain, background, and environmental factors. In this review, we compare aging-related factors in mouse models for breast cancer. We introduce databases of GEMM attributes and colony functional variations.
Collapse
Affiliation(s)
- Hidetoshi Mori
- Center for Comparative Medicine, University of California, Davis, Davis, CA, United States
| | - Robert D Cardiff
- Center for Comparative Medicine, University of California, Davis, Davis, CA, United States.,Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Alexander D Borowsky
- Center for Comparative Medicine, University of California, Davis, Davis, CA, United States.,Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
9
|
Impact of Ospemifene on Quality of Life and Sexual Function in Young Survivors of Cervical Cancer: A Prospective Study. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7513610. [PMID: 28781968 PMCID: PMC5525090 DOI: 10.1155/2017/7513610] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/08/2017] [Accepted: 06/01/2017] [Indexed: 12/16/2022]
Abstract
Background Cervical cancer (CC) treatments impact quality of life (QoL) and sexual function (SF) of survivors. Treatment options to reduce sexual dysfunction are limited. The aim of this study was to assess the effectiveness of ospemifene in CC survivors with clinical signs and symptoms of vulvovaginal atrophy (VVA) focusing on their QoL and SF. Materials and Methods Fifty-two patients with previous diagnosis of stage I-IIa CC suffering from VVA and treated with ospemifene were enrolled into a single arm prospective study. Patient underwent 6 months of therapy. At baseline and after 6 months all subjects performed Vaginal Health Index (VHI). The SF and QoL were measured by The European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire (QLQ) and the Cervical Cancer Module (CXC-24). Results After treatment a significant improvement of each parameter of VHI has been demonstrated. Global health status and emotional and social functioning scores improved significantly. On the contrary, general symptoms scales did not show significant difference from baseline data. Sexual activity, sexual vaginal functioning, body image, and sexual enjoyment scores increased significantly. Conclusion Ospemifene seems to be effective in decreasing the VVA symptoms in CC survivors.
Collapse
|
10
|
Prossnitz ER, Hathaway HJ. What have we learned about GPER function in physiology and disease from knockout mice? J Steroid Biochem Mol Biol 2015; 153:114-26. [PMID: 26189910 PMCID: PMC4568147 DOI: 10.1016/j.jsbmb.2015.06.014] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/24/2015] [Accepted: 06/25/2015] [Indexed: 12/16/2022]
Abstract
Estrogens, predominantly 17β-estradiol, exert diverse effects throughout the body in both normal and pathophysiology, during development and in reproductive, metabolic, endocrine, cardiovascular, nervous, musculoskeletal and immune systems. Estrogen and its receptors also play important roles in carcinogenesis and therapy, particularly for breast cancer. In addition to the classical nuclear estrogen receptors (ERα and ERβ) that traditionally mediate predominantly genomic signaling, the G protein-coupled estrogen receptor GPER has become recognized as a critical mediator of rapid signaling in response to estrogen. Mouse models, and in particular knockout (KO) mice, represent an important approach to understand the functions of receptors in normal physiology and disease. Whereas ERα KO mice display multiple significant defects in reproduction and mammary gland development, ERβ KO phenotypes are more limited, and GPER KO exhibit no reproductive deficits. However, the study of GPER KO mice over the last six years has revealed that GPER deficiency results in multiple physiological alterations including obesity, cardiovascular dysfunction, insulin resistance and glucose intolerance. In addition, the lack of estrogen-mediated effects in numerous tissues of GPER KO mice, studied in vivo or ex vivo, including those of the cardiovascular, endocrine, nervous and immune systems, reveals GPER as a genuine mediator of estrogen action. Importantly, GPER KO mice have also demonstrated roles for GPER in breast carcinogenesis and metastasis. In combination with the supporting effects of GPER-selective ligands and GPER knockdown approaches, GPER KO mice demonstrate the therapeutic potential of targeting GPER activity in diseases as diverse as obesity, diabetes, multiple sclerosis, hypertension, atherosclerosis, myocardial infarction, stroke and cancer.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, United States; University of New Mexico Cancer Center, Albuquerque, NM 87131, United States.
| | - Helen J Hathaway
- Department of Cell Biology & Physiology, University of New Mexico, Albuquerque, NM 87131, United States; University of New Mexico Cancer Center, Albuquerque, NM 87131, United States.
| |
Collapse
|
11
|
Abstract
Breast cancer survivors (BCSs) often suffer from menopausal symptoms induced by systemic treatments, with a consequent negative effect on quality of life. Since the introduction of aromatase inhibitors as the standard therapy for hormone-dependent tumors, genitourinary syndrome of menopause (GSM) has become a main problem for BCSs. This new terminology refers to the wide range of vaginal and urinary symptoms related to menopause, which can be relieved by estrogen therapy. Unfortunately, systemic hormone therapy is contraindicated for BCSs and also vaginal estrogens at standard dosage might influence the risk of recurrence because they cause a significant increase of circulating estrogens. Nonhormonal vaginal moisturizers or lubricants are the first choice for BCSs but only have limited and short-term efficacy. New strategies of management of GSM are now available, including: (1) low-dose or ultra low-dose vaginal estrogens; (2) oral selective estrogen receptor modulators (ospemifene); (3) androgen therapy; (4) physical treatment with vaginal laser; and (5) psychosocial interventions. In this review we discuss and analyze these different options.
Collapse
|
12
|
Wurz GT, Kao CJ, DeGregorio MW. Safety and efficacy of ospemifene for the treatment of dyspareunia associated with vulvar and vaginal atrophy due to menopause. Clin Interv Aging 2014; 9:1939-50. [PMID: 25419123 PMCID: PMC4235480 DOI: 10.2147/cia.s73753] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
During the menopausal transition, women experience a number of symptoms due to declining estrogen levels, including vasomotor symptoms and vulvar and vaginal atrophy (VVA). Unlike vasomotor symptoms, vaginal dryness and dyspareunia, the main symptoms of VVA, typically worsen without treatment and can significantly impact the quality of life. Up to 60% of postmenopausal women may be affected by VVA, but many women unfortunately do not seek treatment due to embarrassment or other factors. After 20+ years in development, ospemifene (Osphena™) was approved by the US Food and Drug Administration in 2013 for treatment of moderate-to-severe dyspareunia associated with VVA due to menopause. As the first non-hormonal alternative to estrogen-based products for this indication, the approval of ospemifene represents a significant milestone in postmenopausal women’s health. Ospemifene is a non-steroidal estrogen receptor agonist/antagonist, also known as a selective estrogen receptor modulator (SERM), from the same chemical class as the breast cancer drugs tamoxifen and toremifene. Unlike other selective estrogen receptor modulators, ospemifene exerts a strong, nearly full estrogen agonist effect in the vaginal epithelium, making it well suited for the treatment of dyspareunia in postmenopausal women. Results of Phase III clinical trials showed that ospemifene significantly improved the vaginal maturation index (decreased parabasal cells and increased superficial cells), decreased vaginal pH, and decreased severity of the self-identified most bothersome symptom (dyspareunia or vaginal dryness) compared to placebo. Long-term safety studies revealed that 60 mg ospemifene given daily for 52 weeks was well tolerated and was not associated with any endometrium or breast-related safety concerns. This review discusses the preclinical and clinical data supporting the use of ospemifene for the treatment of dyspareunia associated with VVA due to menopause and provides an overview of its clinical safety.
Collapse
Affiliation(s)
- Gregory T Wurz
- Department of Internal Medicine, Division of Hematology and Oncology, University of California Davis, Sacramento, CA, USA
| | - Chiao-Jung Kao
- Department of Internal Medicine, Division of Hematology and Oncology, University of California Davis, Sacramento, CA, USA
| | - Michael W DeGregorio
- Department of Internal Medicine, Division of Hematology and Oncology, University of California Davis, Sacramento, CA, USA
| |
Collapse
|
13
|
DeGregorio MW, Zerbe RL, Wurz GT. Ospemifene: a first-in-class, non-hormonal selective estrogen receptor modulator approved for the treatment of dyspareunia associated with vulvar and vaginal atrophy. Steroids 2014; 90:82-93. [PMID: 25087944 DOI: 10.1016/j.steroids.2014.07.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ospemifene is a selective estrogen receptor modulator (SERM) approved for the treatment of dyspareunia associated with vulvar and vaginal atrophy (VVA) due to menopause. As the first non-hormonal treatment for this indication, the approval of ospemifene represents a significant milestone in postmenopausal women's health. Ospemifene is a triphenylethylene similar in chemical structure to tamoxifen and toremifene. Consistent with other SERMs such as tamoxifen, toremifene, and raloxifene, ospemifene possesses a distinctive mix of estrogenic and antiestrogenic tissue-specific effects in bone, breast tissue, serum lipids, and the vagina. Among the approved SERMs, ospemifene is the only agent with a nearly full estrogen agonist effect on the vaginal epithelium while having neutral to slight estrogenic effects in the endometrium, making ospemifene uniquely suited for the treatment of dyspareunia associated with VVA, also known as atrophic vaginitis, which affects up to 50% of postmenopausal women. This review begins with a brief history of the discovery of ospemifene, its mechanism of action, and its preclinical development, with an emphasis on its tissue-specific effects on bone, breast, uterus and endometrium, serum lipids and vagina. A brief discussion on the genotoxicity of ospemifene compared to tamoxifen and toremifene is included. The focus then shifts to the clinical development of ospemifene from Phase I through Phase III. We will close with the FDA approval of ospemifene and a justification of the future clinical evaluation of ospemifene as a potential breast cancer chemopreventive agent, where several preclinical studies in different rodent breast cancer models strongly suggest ospemifene is as effective as tamoxifen.
Collapse
Affiliation(s)
- Michael W DeGregorio
- University of California, Davis, Department of Internal Medicine, Division of Hematology and Oncology, 4501 X Street Suite 3016, Sacramento, CA 95817, USA.
| | - Robert L Zerbe
- QuatRx Pharmaceuticals, 777 East Eisenhower Parkway Suite 100, Ann Arbor, MI 48108, USA.
| | - Gregory T Wurz
- University of California, Davis, Department of Internal Medicine, Division of Hematology and Oncology, 4501 X Street Suite 3016, Sacramento, CA 95817, USA.
| |
Collapse
|
14
|
Cui Y, Zong H, Yan H, Li N, Zhang Y. The Efficacy and Safety of Ospemifene in Treating Dyspareunia Associated with Postmenopausal Vulvar and Vaginal Atrophy: A Systematic Review and Meta‐Analysis. J Sex Med 2014; 11:487-97. [DOI: 10.1111/jsm.12377] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
15
|
Kangas L, Unkila M. Tissue selectivity of ospemifene: pharmacologic profile and clinical implications. Steroids 2013; 78:1273-80. [PMID: 24055829 DOI: 10.1016/j.steroids.2013.09.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 09/03/2013] [Accepted: 09/06/2013] [Indexed: 11/21/2022]
Abstract
The multifactorial consequences of menopausal estrogen deficiency affect numerous tissues throughout the body. Supplemental hormonal therapies carry the burden of a risk/benefit ratio that must be highly individualized. Selective estrogen receptor modulators (SERMs) are estrogen receptor (ER) agonist/antagonists designed to induce benefits comparable with estrogen while minimizing adverse effects. Here, we review the estrogen agonist/antagonist profile of ospemifene, a novel triphenylethylene derivative recently approved to treat dyspareunia, a symptom of vulvar and vaginal atrophy (VVA) due to menopause, both preclinically and clinically. Ospemifene binds ERα and ERβ with approximately equal affinities. In preclinical models, ospemifene increased vaginal and uterine epithelial thickness and mucification to the same extent as estrogen. Ospemifene did not induce endometrial hyperplasia in animal models; there also was no stimulatory effect on endometrial cells. In rat and human mammary cells in vitro, ospemifene evokes a dose-dependent inhibition on estrogen-induced cell responses and cell proliferation, supporting an antiestrogenic effect in breast. In contrast, ospemifene has an estrogenic effect on bone, as seen by improved bone mineral density, strength, mass, and histomorphometry in preclinical models, consistent with improvements in markers of bone resorption and formation in postmenopausal women. Based on the preclinical evidence, ospemifene has beneficial estrogen-like effects on the vaginal epithelium, preliminary evidence to support a neutral endometrial profile, antiproliferative effects in breast, and estrogenic effects in bone. Taken together, especially regarding estrogen-like effects on the vaginal epithelium, ospemifene presents a profile of tissue-specific effects that appear novel among available SERMs and well-suited for the treatment of VVA.
Collapse
|
16
|
Soe LH, Wurz GT, Kao CJ, Degregorio MW. Ospemifene for the treatment of dyspareunia associated with vulvar and vaginal atrophy: potential benefits in bone and breast. Int J Womens Health 2013; 5:605-11. [PMID: 24109197 PMCID: PMC3792833 DOI: 10.2147/ijwh.s39146] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ospemifene is a selective estrogen receptor modulator (SERM), or estrogen receptor agonist/antagonist, that was recently approved by the US Food and Drug Administration for the treatment of dyspareunia associated with vulvar and vaginal atrophy, a chronic condition that affects up to 60% of postmenopausal women. Ospemifene is the first and only nonestrogen compound approved for this indication. Compared with other approved SERMs, such as tamoxifen, toremifene, bazedoxifene, and raloxifene, the estrogen-like effects of ospemifene in the vaginal epithelium are unique. This review first discusses the rationale for developing ospemifene, including its mechanism of action, and then focuses on the clinical development of ospemifene for the treatment of dyspareunia associated with vulvar and vaginal atrophy. Included are discussions of the effects of ospemifene on the endometrium, serum lipids, coagulation markers, bone, and breast cancer. In conclusion, ospemifene is a SERM with a unique estrogen agonist/antagonist tissue profile that was recently approved in the US for the treatment of dyspareunia associated with vulvar and vaginal atrophy in postmenopausal women. Ospemifene warrants further clinical investigation for the treatment and prevention of osteoporosis and breast cancer.
Collapse
Affiliation(s)
- Lin H Soe
- Department of Internal Medicine, Division of Hematology and Oncology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | | | | | | |
Collapse
|
17
|
Abstract
Vulvar and vaginal atrophy (VVA) is a chronic, progressive medical condition prevalent among postmenopausal women, which produces symptoms such as dyspareunia, vaginal dryness, and vaginal irritation. Currently, the only prescription options are systemic and vaginal estrogen therapies that may be limited by concerns about long-term safety and breast cancer risk. Ospemifene is a tissue-selective estrogen agonist/antagonist (a selective estrogen receptor modulator) recently approved by the US Food and Drug Administration for treatment of dyspareunia, a symptom of VVA, due to menopause. Ospemifene, the first nonestrogen oral treatment for this indication, may provide an alternative to treatment with estrogen. Animal models with ospemifene suggest an inhibitory effect on growth of malignant breast tissue, but animal data cannot necessarily be extrapolated to humans. Clinical trials, including 3 long-term studies assessing the overall safety of ospemifene, support that ospemifene is generally well tolerated, with beneficial effects on the vagina, neutral effects on the breast, and minimal effects on the endometrium.
Collapse
Affiliation(s)
- Sarah L Berga
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
18
|
Maximov PY, Lee TM, Jordan VC. The discovery and development of selective estrogen receptor modulators (SERMs) for clinical practice. CURRENT CLINICAL PHARMACOLOGY 2013; 8:135-55. [PMID: 23062036 PMCID: PMC3624793 DOI: 10.2174/1574884711308020006] [Citation(s) in RCA: 251] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 07/11/2012] [Accepted: 10/03/2012] [Indexed: 01/13/2023]
Abstract
Selective estrogen receptor modulators (SERMs) are structurally different compounds that interact with intracellular estrogen receptors in target organs as estrogen receptor agonists or antagonists. These drugs have been intensively studied over the past decade and have proven to be a highly versatile group for the treatment of different conditions associated with postmenopausal women's health, including hormone responsive cancer and osteoporosis. Tamoxifen, a failed contraceptive is currently used to treat all stages of breast cancer, chemoprevention in women at high risk for breast cancer and also has beneficial effects on bone mineral density and serum lipids in postmenopausal women. Raloxifene, a failed breast cancer drug, is the only SERM approved internationally for the prevention and treatment of postmenopausal osteoporosis and vertebral fractures. However, although these SERMs have many benefits, they also have some potentially serious adverse effects, such as thromboembolic disorders and, in the case of tamoxifen, uterine cancer. These adverse effects represent a major concern given that long-term therapy is required to prevent osteoporosis or prevent and treat breast cancer. The search for the 'ideal' SERM, which would have estrogenic effects on bone and serum lipids, neutral effects on the uterus, and antiestrogenic effects on breast tissue, but none of the adverse effects associated with current therapies, is currently under way. Ospemifene, lasofoxifene, bazedoxifene and arzoxifene, which are new SERM molecules with potentially greater efficacy and potency than previous SERMs, have been investigated for use in the treatment and prevention of osteoporosis. These drugs have been shown to be comparably effective to conventional hormone replacement therapy in animal models, with potential indications for an improved safety profile. Clinical efficacy data from ongoing phase III trials are available or are awaited for each SERM so that a true understanding of the therapeutic potential of these compounds can be obtained. In this article, we describe the discovery and development of the group of medicines called SERMs. The newer SERMs in late development: ospemifene, lasofoxifene, bazedoxifene, are arzoxifene are described in detail.
Collapse
Affiliation(s)
- Philipp Y Maximov
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3970 Reservoir Rd NW, Research Building, Suite E204A, Washington, DC 20057, USA
| | - Theresa M Lee
- Division of Hematology and Oncology, Georgetown University Medical Center, 3970 Reservoir Rd NW, Washington, DC 20057, USA
| | - V. Craig Jordan
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3970 Reservoir Rd NW, Research Building, Suite E204A, Washington, DC 20057, USA
| |
Collapse
|
19
|
Abstract
The incidence and severity of vulvovaginal atrophy (VVA) in postmenopausal breast cancer patients has a significant impact on quality of life. While the etiology of VVA is primarily related to low estrogen levels seen in menopause, women with breast cancer have an added risk of VVA induced by a combination of chemotherapy, hormonal therapy, and menopause. Ospemifene is a new, non-hormonal selective estrogen receptor modulator (SERM) triphenylethylene derivative that is effective in treating VVA in postmenopausal women. Although other SERMs have antagonistic effects on the vagina, ospemifene exerts an estrogen-like effect on the vaginal epithelium. This review will focus on data demonstrating the antiestrogenic activity of ospemifene in several unique breast cancer animal models, and the implications for utilizing ospemifene in patients with breast cancer suffering from VVA. Additional research addressing the expanded use of ospemifene in breast cancer patients is also warranted.
Collapse
|
20
|
Buckle T, Kuil J, van den Berg NS, Bunschoten A, Lamb HJ, Yuan H, Josephson L, Jonkers J, Borowsky AD, van Leeuwen FWB. Use of a single hybrid imaging agent for integration of target validation with in vivo and ex vivo imaging of mouse tumor lesions resembling human DCIS. PLoS One 2013; 8:e48324. [PMID: 23326303 PMCID: PMC3543428 DOI: 10.1371/journal.pone.0048324] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 09/24/2012] [Indexed: 12/11/2022] Open
Abstract
Screening of biomarker expression levels in tumor biopsy samples not only provides an assessment of prognostic and predictive factors, but may also be used for selection of biomarker-specific imaging strategies. To assess the feasibility of using a biopsy specimen for a personalized selection of an imaging agent, the chemokine receptor 4 (CXCR4) was used as a reference biomarker.
Collapse
MESH Headings
- Animals
- Carcinoma, Intraductal, Noninfiltrating/diagnosis
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Cell Line, Tumor
- Diagnosis, Differential
- Diagnostic Imaging/methods
- Feasibility Studies
- Flow Cytometry
- Fluorescent Dyes/chemistry
- Fluorescent Dyes/metabolism
- Humans
- Immunohistochemistry
- Mammary Neoplasms, Experimental/diagnosis
- Mammary Neoplasms, Experimental/metabolism
- Mice
- Molecular Structure
- Peptides/chemistry
- Peptides/metabolism
- Receptors, CXCR4/metabolism
- Reproducibility of Results
- Sensitivity and Specificity
- Tomography, Emission-Computed, Single-Photon
- Tomography, X-Ray Computed
Collapse
Affiliation(s)
- Tessa Buckle
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands
- Departments of Radiology and Nuclear Medicine, Netherlands Cancer Institute- Antoni van Leeuwenhoekhuis, Amsterdam, The Netherlands
| | - Joeri Kuil
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands
- Departments of Radiology and Nuclear Medicine, Netherlands Cancer Institute- Antoni van Leeuwenhoekhuis, Amsterdam, The Netherlands
| | - Nynke S. van den Berg
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands
- Departments of Radiology and Nuclear Medicine, Netherlands Cancer Institute- Antoni van Leeuwenhoekhuis, Amsterdam, The Netherlands
| | - Anton Bunschoten
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands
- Departments of Radiology and Nuclear Medicine, Netherlands Cancer Institute- Antoni van Leeuwenhoekhuis, Amsterdam, The Netherlands
| | - Hildo J. Lamb
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hushan Yuan
- Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, United States
| | - Lee Josephson
- Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, United States
| | - Jos Jonkers
- Division of Cell Biology, Netherlands Cancer Institute–Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Alexander D. Borowsky
- Department of Pathology and Laboratory Medicine, Center for Comparative Medicine, School of Medicine, University of California at Davis, Sacramento, California, United States of America
| | - Fijs W. B. van Leeuwen
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands
- Departments of Radiology and Nuclear Medicine, Netherlands Cancer Institute- Antoni van Leeuwenhoekhuis, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
21
|
Caligiuri I, Rizzolio F, Boffo S, Giordano A, Toffoli G. Critical choices for modeling breast cancer in transgenic mouse models. J Cell Physiol 2012; 227:2988-91. [PMID: 22170180 DOI: 10.1002/jcp.24031] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Modeling breast cancer in the mouse has helped to better define the heterogeneity of human breast cancer. In the recent past, it has become evident that some limitations have restricted the potential benefits that can be achieved with this approach. In this review, we highlight some key points that should be taken into account when the mouse is used, with special emphasis on transgenic models.
Collapse
Affiliation(s)
- Isabella Caligiuri
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
22
|
Ospemifene and 4-hydroxyospemifene effectively prevent and treat breast cancer in the MTag.Tg transgenic mouse model. Menopause 2012; 19:96-103. [PMID: 21926925 DOI: 10.1097/gme.0b013e318223e82a] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Ospemifene, a new drug indicated for the treatment of vulvovaginal atrophy, has completed phase III clinical trials. A condition affecting millions of women worldwide, vulvovaginal atrophy has long been treated with estrogen therapy. Estrogen treatment carries with it risks of thromboembolism, endometrial proliferative effects, and breast cancer promotion. In this study, we test the effects of three dosing levels of ospemifene in both the prevention and treatment of breast cancer in the MTag.Tg mouse model. METHODS The polyomavirus middle-T transgenic mouse model (MTag.Tg), which produces synchronized, multifocal mammary tumors in the immunologically intact C57BL/6 background, was used to examine the impact of ospemifene treatment. First, a cell line derived from an MTag.Tg mouse tumor (MTag 34) was treated in vitro with ospemifene and its major metabolite, 4-hydroxyospemifene (4-OH ospemifene). MTag.Tg mice were treated daily by gavage with three different doses of ospemifene (5, 25, and 50 mg/kg) before or after the development of mammary tumors. Survival and tumor development results were used to determine the effect of ospemifene treatment on mammary tumors in both the preventive and treatment settings. RESULTS Tumors and the MTag 34 cell line were positive for estrogen receptor expression. The MTag 34 line was not stimulated by ospemifene or its major, active metabolite 4-OH ospemifene in vitro. Ospemifene increased survival time and exerted an antitumor effect on the development and growth of estrogen receptor-positive mammary tumors in the MTag.Tg mouse model at the 50-mg/kg dose. The levels of ospemifene and 4-OH ospemifene in both the tumors and plasma of mice confirmed the dosing. Ospemifene did not exert an estrogenic effect in the breast tissue at doses equivalent to human dosing. CONCLUSIONS Ospemifene prevents and treats estrogen receptor-positive MTag.Tg mammary tumors in this immune-intact mouse model in a dose-dependent fashion. Ospemifene drug levels in the plasma of treated mice were comparable with those found in humans. Combined with our previous data, ospemifene does not seem to pose a breast cancer risk in animals and slows down cancer development and progression in the MTag.Tg model.
Collapse
|
23
|
Buckle T, van Berg NS, Kuil J, Bunschoten A, Oldenburg J, Borowsky AD, Wesseling J, Masada R, Oishi S, Fujii N, van Leeuwen FWB. Non-invasive longitudinal imaging of tumor progression using an (111)indium labeled CXCR4 peptide antagonist. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2011; 2:99-109. [PMID: 23133805 PMCID: PMC3478110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 11/05/2011] [Indexed: 06/01/2023]
Abstract
The chemokine receptor 4 (CXCR4) is a biomarker that is over-expressed in ductal carcinoma in situ (DCIS). Hence, CXCR4-targeted (molecular) imaging approaches may have diagnostic value in such a challenging, premalignant lesion. The indium labeled CXCR4 peptide-antagonist, (111)In-DTPA-Ac-TZ14011, was used to visualize CXCR4-expression in a mammary intraepithelial neoplastic outgrowth (MIN-O) mouse tumor model resembling human DCIS. MIN-O lesion development was longitudinally monitored using SPET/CT and tracer uptake was compared to uptake in control lesions. Expression of CXCR4 was validated using immunohistochemistry and flow cytometric analysis. The uptake of (111)In-DTPA-Ac-TZ14011 was related to tumor angiogenesis using (111)In-cDTPA-[RGDfK]. Twenty-four hours after tracer injection, MIN-O lesions could be discriminated from low CXCR4-expressing control tumors, while the degree of angiogenesis based on the α(v)β(3) integrin expression in both tumor types was similar. The uptake of (111)In-DTPA-Ac-TZ14011 in early MIN-O lesions was significantly lower than in larger intermediate and late-stage lesions, two-and-a-half-times (p=0.03) and seven-times (p=0.002), respectively. Intermediate and late stage lesions show a higher degree of membranous CXCR4-staining at immunohistochemistry and flow cytometric analysis. From this study we can conclude that (111)In-DTPA-Ac-TZ14011 can be used to visualize the CXCR4-expression in MIN-O lesions longitudinally.
Collapse
Affiliation(s)
- Tessa Buckle
- Departments of Radiology and Nuclear Medicine, The Netherlands
Cancer Institute - Antoni van Leeuwenhoek Hospital (NKI-AvL)Amsterdam, The Netherlands
- Department of Radiology, Interventional Molecular Imaging group,
Leiden University Medical Center (LUMC)Leiden, The Netherlands
| | - Nynke S van Berg
- Departments of Radiology and Nuclear Medicine, The Netherlands
Cancer Institute - Antoni van Leeuwenhoek Hospital (NKI-AvL)Amsterdam, The Netherlands
- Department of Radiology, Interventional Molecular Imaging group,
Leiden University Medical Center (LUMC)Leiden, The Netherlands
| | - Joeri Kuil
- Departments of Radiology and Nuclear Medicine, The Netherlands
Cancer Institute - Antoni van Leeuwenhoek Hospital (NKI-AvL)Amsterdam, The Netherlands
- Department of Radiology, Interventional Molecular Imaging group,
Leiden University Medical Center (LUMC)Leiden, The Netherlands
| | - Anton Bunschoten
- Departments of Radiology and Nuclear Medicine, The Netherlands
Cancer Institute - Antoni van Leeuwenhoek Hospital (NKI-AvL)Amsterdam, The Netherlands
- Department of Radiology, Interventional Molecular Imaging group,
Leiden University Medical Center (LUMC)Leiden, The Netherlands
| | - Joppe Oldenburg
- Departments of Radiology and Nuclear Medicine, The Netherlands
Cancer Institute - Antoni van Leeuwenhoek Hospital (NKI-AvL)Amsterdam, The Netherlands
| | - Alexander D Borowsky
- Department of Pathology and Laboratory Medicine, Center for
Comparative Medicine, School of Medicine, University of California at
DavisSacramento, US
| | - Jelle Wesseling
- Department of Pathology, The Netherlands Cancer Institute - Antoni
van Leeuwenhoek Hospital (NKI-AvL)Amsterdam, The Netherlands
| | - Ryo Masada
- Graduate School of Pharmaceutical Sciences, Kyoto
UniversityKyoto, Japan
| | - Shinya Oishi
- Graduate School of Pharmaceutical Sciences, Kyoto
UniversityKyoto, Japan
| | - Nobutaka Fujii
- Graduate School of Pharmaceutical Sciences, Kyoto
UniversityKyoto, Japan
| | - Fijs WB van Leeuwen
- Departments of Radiology and Nuclear Medicine, The Netherlands
Cancer Institute - Antoni van Leeuwenhoek Hospital (NKI-AvL)Amsterdam, The Netherlands
- Department of Radiology, Interventional Molecular Imaging group,
Leiden University Medical Center (LUMC)Leiden, The Netherlands
| |
Collapse
|
24
|
Kuil J, Buckle T, Oldenburg J, Yuan H, Josephson L, van Leeuwen FW. Hybrid peptide dendrimers for imaging of chemokine receptor 4 (CXCR4) expression. Mol Pharm 2011; 8:2444-53. [PMID: 22085282 PMCID: PMC3711081 DOI: 10.1021/mp200401p] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The chemokine receptor 4 (CXCR4), which is overexpressed in many types of cancer, is an emerging target in the field of molecular imaging and therapeutics. The CXCR4 binding of several peptides, including the cyclic Ac-TZ14011, has already been validated. In this study mono-, di- and tetrameric Ac-TZ14011-containing dendrimers were prepared and functionalized with a multimodal (hybrid) label, consisting of a Cy5.5-like fluorophore and a DTPA chelate. Confocal microscopy revealed that all three dendrimers were membrane bound at 4 °C, consistent with CXCR4 binding in vitro. The unlabeled dimer and tetramer had a somewhat lower affinity for CXCR4 than the unlabeled monomer. However, when labeled with the multimodal label the CXCR4 affinity of the dimer and tetramer was considerably higher compared to that of the labeled monomer. On top of that, biodistribution studies revealed that the additional peptides in the dimer and tetramer reduced nonspecific muscle uptake. Thus, multimerization of the cyclic Ac-TZ14011 peptide reduces the negative influence of the multimodal label on the receptor affinity and the biodistribution.
Collapse
Affiliation(s)
- Joeri Kuil
- Division of Diagnostic Oncology, the Netherlands Cancer Institute Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands
| | - Tessa Buckle
- Division of Diagnostic Oncology, the Netherlands Cancer Institute Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands
| | - Joppe Oldenburg
- Division of Diagnostic Oncology, the Netherlands Cancer Institute Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands
| | - Hushan Yuan
- Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Building 149, 13th Street, Charlestown, MA 02129, USA
| | - Lee Josephson
- Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Building 149, 13th Street, Charlestown, MA 02129, USA
| | - Fijs W.B. van Leeuwen
- Division of Diagnostic Oncology, the Netherlands Cancer Institute Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
25
|
Borowsky AD. Choosing a mouse model: experimental biology in context--the utility and limitations of mouse models of breast cancer. Cold Spring Harb Perspect Biol 2011; 3:a009670. [PMID: 21646376 DOI: 10.1101/cshperspect.a009670] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Genetically engineered mice are critical experimental models for the study of breast cancer biology. Transgenic mice, employing strong mammary epithelial promoters to drive oncogenes, develop carcinomas with phenotypes corresponding to the molecular pathway activated. Gene-targeted (knockout) mice, in which tumor suppressors are deleted, develop mammary neoplasms with phenotypes primarily including patterns seen in spontaneous mouse mammary tumors, albeit at higher rates. Improved genetic engineering, using inducible gene expression, somatic gene transduction, conditional alleles, and crossbreeding for combined/compound genetic engineering yields precise molecular models with exquisite experimental control and phenotypes with comparative pathologic validity. Mammary gland transplantation technology adds a practical and validated method for assessing biologic behavior of selected mammary tissues. Overall, the many mouse models available are a rich resource for experimental biology with phenocopies of breast cancer subtypes, and a variety of practical advantages. The challenge is matching the model to the experimental question.
Collapse
Affiliation(s)
- Alexander D Borowsky
- Department of Pathology and Center for Comparative Medicine, University of California at Davis, Davis, California 95616, USA.
| |
Collapse
|
26
|
Estévez LG, Álvarez I, Seguí MÁ, Muñoz M, Margelí M, Miró C, Rubio C, Lluch A, Tusquets I. Current perspectives of treatment of ductal carcinoma in situ. Cancer Treat Rev 2010; 36:507-17. [DOI: 10.1016/j.ctrv.2010.03.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 03/15/2010] [Accepted: 03/21/2010] [Indexed: 11/16/2022]
|
27
|
Ospemifene effectively treats vulvovaginal atrophy in postmenopausal women: results from a pivotal phase 3 study. Menopause 2010; 17:480-6. [PMID: 20032798 DOI: 10.1097/gme.0b013e3181c1ac01] [Citation(s) in RCA: 182] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE The aim of this study was to study the efficacy and safety of ospemifene, a new selective estrogen receptor modulator, in the treatment of vulvovaginal atrophy in postmenopausal women. METHODS A randomized, double-blind phase 3 study in which 826 postmenopausal women were randomized 1:1:1 to receive treatment with ospemifene 30 or 60 mg/day or placebo orally for 12 weeks was conducted. The primary inclusion criteria were having 5% or less superficial cells on the vaginal smear (maturation index), vaginal pH greater than 5.0, and at least one moderate or severe symptom of vulvovaginal atrophy. The four coprimary endpoints were the change from baseline to 12 weeks in the percentage of superficial and parabasal cells on the vaginal smear, change in vaginal pH, and change in severity of most bothersome symptom (vaginal dryness or dyspareunia) compared with placebo. All participants were given a nonhormonal vaginal lubricant for use as needed. RESULTS Ospemifene was statistically significantly superior to placebo in each of the coprimary endpoints at the 60-mg dose. Statistically significant results were achieved for all coprimary endpoints with the 30-mg dose except for dyspareunia. Ospemifene was well tolerated at both doses and demonstrated a favorable safety profile. CONCLUSIONS Ospemifene was shown to be effective and well tolerated for the treatment of the symptoms of vaginal dryness and dyspareunia associated with vulvovaginal atrophy over and above the use of provided lubricants.
Collapse
|
28
|
Mishra R, Tiwari A, Bhadauria S, Mishra J, Murthy PK, Murthy PSR. Therapeutic effect of centchroman alone and in combination with glycine soya on 7,12-dimethylbenz[alpha]anthracene-induced breast tumor in rat. Food Chem Toxicol 2010; 48:1587-91. [PMID: 20332012 DOI: 10.1016/j.fct.2010.03.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 03/11/2010] [Accepted: 03/17/2010] [Indexed: 11/22/2022]
Abstract
Centchroman is a non-steroidal oral contraceptive and has been found to be a candidate drug for breast cancer exhibiting partial to complete remission of lesions in 40.5% of breast cancer patients. The therapeutic efficacy of centchroman was monitored alone and together with glycine soya on growth of 7,12-dimethylbenz[alpha]anthracene-induced breast tumor in rat. The tumor regression was monitored at different doses of centchroman alone ranging from 0 to 10 mg kg(-1) and with glycine soya from 1x10(4) to 5x10(4) mg kg(-1) per day until 5weeks treatment. An optimum tumor treatment opus was established with varying treatment parameters including doses of therapeutic agents and treatment period. The tumors were found to be static with a strong anti-estrogenic effect. Overall our study shows that both centchroman and glycine soya alone and jointly combat with breast cancer.
Collapse
Affiliation(s)
- Rajeev Mishra
- Division of Toxicology, Central Drug Research Institute, CSIR, Lucknow, India.
| | | | | | | | | | | |
Collapse
|
29
|
|
30
|
Sasano H, Miki Y, Shibuya R, Suzuki T. Aromatase and in situ estrogen production in DCIS (ductal carcinoma in situ) of human breast. J Steroid Biochem Mol Biol 2010; 118:242-5. [PMID: 19782135 DOI: 10.1016/j.jsbmb.2009.09.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 09/12/2009] [Accepted: 09/15/2009] [Indexed: 11/29/2022]
Abstract
Ductal carcinoma in situ or DCIS belongs to intraductal proliferative lesions, which are a group of cytologically and architecturally diverse ductal proliferations, typically originating from the terminal duct-lobular units. In these intraductal proliferative diseases, estrogens are considered to be involved in the progression of the disease especially from ductal non-neoplastic hyperplasia to DCIS and possibly development of invasive carcinoma from DCIS. Estrogen receptor (ER) alpha is abundantly expressed in atypical ductal hyperplasia and low grade DCIS. Suppression of estrogenic actions using tamoxifen resulted in inhibition of recurrence of DCIS and/or of progression into invasive carcinoma. Intratumoral estrogen concentration in DCIS determined by liquid chromatography/electrospray tandem mass spectrometry is significantly higher than that in non-neoplastic breast tissues with statistically not lower than that in invasive carcinoma. Aromatase mRNA expression in both stromal and parenchymal cells of DCIS determined by quantitative RT-PCR following laser capture microdissection was also much higher than that in non-neoplastic breast, although lower than that in invasive carcinoma. Immunohistochemistry of aromatase also revealed the similar patterns of immunolocalization as in invasive carcinoma. Aromatase is overexpressed in noninvasive breast malignancies including DCIS and results in elevated concentrations of intratumoral estradiol. These findings could provide the scientific rationale as to employing aromatase inhibitors in the management of ER positive DCIS patients.
Collapse
Affiliation(s)
- Hironobu Sasano
- Department of Pathology, Tohoku University School of Medicine, 2-1 Seiryou-machi, Aoba-ku, Sendai 980-8575, Japan.
| | | | | | | |
Collapse
|
31
|
Adenosine A1 receptor, a target and regulator of estrogen receptoralpha action, mediates the proliferative effects of estradiol in breast cancer. Oncogene 2009; 29:1114-22. [PMID: 19935720 PMCID: PMC2829108 DOI: 10.1038/onc.2009.409] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Estrogen receptor-alpha (ERalpha) and its ligand estradiol (E2) has critical roles in breast cancer growth and are key therapeutic targets. In this study, we report a novel dual role of the adenosine A1 receptor (Adora1) as an E2/ERalpha target and a regulator of ERalpha transcriptional activity. In ERalpha-positive breast cancer cells, E2 upregulated Adora1 messenger RNA (mRNA) and protein levels, an effect that was reversed by the E2 antagonist ICI 182 780. Small interference RNA ablation of Adora1 in ERalpha-positive cells reduced basal and E2-dependent proliferation, whereas Adora1 over-expression in an ERalpha-negative cell line induced proliferation. The selective Adora1 antagonist, DPCPX, reduced proliferation, establishing Adora1 as a mediator of E2/ERalpha-dependent breast cancer growth. Intriguingly, Adora1 ablation decreased both mRNA and protein levels of ERalpha and, consequently, estrogen-responsive element-dependent ERalpha transcriptional activity. Moreover, Adora1 ablation decreased binding activity of ERalpha to the promoter of its target gene TFF1 and led to reduced TFF1 promoter activity and mRNA levels, suggesting that Adora1 is required for full transcriptional activity of ERalpha on E2 stimulation. Taken together, we showed a short feed-forward loop involving E2, ERalpha and Adora1 that favors breast cancer growth. These data suggest that Adora1 may represent an important target for therapeutic intervention in hormone-dependent breast cancer.
Collapse
|
32
|
Fluck MM, Schaffhausen BS. Lessons in signaling and tumorigenesis from polyomavirus middle T antigen. Microbiol Mol Biol Rev 2009; 73:542-63, Table of Contents. [PMID: 19721090 PMCID: PMC2738132 DOI: 10.1128/mmbr.00009-09] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The small DNA tumor viruses have provided a very long-lived source of insights into many aspects of the life cycle of eukaryotic cells. In recent years, the emphasis has been on cancer-related signaling. Here we review murine polyomavirus middle T antigen, its mechanisms, and its downstream pathways of transformation. We concentrate on the MMTV-PyMT transgenic mouse, one of the most studied models of breast cancer, which permits the examination of in situ tumor progression from hyperplasia to metastasis.
Collapse
Affiliation(s)
- Michele M Fluck
- Department of Microbiology and Molecular Genetics, Interdepartmental Program in Cell and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA.
| | | |
Collapse
|
33
|
Gennari L, Merlotti D, Valleggi F, Nuti R. Ospemifene use in postmenopausal women. Expert Opin Investig Drugs 2009; 18:839-49. [DOI: 10.1517/13543780902953715] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
34
|
Denzel MS, Hebbard LW, Shostak G, Shapiro L, Cardiff RD, Ranscht B. Adiponectin deficiency limits tumor vascularization in the MMTV-PyV-mT mouse model of mammary cancer. Clin Cancer Res 2009; 15:3256-64. [PMID: 19447866 DOI: 10.1158/1078-0432.ccr-08-2661] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE High levels of the fat-secreted cytokine adiponectin (APN) are present in the circulation of healthy people, whereas low levels correlate with an increased incidence of breast cancer in women. The current study experimentally probes the physiologic functions of APN in mammary cancer in a newly generated genetic mouse model. EXPERIMENTAL DESIGN We established an APN null mouse model of mammary cancer by introducing the polyoma virus middle T (PyV-mT) oncogene expressed from mouse mammary tumor virus (MMTV) regulatory elements into APN null mice. MMTV-PyV-mT-induced tumors resemble ErbB2-amplified human breast cancers. We monitored tumor onset, kinetics, and animal survival, and analyzed vascular coverage, apoptosis, and hypoxia in sections from the primary tumors. Metastatic spreading was evaluated by analyses of the lungs. RESULTS APN prominently localized to the vasculature in human and mouse mammary tumors. In APN null mice, MMTV-PyV-mT-induced tumors appeared with delayed onset and exhibited reduced growth rates. Affected animals survived control tumor-bearing mice by an average of 21 days. Pathologic analyses revealed reduced vascularization of APN null tumors along with increased hypoxia and apoptosis. At the experimental end point, APN null transgenic mice showed increased frequency of pulmonary metastases. CONCLUSION The current work identifies a proangiogenic contribution of APN in mammary cancer that, in turn, affects tumor progression. APN interactions with vascular receptors may be useful targets for developing therapies aimed at controlling tumor vascularization in cancer patients.
Collapse
Affiliation(s)
- Martin S Denzel
- Burnham Institute for Medical Research, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
35
|
Medina D, Kittrell F, Hill J, Zhang Y, Hilsenbeck SG, Bissonette R, Brown PH. Prevention of tumorigenesis in p53-null mammary epithelium by rexinoid bexarotene, tyrosine kinase inhibitor gefitinib, and celecoxib. Cancer Prev Res (Phila) 2009; 2:168-74. [PMID: 19174577 DOI: 10.1158/1940-6207.capr-08-0107] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The chemopreventive effects of three agents, rexinoid bexarotene, tyrosine kinase inhibitor gefitinib, and celecoxib, were tested on mammary tumor development arising in p53-null mammary epithelium. The rexinoid bexarotene was the most efficacious inhibitor as it reduced mammary tumor development by 75% in virgin mice and significantly delayed mean tumor development by 98 days in hormone-stimulated mice. The tyrosine kinase inhibitor gefitinib reduced mammary tumor incidence by 50% in virgin mice but did not significantly delay mean tumor latency in hormone-stimulated mice. Celecoxib did not reduce tumor incidence or mean tumor latency in either of the two models. The high doses of the rexinoid and the tyrosine kinase inhibitor did not affect the progression of tumors arising from the premalignant mammary outgrowth line, PN8a. A comparison of these agents with tamoxifen shows the superiority of tamoxifen in preventing tumor development in p53-null mammary cells. Similarly, a comparison of the results of the p53 model with other transgenic models in their response to the chemopreventive agents showed that mammary tumors arising from different oncogenic events will respond differently to the different agents.
Collapse
Affiliation(s)
- Daniel Medina
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
|
37
|
Allred DC, Medina D. The relevance of mouse models to understanding the development and progression of human breast cancer. J Mammary Gland Biol Neoplasia 2008; 13:279-88. [PMID: 18704660 DOI: 10.1007/s10911-008-9093-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Accepted: 07/04/2008] [Indexed: 12/21/2022] Open
Abstract
Mouse modeling of human breast cancer has developed tremendously over the past ten years. Human breast cancer is characterized by enormous biological diversity and, collectively, the new models have come much closer to encompassing this diversity. They have provided a deeper understanding of the fundamental events that mediate the initiation, development, and progression of breast cancer, and they offer new opportunities to develop and test strategies to treat and, perhaps, even prevent the disease. This chapter reviews the historical development of mouse models of breast cancer and highlights some of their major strengths, weaknesses, and contributions.
Collapse
Affiliation(s)
- D Craig Allred
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | | |
Collapse
|
38
|
Abstract
The mouse model for breast cancer has developed into a most effective means of dissecting and understanding this devastating disease. The inbred transgenic mouse lends itself to biological, molecular, immunological, and genetic studies. The observation, dissection, transplantation, and subsequent amplification of precancerous mammary lesions and tumors give the scientist the means to readily study the tissues and design interventions and therapeutic drugs for the future eventual control of breast cancer. There are many inbred strains of mice, selected for specific characteristics. The mouse is easy to handle, breeds well, and does not require extensive facilities, funding, and handling such as monkeys, chimps, and other animal models. A huge advantage is the capability for the transplantation of tissues as well as gene manipulation, which make the transgenic mouse a major research resource. The mouse has served the scientific community well for over a century and will continue to do so in the quest for understanding breast cancer and other diseases.
Collapse
Affiliation(s)
- Lawrence J T Young
- Center for Comparative Medicine, University of California, Hutchison and County Road 98, Davis, CA 95616, USA.
| |
Collapse
|
39
|
Intratumoral estrogen production in breast carcinoma: significance of aromatase. Breast Cancer 2008; 15:270-7. [DOI: 10.1007/s12282-008-0062-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Accepted: 05/12/2008] [Indexed: 10/21/2022]
|
40
|
Wurz GT, Hellmann-Blumberg U, DeGregorio MW. Pharmacologic Effects of Ospemifene in Rhesus Macaques: A Pilot Study. Basic Clin Pharmacol Toxicol 2008; 102:552-8. [DOI: 10.1111/j.1742-7843.2008.00235.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
41
|
Jordan VC. New insights into the metabolism of tamoxifen and its role in the treatment and prevention of breast cancer. Steroids 2007; 72:829-42. [PMID: 17765940 PMCID: PMC2740485 DOI: 10.1016/j.steroids.2007.07.009] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 07/13/2007] [Accepted: 07/20/2007] [Indexed: 12/13/2022]
Abstract
The metabolism of tamoxifen is being redefined in the light of several important pharmacological observations. Recent studies have identified 4-hydroxy N-desmethyltamoxifen (endoxifen) as an important metabolite of tamoxifen necessary for antitumor actions. The metabolite is formed through the enzymatic product of CYP2D6 which also interacts with specific selective serotonin reuptake inhibitors (SSRIs) used to prevent the hot flashes observed in up to 45% of patients taking tamoxifen. Additionally, the finding that enzyme variants of CYP2D6 do not promote the metabolism of tamoxifen to endoxifen means that significant numbers of women might not receive optimal benefit from tamoxifen treatment. Clearly these are particularly important issues not only for breast cancer treatment but also for selecting premenopausal women, at high risk for breast cancer, as candidates for chemoprevention using tamoxifen.
Collapse
Affiliation(s)
- V Craig Jordan
- Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111-2497, USA.
| |
Collapse
|
42
|
Suzuki T, Miki Y, Moriya T, Akahira JI, Hirakawa H, Ohuchi N, Sasano H. In situ production of sex steroids in human breast carcinoma. Med Mol Morphol 2007; 40:121-7. [PMID: 17874044 DOI: 10.1007/s00795-007-0365-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Accepted: 03/06/2007] [Indexed: 10/22/2022]
Abstract
It is well known that sex steroids are closely involved in the growth of human breast carcinomas, and the great majority of breast carcinomas express sex steroid receptors. In particular, recent studies have demonstrated that estrogens and androgens are locally produced and act in breast carcinoma tissues without release into plasma. Blockade of intratumoral estrogen production potentially leads to an improvement in the prognosis of invasive breast carcinoma patients, and, therefore, it is important to obtain a better understanding of sex steroid-producing enzymes in breast carcinoma. In this review, we summarize recent studies on tissue concentration of sex steroids and expression of enzymes related to intratumoral production of estrogens [aromatase, steroid sulfatase (STS), and 17beta-hydroxysteroid dehydrogenase type 1 (17betaHSD1)], and androgens (17betaHSD5 and 5alpha-reductase) in invasive and in situ (noninvasive) breast carcinomas, and discuss the significance of intratumoral production of sex steroids in breast carcinoma.
Collapse
Affiliation(s)
- Takashi Suzuki
- Department of Pathology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | | | | | | | | | | | | |
Collapse
|
43
|
Gennari L, Merlotti D, Valleggi F, Martini G, Nuti R. Selective estrogen receptor modulators for postmenopausal osteoporosis: current state of development. Drugs Aging 2007. [PMID: 17503894 DOI: 10.2165/00002512-200724050.00002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Selective estrogen receptor modulators (SERMs) are structurally different compounds that interact with intracellular estrogen receptors in target organs as estrogen receptor agonists and antagonists. These drugs have been intensively studied over the past decade and have proven to be a highly versatile group for the treatment of different conditions associated with aging, including hormone-responsive cancer and osteoporosis. Tamoxifen and toremifene are currently used to treat advanced breast cancer and also have beneficial effects on bone mineral density and serum lipids in postmenopausal women. Raloxifene is the only SERM approved worldwide for the prevention and treatment of postmenopausal osteoporosis and vertebral fractures. However, although these SERMs have many benefits, they may also be responsible for some potentially very serious adverse effects, such as thromboembolic disorders and, in the case of tamoxifen, uterine cancer. These adverse effects represent a major concern given that long-term therapy is required to prevent osteoporosis. Moreover, both preclinical and clinical reports suggest that tamoxifen, toremifene and raloxifene are considerably less potent than estrogen. The search for the 'ideal' SERM, which would have estrogenic effects on bone and serum lipids, neutral effects on the uterus, and antiestrogenic effects on breast tissue, but none of the adverse effects associated with current therapies, is currently under way. Ospemifene, lasofoxifene, bazedoxifene and arzoxifene, which are new SERM molecules with potential greater efficacy and potency than previous SERMs, are currently under investigation for use in the treatment and prevention of osteoporosis. These drugs have been shown to be comparably effective to conventional hormone replacement therapy in animal models of osteoporosis, with potential indications for an improved safety profile. Clinical efficacy data from ongoing phase III trials are awaited so that a true understanding of the therapeutic potential of these compounds can be obtained.
Collapse
Affiliation(s)
- Luigi Gennari
- Department of Internal Medicine, Endocrine-Metabolic Sciences and Biochemistry, University of Siena, Policlinico Le Scotte, Siena, Italy.
| | | | | | | | | |
Collapse
|
44
|
Cardiff RD, Anver MR, Boivin GP, Bosenberg MW, Maronpot RR, Molinolo AA, Nikitin AY, Rehg JE, Thomas GV, Russell RG, Ward JM. Precancer in mice: animal models used to understand, prevent, and treat human precancers. Toxicol Pathol 2007; 34:699-707. [PMID: 17074738 DOI: 10.1080/01926230600930129] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We present a status report from the NCI Mouse Models of Human Cancers Consortium (MMHCC) Precancers Workshop held November 8 and 9, 2004. An expert panel, the Mouse Models Group (MMG) evaluated the status of mouse models of precancer emphasizing genetically engineered mouse models, especially of lining epithelium and their utilitarian value to human carcinogenesis. An outline of the background for the panel's considerations is provided with examples of past and current precancerous lesions in mice. The experimental use of oncogenic viruses and chemical carcinogens in mice led to operational definitions of initiation, promotion, and preneoplasia Preneoplastic and precancerous lesions are found in these models. In this precancer concept, most preneoplastic lesions are considered as potentially precancerous or at least an earlier stage in cancer development than typical pre-invasive epithelial lesions, which are often seen in these mouse models. Genetically engineered mice, used to test the oncogenicity of individual genes, develop precancers that are initiated by defined molecular and histopathologic changes. The mouse can be used to isolate and study precancers in detail, thereby providing a level of biological understanding not readily available in clinical disease. These studies suggest that genetically engineered mice are very useful preclinical models for chemoprevention and therapy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Carcinogens
- Carcinoma in Situ/chemically induced
- Carcinoma in Situ/genetics
- Cell Transformation, Neoplastic/chemically induced
- Cell Transformation, Neoplastic/genetics
- Colonic Neoplasms/chemically induced
- Colonic Neoplasms/genetics
- Drug Screening Assays, Antitumor
- Epithelial Cells/pathology
- Humans
- Mammary Neoplasms, Experimental/chemically induced
- Mammary Neoplasms, Experimental/genetics
- Mice
- Mice, Transgenic
- Neoplasms, Experimental/chemically induced
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/prevention & control
- Oncogenes/genetics
- Oncogenic Viruses/genetics
- Precancerous Conditions/chemically induced
- Precancerous Conditions/drug therapy
- Precancerous Conditions/genetics
- Precancerous Conditions/prevention & control
- Skin Neoplasms/chemically induced
- Skin Neoplasms/genetics
- Tumor Virus Infections/genetics
- Tumor Virus Infections/pathology
Collapse
Affiliation(s)
- Robert D Cardiff
- The UCD Center for Comparative Medicine, University of California, Davis, Davis, California 95616, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
For over a century, mouse mammary tumor biology and the associated Mouse mammary tumor virus (MMTV) have served as the foundation for experimental cancer research, in general, and, in particular, experimental breast cancer research. Spontaneous mouse mammary tumors were the basis for studies of the natural history of neoplasia, oncogenic viruses, host responses, endocrinology, and neoplastic progression. However, lacking formal proof of a human mammary tumor virus, the preeminence of the mouse model faded in the 1980s. Since the late 1980s, genetically engineered mice (GEM) have proven extremely useful for studying breast cancer and have become the animal model for human breast cancer. Hundreds of mouse models of human breast cancer have been developed since the first demonstration, in 1984, that the mouse mammary gland could be molecularly targeted and used to test the oncogenicity of candidate human genes. Now, very few scientists can avoid using a mouse model to test the biology of their favorite gene. The GEM have attracted a new generation of molecular and cellular biologists eager to apply their skills to these surrogates of the human disease. Newcomers often enter the field without an appreciation of the origins of mouse mammary tumor biology and the basis for many of the prevailing concepts. Our purpose in writing this short history of mouse mammary tumor biology is to provide a historical perspective for the benefit of the newcomers. If Einstein was correct in that "we stand on the shoulders of giants," the neophytes should meet their giants.
Collapse
Affiliation(s)
- Robert D Cardiff
- Center for Comparative Medicine, Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, California 95616, USA
| | | |
Collapse
|
46
|
Gennari L, Merlotti D, Valleggi F, Martini G, Nuti R. Selective Estrogen Receptor Modulators for Postmenopausal Osteoporosis. Drugs Aging 2007; 24:361-79. [PMID: 17503894 DOI: 10.2165/00002512-200724050-00002] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Selective estrogen receptor modulators (SERMs) are structurally different compounds that interact with intracellular estrogen receptors in target organs as estrogen receptor agonists and antagonists. These drugs have been intensively studied over the past decade and have proven to be a highly versatile group for the treatment of different conditions associated with aging, including hormone-responsive cancer and osteoporosis. Tamoxifen and toremifene are currently used to treat advanced breast cancer and also have beneficial effects on bone mineral density and serum lipids in postmenopausal women. Raloxifene is the only SERM approved worldwide for the prevention and treatment of postmenopausal osteoporosis and vertebral fractures. However, although these SERMs have many benefits, they may also be responsible for some potentially very serious adverse effects, such as thromboembolic disorders and, in the case of tamoxifen, uterine cancer. These adverse effects represent a major concern given that long-term therapy is required to prevent osteoporosis. Moreover, both preclinical and clinical reports suggest that tamoxifen, toremifene and raloxifene are considerably less potent than estrogen. The search for the 'ideal' SERM, which would have estrogenic effects on bone and serum lipids, neutral effects on the uterus, and antiestrogenic effects on breast tissue, but none of the adverse effects associated with current therapies, is currently under way. Ospemifene, lasofoxifene, bazedoxifene and arzoxifene, which are new SERM molecules with potential greater efficacy and potency than previous SERMs, are currently under investigation for use in the treatment and prevention of osteoporosis. These drugs have been shown to be comparably effective to conventional hormone replacement therapy in animal models of osteoporosis, with potential indications for an improved safety profile. Clinical efficacy data from ongoing phase III trials are awaited so that a true understanding of the therapeutic potential of these compounds can be obtained.
Collapse
Affiliation(s)
- Luigi Gennari
- Department of Internal Medicine, Endocrine-Metabolic Sciences and Biochemistry, University of Siena, Policlinico Le Scotte, Siena, Italy.
| | | | | | | | | |
Collapse
|
47
|
Heterogeneity of mammary lesions represent molecular differences. BMC Cancer 2006; 6:275. [PMID: 17147824 PMCID: PMC1762020 DOI: 10.1186/1471-2407-6-275] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2006] [Accepted: 12/05/2006] [Indexed: 11/26/2022] Open
Abstract
Background Human breast cancer is a heterogeneous disease, histopathologically, molecularly and phenotypically. The molecular basis of this heterogeneity is not well understood. We have used a mouse model of DCIS that consists of unique lines of mammary intraepithelial neoplasia (MIN) outgrowths, the premalignant lesion in the mouse that progress to invasive carcinoma, to understand the molecular changes that are characteristic to certain phenotypes. Each MIN-O line has distinguishable morphologies, metastatic potentials and estrogen dependencies. Methods We utilized oligonucleotide expression arrays and high resolution array comparative genomic hybridization (aCGH) to investigate whole genome expression patterns and whole genome aberrations in both the MIN-O and tumor from four different MIN-O lines that each have different phenotypes. From the whole genome analysis at 35 kb resolution, we found that chromosome 1, 2, 10, and 11 were frequently associated with whole chromosome gains in the MIN-Os. In particular, two MIN-O lines had the majority of the chromosome gains. Although we did not find any whole chromosome loss, we identified 3 recurring chromosome losses (2F1-2, 3E4, 17E2) and two chromosome copy number gains on chromosome 11. These interstitial deletions and duplications were verified with a custom made array designed to interrogate the specific regions at approximately 550 bp resolution. Results We demonstrated that expression and genomic changes are present in the early premalignant lesions and that these molecular profiles can be correlated to phenotype (metastasis and estrogen responsiveness). We also identified expression changes associated with genomic instability. Progression to invasive carcinoma was associated with few additional changes in gene expression and genomic organization. Therefore, in the MIN-O mice, early premalignant lesions have the major molecular and genetic changes required and these changes have important phenotypic significance. In contrast, the changes that occur in the transition to invasive carcinoma are subtle, with few consistent changes and no association with phenotype. Conclusion We propose that the early lesions carry the important genetic changes that reflect the major phenotypic information, while additional genetic changes that accumulate in the invasive carcinoma are less associated with the overall phenotype.
Collapse
|
48
|
Cardiff RD, Gregg JP, Miller JW, Axelrod DE, Borowsky AD. Histopathology as a predictive biomarker: strengths and limitations. J Nutr 2006; 136:2673S-5S. [PMID: 16988145 DOI: 10.1093/jn/136.10.2673s] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Robert D Cardiff
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis CA 95616, USA.
| | | | | | | | | |
Collapse
|
49
|
Namba R, Young LJT, Abbey CK, Kim L, Damonte P, Borowsky AD, Qi J, Tepper CG, MacLeod CL, Cardiff RD, Gregg JP. Rapamycin Inhibits Growth of Premalignant and Malignant Mammary Lesions in a Mouse Model of Ductal Carcinoma In situ. Clin Cancer Res 2006; 12:2613-21. [PMID: 16638874 DOI: 10.1158/1078-0432.ccr-05-2170] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Rapamycin has been shown to have antitumor effects in various tumor models. To study the effect of rapamycin at different stages of breast cancer development, we used two unique mouse models of breast cancer with activated phosphatidylinositol 3-kinase (PI3K) pathway. Met-1 tumors are highly invasive and metastatic, and mammary intraepithelial neoplasia-outgrowths (MIN-O), a model for human ductal carcinoma in situ, are transplantable premalignant mammary lesions that develop invasive carcinoma with predictable latencies. Both of these models were derived from mammary lesions in Tg(MMTV-PyV-mT) mice. EXPERIMENTAL DESIGN Met-1 tumors were used to study the effect of rapamycin treatment on invasive disease. Transplanted MIN-O model was used to study the effect of rapamycin on premalignant mammary lesions. Animals were in vivo micro-positron emission tomography imaged to follow the lesion growth and transformation to tumor during the treatment. Cell proliferation, angiogenesis, and apoptosis was assayed by immunohistochemistry. RESULTS Rapamycin inhibited in vitro tumor cell proliferation and in vivo Met-1 tumor growth. The growth inhibition was correlated with dephosphorylation of mammalian target of rapamycin (mTOR) targets. Rapamycin treatment significantly reduced the growth of the premalignant MIN-O lesion, as well as tumor incidence and tumor burden. Growth inhibition was associated with reduced cell proliferation and angiogenesis and increased apoptosis. CONCLUSIONS In PyV-mT mouse mammary models, rapamycin inhibits the growth of premalignant lesions and invasive tumors. Although the inhibitory effect of rapamycin was striking, rapamycin treatment did not completely obliterate the lesions.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/pharmacology
- Antigens, Polyomavirus Transforming/genetics
- Antigens, Polyomavirus Transforming/physiology
- Apoptosis/drug effects
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/prevention & control
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Female
- Male
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Transgenic
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphorylation/drug effects
- Positron-Emission Tomography
- Precancerous Conditions/blood supply
- Precancerous Conditions/pathology
- Precancerous Conditions/prevention & control
- Protein Kinases/metabolism
- Signal Transduction/drug effects
- Sirolimus/pharmacology
- TOR Serine-Threonine Kinases
- Time Factors
Collapse
Affiliation(s)
- Ruria Namba
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abbey CK, Borowsky AD, Gregg JP, Cardiff RD, Cherry SR. Preclinical imaging of mammary intraepithelial neoplasia with positron emission tomography. J Mammary Gland Biol Neoplasia 2006; 11:137-49. [PMID: 17091397 DOI: 10.1007/s10911-006-9020-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Small-animal imaging with positron emission tomography (PET) has become a valuable tool for evaluating preclinical models of breast cancer and other diseases. In this review, we examine a number of issues related to preclinical imaging studies with PET, using transgenic models of ductal carcinoma in situ and metastasis as specific examples. We discuss imaging components such as reconstruction, normalization, and extraction of quantitative parameters. We also analyze the effect of longitudinal correlations on cohort size and present some simple statistical techniques for determining cohort sizes that may be helpful in designing preclinical imaging studies. We describe studies that are greatly facilitated by access to non-invasive imaging data including a study involving multiple endpoints and another investigating metastasis. We conclude with a brief survey of emerging approaches in small-animal PET imaging.
Collapse
Affiliation(s)
- Craig K Abbey
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | | | | | | | | |
Collapse
|