1
|
Negi A, Kesari KK, Voisin-Chiret AS. Estrogen Receptor-α Targeting: PROTACs, SNIPERs, Peptide-PROTACs, Antibody Conjugated PROTACs and SNIPERs. Pharmaceutics 2022; 14:pharmaceutics14112523. [PMID: 36432713 PMCID: PMC9699327 DOI: 10.3390/pharmaceutics14112523] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Targeting selective estrogen subtype receptors through typical medicinal chemistry approaches is based on occupancy-driven pharmacology. In occupancy-driven pharmacology, molecules are developed in order to inhibit the protein of interest (POI), and their popularity is based on their virtue of faster kinetics. However, such approaches have intrinsic flaws, such as pico-to-nanomolar range binding affinity and continuous dosage after a time interval for sustained inhibition of POI. These shortcomings were addressed by event-driven pharmacology-based approaches, which degrade the POI rather than inhibit it. One such example is PROTACs (Proteolysis targeting chimeras), which has become one of the highly successful strategies of event-driven pharmacology (pharmacology that does the degradation of POI and diminishes its functions). The selective targeting of estrogen receptor subtypes is always challenging for chemical biologists and medicinal chemists. Specifically, estrogen receptor α (ER-α) is expressed in nearly 70% of breast cancer and commonly overexpressed in ovarian, prostate, colon, and endometrial cancer. Therefore, conventional hormonal therapies are most prescribed to patients with ER + cancers. However, on prolonged use, resistance commonly developed against these therapies, which led to selective estrogen receptor degrader (SERD) becoming the first-line drug for metastatic ER + breast cancer. The SERD success shows that removing cellular ER-α is a promising approach to overcoming endocrine resistance. Depending on the mechanism of degradation of ER-α, various types of strategies of developed.
Collapse
Affiliation(s)
- Arvind Negi
- Department of Bioproduct and Biosystems, Aalto University, 00076 Espoo, Finland
- Correspondence: or (A.N.); or (K.K.K.); (A.S.V.-C.)
| | - Kavindra Kumar Kesari
- Department of Bioproduct and Biosystems, Aalto University, 00076 Espoo, Finland
- Department of Applied Physics, School of Science, Aalto University, 02150 Espoo, Finland
- Correspondence: or (A.N.); or (K.K.K.); (A.S.V.-C.)
| | - Anne Sophie Voisin-Chiret
- CERMN (Centre d’Etudes et de Recherche sur le Médicament de Normandie), Normandie University UNICAEN, 14000 Caen, France
- Correspondence: or (A.N.); or (K.K.K.); (A.S.V.-C.)
| |
Collapse
|
2
|
Abstract
Background: The management of patients with localized and advanced breast cancer continues to evolve. Chemotherapy, endocrine therapy, and trastuzumab are effective therapies but leave considerable room for improvement. As the cellular aberrations inherent to cancer cells in general and breast cancer cells specifically are better understood, therapies to target specific cellular pathways continue to be developed with the goal of expanding available effective therapy through better patient selection. Methods: We conducted a computerized search of the medical literature as well as a manual search of selected meeting abstracts. Results: Several targeted therapies are in phase III clinical trials testing their promise in the treatment of breast cancer. Many other agents are completing phase I and II testing. An overview of the most promising agents in clinical development is discussed herein. Conclusions: Targeted therapy for breast cancer is a reality at this time, and several new agents hold promise for expanding and refining the pool of patients likely to further benefit from this approach in the near future.
Collapse
Affiliation(s)
- Timothy J Hobday
- Department of Oncology, Mayo Clinic, Jacksonville, FL 32224, USA
| | | |
Collapse
|
3
|
Mancuso MR, Massarweh SA. Endocrine therapy and strategies to overcome therapeutic resistance in breast cancer. Curr Probl Cancer 2016; 40:95-105. [PMID: 27839747 DOI: 10.1016/j.currproblcancer.2016.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 09/01/2016] [Indexed: 01/04/2023]
Abstract
Despite the remarkable success of endocrine therapy in the treatment of patients with estrogen receptor (ER)- positive breast cancer, not all patients derive benefit from such therapy, or may benefit only temporarily before disease progression or relapse occurs. The value of endocrine therapy, which blocks ER signaling by a variety of strategies, lies in its simplicity, lower toxicity, and better alignment with preserved quality of life, particularly when compared to chemotherapy, which is more toxic and has only modest benefits for many patients with ER-positive breast cancer. It is therefore critical that we discover ways to extend endocrine therapy benefit in patients and prevent therapeutic resistance whenever possible. The tremendous evolution in our understanding of endocrine resistance mechanisms, coupled with the increasing availability of novel agents that target resistance pathways, has led to enhanced treatment approaches for patients with ER-positive breast cancer, primarily through combinations of endocrine agents with a variety of pathway inhibitors. Despite these treatment advances and our changing view of ER-positive breast cancer, there is much work that needs to be done. It remains a problem that we cannot reliably predict which subsets of patients will experience disease relapse or progression on endocrine therapy, and as such, combination strategies with targeted agents have largely been used in unselected patients with ER-positive breast cancer, including those who continue to have endocrine-sensitive disease. Patient selection is a significant issue since most of the targeted therapeutics that we use with endocrine therapy are expensive and can be toxic, and we may be inadvertently overtreating patients whose disease can still be controlled with endocrine therapy alone. In this article, we will review current and future strategies in the treatment of ER-positive breast cancer, as well as the evolving role of targeted therapy in the management of endocrine-resistance.
Collapse
Affiliation(s)
| | - Suleiman Alfred Massarweh
- Division of Oncology, Stanford University School of Medicine; Stanford Cancer Institute, Stanford, CA, 94305.
| |
Collapse
|
4
|
Guérin A, Hao Y, Tang D, Peeples M, Fang A, Kageleiry A, Koo V, Li N, Wu EQ. Treatment patterns and factors associated with the use of everolimus among post-menopausal women with HR+/HER2- metastatic breast cancer: a retrospective US claims study. Expert Opin Pharmacother 2016; 17:1189-96. [PMID: 27052735 DOI: 10.1080/14656566.2016.1176148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVE To assess the real-world use of everolimus in the treatment of hormone-receptor-positive/human-epidermal-growth-factor-receptor-2-negative (HR+/HER2-) metastatic-breast-cancer (mBC). METHODS Postmenopausal women with HR+/HER2- mBC who initiated a new therapy for mBC between 20 July 2012 and 31 March 2014 after a non-steroidal-aromatase-inhibitor were identified from two commercial claims databases. Multivariate logistic regressions were used to identify factors associated with everolimus use versus endocrine-monotherapy or chemotherapy. Dosing patterns and adherence to everolimus were summarized. RESULTS A total of 940 everolimus, 6,134 endocrine-monotherapy, and 3,410 chemotherapy regimens were included across patients' first four lines of therapy. Patients with bone and visceral metastases were more likely to use everolimus versus endocrine-monotherapy. Patients with more comorbidities, visceral or central-nervous-system metastases, and prior chemotherapy use for mBC were less likely to use everolimus versus chemotherapy. Approximately 80% of patients initiated everolimus at label-recommended-dose of 10 mg daily; 60-70% of patients had a medical possession ratio >0.8 to everolimus, and consistently high adherence was observed across lines of therapy. CONCLUSIONS For HR+/HER2- mBC, patients treated with everolimus had more severe disease than patients treated with endocrine-monotherapy but less severe disease than patients treated with chemotherapy. Most patients used everolimus according to label-recommended dose and adherence was high across lines of therapy.
Collapse
Affiliation(s)
| | - Yanni Hao
- b Novartis Pharmaceuticals Corporation , East Hanover , NJ , USA
| | - Derek Tang
- b Novartis Pharmaceuticals Corporation , East Hanover , NJ , USA
| | | | - Anna Fang
- c Analysis Group, Inc ., Boston , MA , USA
| | | | | | - Nanxin Li
- c Analysis Group, Inc ., Boston , MA , USA
| | - Eric Q Wu
- c Analysis Group, Inc ., Boston , MA , USA
| |
Collapse
|
5
|
Shtivelband MI. Everolimus in hormone receptor–positive advanced breast cancer: Targeting receptor-based mechanisms of resistance. Breast 2013; 22:405-10. [DOI: 10.1016/j.breast.2013.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 01/15/2013] [Accepted: 02/11/2013] [Indexed: 01/14/2023] Open
|
6
|
Topcul M, Topcul F, Cetin I. Effects of femara and tamoxifen on proliferation of FM3A cells in culture. Asian Pac J Cancer Prev 2013; 14:2819-22. [PMID: 23803037 DOI: 10.7314/apjcp.2013.14.5.2819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In this study, antiproliferative effects of the selective estrogen receptor modulator Tamoxifen and the aromatase inhibitor letrozole (Femara) were evaluated and compared using the FM3A cell line, originating from a C3H mouse mammary carcinoma and positive in terms of estrogen receptor (ER) expression. Cell kinetic parameters including labelling index, mitotic index and labelling index were assessed after exposure of the. FM3A cell line to 0.001μg/ml of Tamoxifen and 0.25μg/ml of Femara for 4, 8, 16 and 32 h for all parameters. The results showed that cell growth was inhibited by both agents. There was a significant decrease in labelling index and mitotic index and significant increase in apoptotic index for all experimental groups. The differences between control and all experimental groups were statistically significant (p<0.001) for all applications.
Collapse
Affiliation(s)
- Mehmet Topcul
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey.
| | | | | |
Collapse
|
7
|
Zhou J, Teng R, Wang Q, Xu C, Guo J, Yuan C, Shen J, Hu W, Wang L, Xie S. Endocrine resistance in breast cancer: Current status and a perspective on the roles of miRNAs (Review). Oncol Lett 2013; 6:295-305. [PMID: 24137320 PMCID: PMC3789028 DOI: 10.3892/ol.2013.1405] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 05/03/2013] [Indexed: 12/11/2022] Open
Abstract
Current endocrine therapies for females with estrogen receptor-positive breast cancer have facilitated substantial improvements in outcomes. The effectiveness of endocrine therapy is limited by either initial de novo resistance or acquired endocrine resistance. Multiple mechanisms responsible for endocrine resistance have been proposed, including deregulation of various components of the estrogen receptor (ER) pathway, alterations in cell cycle and cell survival signaling molecules, and the activation of escape pathways. Dysregulation of miRNA expression has been associated with experimental and clinical endocrine therapy resistance. miRNAs are pivotal to understanding the complex biological mechanism of endocrine resistance, and may serve as novel candidate predictive and prognostic surrogates and therapeutic targets. This review focuses on current progress concerning the roles of miRNAs in endocrine resistance, and discusses the challenges and opportunities for implementing miRNA-based assays and treatment for patients with endocrine-resistant breast cancer.
Collapse
Affiliation(s)
- Jichun Zhou
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Lu DF, Yang LJ, Wang F, Zhang GL. Inhibitory effect of luteolin on estrogen biosynthesis in human ovarian granulosa cells by suppression of aromatase (CYP19). JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:8411-8418. [PMID: 22838964 DOI: 10.1021/jf3022817] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Inhibition of aromatase, the key enzyme in estrogen biosynthesis, is an important strategy in the treatment of breast cancer. Several dietary flavonoids show aromatase inhibitory activity, but their tissue specificity and mechanism remain unclear. This study found that the dietary flavonoid luteolin potently inhibited estrogen biosynthesis in a dose- and time-dependent manner in KGN cells derived from human ovarian granulosa cells, the major source of estrogens in premenopausal women. Luteolin decreased aromatase mRNA and protein expression in KGN cells. Luteolin also promoted aromatase protein degradation and inhibited estrogen biosynthesis in aromatase-expressing HEK293A cells, but had no effect on recombinant expressed aromatase. Estrogen biosynthesis in KGN cells was inhibited with differing potencies by extracts of onion and bird chili and by four other dietary flavonoids: kaempferol, quercetin, myricetin, and isorhamnetin. The present study suggests that luteolin inhibits estrogen biosynthesis by decreasing aromatase expression and destabilizing aromatase protein, and it warrants further investigation as a potential treatment for estrogen-dependent cancers.
Collapse
Affiliation(s)
- Dan-feng Lu
- Chengdu Institute of Biology, Chinese Academy of Sciences , Chengdu 610041, China
| | | | | | | |
Collapse
|
9
|
Selig BP, Furr JR, Huey RW, Moran C, Alluri VN, Medders GR, Mumm CD, Hallford HG, Mulvihill JJ. Cancer chemotherapeutic agents as human teratogens. ACTA ACUST UNITED AC 2012; 94:626-50. [DOI: 10.1002/bdra.23063] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 06/29/2012] [Accepted: 07/04/2012] [Indexed: 12/11/2022]
|
10
|
Multicenter, phase II, nonrandomized study of docetaxel plus trastuzumab every 21 days as the primary therapy in metastatic breast cancer overexpressing HER2. Anticancer Drugs 2012; 23:239-46. [PMID: 22112931 DOI: 10.1097/cad.0b013e32834e2fe4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Different anthracycline-free regimens have demonstrated activity, without serious cardiac events. This study was conducted to evaluate the activity and toxicity of docetaxel and trastuzumab given every 21 days in patients with metastatic breast cancer (MBC). The primary endpoint was time to progression and the secondary aims included response rate, safety, duration of response, and overall survival. Eligible patients were those with MBC human epidermal growth factor receptor-2+ (HER2+) with no previous chemotherapy for advanced disease. Patients received six cycles of docetaxel (100 mg/m) plus trastuzumab (8 mg/kg loading dose and 6 mg/kg every 21 days thereafter), followed by maintenance treatment with trastuzumab monotherapy every 21 days until disease progression. Forty-nine patients with HER2+ MBC were included. The overall response rate was 44.9% (22/49). With a median follow-up of 16.6 months, the median time to progression was 8.3 months and the median overall survival was 25.7 months. Nineteen patients did not receive treatment continuation with trastuzumab monotherapy. The most common toxicity was febrile neutropenia. A total of 10 patients were taken off the study due to treatment-related toxicity, mainly cardiac events. First-line trastuzumab combined with docetaxel is an effective and well tolerated regimen for HER2+ MBC.
Collapse
|
11
|
Liu X, Jutooru I, Lei P, Kim K, Lee SO, Brents LK, Prather PL, Safe S. Betulinic acid targets YY1 and ErbB2 through cannabinoid receptor-dependent disruption of microRNA-27a:ZBTB10 in breast cancer. Mol Cancer Ther 2012; 11:1421-31. [PMID: 22553354 DOI: 10.1158/1535-7163.mct-12-0026] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Treatment of ErbB2-overexpressing BT474 and MDA-MB-453 breast cancer cells with 1 to 10 μmol/L betulinic acid inhibited cell growth, induced apoptosis, downregulated specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4, and decreased expression of ErbB2. Individual or combined knockdown of Sp1, Sp3, Sp4 by RNA interference also decreased expression of ErbB2 and this response was because of repression of YY1, an Sp-regulated gene. Betulinic acid-dependent repression of Sp1, Sp3, Sp4, and Sp-regulated genes was due, in part, to induction of the Sp repressor ZBTB10 and downregulation of microRNA-27a (miR-27a), which constitutively inhibits ZBTB10 expression, and we show for the first time that the effects of betulinic acid on the miR-27a:ZBTB10-Sp transcription factor axis were cannabinoid 1 (CB1) and CB2 receptor-dependent, thus identifying a new cellular target for this anticancer agent.
Collapse
Affiliation(s)
- Xinyi Liu
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, USA
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Buzdar A, Vogel C, Schwartzberg L, Garin A, Perez A, Ingle J, Houghton M, Zergebel C, Kimball B. Randomized double-blind phase 2 trial of 3 doses of TAS-108 in patients with advanced or metastatic postmenopausal breast cancer. Cancer 2011; 118:3244-53. [DOI: 10.1002/cncr.26419] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 06/13/2011] [Accepted: 06/14/2011] [Indexed: 11/07/2022]
|
13
|
Del Re M, Michelucci A, Simi P, Danesi R. Pharmacogenetics of anti-estrogen treatment of breast cancer. Cancer Treat Rev 2011; 38:442-50. [PMID: 21917382 DOI: 10.1016/j.ctrv.2011.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 08/08/2011] [Accepted: 08/12/2011] [Indexed: 10/17/2022]
Abstract
A major effort is underway to select genetic polymorphisms potentially relevant to the clinical efficacy and safety of endocrine treatment of breast cancer. Genetic factors of the host that affect the metabolism of tamoxifen, a widely used drug for the adjuvant treatment of breast cancer, have received particular attention. Cytochrome P450 isoform 2D6 (CYP2D6) is a key step in the metabolism of tamoxifen to its active moiety endoxifen. Women with functionally deficient genetic variants of CYP2D6 who are given drugs that inhibit CYP2D6 are exposed to low endoxifen plasma levels and may enjoy reduced benefits from tamoxifen treatment. Therefore, CYP2D6 status may be an important predictor of the benefits of tamoxifen to an individual; unfortunately, the data are not uniformly concordant, and definitive evidence that would suggest the routine analysis of CYP2D6 before commencing tamoxifen treatment is not yet available. Recent research has focused on the role UDP-glucuronosyltransferases, a family of metabolizing enzymes that play an important role in the metabolic clearance of tamoxifen and of the aromatase inhibitors as well, and how interindividual differences in these enzymes may play a role in the clinical outcome upon administration of anti-estrogen treatment. In conclusion, whether a pharmacogenetic profile should be obtained prior to initiating tamoxifen therapy is currently a matter of debate, although summing up all the scientific evidence available on this issue it appears that the genetic screening would be an useful support for clinical decision making in selected patients.
Collapse
Affiliation(s)
- Marzia Del Re
- Division of Pharmacology, Department of Internal Medicine, University of Pisa, Italy
| | | | | | | |
Collapse
|
14
|
Verma MK, Miki Y, Sasano H. Endocrine therapy after aromatase inhibitor therapy in breast cancer. Expert Rev Endocrinol Metab 2011; 6:309-312. [PMID: 30754118 DOI: 10.1586/eem.11.21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Mohit K Verma
- a Department of Pathology, Tohoku University Graduate School of Medicine, 2-1 Seriyo-machi, Aoba-ku, Sendai, Miyagi-ken 980-8575, Japan
| | - Yasuhiro Miki
- a Department of Pathology, Tohoku University Graduate School of Medicine, 2-1 Seriyo-machi, Aoba-ku, Sendai, Miyagi-ken 980-8575, Japan
| | - Hironobu Sasano
- b Department of Pathology, Tohoku University Graduate School of Medicine, 2-1 Seriyo-machi, Aoba-ku, Sendai, Miyagi-ken 980-8575, Japan.
| |
Collapse
|
15
|
Chougule MB, Patel AR, Jackson T, Singh M. Antitumor activity of Noscapine in combination with Doxorubicin in triple negative breast cancer. PLoS One 2011; 6:e17733. [PMID: 21423660 PMCID: PMC3057970 DOI: 10.1371/journal.pone.0017733] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 02/09/2011] [Indexed: 01/08/2023] Open
Abstract
Background The aim of this study was to investigate the anticancer activity and mechanism of action of Noscapine alone and in combination with Doxorubicin against triple negative breast cancer (TNBC). Methods TNBC cells were pretreated with Noscapine or Doxorubicin or combination and combination index values were calculated using isobolographic method. Apoptosis was assessed by TUNEL staining. Female athymic Nu/nu mice were xenografted with MDA-MB-231 cells and the efficacy of Noscapine, Doxorubicin and combination was determined. Protein expression, immunohistochemical staining were evaluated in harvested tumor tissues. Results Noscapine inhibited growth of MDA-MB-231 and MDA-MB-468 cells with the IC50 values of 36.16±3.76 and 42.7±4.3 µM respectively. The CI values (<0.59) were suggestive of strong synergistic interaction between Noscapine and Doxorubicin and combination treatment showed significant increase in apoptotic cells. Noscapine showed dose dependent reduction in the tumor volumes at a dose of 150–550 mg/kg/day compared to controls. Noscapine (300 mg/kg), Doxorubicin (1.5 mg/kg) and combination treatment reduced tumor volume by 39.4±5.8, 34.2±5.7 and 82.9±4.5 percent respectively and showed decreased expression of NF-KB pathway proteins, VEGF, cell survival, and increased expression of apoptotic and growth inhibitory proteins compared to single-agent treatment and control groups. Conclusions Noscapine potentiated the anticancer activity of Doxorubicin in a synergistic manner against TNBC tumors via inactivation of NF-KB and anti-angiogenic pathways while stimulating apoptosis. These findings suggest potential benefit for use of oral Noscapine and Doxorubicin combination therapy for treatment of more aggressive TNBC.
Collapse
Affiliation(s)
- Mahavir B. Chougule
- College of Pharmacy, University of Hawaii, Hilo, Hawaii, United States of America
| | - Apurva R. Patel
- College of Pharmacy, Florida A&M University, Tallahassee, Florida, United States of America
| | - Tanise Jackson
- College of Pharmacy, Florida A&M University, Tallahassee, Florida, United States of America
| | - Mandip Singh
- College of Pharmacy, Florida A&M University, Tallahassee, Florida, United States of America
- * E-mail:
| |
Collapse
|
16
|
Orlando L, Schiavone P, Fedele P, Calvani N, Nacci A, Rizzo P, Marino A, D'Amico M, Sponziello F, Mazzoni E, Cinefra M, Fazio N, Maiello E, Silvestris N, Colucci G, Cinieri S. Molecularly targeted endocrine therapies for breast cancer. Cancer Treat Rev 2011; 36 Suppl 3:S67-71. [PMID: 21129614 DOI: 10.1016/s0305-7372(10)70023-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The identification of the estrogen receptor (ER) provided the first target for antiestrogenic therapeutic agents. Endocrine therapies, either by blocking or downregulating the receptor or by suppressing the estrogen production, inhibit the proliferative effect of estradiol on ER. While the activity on ER is considered a real target-mediated therapy, the effect on enzymatic activity involved in estrogen production (mainly inhibition of aromatase by aromatase inhibitors, AIs, and ovarian ablation) could be considered an "indirect" targeted strategy. In addiction to the direct ligand-ER signal, the complexity of endocrine and non endocrine pathways has led to combination therapies against different targets. Tamoxifen is the widely investigated, most used and representative of drugs blocking the ER and has been introduced in the advanced disease, in neoadjuvant and adjuvant setting and for chemo-prevention of high risk women. Its role has been challenged in the last years by the introduction of third generation aromatase inhibitors that have proven a higher activity than tamoxifen and different toxicity. Several other SERMs (selective estrogen receptor modulators) have been investigated, but none of them was clearly superior to tamoxifen. SERDs (selective estrogen receptor downregulators) act as pure estrogen antagonist. They are used in the treatment of advanced breast cancers and their role in other settings still needs further investigation. Here we discuss the well established data with SERMs, SERDs and AIs, mechanisms underlying resistance and rationale for recycling endocrine compounds and for simultaneously targeting different pathways.
Collapse
Affiliation(s)
- L Orlando
- Medical Oncology Division and Breast Unit, A. Perrino Hospital, Strada Statale 7 (Via Appia), Brindisi, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Epithelial breast malignancies are a group of several disease entities that vary in their biology and response to specific therapies. Historically, classification of different molecular types of breast cancer was done through the use of conventional methods such as tumor morphology, grade, and immunophenotyping for estrogen, progesterone, and HER-2/neu receptor expression. Such techniques, although helpful, are not sufficient to accurately predict biologic behavior of breast cancers. Over the last several years, much progress has been made in more precise identification of molecular breast cancer subtypes. Such advances hold a great promise in improving estimation of prognosis and assigning most appropriate therapies. Thanks to use of cDNA microarrays expression technology and quantitative reverse transcriptase polymerase chain reaction (RT-PCR), tumors with specific gene expression patterns can now be identified. This process is presently reshaping perceptions of how breast cancer should be classified and treated. Categorization of breast cancers by gene expression is only beginning to make its way into the daily clinical practice and likely will complement, but not replace, the conventional methods of classification.
Collapse
Affiliation(s)
- Robert Wesolowski
- Division of Medical Oncology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Bhuvaneswari Ramaswamy
- Division of Medical Oncology, College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
18
|
Gastaldi S, Comoglio PM, Trusolino L. The Met oncogene and basal-like breast cancer: another culprit to watch out for? Breast Cancer Res 2010; 12:208. [PMID: 20804567 PMCID: PMC2949647 DOI: 10.1186/bcr2617] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Recent findings suggest the involvement of the MET oncogene, encoding the tyrosine kinase receptor for hepatocyte growth factor, in the onset and progression of basal-like breast carcinoma. The expression profiles of basal-like tumors - but not those of other breast cancer subtypes - are enriched for gene sets that are coordinately over-represented in transcriptional signatures regulated by Met. Consistently, tissue microarray analyses have revealed that Met immunoreactivity is much higher in basal-like cases of human breast cancer than in other tumor types. Finally, mouse models expressing mutationally activated forms of Met develop a high incidence of mammary tumors, some of which exhibit basal characteristics. The present review summarizes current knowledge on the role and activity of Met in basal-like breast cancer, with a special emphasis on the correlation between this tumor subtype and the cellular hierarchy of the normal mammary gland.
Collapse
Affiliation(s)
- Stefania Gastaldi
- Institute for Cancer Research and Treatment (IRCC), University of Torino Medical School, 10060 Candiolo, Torino, Italy
| | | | | |
Collapse
|
19
|
Untch M. Targeted Therapy for Early and Locally Advanced Breast Cancer. ACTA ACUST UNITED AC 2010; 5:144-152. [PMID: 20847827 DOI: 10.1159/000315047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The treatment of patients with breast cancer continues to evolve, with cytotoxic chemotherapy, endocrine therapy, and molecular targeted therapies representing the backbones of modern systemic breast cancer treatment. As we learn better to understand the biology of breast cancer cells, therapies to target specific pathways continue to be developed with the goal of expanding available effective therapy in specific populations. Several targeted drugs with different molecular pathways have achieved approval for metastatic breast cancer, but for early breast cancer trastuzumab is the only one that is currently approved in combination with chemotherapy for adjuvant or neoadjuvant treatment in women with HER2-positive breast cancer. Lapatinib and bevacizumab are both approved for the treatment of metastatic breast cancer and are now investigated in phase III clinical trials testing their effectiveness in the treatment of early breast cancer. In this publication, we review the current status in the treatment of early and locally advanced breast cancer with molecular targeted therapies that are currently approved or in advanced clinical development.
Collapse
Affiliation(s)
- Michael Untch
- Klinik für Gynäkologie und Geburtshilfe, Helios Klinikum Berlin Buch, Germany
| |
Collapse
|
20
|
Early and late tamoxifen resistance in breast cancer. ARCH BIOL SCI 2010. [DOI: 10.2298/abs1002249a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
In our study we investigated the role of the estrogen receptor (ER), progesterone receptor (PR) and clinicohistological parameters in breast cancer patients treated with tamoxifen during the early (2.5 years) vs. late (2.5-5 years) follow-up. The negative status of both ER and PR and tumors equal to or bigger than 2 cm defined the phenotypes and consequently the groups of patients with the worst clinical course of the disease: ER-negative PR-negative, ER-negative pT2 and PR-negative pT2. These high-risk subgroups were related to early follow-up indicating de novo resistance. It is relevant to point out that examined predictive indicators did not show significant importance in the late follow-up study.
Collapse
|
21
|
Prediction of outcome of patients with metastatic breast cancer: evaluation with prognostic factors and Nottingham prognostic index. Support Care Cancer 2009; 18:1553-64. [DOI: 10.1007/s00520-009-0778-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Accepted: 10/28/2009] [Indexed: 11/26/2022]
|
22
|
Zhang S, Lei P, Liu X, Li X, Walker K, Kotha L, Rowlands C, Safe S. The aryl hydrocarbon receptor as a target for estrogen receptor-negative breast cancer chemotherapy. Endocr Relat Cancer 2009; 16:835-44. [PMID: 19447902 PMCID: PMC2766348 DOI: 10.1677/erc-09-0054] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and the relatively non-toxic selective aryl hydrocarbon receptor (AhR) modulator 6-methyl-1,3,8-trichlorodibenzo-furan (MCDF) induced CYP1A1-dependent ethoxyresorufin O-deethylase activity and inhibited proliferation of seven estrogen receptor (ER) negative breast cancer cell lines. MCDF, TCDD and structurally related 2,3,7,8-tetrachlorodibenzofuran, 1,2,3,7,8-pentachlorodibenzo-p-dioxin, 2,3,4,7,8-pentachlorodibenzofuran, and 3,3',4,4',5-pentachlorobiphenyl induced CYP1A1 and inhibited proliferation of BT-474 and MDA-MB-468 cells. In BT474 and MDA-MB-468 cells transfected with a small inhibitory RNA for the AhR, the antiproliferative activity of the chlorinated aromatic compounds was reversed, whereas for MCDF, only partial reversal was observed, suggesting that this compound acts through both AhR-dependent and AhR-independent pathways in these two cell lines. MCDF also inhibited tumor growth in athymic nude mice in which MDA-MB-468 cells were injected directly into the mammary fat pad. These results suggest that the AhR is a potential drug target for treatment of ER-negative breast cancer.
Collapse
Affiliation(s)
- Shu Zhang
- Institute of Biosciences and Technology, Texas A&M Health Science Center, 2121 Holcombe Blvd., Houston, TX 77030
| | - Ping Lei
- Institute of Biosciences and Technology, Texas A&M Health Science Center, 2121 Holcombe Blvd., Houston, TX 77030
| | - Xinyi Liu
- Institute of Biosciences and Technology, Texas A&M Health Science Center, 2121 Holcombe Blvd., Houston, TX 77030
| | - Xiangrong Li
- Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466
| | - Kelcey Walker
- Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466
| | - Leela Kotha
- Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466
| | - Craig Rowlands
- Dow Chemical Company, Toxicology and Environmental Research and Consulting, Midland, MI 48674
| | - Stephen Safe
- Institute of Biosciences and Technology, Texas A&M Health Science Center, 2121 Holcombe Blvd., Houston, TX 77030
- Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466
| |
Collapse
|
23
|
Jaspers JE, Rottenberg S, Jonkers J. Therapeutic options for triple-negative breast cancers with defective homologous recombination. Biochim Biophys Acta Rev Cancer 2009; 1796:266-80. [PMID: 19616605 DOI: 10.1016/j.bbcan.2009.07.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 07/05/2009] [Accepted: 07/08/2009] [Indexed: 12/27/2022]
Abstract
Breast cancer is the most common malignancy among women in developed countries, affecting more than a million women per year worldwide. Over the last decades, our increasing understanding of breast cancer biology has led to the development of endocrine agents against hormone receptor-positive tumors and targeted therapeutics against HER2-expressing tumors. However, no targeted therapy is available for patients with triple-negative breast cancer, lacking expression of hormone receptors and HER2. Overlap between BRCA1-mutated breast cancers and triple-negative tumors suggests that an important part of the triple-negative tumors may respond to therapeutics targeting BRCA1-deficient cells. Here, we review the features shared between triple-negative, basal-like and BRCA1-related breast cancers. We also discuss the development of novel therapeutic strategies to target BRCA1-mutated tumors and triple-negative tumors with BRCA1-like features. Finally, we highlight the utility of mouse models for BRCA1-mutated breast cancer to optimize (combination) therapy and to understand drug resistance.
Collapse
Affiliation(s)
- Janneke E Jaspers
- Division of Molecular Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | |
Collapse
|
24
|
Liu X, Abdelrahim M, Abudayyeh A, Lei P, Safe S. The nonsteroidal anti-inflammatory drug tolfenamic acid inhibits BT474 and SKBR3 breast cancer cell and tumor growth by repressing erbB2 expression. Mol Cancer Ther 2009; 8:1207-17. [PMID: 19435870 DOI: 10.1158/1535-7163.mct-08-1097] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tolfenamic acid (TA) is a nonsteroidal anti-inflammatory drug that inhibits pancreatic cancer cell and tumor growth through decreasing expression of specificity protein (Sp) transcription factors. TA also inhibits growth of erbB2-overexpressing BT474 and SKBR3 breast cancer cells; however, in contrast to pancreatic cancer cells, TA induced down-regulation of erbB2 but not Sp proteins. TA-induced erbB2 down-regulation was accompanied by decreased erbB2-dependent kinase activities, induction of p27, and decreased expression of cyclin D1. TA also decreased erbB2 mRNA expression and promoter activity, and this was due to decreased mRNA stability in BT474 cells and, in both cell lines, TA decreased expression of the YY1 and AP-2 transcription factors required for basal erbB2 expression. In addition, TA also inhibited tumor growth in athymic nude mice in which BT474 cells were injected into the mammary fat pad. TA represents a novel and promising new anticancer drug that targets erbB2 by decreasing transcription of this oncogene.
Collapse
Affiliation(s)
- Xinyi Liu
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843-4466, USA
| | | | | | | | | |
Collapse
|
25
|
Richert MM, Welch DR. Metastasis of hormone receptor positive breast cancer. Cancer Treat Res 2009; 147:1-22. [PMID: 21461826 DOI: 10.1007/978-0-387-09463-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
|
26
|
Rodriguez-Gonzalez A, Cyrus K, Salcius M, Kim K, Crews CM, Deshaies RJ, Sakamoto KM. Targeting steroid hormone receptors for ubiquitination and degradation in breast and prostate cancer. Oncogene 2008; 27:7201-11. [PMID: 18794799 DOI: 10.1038/onc.2008.320] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Proteolysis targeting chimeric molecules (Protacs) target proteins for destruction by exploiting the ubiquitin-dependent proteolytic system of eukaryotic cells. We designed two Protacs that contain the peptide 'degron' from hypoxia-inducible factor-1alpha, which binds to the Von-Hippel-Lindau (VHL) E3 ubiquitin ligase complex, linked to either dihydroxytestosterone that targets the androgen receptor (AR; Protac-A), or linked to estradiol (E2) that targets the estrogen receptor-alpha (ERalpha; Protac-B). We hypothesized that these Protacs would recruit hormone receptors to the VHL E3 ligase complex, resulting in the degradation of receptors, and decreased proliferation of hormone-dependent cell lines. Treatment of estrogen-dependent breast cancer cells with Protac-B induced the degradation of ERalpha in a proteasome-dependent manner. Protac-B inhibited the proliferation of ERalpha-dependent breast cancer cells by inducing G(1) arrest, inhibition of retinoblastoma phosphorylation and decreasing expression of cyclin D1, progesterone receptors A and B. Protac-B treatment did not affect the proliferation of estrogen-independent breast cancer cells that lacked ERalpha expression. Similarly, Protac-A treatment of androgen-dependent prostate cancer cells induced G(1) arrest but did not affect cells that do not express AR. Our results suggest that Protacs specifically inhibit the proliferation of hormone-dependent breast and prostate cancer cells through degradation of the ERalpha and AR, respectively.
Collapse
Affiliation(s)
- A Rodriguez-Gonzalez
- Department of Pediatrics, Gwynne Hazen Cherry Laboratories, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Brandes LJ. N,N-diethyl-2-[4-(phenylmethyl) phenoxy] ethanamine (DPPE; tesmilifene), a chemopotentiating agent with hormetic effects on DNA synthesis in vitro, may improve survival in patients with metastatic breast cancer. Hum Exp Toxicol 2008; 27:143-7. [PMID: 18480139 DOI: 10.1177/0960327108090751] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
N,N-diethyl-2-[4-(phenylmethyl) phenoxy] ethanamine (DPPE; tesmilifene) is a novel anti-histaminic and chemopotentiating agent that has a hormetic effect on DNA synthesis in MCF (Michigan Cancer Foundation)-7 human breast cancer cells in vitro and stimulates the growth of experimental tumors in rodents. In a prospectively randomized phase three trial (NCIC MA.19), 152 patients who were co-administered DPPE and doxorubicin survived 50% longer (P < 0.03) than 153 patients who were administered the same dose and schedule of doxorubicin alone. At clinically relevant in vitro concentrations that do not inhibit the P-glycoprotein (P-gp) pump, DPPE selectively sensitizes the cancer cells that express the multiple drug resistance phenotype, making them more susceptible to the cytotoxic effects of chemotherapeutic agents, including anthracyclines and taxanes. Based on its previously demonstrated interaction with histamine at CYP3A4, a P450 that metabolizes arachidonic acid, and its induction of high levels of prostacyclin in the gut of rodents, modulation by DPPE of the intracellular concentration of arachidonate products, such as hydroxyeicosatetraeinoic acids, implicated in increased cancer cell proliferation and metastasis, is postulated.
Collapse
Affiliation(s)
- Lorne J Brandes
- Department of Medicine and Pharmacology/Therapeutics, University of Manitoba and Section of Hematology/Oncology, CancerCare Manitoba, 675 McDermot Avenue, Winnipeg MB R3E0V9, Canada.
| |
Collapse
|
28
|
Tonini G, Schiavon G, Fratto ME, Vincenzi B, Santini D. Hormono-biological therapy in metastatic breast cancer: preclinical evidence, clinical studies and future directions. Expert Opin Biol Ther 2008; 8:221-34. [PMID: 18194078 DOI: 10.1517/14712598.8.2.221] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Breast cancer growth is regulated by coordinated actions of the estrogen receptor (ER) and various growth factor receptor signalling pathways. This complex interactive signalling potentially explains some of the reasons behind endocrine therapy action and resistance. Recent research into the molecular biology of ER signalling has revealed new molecular targets which, if present in cancer cells, might be additionally targeted using various signal transduction inhibitors to overcome or prevent resistance to endocrine therapy. The dynamic inverse relationship between the expression of ER and growth factor receptors brings more excitement to the potential of restoring ER expression in apparently ER-negative cells by inhibition of growth factor signalling. The multiple pathways involved in activating ERs also provide a rationale for combining endocrine and non-endocrine therapies that block different signalling pathways. Ongoing clinical trials promise to further improve the present care for breast cancer patients.
Collapse
Affiliation(s)
- Giuseppe Tonini
- University Campus Bio-Medico, Medical Oncology, Via Emilio Longoni, 83 00155 Rome, Italy.
| | | | | | | | | |
Collapse
|
29
|
Mlineritsch B, Tausch C, Singer C, Luschin-Ebengreuth G, Jakesz R, Ploner F, Stierer M, Melbinger E, Menzel C, Urbania A, Fridrik M, Steger G, Wohlmuth P, Gnant M, Greil R. Exemestane as primary systemic treatment for hormone receptor positive post-menopausal breast cancer patients: a phase II trial of the Austrian Breast and Colorectal Cancer Study Group (ABCSG-17). Breast Cancer Res Treat 2007; 112:203-13. [PMID: 18158620 DOI: 10.1007/s10549-007-9843-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Accepted: 11/28/2007] [Indexed: 11/29/2022]
Abstract
BACKGROUND A multicenter phase II study was conducted to analyze the clinical activity of the steroidal aromatase inhibitor exemestane in the neoadjuvant treatment of post-menopausal women with strongly ER- and/or PgR- positive operable breast cancer. PATIENTS AND METHODS From September 2000 to December 2003, 80 women were recruited for treatment with exemestane 25 mg once daily for 4 months. The primary end-point was the clinical response rate according the WHO criteria; the secondary end-points included toxicity and the number of patients who qualified for breast conserving surgery at the end of treatment, comparability of evaluation methods for response, potential alterations of hormone receptor and Her2/neu status during treatment. RESULTS On an intention to evaluate analysis, according to the prespecified criteria the overall clinical objective response rate was 34%, the pCR rate was 3% and the rate of breast conserving surgery was 76%. When sonographic and mammographic longitudinal measurements were included in patients with missing palpation data, response rates were 38% and 41%, respectively. The tumor response was independent of the Her2/neu status which remained unchanged during treatment. In contrast, while the ER expression remained unaltered, downregulation of the PgR was observed. The treatment was well tolerated with no grade 3 and 4 toxicities except gastrointestinal (one grade 3 case) and hot flushes (two grade 3 cases). CONCLUSION This study shows that exemestane is effective and safe as a preoperative therapy in post-menopausal patients with strongly hormone receptor-positive breast cancer.
Collapse
Affiliation(s)
- Brigitte Mlineritsch
- IIIrd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectious Disease, Laboratory of Immunological and Molecular Cancer Research, Paracelsus Medical University Salzburg, Salzburg, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Breast cancer is the most commonly diagnosed cancer in American women and is the second leading cause of cancer-related deaths in this population. This review highlights how the use of a multidisciplinary approach to the management of diseases of the breast and the introduction of novel systemic therapies have improved the quality of life and survival in patients with breast cancer.
Collapse
|
31
|
Salman T, Massiah N, Burns S, Mills S. Metastatic breast cancer to the cervix and myometrium. J OBSTET GYNAECOL 2007; 27:753-4. [PMID: 17999323 DOI: 10.1080/01443610701631060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- T Salman
- Department of Obstetrics and Gynaecology, Royal Albert Edward Infirmary, Wigan, UK.
| | | | | | | |
Collapse
|
32
|
Naeim A, Sawhney R, MacLean CH, Sanati H. Quality Indicators for the Care of Breast Cancer in Vulnerable Elders. J Am Geriatr Soc 2007; 55 Suppl 2:S258-69. [PMID: 17910546 DOI: 10.1111/j.1532-5415.2007.01331.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
MESH Headings
- Aged
- Aromatase Inhibitors/therapeutic use
- Bone Neoplasms/diagnosis
- Bone Neoplasms/secondary
- Bone Neoplasms/therapy
- Breast Neoplasms/diagnosis
- Breast Neoplasms/metabolism
- Breast Neoplasms/therapy
- Carcinoma, Ductal, Breast/diagnosis
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/therapy
- Carcinoma, Lobular/diagnosis
- Carcinoma, Lobular/metabolism
- Carcinoma, Lobular/therapy
- Chemotherapy, Adjuvant
- Comorbidity
- Diphosphonates/therapeutic use
- Evidence-Based Medicine
- Female
- Frail Elderly
- Humans
- Lymphatic Metastasis
- Mammography
- Mastectomy
- Neoplasm Staging
- Process Assessment, Health Care
- Quality Indicators, Health Care
- Receptor, ErbB-2/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
Collapse
Affiliation(s)
- Arash Naeim
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA.
| | | | | | | |
Collapse
|
33
|
Rugo HS. The breast cancer continuum in hormone-receptor-positive breast cancer in postmenopausal women: evolving management options focusing on aromatase inhibitors. Ann Oncol 2007; 19:16-27. [PMID: 17693420 DOI: 10.1093/annonc/mdm282] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
There are now a number of highly effective options for the treatment of hormone-receptor-positive breast cancer. Although tamoxifen was the standard hormonal treatment for many years, we now have another option for postmenopausal women: the third-generation aromatase inhibitors (AIs) anastrozole, exemestane and letrozole. A number of trials have investigated the use of third-generation AIs compared with tamoxifen throughout the continuum of treatment settings for postmenopausal women with breast cancer. In the neoadjuvant setting, letrozole, given for 4 months, resulted in better overall clinical response and breast-conserving surgery rates than tamoxifen. The Immediate Preoperative Anastrozole Tamoxifen or Combined with Tamoxifen trial gave anastrozole for 3 months with no difference in clinical response but significantly improved breast-conserving surgery rates. Compared with tamoxifen, anastrozole and letrozole significantly improved disease-free survival as early adjuvant treatment for hormone-receptor-positive disease. Switching to anastrozole or exemestane after 2 to 3 years of adjuvant tamoxifen for a total of 5 years of therapy was also more effective than continued tamoxifen. All three agents are approved in the early adjuvant or switching setting in the USA. Letrozole following 5 years of tamoxifen as extended adjuvant treatment improved disease-free survival and, in the node-positive subgroup, overall survival when compared with placebo. Anastrozole and letrozole are both approved for the first-line treatment of hormone-sensitive advanced breast cancer in postmenopausal women; letrozole showed an improved response rate compared with tamoxifen. Anastrozole, letrozole and exemestane are all indicated for the second-line treatment of advanced breast cancer. In summary, third-generation AIs have been shown to have superior efficacy over tamoxifen in the metastatic, neoadjuvant and adjuvant settings and to improve outcome as extended adjuvant therapy following 5 years of tamoxifen. Ongoing studies will further define the role of sequential adjuvant treatment. Appropriate duration of treatment is another important area of investigation. This review will cover hormonal therapy for postmenopausal women with breast cancer and will not address the treatment of premenopausal women.
Collapse
Affiliation(s)
- H S Rugo
- University of California San Francisco, Comprehensive Cancer Center, San Francisco, USA.
| |
Collapse
|
34
|
Winters L, Habin K, Gallagher J. Aromatase Inhibitors and Musculoskeletal Pain in Patients With Breast Cancer. Clin J Oncol Nurs 2007; 11:433-9. [PMID: 17623627 DOI: 10.1188/07.cjon.433-439] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Aromatase inhibitors are recommended for use by postmenopausal women who have estrogen receptor-positive early-stage breast cancer. They reduce local and distant recurrence more effectively than tamoxifen. Anastrozole (Arimidex, AstraZeneca Pharmaceuticals LP), letrozole (Femara, Novartis Pharmaceuticals Corporation), and exemestane (Aromasin, Pfizer Inc.) inhibit aromatase activity, thus significantly decreasing estrogen production in tissues such as liver, muscle, and fat. Very low levels of estrogen may be one cause of musculoskeletal pain, a common side effect associated with the drugs. In the major adjuvant aromatase inhibitor clinical trials, 25%-30% of the patients enrolled experienced musculoskeletal pain. Although quality-of-life studies demonstrate that aromatase inhibitors are well tolerated overall, some women discontinue this treatment because of musculoskeletal pain. Little is known about how to predict, measure, or manage musculoskeletal pain caused by aromatase inhibition. Oncology nurses play an important role in the assessment and management of side effects related to cancer. This article provides an overview of the current knowledge about musculoskeletal pain in patients with breast cancer receiving aromatase inhibitor therapy.
Collapse
Affiliation(s)
- Loren Winters
- Cancer Center, Massachusetts General Hospital, Boston, USA.
| | | | | |
Collapse
|
35
|
Abstract
OBJECTIVES To review the chemotherapy and targeted biologic options for treatment of advanced and metastatic breast cancer. DATA SOURCES Clinical and research articles and textbook chapters. CONCLUSION Recent clinical research has led to the use of novel targeted therapies in the management of locally advanced and metastatic breast cancer. Although metastatic breast cancer remains incurable, increases in disease-free and overall survival has been achieved. IMPLICATIONS FOR NURSING PRACTICE Recent advances in the biological targeted therapies against Her2/neu, VEGF, and EGFR are now either approved therapies or are in the final stages of clinical testing. Oncology nurses can help decrease toxicities and maintain better QOL by fully understanding the mechanism of action of the drugs, expected side effects, and anticipated response to the novel regimen.
Collapse
|
36
|
Jørgensen JT, Nielsen KV, Ejlertsen B. Pharmacodiagnostics and Targeted Therapies—A Rational Approach for Individualizing Medical Anticancer Therapy in Breast Cancer. Oncologist 2007; 12:397-405. [PMID: 17470682 DOI: 10.1634/theoncologist.12-4-397] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The selection of therapy for a particular breast cancer patient is traditionally based on average results from randomized clinical trials. Rational pharmacotherapy is in essence about selecting the right drug(s) for the right patient, and in order to guide this selection process pharmacodiagnostic tests are indispensable. A number of tests have been developed or are under development for targeted therapies, such as antiestrogens, human epidermal growth factor receptor 2 inhibitors, and topoisomerase inhibitors. Based on a biopsy from the tumor, the tests are able to identify patients with a high probability to benefit from these therapies. The detection of the predictive biomarkers is based on different technologies, such as immunohistochemistry, fluorescence in situ hybridization, and chromogenic in situ hybridization. Pharmacodiagnostic tests will play an important role in the further development of targeted therapies and may be seen as a prerequisite for the introduction of individualized medicine in oncology.
Collapse
Affiliation(s)
- Jan Trøst Jørgensen
- Clinical Research, Dako Denmark A/S, Produktionsvej 42, DK-2600 Glostrup, Denmark.
| | | | | |
Collapse
|
37
|
Harwood KV. Advances in Endocrine Therapy for Breast Cancer: Considering Efficacy, Safety, and Quality of Life. Clin J Oncol Nurs 2007; 8:629-37. [PMID: 15637958 DOI: 10.1188/04.cjon.629-637] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Breast cancer is the most common cancer found in women in the United States. Endocrine therapy is the standard of care for most women with hormone receptor-positive tumors in adjuvant and metastatic settings. The selective estrogen response modifier tamoxifen has been the standard treatment for postmenopausal patients for many years. Numerous new endocrine therapy agents provide women with novel treatment options, including the nonsteroidal aromatase inhibitors anastrozole and letrozole, the steroidal aromatase inhibitor exemestane, and the estrogen receptor antagonist fulvestrant. Clinical trials have begun to define the role of these agents and their unique side-effect profiles. Nurses are vital in supporting patients in the decision-making process, managing side effects of treatment, and making observations to enhance understanding of the patient experience with new treatments. This article will assist nurses in educating patients about endocrine therapy options and their associated potential short- and long-term side effects, as well as treatment demands.
Collapse
Affiliation(s)
- Kerry V Harwood
- Cancer Patient Education Program, Duke University Health System, Durham, NC, USA.
| |
Collapse
|
38
|
Viale PH. Aromatase Inhibitor Agents in Breast Cancer: Evolving Practices in Hormonal Therapy Treatment. Oncol Nurs Forum 2007; 32:343-53. [PMID: 15759071 DOI: 10.1188/05.onf.343-353] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE/OBJECTIVES To review the role of aromatase inhibitor agents with regard to current treatment strategies with hormonal therapy for women with breast cancer. DATA SOURCES Published articles and books. DATA SYNTHESIS Hormonal therapy is an essential component of the treatment of most women with breast cancer. Aromatase inhibitor agents are becoming an integral part of treatment for women with metastatic breast cancer and recently have become much more prominent in the treatment of women with early-stage breast cancer. The exact role of these agents in adjuvant therapy of breast cancer, either sequentially with the "gold standard" tamoxifen or for the duration of therapy, has yet to be determined. CONCLUSIONS Recent studies with aromatase inhibitor agents are intriguing and suggest an improved side-effect profile and efficacy. The approval of these agents for the adjuvant treatment of breast cancer has led to a significant change in practice. IMPLICATIONS FOR NURSING Breast cancer is an extremely common cancer in women, and oncology nurses take care of large numbers of patients with this disease. Oncology nurses need the most recent information so they can discuss aromatase inhibitor agents and therapy with their patients.
Collapse
|
39
|
Lu J, Li H, Cao D, Di G, Wu J, Sheng K, Han Q, Shen Z, Shao Z. Clinical significance of aromatase protein expression in axillary node negative breast cancer. J Cancer Res Clin Oncol 2007; 133:401-9. [PMID: 17219197 DOI: 10.1007/s00432-006-0186-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2006] [Accepted: 12/05/2006] [Indexed: 11/29/2022]
Abstract
PURPOSE Aromatase catalyzes the conversion of androgens to estrogens; its high expression in breast cancers may be responsible for the local high levels of estrogen and may promote tumor growth and progression; however, the clinical importance of aromatase remains unclear and needs to be further researched. METHODS By immunochemistry, we detected aromatase, MMP2 and MMP9 immunoreactivity in 244 axillary lymph node negative breast cancers. RESULTS Aromatase immunoreactivity was positively associated with co-expression of MMP2 and MMP9 (MMP2/9) in the estrogen receptor and/or progestin receptor- (ER/PR) positive patients (P < 0.05), but not in the ER and PR negative patients (P > 0.05); aromatase status positively associated with tumor size in the postmenopausal patients (P < 0.05) but not in the premenopausal patients (P > 0.05). The proportional hazards assumption was violated for aromatase status (global test, P < 0.05), and aromatase was an unfavorable prognostic factor for disease-free survival (DFS) (P = 0.04) in multivariate analysis of time-dependent non-proportional Cox regression. In the ER/PR-positive patients, positive aromatase staining was significantly associated with decreased overall survival (OS) (P = 0.04), but there was no such association in the ER and PR negative patients (P > 0.05). CONCLUSIONS Our study suggested that local estrogen production by aromatase plays important roles in the growth and invasiveness of breast cancer; tumor aromatase status may be indicative of breast cancer prognosis in some patients.
Collapse
Affiliation(s)
- Jingsong Lu
- Department of Breast Surgery, Cancer Hospital/Cancer Institute, Fudan University, 399 Ling-Ling Road, Shanghai 200032, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstracts of the XXVII Italian Society for the Study of Connective Tissues (SISC) Meeting, Bologna, Italy, 8-10 November 2007. Connect Tissue Res 2007; 48:338-63. [PMID: 18075821 DOI: 10.1080/03008200701726970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
41
|
Paulmurugan R, Gambhir SS. An intramolecular folding sensor for imaging estrogen receptor-ligand interactions. Proc Natl Acad Sci U S A 2006; 103:15883-8. [PMID: 17043219 PMCID: PMC1635097 DOI: 10.1073/pnas.0607385103] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Strategies for high-throughput analysis of interactions between various hormones and drugs with the estrogen receptor (ER) are crucial for accelerating the understanding of ER biology and pharmacology. Through careful analyses of the crystal structures of the human ER (hER) ligand-binding domain (hER-LBD) in complex with different ligands, we hypothesized that the hER-LBD intramolecular folding pattern could be used to distinguish ER agonists from selective ER modulators and pure antiestrogens. We therefore constructed and validated intramolecular folding sensors encoding various hER-LBD fusion proteins that could lead to split Renilla/firefly luciferase reporter complementation in the presence of the appropriate ligands. A mutant hER-LBD with low affinity for circulating estradiol was also identified for imaging in living subjects. Cells stably expressing the intramolecular folding sensors expressing wild-type and mutant hER-LBD were used for imaging ligand-induced intramolecular folding in living mice. This is the first hER-LBD intramolecular folding sensor suited for high-throughput quantitative analysis of interactions between hER with hormones and drugs using cell lysates, intact cells, and molecular imaging of small living subjects. The strategies developed can also be extended to study and image other important protein intramolecular folding systems.
Collapse
Affiliation(s)
- Ramasamy Paulmurugan
- Departments of Radiology and Bioengineering, Bio-X Program, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, James H. Clark Center, 318 Campus Drive, East Wing, First Floor, Stanford, CA 94305-5427
- *To whom correspondence may be addressed. E-mail:
or
| | - Sanjiv S. Gambhir
- Departments of Radiology and Bioengineering, Bio-X Program, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, James H. Clark Center, 318 Campus Drive, East Wing, First Floor, Stanford, CA 94305-5427
- *To whom correspondence may be addressed. E-mail:
or
| |
Collapse
|
42
|
Ballester Viéitez A, Ferriols Lisart F, Magraner Gil J. [A cost-effectiveness study of first-line hormone therapy in post-menopausal patients with metastatic breast cancer on neoadjuvant treatment]. FARMACIA HOSPITALARIA 2006; 30:71-7. [PMID: 16796419 DOI: 10.1016/s1130-6343(06)73950-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
OBJECTIVE Breast cancer is a hormone-dependent tumor, which expands therapy options. The aim of this study was to perform a pharmacoeconomic assessment of these drugs in post-menopausal patients with metastatic breast cancer or on neoadjuvant therapy. METHOD To assess the efficacy of both drug classes a meta-analysis was carried out from papers obtained in a Medline literature search (1997 - June 2004) using the following keywords: breast cancer, postmenopausal, aromatase inhibitors, antiestrogens, tamoxifen, neoadjuvant. Effectiveness parameters included: objective response, percentage with conservative surgery in neoadjuvancy; in metastatic breast cancer, time to progression. Costs were considered from drug acquisition data. To determine result strength a sensitivity analysis was undertaken, with modified costs and effectiveness. RESULTS Results show a higher effectiveness for aromatase inhibitors as compared to antiestrogens. In neoadjuvant therapy: 56.38% versus 36% for objective response rate, and 47.64% versus 35% for conservative surgery. In metastatic cancer: 9.96 versus 6.61 months for time to progression (p <or= 0.05). The pharmacoeconomic analysis showed that selecting anastrozole represents a cost of 969 euros and 779 euros per extra effectiveness unit regarding objective response and conservative surgery, respectively, whereas in metastatic cancer patients it represents 56,525 euros per extra effectiveness unit. CONCLUSIONS Selecting an aromatase inhibitor is a valid option in neoadjuvancy, even if such selection would represent a very high cost for metastatic cancer.
Collapse
|
43
|
Sato N, Sano M, Tabei T, Asaga T, Ando J, Fujii H, Yamamoto N, Kurosumi M, Inoue K, Kimura M. Combination docetaxel and trastuzumab treatment for patients with her-2-overexpressing metastatic breast cancer: A multicenter, phase-II study. Breast Cancer 2006; 13:166-71. [PMID: 16755112 DOI: 10.2325/jbcs.13.166] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Pre-clinical and clinical studies indicate that a combination of docetaxel and trastuzumab may effectively treat patients with human epidermal growth factor receptor-2 (HER-2) overexpressing metastatic breast cancer. We evaluated the efficacy and safety of this combination in a multicenter, open-label phase II study in Japan. METHODS Women with metastatic breast cancer whose tumors overexpressed HER-2, as assessed by immunohistochemistry and by fluorescence in situ hybridisation, received 2 to 6 cycles of docetaxel (70 mg/m2, every 3 weeks) and trastuzumab (4 mg/kg loading dose, 2 mg/kg weekly thereafter). The primary endpoint was tumor response. Secondary endpoints were time to disease progression and adverse events. RESULTS Of the 40 women enrolled in the study, 27 (68%) completed 6 cycles of treatment. Three patients discontinued the study before the second cycle. Median follow-up was 20.8 months (range, 0.6 to 30.9 months). The overall response rate was 65% (26/40; 95% CI, 48% to 79%). The median time to progression was 6.8 months (range, 0.6 to 21.2 months). Of the 40 patients, 35 (88%) had grade 3 or 4 leukopenia, and 33 (83%) had grade 3 or 4 neutropenia. Most instances of leukopenia and neutropenia were manageable by reducing the dose of docetaxel or by treatment with granulocyte colony-stimulating factor. In 4 patients, left ventricular ejection fraction decreased by more than 10% from baseline. CONCLUSIONS The combination of docetaxel and trastuzumab was as effective as reported in other similar studies and was well tolerated in these patients.
Collapse
Affiliation(s)
- Nobuaki Sato
- Department of Surgery, Niigata Cancer Center Hospital, Niigata, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kim HJ, Cui X, Hilsenbeck SG, Lee AV. Progesterone receptor loss correlates with human epidermal growth factor receptor 2 overexpression in estrogen receptor-positive breast cancer. Clin Cancer Res 2006; 12:1013s-1018s. [PMID: 16467118 DOI: 10.1158/1078-0432.ccr-05-2128] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Response to endocrine therapy in breast cancer correlates with estrogen receptor (ER) and progesterone receptor (PR) status. It was originally hypothesized that the ability of PR to predict response to endocrine therapy was due to the fact that PR is an estrogen-regulated gene and that its levels represented a marker of functional ER activity. However, it is now known that loss of PR can occur via multiple mechanisms, many of which do not include ER function, e.g., hypermethylation of the PR promoter and loss of heterozygosity of the PR gene. We have shown that growth factor signaling pathways can directly down-regulate PR levels via the phosphatidylinositol 3'-kinase (PI3K)/Akt/mTOR pathway, and that this can occur independent of ER. For example, overexpression of myr-Akt in MCF-7 cells causes complete loss of PR protein and mRNA but does not reduce ER levels or activity, thus generating ER+/PR- MCF-7 cells. Therefore, the absence of PR may not simply reflect a lack of ER activity but rather may reflect hyperactive cross-talk between ER and growth factor signaling pathways. Consistent with this hypothesis, several recent clinical studies have found that ER+/PR- breast cancers overexpress human epidermal growth factor receptor (HER) 1 and HER2 compared with ER+/PR+ breast cancers. Although HER receptors can lower ER levels, one study showed that loss of PR correlated with high HER2 levels in a multivariate analysis. Furthermore, loss of PTEN, a negative regulator of the PI3K/Akt signaling pathway, has been shown to be associated with specific loss of PR and no change in ER levels. Given the well-recognized resistance of ER+/PR- breast cancer to antiestrogens, more studies are needed to better understand the etiology of ER+/PR- breast cancer, particularly the analysis of other growth factor receptors and their downstream signaling intermediates with respect to PR status.
Collapse
Affiliation(s)
- Hyun-Jung Kim
- Breast Center, Baylor College of Medicine and the Methodist Hospital, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
45
|
Cui X, Schiff R, Arpino G, Osborne CK, Lee AV. Biology of progesterone receptor loss in breast cancer and its implications for endocrine therapy. J Clin Oncol 2005; 23:7721-35. [PMID: 16234531 DOI: 10.1200/jco.2005.09.004] [Citation(s) in RCA: 361] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The response to endocrine therapy in breast cancer correlates with estrogen receptor (ER) and progesterone receptor (PR) status. ER-positive/PR-negative breast cancers respond less well to selective ER modulator (SERM) therapy than ER-positive/PR-positive tumors. The predictive value of PR has long been attributed to the dependence of PR expression on ER activity, with the absence of PR reflecting a nonfunctional ER and resistance to hormonal therapy. However, recent clinical and laboratory evidence suggests that ER-positive/PR-negative breast cancers may be specifically resistant to SERMs, whereas they may be less resistant to estrogen withdrawal therapy with aromatase inhibitors, which is a result inconsistent with the nonfunctional ER theory. Novel alternative molecular mechanisms potentially explaining SERM resistance in ER-positive/PR-negative tumors have been suggested by recent experimental indications that growth factors may downregulate PR levels. Thus, the absence of PR may not simply indicate a lack of ER activity, but rather may reflect hyperactive cross talk between ER and growth factor signaling pathways that downregulate PR even as they activate other ER functions. Therefore, ER-positive/PR-negative breast tumors might best be treated by completely blocking ER action via estrogen withdrawal with aromatase inhibitors, by targeted ER degradation, or by combined therapy targeting both ER and growth factor signaling pathways. In this review, we will discuss the biology and etiology of ER-positive/PR-negative breast cancer, highlighting recent data on molecular cross talk between ER and growth factor signaling pathways and demonstrating how PR might be a useful marker of these activities. Finally, we will consider the clinical implications of these observations.
Collapse
Affiliation(s)
- Xiaojiang Cui
- Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
46
|
Osborne CK, Schiff R, Arpino G, Lee AS, Hilsenbeck VG. Endocrine responsiveness: understanding how progesterone receptor can be used to select endocrine therapy. Breast 2005; 14:458-65. [PMID: 16236516 DOI: 10.1016/j.breast.2005.08.024] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The receptor for the female hormone progesterone (PR), like that for estrogen (ER), is an important predictive marker for response to endocrine therapy in patients with breast cancer. PR exists as two isoforms, A and B. PR is important in mammary gland development and excess production of PRB is associated with breast cancer risk. Overabundance of PRA is related to resistance to tamoxifen. Total loss of PR is linked to reduced benefit from tamoxifen in both the adjuvant and metastatic settings. The predictive significance of PR expression was originally explained on the basis that PR is an ER-regulated gene and its presence indicates a functioning ER pathway and, therefore, an endocrine-responsive tumor. More recent data, however, suggest an alternative explanation. While many studies show that loss of PR predicts relative resistance to the antiestrogen tamoxifen, a recent study suggests that PR loss may not indicate resistance to aromatase inhibition. The finding that PR loss may not correlate with resistance to aromatase inhibition may be related to crosstalk between ER and PR and growth factor receptor pathways such as HER2. PR loss in some tumors is due to excessive growth factor receptor signaling (overexpression of HER2), which downregulates expression of the PR gene. Neoadjuvant studies also show that HER2 signaling is associated with tamoxifen resistance, but not resistance to aromatase inhibitors. Therefore, high HER2 signaling could explain both PR loss and resistance to tamoxifen while the response to aromatase inhibitors is maintained. In this way, PR loss in some tumors may be a surrogate marker for increased signaling through the growth factor receptor tyrosine kinase pathway and it may help clinicians decide between initial use of an aromatase inhibitor or tamoxifen in the individual patient.
Collapse
Affiliation(s)
- C Kent Osborne
- The Breast Center, Baylor College of Medicine, One Baylor Plaza, BCM 600, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
47
|
Abstract
Considerable data implicate estrogens in breast cancer carcinogenesis and progression. In the postmenopausal woman, estrogens are produced in breast tissues and many other sites throughout the body when androgen precursors are converted into estrogens via the enzyme aromatase. Inhibition of this enzyme with aromatase inhibitors (AIs) has demonstrated reductions in systemic as well as intratumoral estrogens. These drugs have now been utilized in large phase 3 randomized trials and have led to greater improved clinical benefit than the "gold standard," tamoxifen. Questions remain about the long-term side effects and safety profile of AIs. They are associated with increasing incidence of osteoporosis and bone fractures. Nevertheless, AIs add to our armamentarium for therapy and possible prevention of breast cancer.
Collapse
Affiliation(s)
- Cynthia Osborne
- University of Texas Southwestern Medical Center, Dallas, Texas 75390-8852, USA.
| | | |
Collapse
|
48
|
Howard-McNatt M, Hughes KS, Schnaper LA, Jones JL, Gadd M, Smith BL. Breast cancer treatment in older women. Surg Oncol Clin N Am 2005; 14:85-102, vi. [PMID: 15542001 DOI: 10.1016/j.soc.2004.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This article presents the author's current approaches to the management of breast cancer in older women, with emphasis on clinical and surgical treatment of the disease in this population. There are controversies surrounding the management of breast cancer in this population regarding adjuvant therapy, radiation therapy and surgical options. We endeavor to address these issues in the article.
Collapse
Affiliation(s)
- Marissa Howard-McNatt
- Division of Surgical Oncology, Massachusetts General Hospital, 100 Blossom Street, Cox 626, Boston, MA 02114, USA
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Estradiol, the most potent endogenous estrogen, is biosynthesized from androgens by the cytochrome P450 enzyme complex called aromatase. Aromatase is present in breast tissue, and intratumoral aromatase is the source of local estrogen production in breast cancer tissues. Inhibition of aromatase is an important approach for reducing growth-stimulatory effects of estrogens in estrogen-dependent breast cancer. Steroidal inhibitors that have been developed to date build upon the basic androstenedione nucleus and incorporate chemical substituents at varying positions on the steroid. Nonsteroidal aromatase inhibitors can be divided into three classes: aminoglutethimide-like molecules, imidazole/triazole derivatives, and flavonoid analogs. Mechanism-based aromatase inhibitors are steroidal inhibitors that mimic the substrate, are converted by the enzyme to a reactive intermediate, and result in the inactivation of aromatase. Both steroidal and nonsteroidal aromatase inhibitors have shown clinical efficacy in the treatment of breast cancer. The potent and selective third-generation aromatase inhibitors, anastrozole, letrozole, and exemestane, were introduced into the market as endocrine therapy in postmenopausal patients failing antiestrogen therapy alone or multiple hormonal therapies. These agents are currently approved as first-line therapy for the treatment of postmenopausal women with metastatic estrogen-dependent breast cancer. Several clinical studies of aromatase inhibitors are currently focusing on the use of these agents in the adjuvant setting for the treatment of early breast cancer. Use of an aromatase inhibitor as initial therapy or after treatment with tamoxifen is now recommended as adjuvant hormonal therapy for a postmenopausal woman with hormone-dependent breast cancer.
Collapse
Affiliation(s)
- Robert W Brueggemeier
- College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, Ohio 43210-1291, USA.
| | | | | |
Collapse
|
50
|
Abstract
Aromatase inhibitors are rapidly becoming the first choice for hormonal treatment of steroid receptor positive breast cancer in postmenopausal women. An understanding of the resistance mechanisms to these agents is, therefore, important for the appropriate delivery of treatment to responsive patients and the rational development of new agents targeted at the resistance pathways. De novo resistance appears to be a quantitative rather than qualitative phenomenon with virtually all oestrogen receptor positive tumours showing an anti-proliferative response to the aromatase inhibitor anastrozole. While the expression of type 1 growth factor receptors reduces response to tamoxifen this appears to have little detrimental effect on response to aromatase inhibitors. Studies of acquired resistance in vitro have indicated that acquisition of hypersensitivity to oestrogenic stimulation is a key mechanism that is dependent on enhanced cross-talk of growth factor and oestrogen signaling pathways. Collection of resistant biopsy tissues from patients is important to determine if this mechanism is clinically relevant.
Collapse
Affiliation(s)
- Mitch Dowsett
- Academic Department of Biochemistry, Royal Marsden Hospital, London SW3 6JJ, UK.
| | | | | | | |
Collapse
|