1
|
Babyshkina N, Dronova T, Erdyneeva D, Gervas P, Cherdyntseva N. Role of TGF-β signaling in the mechanisms of tamoxifen resistance. Cytokine Growth Factor Rev 2021; 62:62-69. [PMID: 34635390 DOI: 10.1016/j.cytogfr.2021.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/18/2021] [Accepted: 09/19/2021] [Indexed: 12/24/2022]
Abstract
The transforming growth factor beta (TGF-β) signaling pathway plays complex role in the regulation of cell proliferation, apoptosis and differentiation in breast cancer. TGF-β activation can lead to multiple cellular responses mediating the drug resistance evolution, including the resistance to antiestrogens. Tamoxifen is the most commonly prescribed antiestrogen that functionally involved in regulation of TGF-β activity. In this review, we focus on the role of TGF-β signaling in the mechanisms of tamoxifen resistance, including its interaction with estrogen receptors alfa (ERα) pathway and breast cancer stem cells (BCSCs). We summarize the current reported data regarding TGF-β signaling components as markers of tamoxifen resistance and review current approaches to overcoming tamoxifen resistance based on studies of TGF-β signaling.
Collapse
Affiliation(s)
- Nataliya Babyshkina
- Department of Molecular Oncology and Immunology, Саncеr Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634050, Russian Federation; Siberian State Medical University, Tomsk 634050, Russian Federation.
| | - Tatyana Dronova
- Department of Biology of Tumor Progression, Саncеr Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634050, Russian Federation
| | - Daiana Erdyneeva
- Department of Molecular Oncology and Immunology, Саncеr Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634050, Russian Federation
| | - Polina Gervas
- Department of Molecular Oncology and Immunology, Саncеr Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634050, Russian Federation
| | - Nadejda Cherdyntseva
- Department of Molecular Oncology and Immunology, Саncеr Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634050, Russian Federation
| |
Collapse
|
2
|
Ma G, He J, Yu Y, Xu Y, Yu X, Martinez J, Lonard DM, Xu J. Tamoxifen inhibits ER-negative breast cancer cell invasion and metastasis by accelerating Twist1 degradation. Int J Biol Sci 2015; 11:618-28. [PMID: 25892968 PMCID: PMC4400392 DOI: 10.7150/ijbs.11380] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/09/2015] [Indexed: 12/22/2022] Open
Abstract
Twist1 is a transcription factor driving epithelial-mesenchymal transition, invasion and metastasis of breast cancer cells. Mice with germ-line Twist1 knockout are embryonic lethal, while adult mice with inducible Twist1 knockout have no obvious health problems, suggesting that Twist1 is a viable therapeutic target for the inhibition of invasion and metastasis of breast cancer in adult patients. In this study, we expressed a luciferase protein or a Twist1-luciferase fusion protein in HeLa cells as part of a high throughput system to screen 1280 compounds in the Library of Pharmacologically Active Compounds (LOPAC) from Sigma-Aldrich for their effects on Twist1 protein expression. One of the most interesting compounds identified is tamoxifen, a selective estrogen receptor (ER) modulator used to treat ER-positive breast cancer. Tamoxifen treatment significantly accelerated Twist1 degradation in multiple cell lines including HEK293 human kidney cells, 4T1 and 168FARN mouse mammary tumor cells with either ectopically or endogenously expressed Twist1. Tamoxifen-induced Twist1 degradation could be blocked by the MG132 proteasome inhibitor, suggesting that tamoxifen induces Twist1 degradation through the ubiquitination-proteasome pathway. However, tamoxifen-induced Twist1 degradation was independent of Twist1 mRNA expression, estrogen signaling and MAPK-mediated Twist1 phosphorylation in these cells. Importantly, tamoxifen also significantly inhibited invasive behavior in Matrigel and lung metastasis in SCID-bg mice of ER-negative 4T1 mammary tumor cells, which depend on endogenous Twist1 to invade and metastasize. These results indicate that tamoxifen can significantly accelerate Twist1 degradation to suppress cancer cell invasion and metastasis, suggesting that tamoxifen can be used not only to treat ER-positive breast cancers but also to reduce Twist1-mediated invasion and metastasis in ER-negative breast cancers.
Collapse
Affiliation(s)
- Gang Ma
- 1. Department of Breast and Thyroid Cancer Surgery, The First Affiliated Hospital of Xi'an Jiaotong University Medical School, Xi'an, China; ; 2. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jianjun He
- 1. Department of Breast and Thyroid Cancer Surgery, The First Affiliated Hospital of Xi'an Jiaotong University Medical School, Xi'an, China
| | - Yang Yu
- 2. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yixiang Xu
- 2. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; ; 3. Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, USA
| | - Xiaobin Yu
- 2. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jarrod Martinez
- 2. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - David M Lonard
- 2. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jianming Xu
- 2. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; ; 4. Institute for Cancer Medicine and School of Basic Medical Sciences, Luzhou Medical College, Sichuan, China
| |
Collapse
|
3
|
Nicolini A, Rossi G, Ferrari P, Carpi A. Clinical and laboratory patterns during immune stimulation in hormone responsive metastatic breast cancer. Biomed Pharmacother 2013; 68:171-8. [PMID: 24491318 DOI: 10.1016/j.biopha.2013.11.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 11/11/2013] [Indexed: 11/29/2022] Open
Abstract
This study clarifies the relationship between clinical and laboratory patterns, in endocrine-responsive metastatic breast cancer patients treated with a cyclic beta-interferon and interleukin-2 sequence added to anti-estrogens. In 31 patients, a regular laboratory and immunological assessment was made. During clinical benefit, as opposed to progression, a significant increase in the total number of lymphocytes, CD4+, CD8+, NK cells, CRP and IL-12 was confirmed. Also, a significant CEA, TPA, CA15.3 decrease occurred 24-72h after interleukin-2 administration. At the progression, both basally and after interleukin-2 stimulation, the mean values of CD4+ plus CD25+ cells were more than twice higher than during clinical benefit, with a decrease of CD4+ plus CD8+ (Teffector)/CD4+CD25+ (Treg) ratio. Moreover, a significant increase for CEA and for all 3 markers (standardized values) was found 24-72h after interleukin-2 administration. In patients who survived less than 5years, the Treg cell increase occurred at a significantly shorter time interval than in those who survived longer than 5years (20 vs 45.5months, respectively; P=0.001). These data show laboratory evidence of the effect of immunotherapy as well as that of hormone resistance occuring concomitantly with a laboratory pattern compatible with immune inhibition.
Collapse
Affiliation(s)
| | - Giuseppe Rossi
- Unit of Epidemiology and Biostatistics, Institute of Clinical Physiology, National Council of Research, Pisa, Italy
| | - Paola Ferrari
- Department of Oncology, University of Pisa, Pisa, Italy
| | - Angelo Carpi
- Department of Reproduction and Ageing, University of Pisa, Italy
| |
Collapse
|
4
|
Sun X, Jiang R, Przepiorski A, Reddy S, Palmano KP, Krissansen GW. "Iron-saturated" bovine lactoferrin improves the chemotherapeutic effects of tamoxifen in the treatment of basal-like breast cancer in mice. BMC Cancer 2012; 12:591. [PMID: 23231648 PMCID: PMC3539967 DOI: 10.1186/1471-2407-12-591] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 12/04/2012] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tamoxifen is used in hormone therapy for estrogen-receptor (ER)-positive breast cancer, but also has chemopreventative effects against ER-negative breast cancers. This study sought to investigate whether oral iron-saturated bovine lactoferrin (Fe-Lf), a natural product which enhances chemotherapy, could improve the chemotherapeutic effects of tamoxifen in the treatment of ER-negative breast cancers. METHODS In a model of breast cancer prevention, female Balb/c mice treated with tamoxifen (5 mg/Kg) were fed an Fe-Lf supplemented diet (5 g/Kg diet) or the base diet. At week 2, 4T1 mammary carcinoma cells were injected into an inguinal mammary fat pad. In a model of breast cancer treatment, tamoxifen treatment was not started until two weeks following tumor cell injection. Tumor growth, metastasis, body weight, and levels of interleukin 18 (IL-18) and interferon γ (IFN-γ) were analyzed. RESULTS Tamoxifen weakly (IC(50) ~ 8 μM) inhibited the proliferation of 4T1 cells at pharmacological concentrations in vitro. In the tumor prevention study, a Fe-Lf diet in combination with tamoxifen caused a 4 day delay in tumor formation, and significantly inhibited tumor growth and metastasis to the liver and lung by 48, 58, and 66% (all P < 0.001), respectively, compared to untreated controls. The combination therapy was significantly (all P < 0.05) more effective than the respective monotherapies. Oral Fe-Lf attenuated the loss of body weight caused by tamoxifen and cancer cachexia. It prevented tamoxifen-induced reductions in serum levels of IL-18 and IFN-γ, and intestinal cells expressing IL-18 and IFN-γ. It increased the levels of Lf in leukocytes residing in gut-associated lymphoid tissues. B, T and Natural killer (NK) cells containing high levels of Lf were identified in 4T1 tumors, suggesting they had migrated from the intestine. Similar effects of Fe-Lf and tamoxifen on tumor cell viability were seen in the treatment of established tumors. CONCLUSIONS The results indicate that Fe-Lf is a potent natural adjuvant capable of augmenting the chemotherapeutic activity of tamoxifen. It could have application in delaying relapse in tamoxifen-treated breast cancer patients who are at risk of developing ER-negative tumors.
Collapse
Affiliation(s)
- Xueying Sun
- Department of Molecular Medicine & Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1005, New Zealand
| | | | | | | | | | | |
Collapse
|
5
|
Tamoxifen decreases extracellular TGF-beta1 secreted from breast cancer cells--a post-translational regulation involving matrix metalloproteinase activity. Exp Cell Res 2008; 315:1-9. [PMID: 18996114 DOI: 10.1016/j.yexcr.2008.10.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2008] [Revised: 09/19/2008] [Accepted: 10/07/2008] [Indexed: 11/23/2022]
Abstract
Transforming growth factor-beta1 (TGF-beta1) promotes cancer progression by regulating tumor cell growth and angiogenesis and high levels of TGF-beta1 have been associated with metastatic disease and poor prognosis in breast cancer patients. We have previously reported anti-angiogenic effects of the anti-estrogen tamoxifen in breast cancer, by increased matrix metalloproteinase-9 (MMP-9) activity and generation of endostatin. Here, we show that exposure of tamoxifen to ER-positive breast cancer cells for 7 days, decreased extracellular TGF-beta1. Intracellular TGF-beta1 levels were unaffected by tamoxifen treatment, indicating a post-translational regulation of TGF-beta1. Inhibition of MMP activity restored TGF-beta1 levels, suggesting an involvement of MMP activities in the down-regulation of TGF-beta1 by tamoxifen. Moreover, using an in vivo model of solid MCF-7 tumors in nude mice, we analyzed tumor levels of TGF-beta1 after in vivo treatment with estradiol and tamoxifen. Exposure of tumor-bearing mice to tamoxifen significantly decreased tumor TGF-beta1 protein levels, tumor growth and angiogenesis. In conclusion, our findings suggest a novel mechanism of action of tamoxifen in breast cancer via sex steroid dependent modulation of the proteolytic tumor microenvironment resulting in reduced extracellular TGF-beta1 levels.
Collapse
|
6
|
Tan AR, Alexe G, Reiss M. Transforming growth factor-beta signaling: emerging stem cell target in metastatic breast cancer? Breast Cancer Res Treat 2008; 115:453-95. [PMID: 18841463 DOI: 10.1007/s10549-008-0184-1] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Accepted: 09/02/2008] [Indexed: 12/24/2022]
Abstract
In most human breast cancers, lowering of TGFbeta receptor- or Smad gene expression combined with increased levels of TGFbetas in the tumor microenvironment is sufficient to abrogate TGFbetas tumor suppressive effects and to induce a mesenchymal, motile and invasive phenotype. In genetic mouse models, TGFbeta signaling suppresses de novo mammary cancer formation but promotes metastasis of tumors that have broken through TGFbeta tumor suppression. In mouse models of "triple-negative" or basal-like breast cancer, treatment with TGFbeta neutralizing antibodies or receptor kinase inhibitors strongly inhibits development of lung- and bone metastases. These TGFbeta antagonists do not significantly affect tumor cell proliferation or apoptosis. Rather, they de-repress anti-tumor immunity, inhibit angiogenesis and reverse the mesenchymal, motile, invasive phenotype characteristic of basal-like and HER2-positive breast cancer cells. Patterns of TGFbeta target genes upregulation in human breast cancers suggest that TGFbeta may drive tumor progression in estrogen-independent cancer, while it mediates a suppressive host cell response in estrogen-dependent luminal cancers. In addition, TGFbeta appears to play a key role in maintaining the mammary epithelial (cancer) stem cell pool, in part by inducing a mesenchymal phenotype, while differentiated, estrogen receptor-positive, luminal cells are unresponsive to TGFbeta because the TGFBR2 receptor gene is transcriptionally silent. These same cells respond to estrogen by downregulating TGFbeta, while antiestrogens act by upregulating TGFbeta. This model predicts that inhibiting TGFbeta signaling should drive the differentiation of mammary stem cells into ductal cells. Consequently, TGFbeta antagonists may convert basal-like or HER2-positive cancers to a more epithelioid, non-proliferating (and, perhaps, non-metastatic) phenotype. Conversely, these agents might antagonize the therapeutic effects of anti-estrogens in estrogen-dependent luminal cancers. These predictions need to be addressed prospectively in clinical trials and should inform the selection of patient populations most likely to benefit from this novel anti-metastatic therapeutic approach.
Collapse
Affiliation(s)
- Antoinette R Tan
- Division of Medical Oncology, Department of Internal Medicine, UMDNJ-Robert Wood Johnson Medical School and The Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| | | | | |
Collapse
|
7
|
Cherlet T, Murphy LC. Estrogen receptors inhibit Smad3 transcriptional activity through Ap-1 transcription factors. Mol Cell Biochem 2007; 306:33-42. [PMID: 17660955 DOI: 10.1007/s11010-007-9551-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Accepted: 07/12/2007] [Indexed: 02/07/2023]
Abstract
Breast tumorigenesis and breast cancer progression involves the deregulation or hyperactivation of intracellular signaling proteins that leads to uncontrolled cellular proliferation, invasion and metastasis. For example, the expression and cellular responses to estogen receptor (ER) and transforming growth factor beta (TGFbeta) signaling pathways change during breast tumorigenesis and breast cancer progression. While the expression and activity of ER and TGFbeta maybe significant in the development of breast cancer, alterations in the cross-talk between these pathways may be equally important. Autocrine and paracrine effects of TGFbeta on breast cancer cell growth have been known for some time, but only recently have direct interactions between ER and TGFbeta been described. The purpose of this article was to further characterize the cross-talk between ER and TGFbeta, by examining ER interaction with Smad3, a downstream mediator of TGFbeta signaling. Transient transfection of Cos1 cells with p3TP-lux, demonstrate that ERalpha and ERbeta(1) repress Smad3 transcriptional activity in an estradiol-dependent manner and that this effect is inhibited by antiestrogen treatment. The ERbeta variants, ERbeta(2) and ERbeta(5), did not have any effect on Smad3 transcriptional activity. Further experiments attempted to characterize the molecular mechanism by which activated ER inhibits Smad3 transcriptional activity. Results indicate that ligand-bound ER does not affect Smad3 protein expression levels and that ER does not form direct protein interactions with Smad3. Transient transfection of Cos1 cells with the Ap-1 transcription factor c-Jun but not c-Fos was able to rescue the inhibitory effect of estrogen on Smad3 transcriptional activity. Based on these results, a model is proposed whereby c-Jun is limiting in its ability to act as a Smad3 co-activator in the presence of E(2)-bound ER, possibly due to ER sequestering c-Jun away from the Smad3 responsive promoter.
Collapse
Affiliation(s)
- Tracy Cherlet
- Manitoba Institute of Cell Biology, Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB, Canada R3E 0V9
| | | |
Collapse
|
8
|
Shekhar MPV, Santner S, Carolin KA, Tait L. Direct involvement of breast tumor fibroblasts in the modulation of tamoxifen sensitivity. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:1546-60. [PMID: 17456761 PMCID: PMC1854950 DOI: 10.2353/ajpath.2007.061004] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Using contact-dependent three-dimensional coculture systems and serum-free conditions, we compared the ability of estrogen receptor (ER)-alpha(+) tamoxifen-sensitive premalignant (EIII8) or tumorigenic (MCF-7), ER-alpha(+) tamoxifen-resistant (EIII8-TAM(R)) or ER-alpha(-) MDA-MB-231 breast cancer cells to interact and undergo epithelial morphogenesis on association with breast tumor-derived fibroblasts. Although all breast cancer cell lines interacted with tumor fibroblasts, EIII8 and its intrinsically tamoxifen-resistant counterpart EIII8-TAM(R) cells were most receptive and responded with dramatic, albeit, aberrant epithelial morphogenesis. EIII8 cells underwent epithelial morphogenesis when cocultured with fibroblasts from ER-alpha(-)/PgR(-) or ER-alpha(+)/PgR(+) breast tumors; however, EIII8 cells cocultured with ER-alpha(-)/PgR(-) tumor-derived fibroblasts exhibited decreased tamoxifen sensitivity compared with cells cocultured with ER-alpha(+)/PgR(+) tumor fibroblasts. Fibroblast-induced tamoxifen resistance was accompanied by mitogen-activated protein kinase and Akt hyperactivation, reduced sensitivity to U0126 or LY294002, and ER-alpha hyperphosphorylation in the activation function-1 domain. The intrinsic tamoxifen resistance of EIII8-Tam(R) cells correlated with constitutive ER-alpha hyperphosphorylation that was unaffected by the tumor fibroblasts. Our results suggest that tumor fibroblast-induced tamoxifen resistance of EIII8 cells is not mediated by epidermal growth factor receptor or insulin-like growth factor (IGF)-1R axes because no correlation was found between expression levels of IGF-1, IGF-2, phosphorylated IGF-1R, or epidermal growth factor receptor, and tamoxifen sensitivity of EIII8 fibroblast cultures.
Collapse
Affiliation(s)
- Malathy P V Shekhar
- Breast Cancer Program, Karmanos Cancer Institute, Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
9
|
McClay EF, Bogart J, Herndon JE, Watson D, Evans L, Seagren SL, Green MR. A Phase III Trial Evaluating the Combination of Cisplatin, Etoposide, and Radiation Therapy With or Without Tamoxifen in Patients With Limited-Stage Small Cell Lung Cancer. Am J Clin Oncol 2005; 28:81-90. [PMID: 15685040 DOI: 10.1097/01.coc.0000139940.52625.d0] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Based on both clinical and laboratory data that suggested that tamoxifen (TAM) enhanced the effectiveness of cisplatin (DDP)-based chemotherapy regimens, the Cancer and Leukemia Group B (CALGB) designed and initiated a prospective, randomized phase III trial to test the efficacy of the addition of high-dose TAM to a standard chemoradiation regimen of DDP and etoposide (VP-16) in patients with limited-stage small cell lung cancer (LS-SCLC). Between August 6, 1993, and January 15, 1999, 319 patients with LSSCLC were accrued to CALGB 9235. Patients were randomized to receive chemotherapy with or without high-dose TAM. Treatment on the non-TAM containing arm (arm 1) included DDP (80 mg/m2 intravenously day 1 only) and VP-16 (80 mg/m2 intravenously days 1-3) given every 3 weeks for a total of 5 cycles. Patients treated on arm 2 received the identical chemotherapy regimen as described here with the addition of high-dose TAM (80 mg orally twice per day), which was given for 5 days each cycle starting 1 day before the DDP. Thoracic radiation (XRT) given at 200 cGy 5 days per week to a total dose of 50 Gy began on day 1 of cycle 4 of chemotherapy and overlapped with cycle 5. Prophylactic cranial irradiation was offered to all patients who achieved a complete response or near-complete response. A total of 307 patients are evaluable for response. After the completion of the chemoradiation portion of the treatment, the overall response rate (ORR) was 88% for 154 patients treated without tamoxifen and 84% for 153 patients treated with tamoxifen with complete response (CR) rates of 49% and 50%, respectively. The median failure-free survivals of 12.3 months and 10.5 months and the overall survivals of 20.6 months and 18.4 months, respectively, were not statistically significant between the 2 arms. Toxicity was similar with and without tamoxifen. This phase III trial failed to demonstrate a positive effect on either the response or survival for the addition of TAM to standard etoposide-cisplatin-radiation management for patients with LS-SCLC. However, these data continue to support a positive effect of chemoradiation in the treatment of patients with LS-SCLC.
Collapse
Affiliation(s)
- Edward F McClay
- San Diego Melanoma Research Center, Vista, California 92083, USA.
| | | | | | | | | | | | | |
Collapse
|
10
|
Balfe PJ, McCann AH, Welch HM, Kerin MJ. Estrogen receptor beta and breast cancer. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2004; 30:1043-50. [PMID: 15522549 DOI: 10.1016/j.ejso.2004.06.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2004] [Indexed: 11/15/2022]
Abstract
A second estrogen receptor, estrogen receptor-beta, was identified in 1996 and has led to an intensive re-evaluation of the role of estrogens in normal physiological and disease processes. While much has been learnt about this new receptor, there remain many outstanding questions, particularly regarding its prognostic significance and therapeutic implications.
Collapse
Affiliation(s)
- P J Balfe
- Department of Surgery and BreastCheck, Mater Misericordiae University Hospital, Eccles St., Dublin 7, Ireland.
| | | | | | | |
Collapse
|
11
|
Turner S, A Sherratt J, Cameron D. Tamoxifen treatment failure in cancer and the nonlinear dynamics of TGFbeta. J Theor Biol 2004; 229:101-11. [PMID: 15178188 DOI: 10.1016/j.jtbi.2004.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2003] [Revised: 02/20/2004] [Accepted: 03/03/2004] [Indexed: 10/26/2022]
Abstract
The process of cancer invasion involves a complex interplay between cell-cell and cell-medium adhesion, proteolytic enzyme secretion, cell birth and death processes, random and directed motility, and immune response, as well as many other factors. The growth factor TGF beta is known to have a complex effect on this process. It inhibits mitosis and promotes apoptosis in a concentration-dependent manner in vitro, and it is for this reason that its secretion is thought to be helpful in inhibiting tumour growth. However, recent in vitro and in vivo results have shown a significant effect of this growth factor in promoting the sensitivity of malignantly transformed cells to gradients of extracellular matrix proteins--an effect which tends to increase invasiveness. The drug tamoxifen has been demonstrated to be therapeutically effective in the treatment of patients with breast cancer; however, it is known also that many patients become resistant to the effect of this drug after a few years, and the reasons for this remain controversial. In this work we take our established model of cancer invasion (J. Theor. Biol. 216(1) (2002) 85), and extend it to include the effect of TGF beta. In so doing we demonstrate that a tamoxifen-stimulated upregulation of the secretion of TGF beta may give rise to a tumour which has a smaller number of cells but which has a greater invasiveness, greater metastatic potential, and a tumour histology which is known to correlate with a poorer prognosis. These data suggest that tamoxifen-stimulated secretion of TGF beta might explain treatment failure in some patients.
Collapse
Affiliation(s)
- Stephen Turner
- Centre for Theoretical Modelling in Medicine, Department of Mathematics, Heriot-Watt University, Edinburgh EH14 4AS, Scotland, UK. ,
| | | | | |
Collapse
|
12
|
Abstract
Transforming growth factor (TGF) beta is a pre-eminent negative growth regulator that has antiproliferative effects on a range of epithelial cells. This ability has evoked interest in this growth factor as a tumour suppressor with potential clinical significance. In the early stages of breast carcinogenesis, a growth-inhibitory response to TGFbeta is maintained, which depends on an intact TGFbeta signalling pathway. Tumour development and progression of cells along a neoplastic continuum is accompanied by loss of this growth-inhibitory response to TGFbeta, which might instead promote tumour growth indirectly through a combination of permissive effects on stromal tissue, angiogenesis, and the immune system. This review discusses the complexity of functional pleiotropy and the continually changing roles of TGFbeta as a tumour evolves, along with competing therapeutic strategies. The boosting of local endogenous amounts of TGFbeta in conjunction with enhancement of cellular responsiveness might be appropriate in early malignant disease, and anti-TGFbeta approaches could yield a therapeutic gain in metastatic states.
Collapse
Affiliation(s)
- John R Benson
- Cambridge Breast Unit, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
13
|
Klijn J, Berns E, Foekens J. Prognostic and Predictive Factors and Targets for Therapy in Breast Cancer. Breast Cancer 2002. [DOI: 10.1201/b14039-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
14
|
|
15
|
Abstract
Transforming growth factor (TGF) beta1 is a potent growth inhibitor, with tumor-suppressing activity. Cancers are often refractile to this growth inhibition either because of genetic loss of TGF-beta signaling components or, more commonly, because of downstream perturbation of the signaling pathway, such as by Ras activation. Carcinomas often secrete excess TGF-beta1 and respond to it by enhanced invasion and metastasis. Therapeutic approaches should aim to inhibit the TGF-beta-induced invasive phenotype, but also to retain its growth-inhibitory and apoptosis-inducing effects.
Collapse
Affiliation(s)
- R J Akhurst
- Mt Zion Cancer Research Institute, University of California at San Francisco, San Francisco, CA 94143-0875, USA.
| | | |
Collapse
|
16
|
Abstract
Transforming growth factor (TGF) beta1 is a potent growth inhibitor, with tumor-suppressing activity. Cancers are often refractile to this growth inhibition either because of genetic loss of TGF-beta signaling components or, more commonly, because of downstream perturbation of the signaling pathway, such as by Ras activation. Carcinomas often secrete excess TGF-beta1 and respond to it by enhanced invasion and metastasis. Therapeutic approaches should aim to inhibit the TGF-beta-induced invasive phenotype, but also to retain its growth-inhibitory and apoptosis-inducing effects.
Collapse
Affiliation(s)
- R J Akhurst
- Mt Zion Cancer Research Institute, University of California at San Francisco, San Francisco, CA 94143-0875, USA.
| | | |
Collapse
|
17
|
Achuthan R, Bell SM, Roberts P, Leek JP, Horgan K, Markham AF, MacLennan KA, Speirs V. Genetic events during the transformation of a tamoxifen-sensitive human breast cancer cell line into a drug-resistant clone. CANCER GENETICS AND CYTOGENETICS 2001; 130:166-72. [PMID: 11675139 DOI: 10.1016/s0165-4608(01)00475-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Tamoxifen resistance is a serious clinical problem commonly encountered in the management of patients with breast cancer. The mechanisms leading to its development are unclear. Tamoxifen acts via multiple pathways and has diverse effects. Hence transformation from a tamoxifen-sensitive to a resistant phenotype could involve multiple genetic events. Knowledge of the genetic pathways leading to resistance may facilitate the development of novel therapeutic strategies. In this study, a variation of conventional comparative genomic hybridization (CGH) has been employed to detect genetic alterations associated with tamoxifen resistance. MCF-7, a tamoxifen-sensitive human breast cancer cells line, and its tamoxifen-resistant clone, CL-9 were used. Both cell lines showed extensive areas of concordance but consistent differences were seen with the acquisition of tamoxifen resistance. These differences included the amplification of 2p16.3 approximately p23.2, 2q21 approximately q34, 3p12.3 approximately p14.1, 3p22 approximately p26, 3q, 12q13.2 approximately q22, 13q12 approximately q14, 17q21.3 approximately q23, 20q11.2 approximately q13.1 and 21q11.2 approximately q21 as well as the deletion of 6p21.1, 6p23 approximately p25, 7q11.1 approximately q31, 7q35 approximately q36, 11p15, 11q24, 13q33, 17p, 18q12 approximately q21.1, 19p, 19q13.3, 22q13.1 approximately q13.2. These findings were supported by conventional cytogenetics and chromosome painting. The regions identified by CGH potentially harbor genes that could be important in the development of tamoxifen resistance.
Collapse
Affiliation(s)
- R Achuthan
- Molecular Medicine Unit, University of Leeds, St. James's University Hospital, LS9 7TF, Leeds, UK
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Derynck R, Akhurst RJ, Balmain A. TGF-beta signaling in tumor suppression and cancer progression. Nat Genet 2001; 29:117-29. [PMID: 11586292 DOI: 10.1038/ng1001-117] [Citation(s) in RCA: 1740] [Impact Index Per Article: 75.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epithelial and hematopoietic cells have a high turnover and their progenitor cells divide continuously, making them prime targets for genetic and epigenetic changes that lead to cell transformation and tumorigenesis. The consequent changes in cell behavior and responsiveness result not only from genetic alterations such as activation of oncogenes or inactivation of tumor suppressor genes, but also from altered production of, or responsiveness to, stimulatory or inhibitory growth and differentiation factors. Among these, transforming growth factor beta (TGF-beta) and its signaling effectors act as key determinants of carcinoma cell behavior. The autocrine and paracrine effects of TGF-beta on tumor cells and the tumor micro-environment exert both positive and negative influences on cancer development. Accordingly, the TGF-beta signaling pathway has been considered as both a tumor suppressor pathway and a promoter of tumor progression and invasion. Here we evaluate the role of TGF-beta in tumor development and attempt to reconcile the positive and negative effects of TGF-beta in carcinogenesis.
Collapse
Affiliation(s)
- R Derynck
- Department of Growth and Development, University of California at San Francisco, San Francisco, California, USA.
| | | | | |
Collapse
|
19
|
Harpel JG, Schultz-Cherry S, Murphy-Ullrich JE, Rifkin DB. Tamoxifen and estrogen effects on TGF-beta formation: role of thrombospondin-1, alphavbeta3, and integrin-associated protein. Biochem Biophys Res Commun 2001; 284:11-4. [PMID: 11374863 DOI: 10.1006/bbrc.2001.4922] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have found that the enhanced activation of latent TGF-beta by human breast carcinoma cell lines either treated with tamoxifen or deprived of estrogen is dependent upon thrombospondin-1 (TSP-1) since activation was blocked by anti-TSP-1 antibodies or by a TSP antagonist peptide. However, TGF-beta formation upon tamoxifen exposure to estrogen withdrawal is associated with decreased levels of soluble TSP-1. A concomitant increase in the expression of the TSP-1 receptors alphavbeta3 and integrin-associated protein (IAP) occurs under these conditions, and antibodies to TSP-1 or to these receptors inhibit increased TGF-beta formation. Therefore, increased cell surface associated TSP-1 enhances latent TGF-beta activation.
Collapse
MESH Headings
- Adenocarcinoma/metabolism
- Antibodies, Blocking/pharmacology
- Antibodies, Monoclonal/pharmacology
- Antigens, CD/metabolism
- Blotting, Northern
- Breast Neoplasms/metabolism
- CD36 Antigens/metabolism
- CD47 Antigen
- Carrier Proteins/antagonists & inhibitors
- Carrier Proteins/metabolism
- Culture Media, Conditioned/chemistry
- Culture Media, Conditioned/metabolism
- Electrophoresis, Polyacrylamide Gel
- Estrogen Antagonists/pharmacology
- Estrogens/pharmacology
- Female
- Humans
- Intracellular Signaling Peptides and Proteins
- Latent TGF-beta Binding Proteins
- RNA, Messenger/metabolism
- Receptors, Vitronectin/antagonists & inhibitors
- Receptors, Vitronectin/metabolism
- Tamoxifen/pharmacology
- Thrombospondin 1/antagonists & inhibitors
- Thrombospondin 1/metabolism
- Transforming Growth Factor beta/biosynthesis
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- J G Harpel
- Department of Cell Biology, New York University School of Medicine, 550 First Avenue, New York, New York 10016, USA
| | | | | | | |
Collapse
|
20
|
Albright CD, Kaufman DG. Lactoferrin: a tamoxifen-responsive protein in normal and malignant human endometrial cells in culture. Exp Mol Pathol 2001; 70:71-6. [PMID: 11263949 DOI: 10.1006/exmp.2000.2354] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We investigated the possible role of the estrogen-regulated protein lactoferrin (Lf) in the response of isolated normal human endometrial epithelial cells (NHEC) and established human endometrial carcinoma (EC) cell lines to tamoxifen (TAM). Using confocal laser scanning microscopy and a monospecific antibody, Lf was localized to the cytoplasm of normal and EC cells. Antibody neutralization of secreted Lf inhibited, whereas exogenous Lf (0--100 microg/ml) enhanced, cell proliferation in both classes of cells. Treatment of NHEC with TAM inhibited cell growth via a protein kinase-C-mediated pathway, concomitant with a reduction in the staining intensity for Lf. Importantly, in EC cells, TAM greatly enhanced the staining intensity for Lf, but did not affect cell growth. We propose that stable expression of Lf protein by EC cells may impart a survival advantage to these cells, which may, in part, account for the resistance of these cells to tamoxifen.
Collapse
Affiliation(s)
- C D Albright
- Department of Nutrition, School of Public Health, University of North Carolina, Chapel Hill, 27599-7400, USA
| | | |
Collapse
|
21
|
Nicholson RI, Gee JM. Oestrogen and growth factor cross-talk and endocrine insensitivity and acquired resistance in breast cancer. Br J Cancer 2000; 82:501-13. [PMID: 10682656 PMCID: PMC2363333 DOI: 10.1054/bjoc.1999.0954] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Affiliation(s)
- R I Nicholson
- Tenovus Cancer Research Centre, University of Wales College of Medicine, Cardiff, UK
| | | |
Collapse
|
22
|
Hilsenbeck SG, Friedrichs WE, Schiff R, O'Connell P, Hansen RK, Osborne CK, Fuqua SA. Statistical analysis of array expression data as applied to the problem of tamoxifen resistance. J Natl Cancer Inst 1999; 91:453-9. [PMID: 10070945 DOI: 10.1093/jnci/91.5.453] [Citation(s) in RCA: 135] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Although the emerging complementary DNA (cDNA) array technology holds great promise to discern complex patterns of gene expression, its novelty means that there are no well-established standards to guide analysis and interpretation of the data that it produces. We have used preliminary data generated with the CLONTECH Atlas human cDNA array to develop a practical approach to the statistical analysis of these data by studying changes in gene expression during the development of acquired tamoxifen resistance in breast cancer. METHODS For hybridization to the array, we prepared RNA from MCF-7 human breast cell tumors, isolated from our athymic nude mouse xenograft model of acquired tamoxifen resistance during estrogen-stimulated, tamoxifen-sensitive, and tamoxifen-resistant growth. Principal components analysis was used to identify genes with altered expression. RESULTS AND CONCLUSIONS Principal components analysis yielded three principal components that are interpreted as 1) the average level of gene expression, 2) the difference between estrogen-stimulated gene expression and the average of tamoxifen-sensitive and tamoxifen-resistant gene expression, and 3) the difference between tamoxifen-sensitive and tamoxifen-resistant gene expression. A bivariate (second and third principal components) 99% prediction region was used to identify outlier genes that exhibit altered expression. Two representative outlier genes, erk-2 and HSF-1 (heat shock transcription factor-1), were chosen for confirmatory study, and their predicted relative expression levels were confirmed in western blot analysis, suggesting that semiquantitative estimates are possible with array technology. IMPLICATIONS Principal components analysis provides a useful and practical method to analyze gene expression data from a cDNA array. The method can identify broad patterns of expression alteration and, based on a small simulation study, will likely provide reasonable power to detect moderate-sized alterations in clinically relevant genes.
Collapse
Affiliation(s)
- S G Hilsenbeck
- Department of Medicine, The University of Texas Health Science Center, San Antonio 78248-7884, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Brandt R, Ebert AD. Growth inhibitors for mammary epithelial cells. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 1999; 20:197-248. [PMID: 9928532 DOI: 10.1007/978-3-642-72149-6_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- R Brandt
- Novartis Pharma Inc., Basel, Switzerland
| | | |
Collapse
|
24
|
Arteaga CL, Koli KM, Dugger TC, Clarke R. Reversal of tamoxifen resistance of human breast carcinomas in vivo by neutralizing antibodies to transforming growth factor-beta. J Natl Cancer Inst 1999; 91:46-53. [PMID: 9890169 DOI: 10.1093/jnci/91.1.46] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Overexpression of transforming growth factor (TGF)-beta has been reported in human breast carcinomas resistant to antiestrogen tamoxifen, but the role of TGF-beta in this resistant phenotype is unclear. We investigated whether inhibition of TGF-beta2, which is overexpressed in LCC2 tamoxifen-resistant human breast cancer cells, could modify antiestrogen resistance. METHODS TGF-beta2 expression was evaluated in LCC2 cells and tamoxifen-sensitive LCC1 cells by northern blot analysis. Secreted TGF-beta activity was quantified by use of an 125I-TGF-beta competitive radioreceptor assay. Sensitivity to tamoxifen was measured in a soft agarose colony-forming assay and in a xenograft model in nude and beige/nude mice. Natural killer (NK) cell cytotoxicity was measured by 51Cr release from LCC1 and LCC2 cell targets coincubated with human peripheral blood mononuclear cells. Decrease in TGF-beta2 expression in LCC2 cells was achieved by treatment with antisense oligodeoxynucleotides and confirmed by TGF-beta2 immunoblot analysis. RESULTS AND CONCLUSIONS The proliferative response of LCC2 cells to tamoxifen in vitro was not altered by TGF-beta neutralizing antibodies. However, established LCC2 tumors in nude mice treated with tamoxifen plus TGF-beta antibodies failed to grow, whereas tumors treated with tamoxifen plus a control antibody continued to proliferate. This reversal of tamoxifen resistance by TGF-beta antibodies did not occur in beige/nude mice, which lack NK-cell function, suggesting that immune mechanisms may be involved in the antitumor effects of tamoxifen. Antisense TGF-beta2 oligodeoxynucleotides enhanced the NK sensitivity of LCC2 cells in the presence of tamoxifen. Finally, LCC1 tumors were markedly more sensitive to tamoxifen in NK-active than in NK-deficient mice. IMPLICATIONS These data suggest that host NK function mediates, in part, the antitumor effect of tamoxifen and that TGF-beta2 may abrogate this mechanism, thus contributing to tamoxifen resistance.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antineoplastic Agents, Hormonal/pharmacology
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Cell Division/drug effects
- Cytotoxicity, Immunologic
- Drug Resistance, Neoplasm
- Estrogen Antagonists/pharmacology
- Estrogens
- Female
- Humans
- Killer Cells, Natural/immunology
- Mice
- Mice, Nude
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/immunology
- Neoplasm Proteins/physiology
- Neoplasm Transplantation
- Neoplasms, Hormone-Dependent/drug therapy
- Neoplasms, Hormone-Dependent/pathology
- Oligonucleotides, Antisense/pharmacology
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Neoplasm/biosynthesis
- RNA, Neoplasm/genetics
- Tamoxifen/pharmacology
- Transforming Growth Factor beta/antagonists & inhibitors
- Transforming Growth Factor beta/biosynthesis
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/immunology
- Transforming Growth Factor beta/physiology
- Tumor Cells, Cultured
- Tumor Stem Cell Assay
Collapse
Affiliation(s)
- C L Arteaga
- Department of Medicine, Vanderbilt University School of Medicine, Vanderbilt Cancer Center, Department of Veterans Affairs Medical Center, Nashville, TN 37232-5536, USA.
| | | | | | | |
Collapse
|
25
|
Transforming Growth Factor-β and Breast Cancer. Breast Cancer 1999. [DOI: 10.1007/978-1-59259-456-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
26
|
Ohmori T, Yang JL, Price JO, Arteaga CL. Blockade of tumor cell transforming growth factor-betas enhances cell cycle progression and sensitizes human breast carcinoma cells to cytotoxic chemotherapy. Exp Cell Res 1998; 245:350-9. [PMID: 9851876 DOI: 10.1006/excr.1998.4261] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have examined the effect of neutralizing TGF-beta antibodies on cisplatin-mediated cytotoxicity against MDA-231 human breast tumor cell spheroids. These tridimensional in vitro systems have been shown to recapitulate the drug sensitivity pattern of tumor cells in vivo. MDA-231 tumor cell spheroids exhibit higher protein levels of the cyclin-dependent kinase (Cdk) inhibitors p21 and p27 and >10-fold lower Cdk2 activity compared to adherent cell monolayers, as well as pRb hypophosphorylation, a predominant G1 population, and a cisplatin 1-h IC50 of approximately 100 microM. Treatment of MDA-231 cells in monolayer with cisplatin for 1 h, subsequently grown as spheroids, increased steady-state TGF-beta1 mRNA levels, secretion of active TGF-beta, cellular Cdk2 activity, pRb phosphorylation, and p21 protein levels, while downregulating p27. Accumulation of cells in G2M and progression into S were noted 48 h after treatment with 100 microM cisplatin. We tested whether drug-induced upregulation of TGF-beta1 and p21, perhaps by preventing cell cycle progression, were protective mechanisms against drug-mediated toxicity by using neutralizing anti-TGF-beta antibodies. Anti-TGF-beta antibodies diminished the induction of p21, enhanced the activation of Cdk2, and facilitated progression into S and G2M following cisplatin treatment. This resulted in a >twofold enhancement of drug-induced DNA fragmentation and a shift in the cisplatin 1-h IC50 from 100 to <10 microM. These data suggest that tumor cell TGF-beta1 may protect from DNA damage and that postchemotherapy administration of TGF-beta inhibitors may facilitate progression beyond G1/S, potentially increasing the efficacy of cytotoxic chemotherapy.
Collapse
Affiliation(s)
- T Ohmori
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, 37232-5536, USA
| | | | | | | |
Collapse
|
27
|
McNamara DA, Harmey JH, Walsh TN, Redmond HP, Bouchier-Hayes DJ. Significance of angiogenesis in cancer therapy. Br J Surg 1998; 85:1044-55. [PMID: 9717994 DOI: 10.1046/j.1365-2168.1998.00816.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND For most solid tumours, surgery remains the most effective primary treatment. Despite apparently curative resection, significant numbers of patients develop secondary disease due to growth of undetected micrometastases. The ability of a tumour to metastasize is related to the degree of angiogenesis it induces. In addition, micrometastases rely on new vessel formation to provide the nutrients necessary for growth. A better understanding of how tumours acquire their blood supply may lead to more effective adjuvant therapies and improve survival following surgery. METHODS A systematic review of the literature on angiogenesis between 1971 and 1997 was performed using the Medline database to ascertain current thinking on angiogenesis and its relevance in oncological surgery. RESULTS Angiogenesis is a physiological process subject to autocrine and paracrine regulation which has the potential to become abnormal and play a part in a number of pathological states, including cancer. Increased angiogenic stimuli in the perioperative period, associated with concomitant reduction in tumour-derived antiangiogenic factors following resection of a primary tumour, result in a permissive environment which allows micrometastases to grow. CONCLUSION Recognition of the role of angiogenesis in metastatic tumour growth represents a significant development in our understanding of tumour biology. The development of antiangiogenic agents offers new promise in the treatment of malignancy. Such agents may prevent or control the development and growth of primary and metastatic tumours.
Collapse
Affiliation(s)
- D A McNamara
- Royal College of Surgeons in Ireland, Department of Surgery, Beaumont Hospital, Dublin
| | | | | | | | | |
Collapse
|
28
|
Larsen SS, Madsen MW, Jensen BL, Lykkesfeldt AE. Resistance of human breast-cancer cells to the pure steroidal anti-estrogen ICI 182,780 is not associated with a general loss of estrogen-receptor expression or lack of estrogen responsiveness. Int J Cancer 1997; 72:1129-36. [PMID: 9378550 DOI: 10.1002/(sici)1097-0215(19970917)72:6<1129::aid-ijc31>3.0.co;2-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To elucidate the mechanisms responsible for the development of anti-estrogen resistance, we have cloned and established 3 stable ICI-182,780-resistant sub-lines, MCF-7/182R-1, MCF-7/182R-6 and MCF-7/182R-7 from the estrogen-receptor(ER)-positive and estrogen-responsive human breast-cancer MCF-7 cell line by long-term treatment with 10(-7) M ICI 182,780. The ICI-182,780-resistant MCF-7 sub-lines express ER, but compared with MCF-7 cells the level is significantly lower in all 3 sub-lines. In the MCF-7 cell line we find that ER expression is regulated by estrogen and anti-estrogens at the transcriptional and post-transcriptional level. This is in contrast to the ICI-182,780-resistant sub-lines, in which we find very little hormonal effects on the ER mRNA expression level. The resistant sub-lines also deviate from parent characteristics by the complete lack of expression of progesterone receptor even when grown in the presence of estradiol. All 3 resistant sub-lines have a lower basal expression of cathepsin-D mRNA comparable with the lower ER expression, but, in contrast, they have higher basal expression of the pS2 mRNA than the parent MCF-7 cell line. Although there are different basal expression levels of the pS2 and cathepsin-D genes, the resistant sub-lines behave like the parent MCF-7 cell line with respect to the hormonal regulation of both genes. The estrogen receptors in the resistant sub-lines have also maintained wild-type characteristics with respect to estrogen and anti-estrogen regulation of the estrogen-regulated proteins procathepsin D, alpha1-antitrypsin and a 42-kDa protein. The resistant cells require estrogen for growth in athymic nude mice. Our results clearly demonstrate that the ER in the resistant sub-lines have a normal function for most parameters investigated, supporting our earlier observation that only wild-type ER protein is expressed in these cells. The few observed differences in ER function between the parent MCF-7 cell line and the resistant sub-lines are not likely to be responsible for the ICI-182,780-resistant phenotype.
Collapse
Affiliation(s)
- S S Larsen
- Department of Tumor Endocrinology, Danish Cancer Society, Copenhagen O.
| | | | | | | |
Collapse
|
29
|
Reiss M, Barcellos-Hoff MH. Transforming growth factor-beta in breast cancer: a working hypothesis. Breast Cancer Res Treat 1997; 45:81-95. [PMID: 9285120 DOI: 10.1023/a:1005865812918] [Citation(s) in RCA: 118] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Transforming Growth Factor-beta (TGF beta) is the most potent known inhibitor of the progression of normal mammary epithelial cells through the cell cycle. During the early stages of breast cancer development, the transformed epithelial cells appear to still be sensitive to TGF beta-mediated growth arrest, and TGF beta can act as an anti-tumor promoter. In contrast, advanced breast cancers are mostly refractory to TGF beta-mediated growth inhibition and produce large amounts of TGF beta, which may enhance tumor cell invasion and metastasis by its effects on extracellular matrix. We postulate that this seemingly paradoxical switch in the responsiveness of tumor cells to TGF beta during progression is the consequence of the activation of the latent TGF beta that is produced and deposited into the tumor microenvironment, thereby driving the clonal expansion of TGF beta-resistant tumor cells. While tumor cells themselves may activate TGF beta, recent observations suggest that environmental tumor promoters or carcinogens, such as ionizing radiation, can cause stromal fibroblasts to activate TGF beta by epigenetic mechanisms. As the biological effects of the anti-estrogen tamoxifen may well be mediated by TGF beta, this model has a number of important implications for the clinical uses of tamoxifen in the prevention and treatment of breast cancer. In addition, it suggests a number of novel approaches to the treatment of advanced breast cancer.
Collapse
Affiliation(s)
- M Reiss
- Department of Medicine (Medical Oncology), Yale University School of Medicine, New Haven, CT 06520-8032, USA.
| | | |
Collapse
|
30
|
Koli KM, Ramsey TT, Ko Y, Dugger TC, Brattain MG, Arteaga CL. Blockade of transforming growth factor-beta signaling does not abrogate antiestrogen-induced growth inhibition of human breast carcinoma cells. J Biol Chem 1997; 272:8296-302. [PMID: 9079651 DOI: 10.1074/jbc.272.13.8296] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
We have studied the role of autocrine transforming growth factor-beta (TGF-beta) signaling on antiestrogen-mediated growth inhibition of hormone-dependent T47D and MCF-7 human breast carcinoma cells. Tamoxifen treatment increased the secretion of TGF-beta activity into serum-free cell medium and the cellular content of affinity cross-linked type I and III TGF-beta receptors in both cell lines. Anti-pan-TGF-beta antibodies did not block anti-estrogen-induced recruitment in G1 and inhibition of anchorage-dependent and -independent growth of both cell lines. Early passage MCF-7 cells, which exhibit detectable type II TGF-beta receptors at the cell surface and exquisite sensitivity to exogenous TGF-beta1, were transfected with a tetracycline-controllable dominant-negative TGF-betaRII (DeltaRII) construct. Although the TGF-beta1 response was blocked by removal of tetracycline in MCF-7/DeltaRII cells, tamoxifen-mediated suppression of Rb phosphorylation, recruitment in G1, and inhibition of cell proliferation were identical in the presence and absence of tetracycline. TGF-beta1 treatment up-regulated the Cdk inhibitor p21 and induced its association with Cdk2 in MCF-7 cells; these responses were blocked by the DeltaRII transgene product. In MCF-7 cells with a functional TGF-beta signaling pathway, tamoxifen did not up-regulate p21 nor did it induce association of p21 with Cdk2, suggesting alternative mechanisms for antiestrogen-mediated cytostasis. Finally, transfection of late-passage, TGF-beta1 unresponsive MCF-7 cells with high levels of TGF-betaRII restored TGF-beta1-induced growth inhibition but did not enhance tamoxifen response in culture. Taken together these data strongly argue against any role for TGF-beta signaling on tamoxifen-mediated growth inhibition of hormone-dependent breast cancer cells.
Collapse
Affiliation(s)
- K M Koli
- Department of Medicine,Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
The nonsteroidal antiestrogen tamoxifen is the most widely used anticancer drug. In women with breast cancer, adjuvant therapy with tamoxifen reduces relapse and improves overall survival. In advanced breast cancer, the response rate is more than 50% in hormonal dependent disease. In women treated with adjuvant tamoxifen the incidence of new primary breast cancers is decreased. This latter observation has led to the initiation of prevention trials. In 1989 the first report from a large prospective randomised trial showed a significant increase of endometrial carcinoma among women treated with adjuvant tamoxifen. This effect may be linked to the somewhat paradoxical estrogenic properties of tamoxifen. The endometrial effects should be considered in the long term use of tamoxifen, and should also be taken into account in the evaluation of the prevention trials. Animal data indicate that tamoxifen can induce tumours in other organ systems, for example the liver, but no increase in primary liver cancer has been reported from the randomised trials. In some of these trials an increase in other gastrointestinal cancers (e.g. colon and gastric carcinoma) has been observed. The mechanism behind this may be different from that of the endometrium. In animal systems, tamoxifen has shown to induce DNA damage, with formation of DNA adducts. The risk of secondary gastrointestinal cancer needs to be further evaluated. The adverse effects of tamoxifen have led to the development of new anti-estrogenic drugs and other estrogen reducing agents (e.g. aromatase inhibitors).
Collapse
Affiliation(s)
- N Wilking
- Department of Oncology, Radiumhemmet, Karolinska Hospital, Stockholm, Sweden.
| | | | | |
Collapse
|
32
|
Amoils KD, Bezwoda WR. TGF-beta 1 mRNA expression in clinical breast cancer and its relationship to ER mRNA expression. Breast Cancer Res Treat 1997; 42:95-101. [PMID: 9138609 DOI: 10.1023/a:1005785421815] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Eighty nine primary breast cancers were investigated for the expression of TGF-beta 1 and ER mRNA using PCR of reverse transcribed RNA. PCR products were validated using Southern blots and hybridization with radiolabelled cDNA probes. TGF beta 1 mRNA was found to be expressed in 56/89 (63%) of the breast cancers while ER mRNA was expressed in 23/89 (26%) of the tumours. Using chi-square analysis TGF-beta mRNA expression was found to correlate significantly with ER mRNA expression (p < 0.001), in that virtually all tumours that expressed ER mRNA co-expressed TGF beta 1. In tumours that were ER mRNA negative, TGF beta 1 expression was more variable. These results suggest that during tumour progression, ER expression is lost more frequently than is growth factor expression.
Collapse
Affiliation(s)
- K D Amoils
- Department of Medicine, University of the Witwatersrand, Johannesburg, Republic of South Africa
| | | |
Collapse
|
33
|
Benson JR, Baum M, Colletta AA. Role of TGF beta in the anti-estrogen response/resistance of human breast cancer. J Mammary Gland Biol Neoplasia 1996; 1:381-9. [PMID: 10887512 DOI: 10.1007/bf02017394] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Transforming growth factor beta (TGF beta) has potent inhibitory effects upon epithelial proliferation and malignant progression may be associated with breakdown of the autocrine and paracrine inhibitory loops in which TGF beta participates. The therapeutic effecs of anti-estrogens may be partially attributable to boosting of local endogenous levels of TGF beta. This article reviews the evidence in support of TGF beta being a proximate effector in mediation of the anti-neoplastic effects of anti-estrogens. Both the conventional estrogen receptor (ER) dependent and ER independent mechanisms of action are likely to be involved. Evidence for preferential stromal induction of TGF beta by anti-estrogens is emphasized, together with the therapeutic potential of this strategy for improving outcome in early breast cancer irrespective of ER status.
Collapse
Affiliation(s)
- J R Benson
- Hartwell Laboratory, Institute of Cancer Research, United Kingdom
| | | | | |
Collapse
|
34
|
Abstract
This review considers the various roles of the TGF-beta system in mammary carcinogenesis, tumor progression, and cellular responses to therapeutic measures. The paradigm that has evolved from the work of many investigators suggests that loss of tumor cell responsiveness to the effects of TGF-beta can result in a crucial shift in the net effect of TGF-beta within the context of the tumor-host interaction. Principal elements of host-tumor interactions in which this shift may play out, including immune suppression, angiogenesis, and modification of the surrounding extracellular matrix by tumor cells, are potentially amenable to manipulation. Additional effects of TGF-beta, such as suggested by reports of its ability to alter the drug resistance of tumor cells and the drug sensitivity of normal tissues, suggest that appropriate molecular intervention designed to affect the TGF-beta system might constitute an effective therapeutic strategy.
Collapse
Affiliation(s)
- B A Arrick
- Department of Medicine, Dartmouth Medical School, Hanover, New Hampshire, USA.
| |
Collapse
|
35
|
Koli KM, Arteaga CL. Complex role of tumor cell transforming growth factor (TGF)-beta s on breast carcinoma progression. J Mammary Gland Biol Neoplasia 1996; 1:373-80. [PMID: 10887511 DOI: 10.1007/bf02017393] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Growth inhibition by the TGF-beta s has been extensively studied in both normal and transformed mammary epithelial cells. It has been proposed that loss of autocrine TGF-beta mediated growth regulation is a critical event in breast tumorigenesis and several lines of in vitro and in vivo data support this hypothesis. However, a positive association between the expression of TGF-beta s by tumor cells and the progression or maintenance of breast cancinoma cells has been observed in many studies in in vivo tumor models. Possible mechanisms for these growth enhancing effects of TGF-beta include immunosuppression mediated by tumor TGF-beta s, enhanced angiogenesis, increased peritumoral stroma formation, and cell adhesion. The net effect of tumor cell TGF-beta on the biology of breast carcinogenesis would depend on the balance between autocrine growth inhibition of mammary epithelial cells and these growth enhancing effects.
Collapse
Affiliation(s)
- K M Koli
- Department of Medicine, Vanderbilt University School of Medicine, Vanderbilt Cancer Center, Nashville, Tennessee, USA
| | | |
Collapse
|
36
|
Herman ME, Katzenellenbogen BS. Response-specific antiestrogen resistance in a newly characterized MCF-7 human breast cancer cell line resulting from long-term exposure to trans-hydroxytamoxifen. J Steroid Biochem Mol Biol 1996; 59:121-34. [PMID: 9010327 DOI: 10.1016/s0960-0760(96)00114-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
To understand better the antiestrogen-resistant phenotype that frequently develops in breast cancer patients receiving tamoxifen, we cultured MCF-7 breast cancer cells long-term (>1 yr) in the presence of the antiestrogen trans-hydroxytamoxifen (TOT) to generate a subline refractory to the growth-suppressive effects of TOT. This subline (designated MCF/TOT) showed growth stimulation, rather than inhibition, with TOT and diminished growth stimulation with estradiol (E2), yet remained as sensitive as the parental cells to growth suppression by another antiestrogen, ICI 164,384. Estrogen receptor (ER) levels were maintained at 40% of that in parent MCF-7 cells, but MCF/TOT cells failed to show an increase in progesterone receptor content in response to E2 or TOT treatment. In contrast, the MCF/TOT subline behaved like parental cells in terms of E2 and TOT regulation of ER and pS2 expression and transactivation of a transiently transfected estrogen-responsive gene construct. DNA sequencing of the hormone binding domain of the ER from both MCF-7 and MCF/TOT cells confirmed the presence of wild-type ER and exon 5 and exon 7 deletion splice variants, but showed no point mutations. Compared to the parental cells, the MCF/TOT subline showed reduced sensitivity to the growth-suppressive effects of retinoic acid and complete resistance to exogenous TGF-beta1. The altered growth responsiveness of MCF/TOT cells to TOT and TGF-beta1 was partly to fully reversible following TOT withdrawal for 16 weeks. Our findings underscore the fact that antiestrogen resistance is response-specific; that loss of growth suppression by TOT appears to be due to the acquisition of weak growth stimulation; and that resistance to TOT does not mean global resistance to other more pure antiestrogens such as ICI 164,384, implying that these antiestrogens must act by somewhat different mechanisms. The association of reduced retinoic acid responsiveness and insensitivity to exogenous TGF-beta with antiestrogen growth resistance in these cells supports the increasing evidence for interrelationships among cell regulatory pathways utilized by these three growth-suppressive agents in breast cancer cells. In addition, our findings indicate that one mechanism of antiestrogen resistance, as seen in MCF/TOT cells, may involve alterations in growth factor and other hormonal pathways that affect the ER response pathway.
Collapse
MESH Headings
- Antineoplastic Agents/toxicity
- Breast Neoplasms
- Cell Division/drug effects
- Cell Line
- Chloramphenicol O-Acetyltransferase/biosynthesis
- Clone Cells
- Drug Resistance, Neoplasm
- Estradiol/pharmacology
- Estrogen Antagonists/toxicity
- Female
- Gene Expression
- Genes, Reporter
- Humans
- Polymerase Chain Reaction
- RNA, Messenger/analysis
- RNA, Messenger/biosynthesis
- Receptors, Estrogen/biosynthesis
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Receptors, Transforming Growth Factor beta/metabolism
- Recombinant Proteins/biosynthesis
- Tamoxifen/analogs & derivatives
- Tamoxifen/toxicity
- Transcription, Genetic/drug effects
- Transcriptional Activation
- Transfection
- Transforming Growth Factor beta/biosynthesis
- Transforming Growth Factor beta/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- M E Herman
- Department of Molecular and Integrative Physiology, University of Illinois, Urbana 61801-3704, USA
| | | |
Collapse
|
37
|
Oft M, Peli J, Rudaz C, Schwarz H, Beug H, Reichmann E. TGF-beta1 and Ha-Ras collaborate in modulating the phenotypic plasticity and invasiveness of epithelial tumor cells. Genes Dev 1996; 10:2462-77. [PMID: 8843198 DOI: 10.1101/gad.10.19.2462] [Citation(s) in RCA: 520] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Metastasis of epithelial tumor cells can be associated with the acquisition of fibroblastoid features and the ability to invade stroma and blood vessels. Using matched in vivo and in vitro culture systems employing fully polarized, mammary epithelial cells, we report here that TGF-beta1 brings about these changes in Ras-transformed cells but not in normal cells. When grown in collagen gels in the absence of TGF-beta, both normal and Ras-transformed mammary epithelial cells form organ-like structures in which the cells maintain their epithelial characteristics. Under these conditions, treatment of normal cells with TGF-beta results in growth arrest. The same treatment renders Ras-transformed epithelial cells fibroblastoid, invasive, and resistant to growth inhibition by TGF-beta. After this epithelial-fibroblastoid conversion, the Ras-transformed cells start to secrete TGF-beta themselves, leading to autocrine maintenance of the invasive phenotype and recruitment of additional cells to become fibroblastoid and invasive. More important, this cooperation of activated Ha-Ras with TGF-beta1 is operative during in vivo tumorigenesis and, as in wound healing processes, is dependent on epithelial-stromal interactions.
Collapse
MESH Headings
- Animals
- Cell Line, Transformed
- Cell Polarity
- Cell Transformation, Neoplastic
- Chick Embryo
- Collagen
- Epithelial Cells
- Fibroblasts/pathology
- Gels
- Genes, ras
- Growth Substances/pharmacology
- Heart
- Mammary Glands, Animal/cytology
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Invasiveness
- Neoplasms, Glandular and Epithelial/pathology
- Oncogene Protein p21(ras)/physiology
- RNA, Messenger/analysis
- Receptors, Transforming Growth Factor beta/analysis
- Receptors, Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/analysis
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/pharmacology
- Transforming Growth Factor beta/physiology
- Tumor Cells, Cultured
- Up-Regulation
Collapse
Affiliation(s)
- M Oft
- Forschungsinstitut für Molekulare Pathologie, Wien, Austria
| | | | | | | | | | | |
Collapse
|
38
|
Benson JR, Baum M. Modulation of transforming growth factor beta expression and induction of apoptosis by tamoxifen in ER positive and ER negative breast cancer cells. Br J Cancer 1996; 74:993-4. [PMID: 8826875 PMCID: PMC2074741 DOI: 10.1038/bjc.1996.475] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
39
|
MacCallum J, Keen JC, Bartlett JM, Thompson AM, Dixon JM, Miller WR. Changes in expression of transforming growth factor beta mRNA isoforms in patients undergoing tamoxifen therapy. Br J Cancer 1996; 74:474-8. [PMID: 8695368 PMCID: PMC2074656 DOI: 10.1038/bjc.1996.385] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Tumour was obtained from 37 patients with oestrogen receptor-positive breast cancer, before and during treatment with tamoxifen, and examined qualitatively and semi-qualitatively for mRNA of the three mammalian TGF-beta isoforms. Levels of TGF-beta isoforms were then correlated with tumour response to tamoxifen, as assessed by monthly ultrasound. A high incidence of expression by each isoform was found in tumour material taken both before and during treatment. Semiquantitative assessment of mRNA showed that in the majority of tumours, expression of TGF-beta s did not change markedly with treatment, i.e. beyond that which might have been caused by method reproducibility and tumour heterogeneity (variations of < 100% between pre- and post-treatment samples). In those displaying significant variation with treatment, expression of TGF-beta 1 and -beta 3 increased or decreased in equal numbers, whereas TGF-beta 2 expression tended to increase with treatment. Subdividing tumours by clinical response revealed no significant association between changes in expression of TGF-beta 1 and TGF-beta 3. There was, however, a significant correlation between changes in expression of TGF-beta 2 and response (P = 0.018). Thus, of 15 responding tumours displaying substantial changes, 11 showed an increase in TGF-beta 2 expression with treatment, whereas none of the non-responding tumours were associated with increased expression. While not providing evidence for a generalised increase in TGF-beta expression with tamoxifen treatment, the present study suggests that response to tamoxifen therapy may be associated with an increase in expression of specific TGF-beta isoforms in some, but not all, tumours.
Collapse
Affiliation(s)
- J MacCallum
- University Department of Surgery, Edinburgh Royal Infirmary, UK
| | | | | | | | | | | |
Collapse
|
40
|
Arteaga CL, Dugger TC, Hurd SD. The multifunctional role of transforming growth factor (TGF)-beta s on mammary epithelial cell biology. Breast Cancer Res Treat 1996; 38:49-56. [PMID: 8825122 DOI: 10.1007/bf01803783] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The transforming growth factor-beta s are potent growth inhibitors of normal and transformed breast epithelial cells in culture. In vivo, these peptides modulate the development of the mouse mammary gland. Tissue-specific overexpression of mature TGF-beta 1 in transgenic mice results in mammary gland atrophy and prevention of carcinogen-induced breast tumorigenesis. However, the inhibitory effect of endogenous or exogenous TGF-beta s on established tumor cells is less clear. Several published circumstantial and more direct data argue that, in some cases, the tumor cell TGF-beta s may contribute to the maintenance and/or progression of tumor cells in an intact host by modulating their interaction with host factors. This differential role of the TGF-beta s on mammary cells as determined by their normal or transformed phenotype as well as the biological and clinical implications of these data are discussed.
Collapse
Affiliation(s)
- C L Arteaga
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | |
Collapse
|
41
|
Nørgaard P, Damstrup L, Rygaard K, Spang-Thomsen M, Poulsen HS. Acquired TGF beta 1 sensitivity and TGF beta 1 expression in cell lines established from a single small cell lung cancer patient during clinical progression. Lung Cancer 1996; 14:63-73. [PMID: 8696721 DOI: 10.1016/0169-5002(95)00512-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Three small cell lung cancer cell lines established from a single patient during longitudinal follow-up were examined for in vitro expression of TGF beta and TGF beta receptors, i.e. the components of an autocrine loop. GLC 14 was established prior to treatment, GLC 16 on relapse after chemotherapy and GLC 19 on recurrence after radiotherapy. TGF beta was detected by ELISA and TGF beta receptors by chemical crosslinking to radiolabelled TGF beta 1. Furthermore, TGF beta and TGF beta receptor mRNAs were detected by northern blot analysis. Expression of type II TGF beta receptor mRNA and protein was found in GLC 16 and GLC 19. These cell lines were also growth inhibited by exogenously administrated TGF beta 1. TGF beta 1 mRNA and protein in its latent form was only expressed in the radiotherapy-resistant cell line, GLC 19. The results indicate that disease progression in this patient was paralleled by a gain in sensitivity to the growth inhibition by TGF beta 1 due to type II TGF beta receptor, and a gain of latent TGF beta 1 protein. Lack of type II receptor expression in GLC 14, which was also resistant to growth inhibition by exogenous TGF beta 1, was not due to gross structural changes in the type II receptor gene, as examined by Southern blotting. Also, the type I receptor could not be detected by ligand binding assay in this cell line, despite expression of mRNA for this receptor. This agrees with previous findings that type I receptor cannot bind TGF beta 1 without co-expression of the type II receptor.
Collapse
MESH Headings
- Blotting, Northern
- Blotting, Southern
- Carcinoma, Small Cell/genetics
- Carcinoma, Small Cell/metabolism
- Carcinoma, Small Cell/pathology
- Cell Division
- DNA, Neoplasm/analysis
- Disease Progression
- Enzyme-Linked Immunosorbent Assay
- Female
- Gene Expression
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Middle Aged
- RNA, Messenger/metabolism
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- P Nørgaard
- Section for Radiation Biology, Finsen Center, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
42
|
Abstract
Retroperitoneal fibrosis, a disease that can occur at any age, is characterized by a periaortic fibrous mass that often surrounds the ureters, leading to ureteral obstruction. Patients who present with this disease may complain of flank pain and acute renal failure. There is a high correlation with atherosclerotic disease of the aorta, although the pathogenesis of this disease remains unknown. Although recommendations for therapy remain empiric, prednisone seems to be efficacious in treatment; tamoxifen also may be effective. Retroperitoneal fibrosis usually does not lead to long-term morbidity or affect survival.
Collapse
Affiliation(s)
- G S Gilkeson
- Duke University Medical Center, Durham, North Carolina, USA
| | | |
Collapse
|
43
|
Koli K, Keski-Oja J. Transforming growth factor-beta system and its regulation by members of the steroid-thyroid hormone superfamily. Adv Cancer Res 1996; 70:63-94. [PMID: 8902054 DOI: 10.1016/s0065-230x(08)60872-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
TGF-beta s and their receptors are expressed ubiquitously, and they act as key regulators of many aspects of cell growth, differentiation, and function. Steroid action on target tissues is often associated with increase in TGF-beta isoforms. Regulation of TGF-beta expression and activation is crucial for normal development and growth control. The loss of responsiveness of different tumor cells to the antiproliferative effects of TGF-beta is a common feature in carcinogenesis. Multiple changes are required for the cells to gain complete resistance to TGF-beta growth inhibition (Fynan and Reiss, 1993; Kimchi et al., 1988; Samuel et al., 1992). Although many tumor cells are not growth inhibited by TGF-beta, they respond to TGF-beta treatment by changes in the expression of matrix components and enhanced proteolytic activity (KeskiOja et al., 1988). Agents that induce TGF-beta production in target tissues can have a chemopreventive or chemotherapeutic value for the management of epithelial malignancies. Conversely, data supporting a positive role for TGF-beta in established tumor progression are beginning to emerge (Arteaga et al., 1993a,b; Barrett-Lee et al., 1990; Arrick et al., 1992 ; E. A. Thompson et al., 1991). In later stages of tumor development, cell proliferation is often not inhibited by TGF-beta, and tumor cells secrete large amounts of this growth factor (Fynan and Reiss, 1993). In vivo TGF-beta secreted by tumor or stromal cells can influence host responses such as a natural killer cell function and thus indirctly support tumor cell viability (Arteaga et al., 1993b). TGF-beta may also affect tumor growth indirectly by stromal effects and promotion of angiogenesis. TGF-beta may also be involved in the progression of breast tumors from the steroid-sensitive to steroid-insensitive state (King et al., 1989). Understanding of the net effect of TGF-beta in different stages of tumor development is critical for the evaluation of its therapeutic value in cancer treatment.
Collapse
Affiliation(s)
- K Koli
- Department of Virology, Haartman Institute, Helsinki, Finland
| | | |
Collapse
|
44
|
Abstract
Endocrine treatment plays an important role in the therapy of breast cancer. While the basic mechanisms are understood, additional mechanisms may be of importance to their action and they may also contribute to the mechanism(s) of acquired resistance. Currently, several novel drugs are entering into clinical trials. Observations of the absence or presence of cross resistance to novel 'pure' steroidal antiestrogens and the non-steroidal tamoxifen may add important information to our understanding of the mechanisms of action of both classes of drugs. Similarly, exploration of different aromatase inhibitors in sequence or concert, as well as the combining of different endocrine treatment options may be warranted. Additionally, alterations in different biochemical parameters such as growth factors should not only be carefully explored in relation to treatment options but should also be followed during the course of treatment to asess alterations over time and in relation to the development of drug resistance.
Collapse
MESH Headings
- Adrenal Cortex/drug effects
- Adrenal Cortex/physiopathology
- Adult
- Aged
- Animals
- Antineoplastic Agents, Hormonal/classification
- Antineoplastic Agents, Hormonal/pharmacology
- Antineoplastic Agents, Hormonal/therapeutic use
- Aromatase Inhibitors
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/physiopathology
- Breast Neoplasms/therapy
- Chemotherapy, Adjuvant
- Combined Modality Therapy
- Drug Resistance, Multiple
- Enzyme Inhibitors/pharmacology
- Enzyme Inhibitors/therapeutic use
- Estrogen Antagonists/adverse effects
- Estrogen Antagonists/pharmacology
- Estrogen Antagonists/therapeutic use
- Estrogens/blood
- Estrogens/physiology
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Gonadotropin-Releasing Hormone/agonists
- Humans
- Insulin-Like Growth Factor I/antagonists & inhibitors
- Insulin-Like Growth Factor I/physiology
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/physiopathology
- Menopause
- Mice
- Middle Aged
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasms, Hormone-Dependent/drug therapy
- Neoplasms, Hormone-Dependent/genetics
- Neoplasms, Hormone-Dependent/physiopathology
- Neoplasms, Hormone-Dependent/therapy
- Progesterone/antagonists & inhibitors
- Progesterone/physiology
- Progestins/antagonists & inhibitors
- Progestins/pharmacology
- Progestins/therapeutic use
- Receptors, Estrogen/drug effects
- Receptors, Estrogen/physiology
- Signal Transduction/drug effects
- Steroids/metabolism
- Tamoxifen/adverse effects
- Tamoxifen/pharmacology
- Tamoxifen/therapeutic use
- Tumor Cells, Cultured/drug effects
Collapse
Affiliation(s)
- E Lønning
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | | |
Collapse
|
45
|
Nørgaard P, Hougaard S, Poulsen HS, Spang-Thomsen M. Transforming growth factor beta and cancer. Cancer Treat Rev 1995; 21:367-403. [PMID: 7585661 DOI: 10.1016/0305-7372(95)90038-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- P Nørgaard
- Section for Radiation Biology, Finsen Center, Righospitalet, Copenhagen, Denmark
| | | | | | | |
Collapse
|
46
|
Kalkhoven E, Kwakkenbos-Isbrücker L, Mummery CL, de Laat SW, van den Eijnden-van Raaij AJ, van der Saag PT, van der Burg B. The role of TGF-beta production in growth inhibition of breast-tumor cells by progestins. Int J Cancer 1995; 61:80-6. [PMID: 7705937 DOI: 10.1002/ijc.2910610114] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We have studied the influence of synthetic progestins on the estrogen-induced proliferation and type-beta transforming-growth-factor (TGF-beta) production of 3 breast-tumor cell lines. In long-term growth experiments, progestins inhibited proliferation of T47D cells, while a specific T47D variant and MCF7 cells were not affected, despite the presence of functional progesterone receptors. The effect of progestins was biphasic, since an initial stimulation of proliferation was followed by a prolonged inhibition. This response suggests the involvement of a progestin-induced negative growth regulator. We show here that TGF-beta s do not fulfill this role since (i) the progestin-induced T47D cells are not sensitive to TGF-beta 1, -beta 2 or -beta 3, (ii) secretion of TGF-beta s is decreased by progestins in all 3 cell lines, and (iii) TGF-beta neutralizing antibodies do not reverse progestin-induced growth inhibition. Furthermore, evidence was obtained that medium conditioned by T47D cells does not contain any other growth inhibitor to which this cell line responds in a negative autocrine manner. In contrast, MCF7 cells are growth-inhibited by all 3 TGF-beta isoforms, but are not growth-inhibited by progestins, suggesting that there is no correlation between growth inhibition by progestins and responsiveness to and production of TGF-beta in vitro. Although TGF-beta is a strong growth inhibitor of normal mammary tissue, recent evidence suggests that, in malignant tissue, enhanced TGF-beta secretion correlates with increased malignancy. Therefore, a progestin-induced decrease in TGF-beta production, as observed here, may lead to enhanced proliferation of normal but not malignant mammary epithelium.
Collapse
Affiliation(s)
- E Kalkhoven
- Hubrecht Laboratory, Netherlands Institute for Developmental Biology, Utrecht
| | | | | | | | | | | | | |
Collapse
|
47
|
Foss AJ, Alexander RA, Guille MJ, Hungerford JL, McCartney AC, Lightman S. Estrogen and progesterone receptor analysis in ocular melanomas. Ophthalmology 1995; 102:431-5. [PMID: 7891981 DOI: 10.1016/s0161-6420(95)31004-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND It has been suggested that pregnancy may promote metastases in melanoma and that the contraceptive pill may be an etiologic factor. The purpose of this study is to determine if uveal or conjunctival melanomas express estrogen or progesterone receptors. METHODS Twenty-seven choroidal and five conjunctival melanomas were investigated. Immunohistochemistry was performed using the antibodies ER-D5, which recognizes heat-shock protein 27 (formerly called the estrogen receptor-associated cytoplasmic antigen); ER-1D5, which recognizes the estrogen receptor; and PgR, which recognizes the progesterone receptor. RESULTS Most of the conjunctival and uveal melanomas stained strongly for heat-shock protein 27 but none of the tumors showed positive nuclear staining for either the estrogen or the progesterone receptor. CONCLUSIONS No evidence was found for either estrogen receptor or progesterone receptor expression in choroidal or conjunctival melanomas. Based on the literature, there is little evidence for these hormones having a role in the development or progression of these tumors.
Collapse
Affiliation(s)
- A J Foss
- Department of Clinical Science, Institute of Ophthalmology, London, England
| | | | | | | | | | | |
Collapse
|
48
|
MacCallum J, Bartlett JM, Thompson AM, Keen JC, Dixon JM, Miller WR. Expression of transforming growth factor beta mRNA isoforms in human breast cancer. Br J Cancer 1994; 69:1006-9. [PMID: 8198961 PMCID: PMC1969445 DOI: 10.1038/bjc.1994.197] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Using an RNAse protection assay, expression of messenger RNA for isoforms of TGF-beta was determined in a series of breast cancers. Of 50 tumours, 45 (90%) expressed TGF-beta 1 mRNA, 39 (78%) expressed TGF-beta 2, and 47 (94%) expressed TGF-beta 3. Patterns of expression varied between different tumours: 37 (74%) cancers expressed all three TGF-beta isoforms, ten (20%) expressed only two isoforms and two expressed TGF-beta 1 alone. One sample showed no evidence of TGF-beta mRNA expression. Although most breast cancers expressed mRNA for at least one isoform of TGF-beta, there were differences in patterns of mRNA expression between individual tumours. The relatively small number of tumours examined precluded detailed analysis between expression and other clinical parameters, but a significant association was identified between one aspect of isoform expression and lymph node status, in that the majority of tumours expressing all three isoforms were associated with lymph node involvement, whereas tumours without one or more isoform were usually lymph node negative (P = 0.025 by Fisher's exact test).
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biopsy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Breast Neoplasms/surgery
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/surgery
- Carcinoma, Lobular/metabolism
- Carcinoma, Lobular/pathology
- Carcinoma, Lobular/surgery
- Female
- Gene Expression
- Humans
- Lymphatic Metastasis
- Mastectomy
- Middle Aged
- Neoplasm Invasiveness
- Polymorphism, Genetic
- RNA, Messenger/analysis
- RNA, Messenger/biosynthesis
- Receptors, Estrogen/analysis
- Transforming Growth Factor beta/biosynthesis
Collapse
Affiliation(s)
- J MacCallum
- University Department of Surgery, Edinburgh Royal Infirmary, UK
| | | | | | | | | | | |
Collapse
|
49
|
Ji H, Stout LE, Zhang Q, Zhang R, Leung HT, Leung BS. Absence of transforming growth factor-beta responsiveness in the tamoxifen growth-inhibited human breast cancer cell line CAMA-1. J Cell Biochem 1994; 54:332-42. [PMID: 8200913 DOI: 10.1002/jcb.240540309] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Tamoxifen has been an effective antiestrogen in suppressing breast cancer growth which is estrogen-responsive or dependent. Early studies have provided circumstantial evidence that transforming growth factor-beta (TGF-beta) may be an autocrine mediator of tamoxifen action. Therefore, it is both fundamentally important and clinically relevant to investigate the relationship between tamoxifen and TGF-beta. In this study, we demonstrated that CAMA-1 cells, which are sensitive to tamoxifen inhibition, did not respond to TGF-beta growth inhibition. The type I and II TGF-beta receptors were undetectable by the radio-ligand affinity labeling technique. Despite the presence of a normal TGF-beta type II receptor gene, the mRNA transcript of the gene was undetectable by the extremely sensitive Intron-differential RNA/PCR method. The possibility that the lack of TGF-beta receptors might be intimately linked to the absence of normal retinoblastoma (Rb) gene products, as suggested by previous studies of retinoblastoma cells, was further investigated. The lack of TGF-beta receptor expression was found due to reasons other than the absence, deletion or abnormality of the Rb gene because a normal Rb gene and its hyper- and hypo-phosphorylated protein products were detected in CAMA-1 cells. In conclusion, our results suggest that the TGF-beta system is not obligatory for antiestrogen growth inhibition of CAMA-1 cells.
Collapse
Affiliation(s)
- H Ji
- Department of Obstetrics and Gynecology, University of Minnesota, Minneapolis 55455
| | | | | | | | | | | |
Collapse
|
50
|
Rubens RD, Bajetta E, Bonneterre J, Klijn JG, Lønning PE, Paridaens R. Treatment of relapse of breast cancer after adjuvant systemic therapy--review and guidelines for future research. Eur J Cancer 1994; 30A:106-11. [PMID: 8142150 DOI: 10.1016/s0959-8049(05)80029-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- R D Rubens
- ICRF Clinical Oncology Unit, Guy's Hospital, London, U.K
| | | | | | | | | | | |
Collapse
|