1
|
Masuyama M, Masuda N, Kawaguchi H, Yamamoto Y, Saji S, Nakayama T, Aogi K, Anan K, Ohtani S, Sato N, Takano T, Tokunaga E, Nakamura S, Hasegawa Y, Hattori M, Fujisawa T, Morita S, Yamaguchi M, Yamashita T, Yotsumoto D, Toi M, Ohno S. Fulvestrant with or without anti-HER2 therapy in patients in a postmenopausal hormonal state and with ER-positive HER2-positive advanced or metastatic breast cancer: A subgroup analysis of data from the Safari study (JBCRG-C06). Cancer Med 2023; 12:17718-17730. [PMID: 37525895 PMCID: PMC10523974 DOI: 10.1002/cam4.6390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/03/2023] [Accepted: 07/19/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND The role of endocrine therapy in the treatment of patients in a postmenopausal hormonal state and with estrogen receptor (ER)-positive, human epidermal growth factor receptor 2 (HER2)-positive advanced or metastatic breast cancer (AMBC) is unclear. METHODS We analyzed the data from 94 patients with ER-positive HER2-positive AMBC enrolled in the Safari study (UMIN000015168), a retrospective cohort study of 1072 ER-positive AMBC patients in a postmenopausal hormonal state who received fulvestrant 500 mg (F500): (1) to compare time to treatment failure (TTF) and overall survival (OS) by treatment group, and TTF by treatment line; (2) in patients who received endocrine therapy (including F500) or anti-HER2 therapy as initial systemic therapy before chemotherapy, to investigate relations between TTF for the first-line therapy or time to chemotherapy (TTC) and OS; (3) to investigate factors associated with OS. RESULTS The TTF was longer in the patients treated with F500 as first- or second-line therapy (n = 20) than in those who received later-line F500 therapy (n = 74) (6.6 vs. 3.7 months; HR, 1.98; p = 0.014). In the 59 patients who received endocrine therapy or anti-HER2 therapy as initial systemic therapy before chemotherapy, those with TTC ≥3 years had longer median OS than those with TTC <3 years (10.5 vs. 5.9 years; HR, 0.32; p = 0.001). Longer TTC was associated with prolonged OS. CONCLUSIONS In patients with ER-positive HER2-positive AMBC enrolled in the Safari study, TTF was longer in patients who received F500 as first- or second-line therapy. In patients who received chemotherapy-free initial systemic therapy, the prolonged OS in those with TTC ≥3 years suggests that this value may be a helpful cut-off for indicating clinical outcomes.
Collapse
Affiliation(s)
- Misato Masuyama
- Department of Breast and Endocrine Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Surgery, Breast OncologyNational Hospital Organization Osaka National HospitalOsakaJapan
| | - Norikazu Masuda
- Department of Breast and Endocrine SurgeryNagoya University Graduate School of MedicineNagoyaJapan
| | | | - Yutaka Yamamoto
- Department of Breast and Endocrine SurgeryKumamoto University HospitalKumamotoJapan
| | - Shigehira Saji
- Department of Medical OncologyFukushima Medical UniversityFukushimaJapan
| | - Takahiro Nakayama
- Department of Breast and Endocrine SurgeryOsaka International Cancer InstituteOsakaJapan
| | - Kenjiro Aogi
- Department of Breast OncologyNHO Shikoku Cancer CenterMatsuyamaJapan
| | - Keisei Anan
- Department of SurgeryKitakyushu Municipal Medical CenterKitakyushuJapan
| | - Shoichiro Ohtani
- Department of Breast SurgeryHiroshima City Hiroshima Citizens HospitalHiroshimaJapan
| | - Nobuaki Sato
- Department of Breast OncologyNiigata Cancer Center HospitalNiigataJapan
| | - Toshimi Takano
- Department of Medical OncologyToranomon HospitalTokyoJapan
| | - Eriko Tokunaga
- Department of Breast OncologyNHO Kyushu Cancer CenterFukuokaJapan
| | - Seigo Nakamura
- Department of Surgery, Division of Breast Surgical OncologyShowa University School of MedicineTokyoJapan
| | - Yoshie Hasegawa
- Department of Breast SurgeryHachinohe City Hospital, HachinoheJapan
| | - Masaya Hattori
- Department of Breast OncologyAichi Cancer Center HospitalNagoyaJapan
| | - Tomomi Fujisawa
- Department of Breast OncologyGunma Prefectural Cancer CenterOhtaJapan
| | - Satoshi Morita
- Department of Biomedical Statistics and BioinformaticsKyoto University Graduate School of MedicineKyotoJapan
| | - Miki Yamaguchi
- Department of Breast SurgeryJCHO Kurume General HospitalKurumeJapan
| | - Toshinari Yamashita
- Department of Breast Surgery and OncologyKanagawa Cancer CenterYokohamaJapan
| | - Daisuke Yotsumoto
- Department of Breast and Thyroid SurgeryHakuaikai Social Medical Corporation, Sagara HospitalKagoshimaJapan
| | - Masakazu Toi
- Department of Breast SurgeryKyoto University Graduate School of MedicineKyotoJapan
| | - Shinji Ohno
- Breast Oncology CenterThe Cancer Institute Hospital of JFCRTokyoJapan
| |
Collapse
|
2
|
Torrisi R, Palumbo R, De Sanctis R, Vici P, Bianchi GV, Cortesi L, Leonardi V, Gueli R, Fabi A, Valerio MR, Gambaro AR, Tagliaferri B, Pizzuti L, Cazzaniga ME, Santoro A. Fulvestrant and trastuzumab in patients with luminal HER2-positive advanced breast cancer (ABC): an Italian real-world experience (HERMIONE 9). Breast Cancer Res Treat 2021; 190:103-109. [PMID: 34453206 DOI: 10.1007/s10549-021-06371-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/20/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE The most appropriate therapy for HR + /HER2-positive (HER2 +) advanced breast cancer (ABC) is a matter of debate. Co-targeting of both receptors represents an attractive strategy to overcome the cross-talk between them. METHODS The HERMIONE 9 is an observational retrospective multicentric study which aimed to describe the clinical outcome of patients with HR + /HER2 + ABC who received the combination of Fulvestrant (F) and Trastuzumab (T) as part of their routine treatment at 10 Italian Institutions. RESULTS Eighty-seven patients were included. Median age was 63 (range, 35-87) years. The median number of previous treatments was 3 (range, 0-10) and F and T were administered as ≥ 3rd line in 67 patients. Among the 86 evaluable patients, 6 (6.9%) achieved CR, 18 (20.7%) PR, and 44 (50.6%) had SD ≥ 24 weeks with an overall CBR of 78.2%. At a median follow-up of 33.6 months, mPFS of the entire cohort was 12.9 months (range, 2.47-128.67). No difference was observed in mPFS between patients treated after progression or as maintenance therapy (mPFS 12.9 and 13.9 months in 64 and 23 patients, respectively), neither considering the number of previous treatment lines (≤ 3 or < 3). CONCLUSION The combination of F and T was active in this cohort at poor prognosis and deserves further investigations possibly in combination with pertuzumab in patients with high ER expression.
Collapse
Affiliation(s)
- Rosalba Torrisi
- Department of Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, via A. Manzoni 56 20089, Rozzano, Milano, Italy.
| | | | - Rita De Sanctis
- Department of Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, via A. Manzoni 56 20089, Rozzano, Milano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Patrizia Vici
- UOC OM2 IRCCS Regina Elena National Cancer Institute, Roma, Italy
| | | | - Laura Cortesi
- Department of Oncology and Haematology, Modena Hospital University, Modena, Italy
| | - Vita Leonardi
- Department of Medical Oncology, ARNAS Civico, Palermo, Italy
| | | | - Alessandra Fabi
- Precision Medicine in Breast Cancer Unit, Scientific Directorate, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Maria Rosaria Valerio
- Medical Oncology Unit, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Anna Rita Gambaro
- Medical Oncology, ASST Fatebenefratelli Sacco PO Sacco, Milano, Italy
| | | | - Laura Pizzuti
- UOC OM2 IRCCS Regina Elena National Cancer Institute, Roma, Italy
| | - Marina Elena Cazzaniga
- Centro Ricerca Fase 1 ASST Monza and Università Degli Studi Milano Bicocca, Milano, Italy
| | - Armando Santoro
- Department of Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, via A. Manzoni 56 20089, Rozzano, Milano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| |
Collapse
|
3
|
McDonnell DP, Wardell SE, Chang CY, Norris JD. Next-Generation Endocrine Therapies for Breast Cancer. J Clin Oncol 2021; 39:1383-1388. [PMID: 33705209 DOI: 10.1200/jco.20.03565] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC
| | - Suzanne E Wardell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC
| | - John D Norris
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC
| |
Collapse
|
4
|
Rusz O, Kószó R, Dobi Á, Csenki M, Valicsek E, Nikolényi A, Uhercsák G, Cserháti A, Kahán Z. Clinical benefit of fulvestrant monotherapy in the multimodal treatment of hormone receptor and HER2 positive advanced breast cancer: a case series. Onco Targets Ther 2018; 11:5459-5463. [PMID: 30233207 PMCID: PMC6129034 DOI: 10.2147/ott.s170736] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Fulvestrant is a pure estrogen receptor (ER) antagonist approved for the treatment of metastatic ER positive breast cancer in postmenopausal women with disease progression following antiestrogen therapy. The clinical results of fulvestrant demonstrated encouraging activity in tumors in spite of HER2 positivity, but data about its use after progression on anti-HER2 agents are limited. Partial responses and durations of response of 12, 25, and 38 months in three cases with multiple metastases of ER positive and HER2 positive breast cancer were observed; all patients had been treated with 1–4 regimens of an anti-HER2 agent in combination with chemotherapy or an aromatase inhibitor before the initiation of fulvestrant. Fulvestrant is a valuable option with limited toxicity and durable response in metastatic HER2 and ER positive breast cancer after progression on anti-HER2 agents as well. Therapeutic benefit even in extensive skin metastases and (irradiated) brain metastases may be expected. Further investigations are warranted to establish where it fits into the multimodal management of ER and HER positive breast cancer.
Collapse
Affiliation(s)
- Orsolya Rusz
- Department of Oncotherapy, University of Szeged, Szeged, Hungary,
| | - Renáta Kószó
- Department of Oncotherapy, University of Szeged, Szeged, Hungary,
| | - Ágnes Dobi
- Department of Oncotherapy, University of Szeged, Szeged, Hungary,
| | - Melinda Csenki
- Department of Oncotherapy, University of Szeged, Szeged, Hungary,
| | | | - Alíz Nikolényi
- Department of Oncotherapy, University of Szeged, Szeged, Hungary,
| | | | | | - Zsuzsanna Kahán
- Department of Oncotherapy, University of Szeged, Szeged, Hungary,
| |
Collapse
|
5
|
Rocca A, Maltoni R, Bravaccini S, Donati C, Andreis D. Clinical utility of fulvestrant in the treatment of breast cancer: a report on the emerging clinical evidence. Cancer Manag Res 2018; 10:3083-3099. [PMID: 30214302 PMCID: PMC6124791 DOI: 10.2147/cmar.s137772] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Fulvestrant is the first selective estrogen receptor (ER) downregulator available in clinical practice. It is a pure antiestrogen with no agonistic effects, leading to degradation of ER alpha, with activity in tamoxifen-resistant breast cancer (BC) models. Pharmacokinetic and pharmacodynamic studies and several postmarketing clinical trials led to the definition of the optimal dose at 500 mg intramuscularly on days 1, 15, and 29 and then every 28 days. Targeting ER alpha, fulvestrant is a cornerstone of treatment in luminal BCs, whose growth is largely driven by the ER pathway. In endocrine therapy-naïve patients with hormone receptor-positive, HER2− advanced BC (ABC), fulvestrant yielded significantly longer progression-free survival compared to anastrozole in the Phase III FALCON study. Due to its mechanism of action and pharmacokinetic properties, fulvestrant is an ideal backbone for combination therapies. Preclinical studies have shown synergism with drugs acting on signaling pathways involved in the development of endocrine resistance, among which the cyclin D/cyclin-dependent kinase 4-6/retinoblastoma pathway and the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin pathway, contributing to overcoming or delaying endocrine resistance. In the Phase III PALOMA-3 trial, a combination of the cyclin-dependent kinase 4/6 inhibitor palbociclib with fulvestrant significantly improved progression-free survival over fulvestrant alone in women with hormone receptor positive, HER2− ABC progressing during prior endocrine therapy. This led to approval of the combination in this clinical setting. Similar results were obtained with abemaciclib and ribociclib. Combination with pan-PI3K inhibitors, though showing some efficacy, was hampered by the toxicity of these agents, and studies in combinations with more selective inhibitors of the α-catalytic subunit of PI3K are ongoing. Fulvestrant has shown partial activity also in patients with tumors harboring mutations of the ESR1 gene. It is thus a key drug in the treatment of ABC, whose role in combination with new targeted agents is still evolving.
Collapse
Affiliation(s)
- Andrea Rocca
- Breast Cancer Unit, Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS,
| | - Roberta Maltoni
- Breast Cancer Unit, Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS,
| | - Sara Bravaccini
- Cyto-Histo-Molecular Pathology, Bioscience Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS
| | - Caterina Donati
- Pharmacy, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS
| | - Daniele Andreis
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
6
|
Chen Q, Weng Z, Lu Y, Jia Y, Ding L, Bai F, Ge M, Lin Q, Wu K. An Experimental Analysis of the Molecular Effects of Trastuzumab (Herceptin) and Fulvestrant (Falsodex), as Single Agents or in Combination, on Human HR+/HER2+ Breast Cancer Cell Lines and Mouse Tumor Xenografts. PLoS One 2017; 12:e0168960. [PMID: 28045951 PMCID: PMC5207527 DOI: 10.1371/journal.pone.0168960] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/08/2016] [Indexed: 11/28/2022] Open
Abstract
PURPOSE To investigate the effects of trastuzumab (herceptin) and fulvestrant (falsodex) either in combination or alone, on downstream cell signaling pathways in lab-cultured human HR+/HER2+ breast cancer cell lines ZR-75-1 and BT-474, as well as on protein expression levels in mouse xenograft tissue. METHODS Cells were cultivated in the presence of trastuzumab or fulvestrant or both. Molecular events that resulted in an inhibition of cell proliferation and cell cycle progression or in an increased rate of apoptosis were studied. The distribution and abundance of the proteins p-Akt and p-Erk expressed in these cells in response to single agents or combinatorial treatment were also investigated. In addition, the effects of trastuzumab and fulvestrant, either as single agents or in combination on tumor growth as well as on expression of the protein p-MED1 expressed in in vivo mouse xenograft models was also examined. RESULTS Cell proliferation was increasingly inhibited by trastuzumab or fulvestrant or both, with a CI<1 and DRI>1 in both human cell lines. The rate of apoptosis increased only in the BT-474 cell line and not in the ZR-75-1 cell line upon treatment with fulvestrant and not trastuzumab as a single agent (P<0.05). Interestingly, fulvestrant, in combination with trastuzumab, did not significantly alter the rate of apoptosis (in comparison with fulvestrant alone), in the BT-474 cell line (P>0.05). Cell accumulation in the G1 phase of cell cycle was investigated in all treatment groups (P<0.05), and the combination of trastuzumab and fulvestrant reversed the effects of fulvestrant alone on p-Akt and p-Erk protein expression levels. Using ZR-75-1 or BT-474 to generate in vivo tumor xenografts in BALB/c athymic mouse models, we showed that a combination of both drugs resulted in a stronger inhibition of tumor growth (P<0.05) and a greater decrease in the levels of activated MED1 (p-MED1) expressed in tumor issues compared with the use of either drug as a single agent. CONCLUSIONS We demonstrate that the administration of trastuzumab and fulvestrant in combination results in positive synergistic effects on both, ZR-75-1 and BT-474 cell lines. This combinatorial approach is likely to reduce physiological side effects of both drugs, thus providing a theoretical basis for the use of such combination treatment in order to resolve HR+/HER2+ triple positive breast cancer that has previously been shown to be resistant to endocrine treatment alone.
Collapse
Affiliation(s)
- Qing Chen
- Department of General Surgery, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyi Weng
- Department of General Surgery, Shanghai International Medical Center, Shanghai, China
| | - Yunshu Lu
- Department of General Surgery, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yijun Jia
- Department of General Surgery, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Longlong Ding
- Department of General Surgery, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Bai
- Department of General Surgery, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meixin Ge
- Department of General Surgery, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Lin
- Department of Radiation Oncology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Kejin Wu
- Department of Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
7
|
Agrawal A, Robertson JFR, Cheung KL, Gutteridge E, Ellis IO, Nicholson RI, Gee JMW. Biological effects of fulvestrant on estrogen receptor positive human breast cancer: short, medium and long-term effects based on sequential biopsies. Int J Cancer 2015; 138:146-59. [PMID: 26178788 PMCID: PMC4879515 DOI: 10.1002/ijc.29682] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 06/07/2015] [Accepted: 06/17/2015] [Indexed: 02/06/2023]
Abstract
We report the first study of the biological effect of fulvestrant on ER positive clinical breast cancer using sequential biopsies through to progression. Thirty-two locally/systemically advanced breast cancers treated with first-line fulvestrant (250 mg/month) were biopsied at therapy initiation, 6 weeks, 6 months and progression and immunohistochemically-analyzed for Ki67, ER, EGFR and HER2 expression/signaling activity. This series showed good fulvestrant responses (duration of response [DoR] = 25.8 months; clinical benefit = 81%). Ki67 fell (p < 0.001) in 79% of tumours by 6 months and lower Ki67 at all preprogression time-points predicted for longer DoR. ER and PR significantly decreased in all tumours by 6 months (p < 0.001), with some declines in ER (serine 118) phosphorylation and Bcl-2 (p = 0.007). There were modest HER2 increases (p = 0.034, 29% tumours) and loss of any detectable EGFR phosphorylation (p = 0.024, 50% tumours) and MAP kinase (ERK1/2) phosphorylation (p = 0.019, 65% tumours) by 6 months. While ER remained low, there was some recovery of Ki67, Bcl-2 and (weakly) EGFR/MAPK activity in 45-67% patients at progression. Fulvestrant's anti-proliferative impact is related to DoR, but while commonly downregulating ER and indicators of its signaling and depleting EGFR/MAPK signaling in some patients, additional elements must determine response duration. Residual ER at fulvestrant relapse explains reported sensitivity to further endocrine therapies. Occasional modest treatment-induced HER2 and weakly detectable EGFR/HER2/MAPK signaling at relapse suggests targeting of such activity might have value alongside fulvestrant in some patients. However, unknown pathways must drive relapse in most. Ki67 has biomarker potential to predict fulvestrant outcome and as a quantitative measure of response.
Collapse
Affiliation(s)
- Amit Agrawal
- Division of Breast Surgery, School of Graduate Entry Medicine and Health, Royal Derby Hospital, University of Nottingham, Derby, DE22 3DT, United Kingdom.,Cambridge Breast Unit, Cambridge University Hospitals, Cambridge, CB2 0QQ, United Kingdom
| | - John F R Robertson
- Division of Breast Surgery, School of Graduate Entry Medicine and Health, Royal Derby Hospital, University of Nottingham, Derby, DE22 3DT, United Kingdom
| | - Kwok L Cheung
- Division of Breast Surgery, School of Graduate Entry Medicine and Health, Royal Derby Hospital, University of Nottingham, Derby, DE22 3DT, United Kingdom
| | - Eleanor Gutteridge
- University of Nottingham, Nottingham City Hospital, NG5 1PB, United Kingdom
| | - Ian O Ellis
- University of Nottingham, Nottingham City Hospital, NG5 1PB, United Kingdom
| | - Robert I Nicholson
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, CF10 3NB, United Kingdom
| | - Julia M W Gee
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, CF10 3NB, United Kingdom
| |
Collapse
|
8
|
Gee JMW, Nicholson RI, Barrow D, Dutkowski CM, Goddard L, Jordan NJ, McClelland RA, Knowlden JM, Francies HE, Hiscox SE, Hutcheson IR. Antihormone induced compensatory signalling in breast cancer: an adverse event in the development of endocrine resistance. Horm Mol Biol Clin Investig 2015; 5:67-77. [PMID: 25961242 DOI: 10.1515/hmbci.2011.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 01/21/2011] [Indexed: 12/16/2022]
Abstract
Using MCF7 breast cancer cells, it has been shown that antihormones promote expression/activity of oestrogen-repressed tyrosine kinases, notably EGFR, HER2 and Src. These inductive events confer responsiveness to targeted inhibitors (e.g., gefitinib, trastuzumab, saracatinib). We observed that these antihormone-induced phenomena are common to ER+HER2- and ER+HER2+ breast cancer models in vitro, where targeting of EGFR, HER2 or Src alongside antihormone improves antitumour response and delays/prevents endocrine resistance. Such targeted inhibitors also subvert acquired endocrine resistant cells which retain increased EGFR, HER2 and Src (e.g., TAMR and FASR models derived after 6-12 months of tamoxifen or Faslodex treatment). Thus, antihormone-induced tyrosine kinases comprise "compensatory signalling" crucial in limiting maximal initial antihormone response and subsequently driving acquired resistance in vitro. However, despite such convincing preclinical findings from our group and others, clinical trials examining equivalent antigrowth factor strategies have proved relatively disappointing. Our new studies deciphering underlying causes reveal that further antihormone-promoted events could be pivotal in vivo. Firstly, Faslodex induces HER3 and HER4 which sensitise ER+ cells to heregulin, a paracrine growth factor that overcomes endocrine response and diminishes antitumour effect of agents targeting EGFR, HER2 or Src alongside antihormone. Secondly, extended antihormone exposure (experienced by ER+ cells prior to adjuvant clinical relapse) can "reprogramme" the compensatory kinase profile in vitro, hindering candidate antigrowth factor targeting of endocrine resistance. Faslodex resistant cells maintained with this antihormone for 3 years in vitro lose EGFR/HER2 dependency, gaining alternative mitogenic/invasion kinases. Deciphering these previously unrecognised antihormone-induced events could provide superior treatments to control endocrine relapse in the clinic.
Collapse
|
9
|
Senkus E, Cardoso F, Pagani O. Time for more optimism in metastatic breast cancer? Cancer Treat Rev 2014; 40:220-8. [DOI: 10.1016/j.ctrv.2013.09.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/21/2013] [Accepted: 09/24/2013] [Indexed: 10/26/2022]
|
10
|
Mendoza-Parra MA, Nowicka M, Van Gool W, Gronemeyer H. Characterising ChIP-seq binding patterns by model-based peak shape deconvolution. BMC Genomics 2013; 14:834. [PMID: 24279297 PMCID: PMC4046686 DOI: 10.1186/1471-2164-14-834] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 11/20/2013] [Indexed: 12/01/2022] Open
Abstract
Background Chromatin immunoprecipitation combined with massive parallel sequencing (ChIP-seq) is widely used to study protein-chromatin interactions or chromatin modifications at genome-wide level. Sequence reads that accumulate locally at the genome (peaks) reveal loci of selectively modified chromatin or specific sites of chromatin-binding factors. Computational approaches (peak callers) have been developed to identify the global pattern of these sites, most of which assess the deviation from background by applying distribution statistics. Results We have implemented MeDiChISeq, a regression-based approach, which - by following a learning process - defines a representative binding pattern from the investigated ChIP-seq dataset. Using this model MeDiChISeq identifies significant genome-wide patterns of chromatin-bound factors or chromatin modification. MeDiChISeq has been validated for various publicly available ChIP-seq datasets and extensively compared with other peak callers. Conclusions MeDiChI-Seq has a high resolution when identifying binding events, a high degree of peak-assessment reproducibility in biological replicates, a low level of false calls and a high true discovery rate when evaluated in the context of gold-standard benchmark datasets. Importantly, this approach can be applied not only to ‘sharp’ binding patterns - like those retrieved for transcription factors (TFs) - but also to the broad binding patterns seen for several histone modifications. Notably, we show that at high sequencing depths, MeDiChISeq outperforms other algorithms due to its powerful peak shape recognition capacity which facilitates discerning significant binding events from spurious background enrichment patterns that are enhanced with increased sequencing depths. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-14-834) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marco-Antonio Mendoza-Parra
- Equipe Labellisée Ligue Contre le Cancer, Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC)/CNRS/INSERM/Université de Strasbourg, BP 10142, Illkirch Cedex 67404, France.
| | | | | | | |
Collapse
|
11
|
Chen HX, Sharon E. IGF-1R as an anti-cancer target--trials and tribulations. CHINESE JOURNAL OF CANCER 2013; 32:242-52. [PMID: 23601239 PMCID: PMC3845553 DOI: 10.5732/cjc.012.10263] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 02/19/2013] [Accepted: 03/15/2013] [Indexed: 12/23/2022]
Abstract
Type I insulin-like growth factor receptor (IGF-1R) has long been recognized for its role in tumorigenesis and growth, but only recently have the tools for targeting the IGF pathway become available. More than 10 IGF/IGF-1R inhibitors have entered clinical trials, and these belong to three main classes: (1) monoclonal antibodies against IGF-1R, (2) monoclonal antibodies against IGF-1R ligands (IGF-1 and IGF-2), and (3) IGF-1R tyrosine kinase inhibitors. These IGF-1R-targeting agents share common effects on IGF-1R signaling but differ in mechanisms of action, spectrum of target inhibition, and pharmacological features. Clinical activity of IGF-1R inhibitors has been demonstrated with sustained responses in a small number of patients with select tumor types, such as Ewing sarcoma and thymoma. However, many large clinical trials involving patients with adult tumors, including non-small cell lung cancer, breast cancer, and pancreatic cancer, failed to show clinical benefit in the overall patient population. Possible reasons for failure include the complexity of the IGF-1R/insulin receptor system and parallel growth and survival pathways, as well as a lack of patient selection markers. While IGF-1R remains a valid target for selected tumor types, identification of predictive markers and rational combinations will be critical to success in future development.
Collapse
Affiliation(s)
- Helen X Chen
- National Cancer Institute, Bethesda, MD 20892, USA.
| | | |
Collapse
|
12
|
Giuliano M, Schifp R, Osborne CK, Trivedi MV. Biological mechanisms and clinical implications of endocrine resistance in breast cancer. Breast 2012; 20 Suppl 3:S42-9. [PMID: 22015292 DOI: 10.1016/s0960-9776(11)70293-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Endocrine therapy represents the first and most efficacious targeted treatment for women with estrogen receptor-positive (ER+) breast cancer. In the last four decades several hormonal agents have been successfully introduced in clinical practice as both palliative therapy for advanced disease and adjuvant treatment for prevention of tumor relapse. Nevertheless, the intrinsic and acquired resistance occurs in a significant proportion of patients, limiting the efficacy of endocrine treatments. Several molecular mechanisms have been proposed to be responsible for endocrine resistance. Loss of ER expression, altered activity of ER coregulators, deregulation of apoptosis and cell cycle signaling, and hyperactive receptor tyrosine kinase (RTK) and stress/cell kinase pathways can collectively orchestrate the development and sustenance of pharmacologic resistance to endocrine therapy. Preclinical and clinical evidence documents the plasticity in ER expression levels and signaling. As such, ER can either drive gene transcription and tumor progression directly or crosstalk with alternate RTK and cellular kinase signaling pathways, resulting in modulation of its own expression levels and transcriptional program. For this reason a therapeutic approach based on the combination of agents targeting both ER and RTK signaling represents a promising strategy to be tested. Among many RTKs, EGFR, HER2, and PI3K have been found to be viable targets for the combination therapy strategy, at least in the preclinical setting. However, early results from clinical trials testing combination strategies have been discordant, suggesting the need for better approaches to simultaneously inhibit multiple escape pathways and to select the patients who may benefit the most from these strategies.
Collapse
Affiliation(s)
- Mario Giuliano
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
13
|
Bartsch R, Bago-Horvath Z, Berghoff A, DeVries C, Pluschnig U, Dubsky P, Rudas M, Mader RM, Rottenfusser A, Fitzal F, Gnant M, Zielinski CC, Steger GG. Ovarian function suppression and fulvestrant as endocrine therapy in premenopausal women with metastatic breast cancer. Eur J Cancer 2012; 48:1932-8. [PMID: 22459763 DOI: 10.1016/j.ejca.2012.03.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 12/29/2011] [Accepted: 03/03/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Endocrine therapy is the preferred treatment for hormone-receptor (HR) positive metastatic breast cancer. In premenopausal patients, ovarian function suppression with goserelin in combination with anastrozole yielded promising results in phase II studies. Fulvestrant, a pure antioestrogen, yields high rates of disease stabilisation in postmenopausal women. Therefore, we investigated the feasibility and safety of fulvestrant plus goserelin in premenopausal women with HR-positive metastatic breast cancer. METHODS Premenopausal patients with metastatic breast cancer eligible for endocrine treatment received fulvestrant 250 mg and goserelin 3.6 mg every four weeks as first- to fourth-line therapy. Clinical benefit rate (CBR; response rate plus disease stabilisation ≥ 6 months) was defined as the primary study end-point. Time to progression (TTP) and overall survival (OS) were estimated using the Kaplan-Meier product limit method. FINDINGS Twenty-six patients received treatment as scheduled. 81% were pre-treated with tamoxifen and 69% had received prior aromatase inhibitors in combination with goserelin. The majority of patients (69%) presented with visceral metastases. Complete response was observed in a single patient, partial response in three and disease stabilisation ≥ 6 months in eleven patients, resulting in a CBR of 58%. Median TTP was 6 months (95%confidence interval (CI), 2.4-9.6) and OS 32 months (95%CI, 14.28-49.72), respectively. INTERPRETATION Results suggest that the combination of fulvestrant and goserelin offers promising activity in premenopausal patients and further investigation is warranted.
Collapse
Affiliation(s)
- Rupert Bartsch
- Comprehensive Cancer Center Vienna, Medical University of Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Krell J, Januszewski A, Yan K, Palmieri C. Role of fulvestrant in the management of postmenopausal breast cancer. Expert Rev Anticancer Ther 2012; 11:1641-52. [PMID: 22050013 DOI: 10.1586/era.11.138] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fulvestrant is a form of endocrine therapy used in the treatment of postmenopausal breast cancer. It has a unique mechanism of action in that it causes the degradation of estrogen receptor and therefore has been labeled a selective estrogen receptor downregulator. Unlike the selective estrogen receptor modulator tamoxifen, it has no agonistic properties and is therefore a pure anti-estrogen. Given its low level of bioavailability and presystemic metabolism, it has been formulated as an intramuscular injection. A number of dosing regimens have been utilized - these include a dose of 250 mg monthly ('approved dose'), an initial 500 mg followed by 250 mg on days 14 and 28, and thereafter 250 mg every 28 days ('loading dose'), or 500 mg on days 0, 14 and 28, and thereafter every 28 days ('high dose'). This article will review its unique mode of action and preclinical data, as well as clinical data for different dosing regimens and data for its combination with aromatase inhibitors. Fulvestrant is a well-tolerated drug and its toxicities will also be reviewed. The optimal position of fulvestrant in sequential endocrine therapy has yet to be defined.
Collapse
Affiliation(s)
- Jonathan Krell
- Department of Surgery and Cancer, MRC Cyclotron Building, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 ONN, UK
| | | | | | | |
Collapse
|
15
|
First-line endocrine therapy alone could be a reasonable treatment option for hormone-positive, HER2-positive metastatic breast cancer. Bull Cancer 2012; 99:E18-25. [DOI: 10.1684/bdc.2011.1537] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
16
|
Oakman C, Moretti E, Santarpia L, Di Leo A. Fulvestrant in the management of postmenopausal women with advanced, endocrine-responsive breast cancer. Future Oncol 2011; 7:173-86. [PMID: 21345137 DOI: 10.2217/fon.10.179] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Fulvestrant is a pure estrogen antagonist that binds, blocks and downgrades the estrogen receptor (ER). Its unique mechanism of action is its antitumor activity after progression on prior endocrine therapy. Fulvestrant has shown activity in ER-dependent cells that are ligand independent. Fulvestrant has been approved at 250 mg/month for postmenopausal women with hormone-sensitive advanced breast cancer after progression or recurrence on antiestrogen therapy. The fulvestrant 500 mg regimen has just received approval by the EMA and the US FDA, supported by dose-dependent ER downregulation and the recent results of the clinical trial CONFIRM. Fulvestrant in combination with systemic lowering of estrogen has shown no improvement over fulvestrant alone. Combination therapy with inhibitors of growth factor signaling may have greater efficacy and is under exploration. To enhance the benefit of fulvestrant and improve outcomes for individuals with ER-positive breast cancer, greater understanding of resistance mechanisms is required. A key issue is identification of patients with ER-positive disease who retain sensitivity to antiestrogen therapy after progression on tamoxifen and/or aromatase inhibitors.
Collapse
Affiliation(s)
- Catherine Oakman
- Sandro Pitigliani Medical Oncology Unit, Hospital of Prato, Istituto Toscano Tumori, Piazza Ospedale 2, Prato, Italy
| | | | | | | |
Collapse
|
17
|
McDonnell DP, Wardell SE. The molecular mechanisms underlying the pharmacological actions of ER modulators: implications for new drug discovery in breast cancer. Curr Opin Pharmacol 2011; 10:620-8. [PMID: 20926342 DOI: 10.1016/j.coph.2010.09.007] [Citation(s) in RCA: 145] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 09/07/2010] [Accepted: 09/10/2010] [Indexed: 01/20/2023]
Abstract
Our understanding of the molecular mechanisms underlying the pharmacological actions of estrogen receptor (ER) ligands has evolved considerably in recent years. Much of this knowledge has come from a detailed dissection of the mechanism(s) of action of the Selective Estrogen Receptor Modulators (SERMs) tamoxifen and raloxifene, so called for their ability to function as ER agonists or antagonists depending on the tissue in which they operate. These mechanistic insights have had a significant impact on the discovery of second generation SERMs, some of which are in late stage clinical development for the treatment/prevention of breast cancer as well as other estrogenopathies. In addition to the SERMs, however, have emerged the Selective Estrogen Degraders (SERDs), which as their name suggests, interact with and facilitate ER turnover in cells. One drug of this class, fulvestrant, has been approved as a third line treatment for ER-positive metastatic breast cancer. Whereas the first generation SERMs/SERDs were discovered in a serendipitous manner, this review will highlight how our understanding of the molecular pharmacology of ER ligands has been utilized in the development of the next generation of SERMs/SERDs, some of which are likely to have a major impact on the pharmacotherapy of breast cancer.
Collapse
Affiliation(s)
- Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA.
| | | |
Collapse
|
18
|
Doroshow JH. Selecting Systemic Cancer Therapy One Patient at a Time: Is There a Role for Molecular Profiling of Individual Patients With Advanced Solid Tumors? J Clin Oncol 2010; 28:4869-71. [DOI: 10.1200/jco.2010.31.1472] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- James H. Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
19
|
Lupien M, Meyer CA, Bailey ST, Eeckhoute J, Cook J, Westerling T, Zhang X, Carroll JS, Rhodes DR, Liu XS, Brown M. Growth factor stimulation induces a distinct ER(alpha) cistrome underlying breast cancer endocrine resistance. Genes Dev 2010; 24:2219-27. [PMID: 20889718 DOI: 10.1101/gad.1944810] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Estrogen receptor α (ERα) expression in breast cancer is predictive of response to endocrine therapy; however, resistance is common in ERα-positive tumors that overexpress the growth factor receptor ERBB2. Even in the absence of estrogen, ERα can be activated by growth factors, including the epidermal growth factor (EGF). EGF induces a transcriptional program distinct from estrogen; however, the mechanism of the stimulus-specific response is unknown. Here we show that the EGF-induced ERα genomic targets, its cistromes, are distinct from those induced by estrogen in a process dependent on the transcription factor AP-1. The EGF-induced ERα cistrome specifically regulates genes found overexpressed in ERBB2-positive human breast cancers. This provides a potential molecular explanation for the endocrine therapy resistance seen in ERα-positive breast cancers that overexpress ERBB2. These results suggest a central role for ERα in hormone-refractory breast tumors dependent on growth factor pathway activation and favors the development of therapeutic strategies completely antagonizing ERα, as opposed to blocking its estrogen responsiveness alone.
Collapse
Affiliation(s)
- Mathieu Lupien
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Streckfus CF, Brown RE, Bull JM. Proteomics, morphoproteomics, saliva and breast cancer: An emerging approach to guide the delivery of individualised thermal therapy, thermochemotherapy and monitor therapy response. Int J Hyperthermia 2010; 26:649-61. [DOI: 10.3109/02656736.2010.506470] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|