1
|
Viswanadhapalli S, Ma S, Sareddy GR, Lee TK, Li M, Gilbreath C, Liu X, Luo Y, Pratap UP, Zhou M, Blatt EB, Kassees K, Arteaga C, Alluri P, Rao M, Weintraub ST, Tekmal RR, Ahn JM, Raj GV, Vadlamudi RK. Estrogen receptor coregulator binding modulator (ERX-11) enhances the activity of CDK4/6 inhibitors against estrogen receptor-positive breast cancers. Breast Cancer Res 2019; 21:150. [PMID: 31878959 PMCID: PMC6933697 DOI: 10.1186/s13058-019-1227-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND CDK4/6 inhibitors in combination with endocrine therapy (AE/AI/SERDs) are approved for the treatment of ER+ advanced breast cancer (BCa). However, not all patients benefit from CDK4/6 inhibitors therapy. We previously reported a novel therapeutic agent, ERX-11, that binds to the estrogen receptor (ER) and modulates ER-coregulator interactions. Here, we tested if the combination of ERX-11 with agents approved for ER+ BCa would be more potent. METHODS We tested the effect of combination therapy using BCa cell line models, including those that have acquired resistance to tamoxifen, letrozole, or CDK4/6 inhibitors or have been engineered to express mutant forms of the ER. In vitro activity was tested using Cell Titer-Glo, MTT, and apoptosis assays. Mechanistic studies were conducted using western blot, reporter gene assays, RT-qPCR, and mass spectrometry approaches. Xenograft, patient-derived explants (PDEs), and xenograft-derived explants (XDE) were used for preclinical evaluation and toxicity. RESULTS ERX-11 inhibited the proliferation of therapy-resistant BCa cells in a dose-dependent manner, including ribociclib resistance. The combination of ERX-11 and CDK4/6 inhibitor was synergistic in decreasing the proliferation of both endocrine therapy-sensitive and endocrine therapy-resistant BCa cells, in vitro, in xenograft models in vivo, xenograft-derived explants ex vivo, and in primary patient-derived explants ex vivo. Importantly, the combination caused xenograft tumor regression in vivo. Unbiased global mass spectrometry studies demonstrated profound decreases in proliferation markers with combination therapy and indicated global proteomic changes in E2F1, ER, and ER coregulators. Mechanistically, the combination of ERX-11 and CDK4/6 inhibitor decreased the interaction between ER and its coregulators, as evidenced by immunoprecipitation followed by mass spectrometry studies. Biochemical studies confirmed that the combination therapy significantly altered the expression of proteins involved in E2F1 and ER signaling, and this is primarily driven by a transcriptional shift, as noted in gene expression studies. CONCLUSIONS Our results suggest that ERX-11 inhibited the proliferation of BCa cells resistant to both endocrine therapy and CDK4/6 inhibitors in a dose-dependent manner and that the combination of ERX-11 with a CDK4/6 inhibitor may represent a viable therapeutic approach.
Collapse
Affiliation(s)
| | - Shihong Ma
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Gangadhara Reddy Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX, 78229, USA
- CDP Program, University of Texas Health Cancer Center, San Antonio, TX, 78229, USA
| | - Tae-Kyung Lee
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Mengxing Li
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX, 78229, USA
| | - Collin Gilbreath
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Xihui Liu
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Yiliao Luo
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX, 78229, USA
| | - Uday P Pratap
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX, 78229, USA
| | - Mei Zhou
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX, 78229, USA
| | - Eliot B Blatt
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Kara Kassees
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Carlos Arteaga
- Simmons Cancer Center, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Prasanna Alluri
- Department of Radiation Oncology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Manjeet Rao
- Department of Cell Systems and Anatomy, University of Texas Health, San Antonio, TX, 78229, USA
| | - Susan T Weintraub
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX, 78229, USA
| | - Rajeshwar Rao Tekmal
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX, 78229, USA
| | - Jung-Mo Ahn
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, 75080, USA.
| | - Ganesh V Raj
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA.
- Simmons Cancer Center, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA.
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX, 78229, USA.
- CDP Program, University of Texas Health Cancer Center, San Antonio, TX, 78229, USA.
| |
Collapse
|
2
|
Gougis P, Carton M, Tchokothe C, Campone M, Dalenc F, Mailliez A, Levy C, Jacot W, Debled M, Leheurteur M, Bachelot T, Hennequin A, Perrin C, Gonçalves A, Uwer L, Eymard JC, Petit T, Mouret-Reynier MA, Chamorey E, Simon G, Saghatchian M, Cailliot C, Le Tourneau C. CinéBreast-factors influencing the time to first metastatic recurrence in breast cancer: Analysis of real-life data from the French ESME MBC database. Breast 2019; 49:17-24. [PMID: 31675683 PMCID: PMC7375625 DOI: 10.1016/j.breast.2019.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 12/25/2022] Open
Abstract
Purpose The Time to First Metastatic Recurrence (TFMR) could be considered as an indirect reflection of the tumour growth kinetics which plays an important role in cancer. Molecular subtypes such as expression of estrogen receptor are known predictive factors of TFMR. The CinéBreast study aimed to identify predictive factors of the time to TFMR. Methods The French Epidemiological Strategy and Medical Economics (ESME) Metastatic Breast Cancer (MBC) Database (NCT03275311) was used, which contains data from a cohort of metastatic breast cancer patients from 2008 to 2016 using retrospective data collection. It is a national multi-centre database. The impact of TFMR on overall survival (OS) since first metastasis was also evaluated. Results Among 16 702 patients recorded in the ESME MBC database, 10 595 had an initially localised breast cancer with hormone receptor (HR) and HER2 status available, with a metastatic recurrence. Median follow up was 56 months. Median TFMR was 59 months (<24: 20%, 24–60: 31%, 60–120: 25%, >120: 24%). HER2+ and TNBC were respectively 4 times and 12 times (p < 0.0001) more likely to have a recurrence within 2 years when compared to the luminal subgroup. Short TFMR and HR-/HER2-subtype significantly correlated with a poor OS in multivariate analysis. Some patients with MBC (20% in HER2+, 10% in ER+/HER2-and <5% in the ER-/HER2-) were long-term survivors in all 3 subgroups. Conclusions In this large-scale real-life data study, patients with a TNBC metastatic recurrence had a shorter TFMR. Short TFMR significantly correlated with worse overall survival. ESME is a large-scale real-life database of 16 702 metastatic breast cancer patients. A short time to first metastatic recurrence is associated with poor overall survival. Triple-negative tumours were more likely to recur early than HR+ and HER2+ tumours.
Collapse
Affiliation(s)
- P Gougis
- Department of Drug Development and Innovation, Institut Curie, Paris, Saint-Cloud, France; Department of Clinical Pharmacology, Centre D'Investigation Clinique Paris-Est, AP-HP, Pitié-Salpêtrière Hospital, PSL University, CLIP² Galilée, Paris, France
| | - M Carton
- Department of Biostatistics, Institut Curie, Saint-Cloud, France
| | - C Tchokothe
- Department of Biostatistics, Institut Curie, Saint-Cloud, France
| | - M Campone
- Department of Medical Oncology, Institut de Cancérologie de L'Ouest, Nantes and Angers, France
| | - F Dalenc
- Department of Medical Oncology, Institut Claudius Regaud, Toulouse, France
| | - A Mailliez
- Department of Breast Cancer, Centre Oscar Lambret, Lille, France
| | - C Levy
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | - W Jacot
- Department of Medical Oncology, Institut Du Cancer de Montpellier, Montpellier, France
| | - M Debled
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - M Leheurteur
- Department of Medical Oncology, Henri Becquerel Centre, Rouen, France
| | - T Bachelot
- Department of Biostatistics, Centre Léon Bérard, Lyon, France
| | - A Hennequin
- Department of Medical Oncology, Center Georges François Leclerc, Dijon, France
| | - C Perrin
- Department of Medical Oncology, Centre Eugène Marquis, Rennes, France
| | - A Gonçalves
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - L Uwer
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Vandoeuvre-lès-Nancy, France
| | - J C Eymard
- Department of Medical Oncology, Centre Jean Godinot, Reims, France
| | - T Petit
- Department of Medical Oncology, Centre Paul Strauss, Strasbourg, France
| | - M A Mouret-Reynier
- Department of Medical Oncology, Centre Jean Perrin, Clermont Ferrand, France
| | - E Chamorey
- Department of Biostatistics, Centre Antoine Lacassagne, Nice, France
| | - G Simon
- Department of Research and Development, R&D Unicancer, Paris, France
| | - M Saghatchian
- Department of Biostatistics, Institut Curie, Saint-Cloud, France
| | - C Cailliot
- Department of Research and Development, R&D Unicancer, Paris, France
| | - C Le Tourneau
- Department of Drug Development and Innovation, Institut Curie, Paris, Saint-Cloud, France; U900 INSERM Research Unit, Saint-Cloud, France.
| |
Collapse
|
3
|
Tumor-Associated Macrophages Induce Endocrine Therapy Resistance in ER+ Breast Cancer Cells. Cancers (Basel) 2019; 11:cancers11020189. [PMID: 30736340 PMCID: PMC6406935 DOI: 10.3390/cancers11020189] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 01/09/2019] [Accepted: 01/15/2019] [Indexed: 02/07/2023] Open
Abstract
Antiestrogenic adjuvant treatments are first-line therapies in patients with breast cancer positive for estrogen receptor (ER+). Improvement of their treatment strategies is needed because most patients eventually acquire endocrine resistance and many others are initially refractory to anti-estrogen treatments. The tumor microenvironment plays essential roles in cancer development and progress; however, the molecular mechanisms underlying such effects remain poorly understood. Breast cancer cell lines co-cultured with TNF-α-conditioned macrophages were used as pro-inflammatory tumor microenvironment models. Proliferation, migration, and colony formation assays were performed to evaluate tamoxifen and ICI 182,780 resistance and confirmed in a mouse-xenograft model. Molecular mechanisms were investigated using cytokine antibody arrays, WB, ELISA, ChIP, siRNA, and qPCR-assays. In our simulated pro-inflammatory tumor microenvironment, tumor-associated macrophages promoted proliferation, migration, invasiveness, and breast tumor growth of ER+ cells, rendering these estrogen-dependent breast cancer cells resistant to estrogen withdrawal and tamoxifen or ICI 182,780 treatment. Crosstalk between breast cancer cells and conditioned macrophages induced sustained release of pro-inflammatory cytokines from both cell types, activation of NF-κB/STAT3/ERK in the cancer cells and hyperphosphorylation of ERα, which resulted constitutively active. Our simulated tumor microenvironment strongly altered endocrine and inflammatory signaling pathways in breast cancer cells, leading to endocrine resistance in these cells.
Collapse
|
4
|
Raj GV, Sareddy GR, Ma S, Lee TK, Viswanadhapalli S, Li R, Liu X, Murakami S, Chen CC, Lee WR, Mann M, Krishnan SR, Manandhar B, Gonugunta VK, Strand D, Tekmal RR, Ahn JM, Vadlamudi RK. Estrogen receptor coregulator binding modulators (ERXs) effectively target estrogen receptor positive human breast cancers. eLife 2017; 6. [PMID: 28786813 PMCID: PMC5548489 DOI: 10.7554/elife.26857] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/05/2017] [Indexed: 12/22/2022] Open
Abstract
The majority of human breast cancer is estrogen receptor alpha (ER) positive. While anti-estrogens/aromatase inhibitors are initially effective, resistance to these drugs commonly develops. Therapy-resistant tumors often retain ER signaling, via interaction with critical oncogenic coregulator proteins. To address these mechanisms of resistance, we have developed a novel ER coregulator binding modulator, ERX-11. ERX-11 interacts directly with ER and blocks the interaction between a subset of coregulators with both native and mutant forms of ER. ERX-11 effectively blocks ER-mediated oncogenic signaling and has potent anti-proliferative activity against therapy-sensitive and therapy-resistant human breast cancer cells. ERX-11 is orally bioavailable, with no overt signs of toxicity and potent activity in both murine xenograft and patient-derived breast tumor explant models. This first-in-class agent, with its novel mechanism of action of disrupting critical protein-protein interactions, overcomes the limitations of current therapies and may be clinically translatable for patients with therapy-sensitive and therapy-resistant breast cancers. DOI:http://dx.doi.org/10.7554/eLife.26857.001 Around 70% of breast cancers in women need one or both of the female hormones (estrogen and progesterone) to grow. To treat these 'hormone-dependent' cancers, patients receive drugs that either block the production of estrogen or directly target a receptor protein that senses estrogen in the cancer cells. Unfortunately, many breast cancers develop resistance to these drugs. This resistance is often caused by genetic mutations that alter the estrogen receptor; for example, the receptor may develop the ability to interact with other proteins in the cell known as coregulators to promote tumor growth. Developing new drugs that prevent estrogen receptors from interacting with coregulators may provide more options for treating hormone-dependent breast cancers. Here, Raj et al. developed a new small molecule named ERX-11 that is able to inhibit the growth of human breast cancer cells that are sensitive to existing drugs as well as cells that have become drug-resistant. For the experiments, hormone-dependent breast cancer cells from humans were transplanted into mice. This procedure usually causes the mice to develop tumors, but giving the mice ERX-11 by mouth stopped estrogen receptors from interacting with coregulators and blocked the growth of tumors. Furthermore, ERX-11 does not appear to have any toxic effects on the mice, indicating that it may also be safe for humans. The findings of Raj et al. suggest that ERX-11 is a promising new drug candidate for treating some breast cancers. The next steps are to examine the effects of ERX-11 on mice and other animals in more detail before deciding whether this molecule is suitable for clinical trials. In the longer term, molecules similar to ERX-11 could also be developed into drugs to treat other types of cancer that are also caused by abnormal interactions of coregulator proteins. DOI:http://dx.doi.org/10.7554/eLife.26857.002
Collapse
Affiliation(s)
- Ganesh V Raj
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| | - Gangadhara Reddy Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, United States.,CDP program, University of Texas Health Cancer Center, San Antonio, United States
| | - Shihong Ma
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| | - Tae-Kyung Lee
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, United States
| | - Suryavathi Viswanadhapalli
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, United States
| | - Rui Li
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| | - Xihui Liu
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| | - Shino Murakami
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, United States.,Laboratory of Signaling and Gene Regulation, Cecil H and Ida Green Center for Reproductive Biology Sciences and Division of Basic Reproductive Biology Research, University of Texas Southwestern Medical Center, Dallas, United States
| | - Chien-Cheng Chen
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| | - Wan-Ru Lee
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| | - Monica Mann
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, United States
| | - Samaya Rajeshwari Krishnan
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, United States
| | - Bikash Manandhar
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, United States
| | - Vijay K Gonugunta
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, United States
| | - Douglas Strand
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| | - Rajeshwar Rao Tekmal
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, United States.,CDP program, University of Texas Health Cancer Center, San Antonio, United States
| | - Jung-Mo Ahn
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, United States
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, United States.,CDP program, University of Texas Health Cancer Center, San Antonio, United States
| |
Collapse
|
5
|
Harrelson JP, Lee MW. Expanding the view of breast cancer metabolism: Promising molecular targets and therapeutic opportunities. Pharmacol Ther 2016; 167:60-73. [DOI: 10.1016/j.pharmthera.2016.07.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/21/2016] [Indexed: 12/23/2022]
|
6
|
Wang W, Belosay A, Yang X, Hartman JA, Song H, Iwaniec UT, Turner RT, Churchwell MI, Doerge DR, Helferich WG. Effects of letrozole on breast cancer micro-metastatic tumor growth in bone and lung in mice inoculated with murine 4T1 cells. Clin Exp Metastasis 2016; 33:475-85. [PMID: 27209469 DOI: 10.1007/s10585-016-9792-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 04/11/2016] [Indexed: 12/11/2022]
Abstract
Breast cancer (BC) is the leading cancer in women worldwide. Metastasis occurs in stage IV BC with bone and lung being common metastatic sites. Here we evaluate the effects of the aromatase inhibitor letrozole on BC micro-metastatic tumor growth in bone and lung metastasis in intact and ovariectomized (OVX) mice with murine estrogen receptor negative (ER-) BC cells inoculated in tibia. Forty-eight BALB/c mice were randomly assigned to one of four groups: OVX, OVX + Letrozole, Intact, and Intact + Letrozole, and injected with 4T1 cells intra-tibially. Letrozole was subcutaneously injected daily for 23 days at a dose of 1.75 µg/g body weight. Tumor progression was monitored by bioluminescence imaging (BLI). Following necropsy, inoculated tibiae were scanned via µCT and bone response to tumor was scored from 0 (no ectopic mineralization/osteolysis) to 5 (extensive ectopic mineralization/osteolysis). OVX mice had higher tibial pathology scores indicative of more extensive bone destruction than intact mice, irrespective of letrozole treatment. Letrozole decreased serum estradiol levels and reduced lung surface tumor numbers in intact animals. Furthermore, mice receiving letrozole had significantly fewer tumor colonies and fewer proliferative cells in the lung than OVX and intact controls based on H&E and Ki-67 staining, respectively. In conclusion, BC-inoculated OVX animals had higher tibia pathology scores than BC-inoculated intact animals and letrozole reduced BC metastases to lungs. These findings suggest that, by lowering systemic estrogen level and/or by interacting with the host organ, the aromatase inhibitor letrozole has the potential to reduce ER- BC metastasis to lung.
Collapse
Affiliation(s)
- Wendan Wang
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, 574 Bevier Hall, 905 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - Aashvini Belosay
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, 574 Bevier Hall, 905 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - Xujuan Yang
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, 574 Bevier Hall, 905 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - James A Hartman
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, 574 Bevier Hall, 905 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - Huaxin Song
- Health Sciences Center, School of Nursing, Texas Tech University, Lubbock, TX, USA
| | - Urszula T Iwaniec
- Skeletal Biology Laboratory, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA.,Center for Healthy Aging Research, Oregon State University, Corvallis, OR, USA
| | - Russell T Turner
- Skeletal Biology Laboratory, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA.,Center for Healthy Aging Research, Oregon State University, Corvallis, OR, USA
| | - Mona I Churchwell
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Daniel R Doerge
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - William G Helferich
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, 574 Bevier Hall, 905 South Goodwin Avenue, Urbana, IL, 61801, USA.
| |
Collapse
|
7
|
Shankar A, Roy S, Rath GK, Julka PK, Kamal VK, Malik A, Patil J, Jeyaraj PA, Mahajan MK. Aromatase Inhibition and Capecitabine Combination as 1stor 2ndLine Treatment for Metastatic Breast Cancer - a Retrospective Analysis. Asian Pac J Cancer Prev 2015; 16:6359-64. [DOI: 10.7314/apjcp.2015.16.15.6359] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
8
|
Madhavan S, Gusev Y, Singh S, Riggins RB. ERRγ target genes are poor prognostic factors in Tamoxifen-treated breast cancer. J Exp Clin Cancer Res 2015; 34:45. [PMID: 25971350 PMCID: PMC4436109 DOI: 10.1186/s13046-015-0150-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 03/26/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND One-third of estrogen (ER+) and/or progesterone receptor-positive (PGR+) breast tumors treated with Tamoxifen (TAM) do not respond to initial treatment, and the remaining 70% are at risk to relapse in the future. Estrogen-related receptor gamma (ESRRG, ERRγ) is an orphan nuclear receptor with broad, structural similarities to classical ER that is widely implicated in the transcriptional regulation of energy homeostasis. We have previously demonstrated that ERRγ induces resistance to TAM in ER+ breast cancer models, and that the receptor's transcriptional activity is modified by activation of the ERK/MAPK pathway. We hypothesize that hyper-activation or over-expression of ERRγ induces a pro-survival transcriptional program that impairs the ability of TAM to inhibit the growth of ER+ breast cancer. The goal of the present study is to determine whether ERRγ target genes are associated with reduced distant metastasis-free survival (DMFS) in ER+ breast cancer treated with TAM. METHODS Raw gene expression data was obtained from 3 publicly available breast cancer clinical studies of women with ER+ breast cancer who received TAM as their sole endocrine therapy. ERRγ target genes were selected from 2 studies that published validated chromatin immunoprecipitation (ChIP) analyses of ERRγ promoter occupancy. Kaplan-Meier estimation was used to determine the association of ERRγ target genes with DMFS, and selected genes were validated in ER+, MCF7 breast cancer cells that express exogenous ERRγ. RESULTS Thirty-seven validated receptor target genes were statistically significantly altered in women who experienced a DM within 5 years, and could classify several independent studies into poor vs. good DMFS. Two genes (EEF1A2 and PPIF) could similarly separate ER+, TAM-treated breast tumors by DMFS, and their protein levels were measured in an ER+ breast cancer cell line model with exogenous ERRγ. Finally, expression of ERRγ and these two target genes are elevated in models of ER+ breast cancer with hyperactivation of ERK/MAPK. CONCLUSIONS ERRγ signaling is associated with poor DMFS in ER+, TAM-treated breast cancer, and ESRRG, EEF1A2, and PPIF comprise a 3-gene signaling node that may contribute to TAM resistance in the context of an active ERK/MAPK pathway.
Collapse
Affiliation(s)
- Subha Madhavan
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, 20057, USA.
| | - Yuriy Gusev
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, 20057, USA.
| | - Salendra Singh
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, 20057, USA.
| | - Rebecca B Riggins
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, 20057, USA.
| |
Collapse
|
9
|
Abstract
Aromatase is an enzyme that converts testosterones to estrogens. Inhibition of this enzyme has been shown to have several clinical utilities in breast cancer. Currently, there are three aromatase inhibitors (AIs) in clinical use, namely anastrozole, letrozole, and exemestane. AIs have been used in various clinical settings for breast cancer, ranging from chemoprevention in breast cancer to treating breast cancer in both early stage in the adjuvant setting and metastatic disease. This article reviews mechanism of action, AI classification, and clinical utilities of AIs in various clinical settings in the context of breast cancer.
Collapse
Affiliation(s)
- Saranya Chumsri
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
10
|
Yoshimaru T, Komatsu M, Miyoshi Y, Honda J, Sasa M, Katagiri T. Therapeutic advances in BIG3-PHB2 inhibition targeting the crosstalk between estrogen and growth factors in breast cancer. Cancer Sci 2015; 106:550-8. [PMID: 25736224 PMCID: PMC4452155 DOI: 10.1111/cas.12654] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/11/2015] [Accepted: 02/28/2015] [Indexed: 12/13/2022] Open
Abstract
Our previous studies demonstrated that specific inhibition of the BIG3-PHB2 complex, which is a critical modulator in estrogen (E2) signaling, using ERAP, a dominant negative peptide inhibitor, leads to suppression of E2-dependent estrogen receptor (ER) alpha activation through the reactivation of the tumor suppressive activity of PHB2. Here, we report that ERAP has significant suppressive effects against synergistic activation caused by the crosstalk between E2 and growth factors associated with intrinsic or acquired resistance to anti-estrogen tamoxifen in breast cancer cells. Intrinsic PHB2 released from BIG3 by ERAP effectively disrupted each interaction of membrane-associated ERα and insulin-like growth factor 1 receptor beta (IGF-1Rβ), EGFR, PI3K or human epidermal growth factor 2 (HER2) in the presence of E2 and the growth factors IGF or EGF, followed by inhibited the activation of IGF-1Rβ, EGFR or HER2, and reduced Akt, MAPK and ERα phosphorylation levels, resulting in significant suppression of proliferation of ERα-positive breast cancer cells in vitro and in vivo. More importantly, combined treatment with ERAP and tamoxifen led to a synergistic suppression of signaling that was activated by crosstalk between E2 and growth factors or HER2 amplification. Taken together, our findings suggest that the specific inhibition of BIG3-PHB2 is a novel potential therapeutic approach for the treatment of tamoxifen-resistant breast cancers activated by the crosstalk between E2 and growth factor signaling, especially in premenopausal women.
Collapse
Affiliation(s)
- Tetsuro Yoshimaru
- Division of Genome Medicine, Institute for Genome Research, The University of Tokushima, Tokushima, Japan
| | - Masato Komatsu
- Division of Genome Medicine, Institute for Genome Research, The University of Tokushima, Tokushima, Japan
| | - Yasuo Miyoshi
- Division of Breast and Endocrine, Department of Surgery, Hyogo College of Medicine, Hyogo, Japan
| | - Junko Honda
- Department of Surgery, National Hospital Organization Higashitokushima Medical Center, Tokushima, Japan
| | - Mitsunori Sasa
- Department of Surgery, Tokushima Breast Care Clinic, Tokushima, Japan
| | - Toyomasa Katagiri
- Division of Genome Medicine, Institute for Genome Research, The University of Tokushima, Tokushima, Japan
| |
Collapse
|
11
|
Chakraborti A, Gulati K, Ray A. Possible role of nitric oxide (NO) in the regulation of gender related differences in stress induced anxiogenesis in rats. Nitric Oxide 2014; 43:74-80. [DOI: 10.1016/j.niox.2014.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 07/28/2014] [Accepted: 08/14/2014] [Indexed: 02/03/2023]
|
12
|
Chumsri S, Schech A, Chakkabat C, Sabnis G, Brodie A. Advances in mechanisms of resistance to aromatase inhibitors. Expert Rev Anticancer Ther 2014; 14:381-93. [PMID: 24559291 DOI: 10.1586/14737140.2014.882233] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Clinically, there are two distinct types of aromatase inhibitor (AI) resistance, namely acquired and innate resistance. Because the underlying mechanisms of these two types of resistance may not be mutually exclusive, strategies to tackle these resistances may not be effective when used interchangeably. Activation of growth factor receptor pathways is the hallmark of acquired AI resistance. These pathways can be targeted either at the cell surface receptor level or their downstream signaling cascades. Currently, everolimus in combination with exemestane represents a new standard of care for patients progressing on non-steroidal AIs. HDAC inhibitors have also shown promising results For innate resistance, the combination of fulvestrant and AI in the front line setting represents a new treatment option, particularly for patients who present with de novo metastatic disease. A Phase III trial is currently ongoing to evaluate the benefit of CDK 4/6 inhibitor, palbociclib, in the first line setting in combination with AI.
Collapse
Affiliation(s)
- Saranya Chumsri
- Department of Medicine, University of Maryland, School of Medicine and the Greenebaum Cancer Center, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
13
|
Xu Y, Sun Q. Headway in resistance to endocrine therapy in breast cancer. J Thorac Dis 2012; 2:171-7. [PMID: 22263039 DOI: 10.3978/j.issn.2072-1439.2010.02.03.9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Accepted: 08/14/2010] [Indexed: 12/13/2022]
Abstract
Resistance to endocrine therapy is the major problem for ERα(+) breast cancer patients. Research in endocrine resistance, mainly based on breast cancer cell lines and transplantation animal models, has indicated that phosphorylation of estrogen receptors, high expression of SRC and high activation of ErbB/MAPK pathway are the 3 main mechanisms for occurrence of endocrine resistance. Restoration of ER expression and exploration of inhibitors to various biological targets are the 2 promising ways to solve this problem. Further research is needed to deeply explore relevant mechanisms and resolvents so as to guide clinical practice.
Collapse
Affiliation(s)
- Yali Xu
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, PR China
| | | |
Collapse
|
14
|
Cortez V, Mann M, Tekmal S, Suzuki T, Miyata N, Rodriguez-Aguayo C, Lopez-Berestein G, Sood AK, Vadlamudi RK. Targeting the PELP1-KDM1 axis as a potential therapeutic strategy for breast cancer. Breast Cancer Res 2012; 14:R108. [PMID: 22812534 PMCID: PMC3680946 DOI: 10.1186/bcr3229] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 07/19/2012] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION The estrogen receptor (ER) co-regulator proline glutamic acid and leucine-rich protein 1 (PELP1) is a proto-oncogene that modulates epigenetic changes on ER target gene promoters via interactions with lysine-specific histone demethylase 1 (KDM1). In this study, we assessed the therapeutic potential of targeting the PELP1-KDM1 axis in vivo using liposomal (1,2-dioleoyl-sn-glycero-3-phosphatidylcholine; DOPC) siRNA to downregulate PELP1 expression and KDM1 inhibitors, pargyline and N-((1S)-3-(3-(trans-2-aminocyclopropyl)phenoxy)-1-(benzylcarbamoyl)propyl)benzamide using preclinical models. METHODS Preclinical xenograft models were used to test the efficacy of drugs in vivo. Ki-67 and terminal deoxynucleotidyl transferase dUTP nick end-labeling immunohistochemical analysis of epigenetic markers was performed on tumor tissues. The in vitro effect of PELP1-KDM axis blockers was tested using proliferation, reporter gene, chromatin immunoprecipitation and real-time RT-PCR assays. The efficacy of the KDM1 targeting drugs alone or in combination with letrozole and tamoxifen was tested using therapy-resistant model cells. RESULTS Treatment of ER-positive xenograft-based breast tumors with PELP1-siRNA-DOPC or pargyline reduced tumor volume by 58.6% and 62%, respectively. In a postmenopausal model, in which tumor growth is stimulated solely by local estrogen synthesis, daily pargyline treatment reduced tumor volume by 78%. Immunohistochemical analysis of excised tumors revealed a combined decrease in cellular proliferation, induction of apoptosis and upregulation of inhibitory epigenetic modifications. Pharmacological inhibition of KDM1 in vitro increased inhibitory histone mark dimethylation of histone H3 at lysine 9 (H3K9me2) and decreased histone activation mark acetylation of H3K9 (H3K9Ac) on ER target gene promoters. Combining KDM1 targeting drugs with current endocrine therapies substantially impeded growth and restored sensitivity of therapy-resistant breast cancer cells to treatment. CONCLUSION Our results suggest inhibition of PELP1-KDM1-mediated histone modifications as a potential therapeutic strategy for blocking breast cancer progression and therapy resistance.
Collapse
|
15
|
Sweeney EE, McDaniel RE, Maximov PY, Fan P, Jordan VC. Models and Mechanisms of Acquired Antihormone Resistance in Breast Cancer: Significant Clinical Progress Despite Limitations. Horm Mol Biol Clin Investig 2012; 9:143-163. [PMID: 23308083 PMCID: PMC3539798 DOI: 10.1515/hmbci-2011-0004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Translational research for the treatment and prevention of breast cancer depends upon the four Ms: models, molecules, and mechanisms in order to create medicines. The process, to target the estrogen receptor (ER) in estrogen-dependent breast cancer, has yielded significant advances in patient survivorship and the first approved medicines (tamoxifen and raloxifene) to reduce the incidence of any cancer in high- or low-risk women. This review focuses on the critical role of the few ER-positive cell lines (MCF-7, T47D, BT474, ZR-75) that continue to advance our understanding of the estrogen-regulated biology of breast cancer. More importantly, the model cell lines have provided an opportunity to document the development and evolution of acquired antihormone resistance. The description of this evolutionary process that occurs in micrometastatic disease during up to a decade of adjuvant therapy would not be possible in the patient. The use of the MCF-7 breast cancer cell line in particular has been instrumental in discovering a vulnerability of ER-positive breast cancer exhaustively treated with antihormone therapy. Physiologic estradiol acts as an apoptotic trigger to cause tumor regression. These unanticipated findings in the laboratory have translated to clinical advances in our knowledge of the paradoxical role of estrogen in the life and death of breast cancer.
Collapse
Affiliation(s)
- Elizabeth E Sweeney
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | | | | | | | | |
Collapse
|
16
|
Gaughan EM, Come SE. Optimizing Endocrine Therapy for Metastatic Breast Cancer. CURRENT BREAST CANCER REPORTS 2012. [DOI: 10.1007/s12609-011-0063-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
17
|
Nair BC, Vallabhaneni S, Tekmal RR, Vadlamudi RK. Roscovitine confers tumor suppressive effect on therapy-resistant breast tumor cells. Breast Cancer Res 2011; 13:R80. [PMID: 21834972 PMCID: PMC3218960 DOI: 10.1186/bcr2929] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 06/28/2011] [Accepted: 08/11/2011] [Indexed: 12/21/2022] Open
Abstract
Introduction Current clinical strategies for treating hormonal breast cancer involve the use of anti-estrogens that block estrogen receptor (ER)α functions and aromatase inhibitors that decrease local and systemic estrogen production. Both of these strategies improve outcomes for ERα-positive breast cancer patients, however, development of therapy resistance remains a major clinical problem. Divergent molecular pathways have been described for this resistant phenotype and interestingly, the majority of downstream events in these resistance pathways converge upon the modulation of cell cycle regulatory proteins including aberrant activation of cyclin dependent kinase 2 (CDK2). In this study, we examined whether the CDK inhibitor roscovitine confers a tumor suppressive effect on therapy-resistant breast epithelial cells. Methods Using various in vitro and in vivo assays, we tested the effect of roscovitine on three hormonal therapy-resistant model cells: (a) MCF-7-TamR (acquired tamoxifen resistance model); (b) MCF-7-LTLTca (acquired letrozole resistance model); and (c) MCF-7-HER2 that exhibit tamoxifen resistance (ER-growth factor signaling cross talk model). Results Hormonal therapy-resistant cells exhibited aberrant activation of the CDK2 pathway. Roscovitine at a dose of 20 μM significantly inhibited the cell proliferation rate and foci formation potential of all three therapy-resistant cells. The drug treatment substantially increased the proportion of cells in G2/M cell cycle phase with decreased CDK2 activity and promoted low cyclin D1 levels. Interestingly, roscovitine also preferentially down regulated the ERα isoform and ER-coregulators including AIB1 and PELP1. Results from xenograft studies further showed that roscovitine can attenuate growth of therapy-resistant tumors in vivo. Conclusions Roscovitine can reduce cell proliferation and survival of hormone therapy-resistant breast cancer cells. Our results support the emerging concept that inhibition of CDK2 activity has the potential to abrogate growth of hormonal therapy-resistant cells.
Collapse
Affiliation(s)
- Binoj C Nair
- Department of Obstetrics and Gynecology, CTRC at UT Health Science Center, San Antonio, Texas 78229, USA
| | | | | | | |
Collapse
|
18
|
Scott SM, Brown M, Come SE. Emerging data on the efficacy and safety of fulvestrant, a unique antiestrogen therapy for advanced breast cancer. Expert Opin Drug Saf 2011; 10:819-26. [PMID: 21699443 DOI: 10.1517/14740338.2011.595560] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Fulvestrant is an antiestrogen therapy with a unique mechanism of action. Unlike the selective estrogen receptor modulator tamoxifen, fulvestrant has no known estrogen agonist activity and is considered a pure antiestrogen. Its primary mechanism of action is thought to result from downregulation of the estrogen receptor (ER). Considerable data have demonstrated the efficacy of fulvestrant in postmenopausal women with ER-positive advanced breast cancer, both in the first-line setting and following disease progression on tamoxifen or aromatase inhibitors. Recent studies report improved benefit with alternative dosing strategies. At all administration schedules, fulvestrant has an excellent safety profile with no significant adverse effects. AREAS COVERED This article provides a review of the mechanism of action of fulvestrant and the preclinical and clinical data evaluating its use as a form of endocrine therapy. The reader will gain insight into the pharmacologic properties of the drug and its role in the treatment of advanced hormone receptor-positive breast cancer in postmenopausal women. EXPERT OPINION Based on data demonstrating the efficacy of fulvestrant, including prolonged clinical benefit in many patients, this well-tolerated antiestrogen is an important therapy for breast cancer. The optimal position of fulvestrant in the sequence of endocrine therapies for postmenopausal women and its role in combination regimens are not yet resolved.
Collapse
Affiliation(s)
- Sarah M Scott
- Beth Israel Deaconess Medical Center, Dana Farber/Harvard Cancer Center, 330 Brookline Ave, Boston, MA 02215, USA
| | | | | |
Collapse
|
19
|
Verma S, Sehdev S, Joy A, Madarnas Y, Younus J, Roy JA. An updated review on the efficacy of adjuvant endocrine therapies in hormone receptor-positive early breast cancer. ACTA ACUST UNITED AC 2011; 16 Suppl 2:S1-13. [PMID: 19672416 PMCID: PMC2722048 DOI: 10.3747/co.v16i0.455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The third-generation aromatase inhibitors (AIS) are largely replacing tamoxifen in the adjuvant treatment of early-stage breast cancer in postmenopausal women with hormone receptor–positive tumours. To date, multiple trials have been conducted comparing tamoxifen treatment with an AI, and all have demonstrated improved disease-free survival with AI treatment. Trials have included direct 5-year comparisons between tamoxifen and an AI, switching to an AI within 5 years after initial tamoxifen treatment, or extending treatment with an AI after 5 years of completed tamoxifen treatment. Some of these trials have been completed; others are ongoing; and head-to-head trial comparisons of individual AIS are also in progress. The present article summarizes the data obtained from various clinical trials of hormonal therapy for early breast cancer. It also reviews recent data so as to shed light on the current status of these therapies. The focus is on the efficacy of treatment with an AI. Toxicity is discussed in the second article in this supplement.
Collapse
Affiliation(s)
- S Verma
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON.
| | | | | | | | | | | |
Collapse
|
20
|
Chumsri S, Howes T, Bao T, Sabnis G, Brodie A. Aromatase, aromatase inhibitors, and breast cancer. J Steroid Biochem Mol Biol 2011; 125:13-22. [PMID: 21335088 PMCID: PMC3104073 DOI: 10.1016/j.jsbmb.2011.02.001] [Citation(s) in RCA: 252] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Revised: 01/31/2011] [Accepted: 02/03/2011] [Indexed: 12/15/2022]
Abstract
Estrogens are known to be important in the growth of breast cancers in both pre and postmenopausal women. As the number of breast cancer patients increases with age, the majority of breast cancer patients are postmenopausal women. Although estrogens are no longer made in the ovaries after menopause, peripheral tissues produce sufficient concentrations to stimulate tumor growth. As aromatase catalyzes the final and rate-limiting step in the biosynthesis of estrogen, inhibitors of this enzyme are effective targeted therapy for breast cancer. Three aromatase inhibitors (AIs) are now FDA approved and have been shown to be more effective than the antiestrogen tamoxifen and are well tolerated. AIs are now a standard treatment for postmenopausal patients. AIs are effective in adjuvant and first-line metastatic setting. This review describes the development of AIs and their current use in breast cancer. Recent research focuses on elucidating mechanisms of acquired resistance that may develop in some patients with long term AI treatment and also in innate resistance. Preclinical data in resistance models demonstrated that the crosstalk between ER and other signaling pathways particularly MAPK and PI3K/Akt is an important resistant mechanism. Blockade of these other signaling pathways is an attractive strategy to circumvent the resistance to AI therapy in breast cancer. Several clinical trials are ongoing to evaluate the role of these novel targeted therapies to reverse resistance to AIs. Article from the special issue on 'Targeted Inhibitors'.
Collapse
Affiliation(s)
- Saranya Chumsri
- Department of Medicine, University of Maryland School of Medicine, and the Greenebaum Cancer Center, Baltimore, MD, 21 USA
| | - Timothy Howes
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, and the Greenebaum Cancer Center, Baltimore, MD, 21 USA
| | - Ting Bao
- Department of Medicine, University of Maryland School of Medicine, and the Greenebaum Cancer Center, Baltimore, MD, 21 USA
| | - Gauri Sabnis
- Department of Pharmacology and Experimental Therapeutics, University of Maryland School of Medicine, and the Greenebaum Cancer Center, Baltimore, MD, 21 USA
| | - Angela Brodie
- Department of Pharmacology and Experimental Therapeutics, University of Maryland School of Medicine, and the Greenebaum Cancer Center, Baltimore, MD, 21 USA
| |
Collapse
|
21
|
Leary AF, Drury S, Detre S, Pancholi S, Lykkesfeldt AE, Martin LA, Dowsett M, Johnston SRD. Lapatinib restores hormone sensitivity with differential effects on estrogen receptor signaling in cell models of human epidermal growth factor receptor 2-negative breast cancer with acquired endocrine resistance. Clin Cancer Res 2010; 16:1486-97. [PMID: 20179226 DOI: 10.1158/1078-0432.ccr-09-1764] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Acquired endocrine resistance in estrogen receptor (ER)alpha+/human epidermal growth factor receptor 2-negative (HER2-) breast cancer has been associated with modest adaptive increases in HER2, although exactly how aberrant HER2 signaling affects the ERalpha pathway is poorly understood. We investigated (a) whether the epidermal growth factor receptor/HER2 inhibitor lapatinib could restore endocrine responsiveness in cell models of acquired endocrine resistance with modest increases in HER2, and (b) the nature of ERalpha-HER2 cross-talk in this process. METHODS Combination growth studies, ERalpha transcription, immunoblot, and gene expression assays were conducted in two models of acquired resistance to (a) estrogen deprivation (long-term estrogen-deprived cells) and (b) tamoxifen (long-term tamoxifen-treated cells), and in hormone sensitive controls. Changes in ERalpha, PgR, and HER2 were assessed in samples from patients treated with tamoxifen. RESULTS Both cell models of acquired endocrine resistance showed modest adaptive upregulation in HER2, and lapatinib restored endocrine sensitivity in both. The effect of lapatinib on ERalpha signaling varied markedly depending on the nature of the HER2/ERalpha cross-talk. In long-term estrogen-deprived cells characterized by enhanced ERalpha function, lapatinib suppressed ERalpha genomic activity (as measured by pERSer118, ERalpha transcriptional activity, and PGR gene expression). In contrast, in long-term tamoxifen-treated cells with reduced ERalpha activation, lapatinib reactivated ERalpha genomic function. Twenty percent of tamoxifen-resistant patients relapsed with modest increases in HER2 and either suppressed or enhanced ERalpha/PgR expression. CONCLUSIONS Aberrant GFR signaling can augment or suppress ERalpha function. Regardless, interrupting the HER2/ERalpha cross-talk with lapatinib can restore endocrine sensitivity and should be investigated as a therapeutic strategy in combination with endocrine therapy in ERalpha+/HER2- patients with acquired endocrine resistance.
Collapse
Affiliation(s)
- Alexandra F Leary
- Royal Marsden Hospital, Institute of Cancer Research, London, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Hayes TB. The one stop shop: chemical causes and cures for cancer. REVIEWS ON ENVIRONMENTAL HEALTH 2009; 24:333-337. [PMID: 20384042 DOI: 10.1515/reveh.2009.24.4.333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Affiliation(s)
- Tyrone B Hayes
- Department of Integrative Biology, University of California Berkeley, Berkeley, California, USA
| |
Collapse
|
23
|
Fox EM, Andrade J, Shupnik MA. Novel actions of estrogen to promote proliferation: integration of cytoplasmic and nuclear pathways. Steroids 2009; 74:622-7. [PMID: 18996136 PMCID: PMC2702758 DOI: 10.1016/j.steroids.2008.10.014] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2008] [Accepted: 10/25/2008] [Indexed: 12/28/2022]
Abstract
Both steroids and growth factors stimulate proliferation of steroid-dependent tumor cells, and interaction between these signaling pathways occurs at several levels. Steroid receptors are classified as ligand-activated transcription factors, and steps by which they activate target gene transcription are well understood. Several steroid responses have now been functionally linked to other intracellular signaling pathways, including c-Src or tyrosine kinase receptors. Steroids such as 17beta-estradiol (E2), via binding to cytoplasmic or membrane-associated receptors, were also shown to rapidly activate intracellular signaling cascades such as ERK, PI3K and STATs. These E2-stimulated phosphorylations can then contribute to altered tumor cell function. ER-positive breast cancer cells, in which proliferation is stimulated by E2 and suppressed by antiestrogens, have been of particular interest in dissecting nuclear and cytoplasmic roles of estrogen receptors (ER). In some cell contexts, ER interacts directly with the intracellular tyrosine kinase c-Src and other cytoplasmic signaling and adaptor molecules, such as Shc, PI3K, MNAR, and p130 Cas. Although the hierarchy among these associations is not known, it is clear that c-Src plays a fundamental role in both growth factor and E2-stimulated cell growth, and this may also require other growth factor receptors such as those for EGF or IGF-1. STAT transcription factors represent one pathway to integrate E2 cytoplasmic and nuclear signaling. STAT5 is phosphorylated in the cytoplasm at an activating tyrosine in response to E2 or EGF, and then is translocated to the nucleus to stimulate target gene transcription. E2 stimulates recruitment of STAT5 and ER to the promoter of several proliferative genes, and STAT5 knockdown prevents recruitment of either protein to these promoters. STAT5 activation by E2 in breast cancer cells requires c-Src and EGF receptor, and inhibition of c-Src or EGFR, or knockdown of STAT5, prevents E2 stimulation of several genes and breast cancer cell proliferation. Hyperactivation of the growth factor receptor-c-Src pathway can in some contexts decrease growth responses to E2, or render cells and tumors resistant to suppressive actions of endocrine therapies. Crosstalk between growth factors and steroids in both the cytoplasm and nucleus may thus have a profound impact on complex biological processes such as cell growth, and may play a significant role in the treatment of steroid-dependent breast cancers.
Collapse
Affiliation(s)
- Emily M. Fox
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22903
| | - Josefa Andrade
- Department of, Medicine, University of Virginia School of Medicine, Charlottesville, VA 22903
| | - Margaret A. Shupnik
- Department of, Medicine, University of Virginia School of Medicine, Charlottesville, VA 22903
| |
Collapse
|
24
|
Bartsch R, Mlineritsch B, Gnant M, Niernberger T, Pluschnig U, Greil R, Wenzel C, Sevelda P, Thaler J, Rudas M, Pober M, Zielinski CC, Steger GG. The Austrian fulvestrant registry: results from a prospective observation of fulvestrant in postmenopausal patients with metastatic breast cancer. Breast Cancer Res Treat 2008; 115:373-80. [DOI: 10.1007/s10549-008-0132-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Accepted: 07/10/2008] [Indexed: 12/23/2022]
|
25
|
Macedo LF, Sabnis GJ, Goloubeva OG, Brodie A. Combination of anastrozole with fulvestrant in the intratumoral aromatase xenograft model. Cancer Res 2008; 68:3516-22. [PMID: 18451180 DOI: 10.1158/0008-5472.can-07-6807] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although the aromatase inhibitor anastrozole has been shown to be very effective in the treatment of hormone-dependent postmenopausal breast cancer, some patients with advanced disease will develop resistance to treatment. To investigate therapeutic strategies to overcome resistance to anastrozole treatment, we have used an intratumoral aromatase model that simulates postmenopausal breast cancer patients with estrogen-dependent tumors. Growth of the tumors in the mice was inhibited by both anastrozole and fulvestrant compared with the control tumors. Nevertheless, tumors had doubled in size at 5 weeks of treatment. We therefore investigated whether switching the original treatments to anastrozole or fulvestrant alone or the combination of anastrozole plus fulvestrant would reduce tumor growth. The results showed that the best strategy to reverse the insensitivity to anastrozole or fulvestrant is to combine the two agents. Additionally, the tumors treated with anastrozole plus fulvestrant from the beginning had only just doubled their size after 14 weeks of treatment, whereas the anastrozole and fulvestrant treatments alone resulted in 9- and 12-fold increases in tumor size, respectively, in the same time period. Anastrozole plus fulvestrant from the beginning or in sequence was associated with down-regulation of signaling proteins involved in the development of hormonal resistance such as insulin-like growth factor type I receptor beta, mitogen-activated protein kinase (MAPK), p-MAPK, AKT, mammalian target of rapamycin (mTOR), p-mTOR, and estrogen receptor alpha compared with tumors treated with anastrozole or fulvestrant alone. These results suggest that blocking the estrogen receptor and aromatase may delay or reverse the development of resistance to aromatase inhibitors in advanced breast cancer patients.
Collapse
Affiliation(s)
- Luciana F Macedo
- Department of Pharmacology and Experimental Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
26
|
Eng-Wong J, Zujewski JA. Current NCI-sponsored Cooperative Group trials of endocrine therapies in breast cancer. Cancer 2008; 112:723-729. [PMID: 18072276 DOI: 10.1002/cncr.23188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Over several decades, investigators working through National Cancer Institute-sponsored Cooperative Groups have contributed to major advances in the endocrine treatment of breast cancer. Accomplishments include the benefit of tamoxifen therapy for early stage invasive and noninvasive breast cancer, the benefit of raloxifene and tamoxifen for prevention of breast cancer, the improved efficacy of tamoxifen after chemotherapy as opposed to concurrent administration, and the ability of letrozole administered after 5 years of tamoxifen to improve disease-free survival. Most recently, Cooperative Group studies have contributed to the development of a molecular profiling test, Oncotype Dx, which identifies women who have an excellent prognosis with hormonal therapy alone. Ongoing phase 3 clinical trials address the following questions: Is prolonged duration of aromatase inhibitor (AI) therapy beneficial? What is the efficacy and toxicity of steroidal versus nonsteroidal AIs in adjuvant treatment? Is combination hormonal therapy with an estrogen receptor down-regulator (fulvestrant) and an AI superior to an AI alone in the treatment of metastatic breast cancer? Does ovarian suppression offer superior benefit to standard therapy in the treatment of premenopausal breast cancer? What is the role of chemotherapy for early stage breast cancer selected via molecular profiling analysis? How can targeted therapies be used effectively in combination? Studies in subsets of patients defined by molecular profiling will be necessary to fully define breast cancer subtypes and realize the promise of personalized medicine. Close research partnerships that promote large-scale translational research are essential to the continuation of rapid achievements in this field.
Collapse
Affiliation(s)
- Jennifer Eng-Wong
- Medical Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - Jo Anne Zujewski
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|