1
|
Parsonson AO, Sarkar S, Brown L, Low GKK, Kiely BE, Vasista A. Beyond the median: Estimating survival times for patients starting endocrine therapy for estrogen receptor-positive, metastatic breast cancer from recent randomized trials. Asia Pac J Clin Oncol 2024; 20:714-722. [PMID: 38881159 DOI: 10.1111/ajco.14096] [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: 09/18/2023] [Revised: 04/19/2024] [Accepted: 05/27/2024] [Indexed: 06/18/2024]
Abstract
AIM To estimate scenarios for survival for patients with estrogen receptor (ER) positive, metastatic breast cancer (MBC) and to help communicate prognosis to patients starting endocrine therapy (ET) METHODS: We searched for randomized trials of ET for ER-positive MBC and extracted the following percentiles (representative survival scenarios) from each overall survival (OS) curve: 90th (worst-case), 75th (lower-typical), 50th (median), 25th (upper-typical), and 10th (best-case). We then assessed the accuracy of estimating these percentiles for each OS curve by multiplying the median OS by four simple multiples: 0.25 (to estimate the 90th percentile), 0.5 (75th), 2 (25th), and 3 (10th). Estimates were deemed accurate if it fell within 0.75-1.33 times the actual value. RESULTS We identified 25 trials with 10,566 patients. The median OS (interquartile range) was: 61.3 months (53.4-64.8) for first-line ET with cyclin-dependant kinase 4/6 inhibitors (four treatment groups); 42.6 months (40.9-50.4) for first-line ET alone (21 treatment groups) and 29.2 months (24.8-33.4) for subsequent line ET (19 treatment groups). Simple multiples of the median OS accurately estimated the 90th percentile in 80%; 75th percentile in 93%; and 25th percentile in 76% of curves. The 10th percentile was only available for four OS curves and could not be evaluated. CONCLUSION Simple multiples of the median OS are a helpful and accurate method to assist in estimating and discussing scenarios for survival for MBC patients starting ET. Longer follow-up of trials is required to help clinicians estimate the best-case scenario.
Collapse
Affiliation(s)
- Andrew O Parsonson
- Nepean Cancer and Wellness Centre, Nepean Hospital, Sydney, Australia
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Sunit Sarkar
- Department of Medical Oncology, Royal Hobart Hospital, Hobart, Australia
| | - Lauren Brown
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, Australia
| | - Gary K K Low
- Research Operations, Nepean Hospital, Sydney, Australia
| | - Belinda E Kiely
- Macarthur Cancer Therapy Centre, Campbelltown Hospital, Sydney, Australia
| | - Anuradha Vasista
- Nepean Cancer and Wellness Centre, Nepean Hospital, Sydney, Australia
| |
Collapse
|
2
|
Okwor V, Okwor CJ, Ukwuoma M, Nweke M. Effectiveness of combined targeted and hormonal therapies for post-menopausal women with hormone receptor-positive and HER2-negative advanced breast cancer: A systematic review and meta-analysis of RCTs. J Oncol Pharm Pract 2024:10781552241279019. [PMID: 39295518 DOI: 10.1177/10781552241279019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
OBJECTIVE we aim to synthesize available evidence on the effectiveness of hormonal plus targeted therapies for post-menopausal women with hormone receptor-positive and HER2-negative advanced breast cancer. DATA SOURCES AND METHODS We searched the following databases: Medline, PubMed, Cochrane Library, CINAHL, Web of Science, Scopus, and African Journal. Only studies that investigated the effectiveness of hormonal therapy combined with targeted therapy for HR+/HER2- advanced breast cancer treatment were included. The outcomes were progression-free survival (PFS), overall survival (OS) and objective response rate (ORR). A random-effect meta-analysis model was employed. Statistical analysis was performed using Comprehensive Meta-analysis version 3. RESULTS 24 studies were included in the meta-analysis with an overall sample size of 7635. Median PFS, OS and ORR were found to be significantly increased in the combination group compared to hormonal monotherapy [SMD = 6.072 (95% CI = 3.785-8.360), p < 0.001], [SMD = 1.614 (95% CI = 0.139-3.089), p = 0.032] and [OR = 1.584 (CI 1.134-2.213), p = 0.007] respectively. Subgroup analysis showed a significant difference in PFS and ORR between patients who received "hormonal therapy + CDK4/6 inhibitors" vs hormonal therapy only [SMD = 6.015 (CI 3.069-8.960), p < 0.001], (OR = 1.828 (CI 1.030-3.243), p = 0.039] respectively. CONCLUSION Compared with hormonal monotherapy, targeted plus hormonal therapy significantly improves PFS, OS and ORR in postmenopausal women with HR+/HER2- advanced breast cancer.
Collapse
Affiliation(s)
- Vitalis Okwor
- Department of Radiation and Clinical Oncology, University of Nigeria Teaching Hospital, Enugu, Nigeria
| | - Chika Juliet Okwor
- Department of Chemical Pathology, College of Medicine, University of Nigeria Ituku-Ozalla Campus, Enugu, Nigeria
| | - Maryjane Ukwuoma
- Department of Physiotherapy, University of Nigeria Teaching Hospital, Enugu, Enugu
| | - Martins Nweke
- Department of Physiotherapy, Evangel University Akaeze, Enugu, Nigeria
| |
Collapse
|
3
|
Lei L, Huang Y, Shi L, Ye W, Lv X, Ying L, Yu X, Cheng SHC, Zheng Y. Palbociclib sensitizes ER-positive breast cancer cells to fulvestrant by promoting the ubiquitin-mediated degradation of ER-α via SNHG17/Hippo-YAP axis. Breast Cancer Res Treat 2024; 203:613-625. [PMID: 37924380 PMCID: PMC10806073 DOI: 10.1007/s10549-023-07138-0] [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: 07/25/2023] [Accepted: 09/23/2023] [Indexed: 11/06/2023]
Abstract
PURPOSE Endocrine therapy is the anti-tumor therapy for human breast cancer but endocrine resistance was a major burden. It has been reported that Palbociclib and fulvestrant can be used in combination for the treatment of patients who are experiencing endocrine resistance. However, the underlying mechanism is unclear. In this study, we aimed to investigate the mechanism by which Palbocicilib affected ER-positive breast cancer, combined with fulvestrant. METHODS We first detected the effect of palbociclib on cell survival, growth and cycle distribution separately by MTT, colony formation and flow cytometry. Then SNHG17 was screened as palbociclib-targeted LncRNA by LncRNA-seq, and the SNHG17-targeted mRNAs were selected by mRNA-seq for further determination. Subsequently, the underlying mechanism by which palbociclib promoted the cytotoxicity of fulvestrant was confirmed by qRT-PCR, western blot, and immunoprecipitation. Eventually, the xenograft model and immunohistochemistry experiments were used to validate the sensitization effect of palbociclib on fulvestrant and its mechanism in vivo. RESULTS Palbociclib significantly enhanced the cytotoxicity of fulvestrant in fulvestrant-resistant breast cancer cell lines. Interestingly, this might be related to the lncRNA SNHG17 and the Hippo signaling pathway. And our subsequent western blotting experiments confirmed that overexpressing SNHG17 induced the down-regulation of LATS1 and up-regulated YAP expression. Furthermore, we found that the increased sensitivity of breast cancer cells was closely associated with the LATS1-mediated degradation of ER-α. The following animal experiments also indicated that overexpressing SNHG17 obviously impaired the anti-cancer effect of co-treatment of palbociclib and fulvestrant accompanied by decreased LATS1 and increased ER-α levels. CONCLUSION Palbociclib might sensitize the cytotoxicity of fulvestrant in ER-positive breast cancer cells by down-regulating SNHG17 expression, and then resulted in the LATS1-inactivated oncogene YAP and LATS1-mediated degradation of ER-α.
Collapse
Affiliation(s)
- Lei Lei
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Yuan Huang
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Lei Shi
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Weiwu Ye
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Xianmei Lv
- Department of Radiation Oncology, Jinhua People's Hospital, Jinhua, 321000, Zhejiang, China
| | - Lisha Ying
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
- Zhejiang Cancer Institute, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xingfei Yu
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Skye Hung-Chun Cheng
- Department of Radiation Oncology, Koo Foundation, Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | - Yabing Zheng
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
4
|
Shao H, Zhao M, Guan AJ, Shao T, Zhou D, Yu G, Tang W. A network meta-analysis of efficacy and safety for first-line and second/further-line therapies in postmenopausal women with hormone receptor-positive, HER2-negative, advanced breast cancer. BMC Med 2024; 22:13. [PMID: 38212842 PMCID: PMC10785354 DOI: 10.1186/s12916-023-03238-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 12/19/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR + /HER2 -) advanced breast cancer is a prevalent subtype among postmenopausal women. Despite the growing number of randomized clinical trials (RCTs) exploring this topic, the efficacy and safety of first-line and second/further-line treatments remain uncertain. Accordingly, our aim was to conduct a comprehensive evaluation of the efficacy and safety of these therapies through network meta-analysis. METHODS RCTs were identified by searching Pubmed, Embase, and major cancer conferences. The efficacy of interventions was assessed using the hazard ratios (HRs) of progression-free survival (PFS) and overall survival (OS), while safety was indicated by the incidence of any grade adverse events (AEs), grade 3-5 AEs, AEs leading to treatment discontinuation, and AEs leading to death. Both time-variant HRs fractional polynomial models and time-invariant HRs Cox-proportional hazards models were considered for handling time-to-event data. Safety indicators were analyzed using Bayesian network meta-analysis. Additionally, subgroup analyses were conducted based on patient characteristics. RESULTS A total of 41 RCTs (first-line 17, second/further-lines 27) were included in the analysis. For first-line treatment, the addition of Cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors to endocrine therapy significantly improved therapeutic efficacy in terms of both PFS and OS, demonstrating the best performance across all mechanisms. Specifically, the combination of Abemaciclib and Letrozole demonstrated the most favorable performance in terms of PFS, while Ribociclib plus Fulvestrant yielded the best outcomes in OS. Incorporating the immune checkpoint inhibitor Avelumab into the regimen with CDK4/6 inhibitors and selective estrogen receptor degraders significantly enhanced both PFS and OS in second-line or later treatments. Regarding safety, endocrine monotherapy performed well. Regarding safety, endocrine monotherapy performed well. There is mounting evidence suggesting that most CDK4/6 inhibitors may demonstrate poorer performance with respect to hematologic AEs. However, additional evidence is required to further substantiate these findings. CONCLUSIONS CDK4/6 inhibitors, combined with endocrine therapy, are pivotal in first-line treatment due to their superior efficacy and manageable AEs. For second/further-line treatment, adding immune checkpoint inhibitors to CDK4/6 inhibitors plus endocrine therapy may produce promising results. However, to reduce the results' uncertainty, further trials comparing these novel treatments are warranted. TRIAL REGISTRATION Registration number: PROSPERO (CRD42022377431).
Collapse
Affiliation(s)
- Hanqiao Shao
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
- Center for Pharmacoeconomics and Outcomes Research & Department of Public Affairs Management, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Mingye Zhao
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
- Center for Pharmacoeconomics and Outcomes Research & Department of Public Affairs Management, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ai-Jia Guan
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Sichuan, China
| | - Taihang Shao
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
- Center for Pharmacoeconomics and Outcomes Research & Department of Public Affairs Management, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Dachuang Zhou
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
- Center for Pharmacoeconomics and Outcomes Research & Department of Public Affairs Management, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Guo Yu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Wenxi Tang
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China.
- Center for Pharmacoeconomics and Outcomes Research & Department of Public Affairs Management, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
5
|
Wang Y, Xu H, Han Y, Wu Y, Sa Q, Wang J. Identifying the optimal therapeutics for patients with hormone receptor-positive, HER2-positive advanced breast cancer: a systematic review and network meta-analysis. ESMO Open 2023; 8:101216. [PMID: 37084609 PMCID: PMC10172889 DOI: 10.1016/j.esmoop.2023.101216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/12/2023] [Accepted: 03/18/2023] [Indexed: 04/23/2023] Open
Abstract
INTRODUCTION Hormone receptor-positive (HR+) and human epidermal growth factor receptor 2-positive (HER2+) breast cancer is a distinct subtype with different prognosis and response to treatment. HER2-targeted therapy is currently recommended for patients with HR+/HER2+ advanced breast cancer. However, there is debate over which drugs to add on the basis of HER2 blockade yield the optimal efficacy. This systematic review and network meta-analysis was conducted to solve the problem. METHODS Eligible randomized controlled trials (RCTs) comparing different interventions in HR+/HER2+ metastatic breast cancer were included. The outcomes of interest included progression-free survival (PFS), overall survival (OS) and treatment-related adverse events (TRAEs). Pooled hazard ratios or odds ratios with credible intervals (CrIs) were calculated to estimate the predefined outcomes. The optimal therapeutics were identified by comparing the surface under the cumulative ranking curves (SUCRA). RESULTS Totally, 23 literatures of 20 RCTs were included. Regarding PFS, significant differences were detected between single or dual HER2 blockade plus endocrine therapy (ET) versus ET alone and dual HER2 blockade plus ET versus physician's choice. Trastuzumab, pertuzumab plus chemotherapy significantly improved PFS than trastuzumab plus chemotherapy (hazard ratio 0.69, 95% CrI 0.50-0.92). The SUCRA values suggested the relatively better efficacy of dual HER2-targeted therapy plus ET (86%-91%) than chemotherapy (62%-81%) in prolonging PFS and OS. The HER2 blockade-containing regimens showed similar safety profiles in eight documented TRAEs. CONCLUSIONS Prominent status of dual-targeted therapy for patients with HR+/HER2+ metastatic breast cancer was revealed. Compared with chemotherapy-containing regimens, the ET-containing ones showed better efficacy and similar safety profiles, which could be recommended in clinical practice.
Collapse
Affiliation(s)
- Y Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - H Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Y Han
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Y Wu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Q Sa
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - J Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Optimizing treatment for HER2-positive HR-positive breast cancer. Cancer Treat Rev 2023; 115:102529. [PMID: 36921556 DOI: 10.1016/j.ctrv.2023.102529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
Triple-positive breast tumors overexpress human epidermal growth factor receptor 2 (HER2) and are positive for hormone receptor (HR) expression. Data from real-life and clinical trials show that estrogen receptor (ER) expression affects the response to combinations of anti-HER2 and associated systemic therapies. Despite triple-positive tumors having decreased response rates compared to HR-negative/HER2-positive breast cancers, optimizing anti-HER2 treatment with dual anti-HER2 blockade remains important for optimal disease control. Preclinical data on the cross-talk between ER and growth factor receptor pathways show the efficacy of combinations of endocrine therapy and anti-HER2 drugs, which is confirmed in the clinic. Molecular dissection of triple-positive breast cancer might provide the rational for additional therapeutic strategies and the identification of promising biomarkers. This review summarizes data on systemic treatment efficacy from major clinical trials and perspectives for future clinical research in triple-positive breast cancer.
Collapse
|
7
|
Petri BJ, Klinge CM. m6A readers, writers, erasers, and the m6A epitranscriptome in breast cancer. J Mol Endocrinol 2023; 70:JME-22-0110. [PMID: 36367225 PMCID: PMC9790079 DOI: 10.1530/jme-22-0110] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/11/2022] [Indexed: 11/13/2022]
Abstract
Epitranscriptomic modification of RNA regulates human development, health, and disease. The true diversity of the transcriptome in breast cancer including chemical modification of transcribed RNA (epitranscriptomics) is not well understood due to limitations of technology and bioinformatic analysis. N-6-methyladenosine (m6A) is the most abundant epitranscriptomic modification of mRNA and regulates splicing, stability, translation, and intracellular localization of transcripts depending on m6A association with reader RNA-binding proteins. m6A methylation is catalyzed by the METTL3 complex and removed by specific m6A demethylase ALKBH5, with the role of FTO as an 'eraser' uncertain. In this review, we provide an overview of epitranscriptomics related to mRNA and focus on m6A in mRNA and its detection. We summarize current knowledge on altered levels of writers, readers, and erasers of m6A and their roles in breast cancer and their association with prognosis. We summarize studies identifying m6A peaks and sites in genes in breast cancer cells.
Collapse
Affiliation(s)
- Belinda J. Petri
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine; Louisville, KY 40292 USA
| | - Carolyn M. Klinge
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine; Louisville, KY 40292 USA
- University of Louisville Center for Integrative Environmental Health Sciences (CIEHS)
| |
Collapse
|
8
|
[Therapeutic strategies for the treatment of endocrine resistant hormone receptor positive advanced breast cancer]. Bull Cancer 2023; 110:69-87. [PMID: 36307325 DOI: 10.1016/j.bulcan.2022.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/06/2022]
Abstract
HR+ breast cancers are defined by the prominence of signaling pathways dependent on the estrogen receptor. Endocrine therapy is the standard treatment for these advanced diseases. Resistance to these treatments, called hormone resistance, appears invariably with biological mechanisms that have led to the development of therapeutic opportunities. An exhaustive literature review was carried out concerning the biology of the hormone resistance pathways, the therapeutic options before the era of CDK4/6 inhibitors, the rise of CDK4/6 inhibitors and the therapeutic prospects in a situation of hormone resistance. Various biological abnormalities have been identified in the mechanisms of hormone resistance such as changes in the estrogen receptor, mutations in the ESR1 gene, aberrant activation of the PI3K pathway or cell cycle deregulations. Historical strategies for circumventing this hormone resistance have been based on hormonal manipulation, on the development of new endocrine therapy such as fulvestrant (selective estrogen receptor inhibitor, SERD), on combinations of treatments such as everolimus, a mTOR inhibitor. This strategy combining endocrine therapy and targeted therapy has led to the development of combinations with CDK4/6 inhibitors which have now become a standard treatment in the hormone resistance phase. The future of this therapeutic era remains to be written with new combinations of hormone therapy and targeted therapy such as PI3K inhibitors or even with the positioning of new SERDs in clinical development.
Collapse
|
9
|
Xiao Y, Ding J, Ma D, Chen S, Li X, Yu K. Predicting Pathological Complete Response in Neoadjuvant Dual Blockade With Trastuzumab and Pertuzumab in HER2 Gene Amplified Breast Cancer. Front Immunol 2022; 13:877825. [PMID: 35663978 PMCID: PMC9161548 DOI: 10.3389/fimmu.2022.877825] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background Dual-targeted therapy is the standard treatment for human epidermal growth factor receptor 2 (HER2)-positive breast cancer, and effective biomarkers to predict the response to neoadjuvant trastuzumab and pertuzumab treatment need further investigation. Here, we developed a predictive model to evaluate the dual-targeted neoadjuvant treatment efficacy in HER2 gene-amplified breast cancer. Method This retrospective study included 159 HER2-amplified patients with locally advanced breast cancer who received neoadjuvant trastuzumab, pertuzumab, and chemotherapy. The correlation between clinicopathological factors and pathological complete response (pCR, in the breast and axilla) was evaluated. Patients were randomly assigned into the training set (n=110) and the testing set (n=49). We used an independent cohort (n=65) for external validation. We constructed our predictive nomogram model with the results of risk variables associated with pCR identified in the multivariate logistic analysis. The area under the curve (AUC) of the receiver operating characteristic (ROC) curve, decision curve analysis, and calibration curves were employed to assess the nomogram's performance. Results We revealed that the HER2/CEP17 ratio (p=0.001), CD8 levels (p=0.005), and histological grade (p=0.007) were independent indicators for pCR in dual-targeted neoadjuvant treatment after multivariate adjustment. The combined prediction efficacy of the three indicators was significantly higher than that of each single indicator alone. The AUCs were 0.819, 0.773, and 0.744 in the training, testing, and external validation sets, respectively. Conclusions The HER2/CEP17 ratio, CD8 levels, and histological grade were significantly correlated with pCR in dual-targeted neoadjuvant treatment. The combined model using these three markers provided a better predictive value for pCR than the HER2/CEP17 ratio, CD8 levels, and the histological grade alone, which showed that an immunological effect partially mediates the predictive impact of neoadjuvant treatment.
Collapse
Affiliation(s)
- Yi Xiao
- Department of Breast Surgery, The First Hospital of Lanzhou University, Lanzhou, China.,Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Jiahan Ding
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Dachang Ma
- Department of Breast Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Sheng Chen
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xun Li
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Keda Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
10
|
Mazumder A, Shiao S, Haricharan S. HER2 Activation and Endocrine Treatment Resistance in HER2-negative Breast Cancer. Endocrinology 2021; 162:6329618. [PMID: 34320193 PMCID: PMC8379900 DOI: 10.1210/endocr/bqab153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Indexed: 11/19/2022]
Abstract
The lethality of estrogen receptor alpha positive (ER+) breast cancer, which is often considered to have better prognosis than other subtypes, is defined by resistance to the standard of care endocrine treatment. Relapse and metastasis are inevitable in almost every patient whose cancer is resistant to endocrine treatment. Therefore, understanding the underlying causes of treatment resistance remains an important biological and clinical focus of research in this area. Growth factor receptor pathway activation, specifically HER2 activation, has been identified as 1 mechanism of endocrine treatment resistance across a range of experimental model systems. However, clinical trials conducted to test whether targeting HER2 benefits patients with endocrine treatment-resistant ER+ breast cancer have consistently and disappointingly shown mixed results. One reason for the failure of these clinical trials could be the complexity of crosstalk between ER, HER2, and other growth factor receptors and the fluidity of HER2 activation in these cells, which makes it challenging to identify stratifiers for this targeted intervention. In the absence of stratifiers that can be assayed at diagnosis to allow prospective tailoring of HER2 inhibition to the right patients, clinical trials will continue to disappoint. To understand stratifiers, it is important that the field invests in key understudied areas of research including characterization of the tumor secretome and receptor activation in response to endocrine treatment, and mapping the ER-HER2 growth factor network in the normal and developing mammary gland. Understanding these mechanisms further is critical to improving outcomes for the hard-to-treat endocrine treatment-resistant ER+ breast cancer cohort.
Collapse
Affiliation(s)
- Aloran Mazumder
- Aging and Cancer Immuno-oncology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Stephen Shiao
- Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Svasti Haricharan
- Aging and Cancer Immuno-oncology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
- Correspondence: Svasti Haricharan, PhD, Sanford Burnham Prebys, 10901 N Torrey Pines Rd, La Jolla, CA, USA.
| |
Collapse
|
11
|
Brief relaxation training is associated with long-term endocrine therapy adherence among women with breast cancer: post hoc analysis of a randomized controlled trial. Breast Cancer Res Treat 2021; 190:79-88. [PMID: 34410568 DOI: 10.1007/s10549-021-06361-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/08/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Despite life-saving potential, many women struggle to adhere to adjuvant endocrine therapy (AET) for their breast cancer (BCa). Prior research has demonstrated that emotional distress is a barrier to AET adherence. We followed women from a trial to test the long-term effects of two 5-week post-surgical group-based stress management interventions, cognitive behavioral therapy (CBT), and relaxation training versus an attention-matched health education control, on AET adherence. METHODS We conducted a long-term follow-up (median = 8 years) of women randomized to CBT, relaxation training, or health education after surgery for stage 0-3 BCa. We measured adherence with the Endocrine Therapy Medication Usage Questionnaire (ETMUQ). First, we established factors on the ETMUQ via confirmatory factor analysis. We then used Bayesian structural equation modeling to regress these factors on study arm, controlling for age and treatments received. RESULTS Of those who completed long-term follow-up (n = 59, 44.7%), over half (n = 33; 55.9%) reported problems with adherence generally. Women receiving relaxation training (n = 15) had better adherence than those receiving health education (n = 24) on the Forgetfulness/Inconsistency [B(SE) = 0.25(0.14), p = 0.049] and Intentional Nonadherence [B(SE) = 0.31(0.14), p = 0.018] factors of the ETMUQ. Similar results were observed for those receiving relaxation training compared to CBT (n = 20): Forgetfulness/Inconsistency [B(SE) = - 0.47(0.25), p = 0.031]; Intentional Nonadherence [B(SE) = - 0.31(0.15), p = 0.027]. CONCLUSION Women receiving relaxation training were less likely to (1) forget to take their AET and (2) intentionally miss doses of AET in the long term compared to women receiving health education or CBT. This is evidence for the need of randomized trials that aim to improve adherence by incorporating theoretically based behavioral change techniques. TRIAL REGISTRATION AND DATES Trial 2R01-CA-064710 was registered March 26, 2006.
Collapse
|
12
|
Anti-HER2 antibody prolongs overall survival disproportionally more than progression-free survival in HER2-Positive metastatic breast cancer patients. Breast 2021; 59:211-220. [PMID: 34298300 PMCID: PMC8321956 DOI: 10.1016/j.breast.2021.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/28/2021] [Accepted: 07/05/2021] [Indexed: 12/01/2022] Open
Abstract
Background This meta-analysis aimed to test the hypothesis that the HER2-positive metastatic breast cancer (mBC) patients treated with anti-HER2 antibodies in trial intervention arms have a greater prolongation of overall survival (OS) than of progression-free survival (PFS) and this extra-prolongation of median survival time in OS relates specifically to the anti-HER2 antibody. Methods The NCBI/Pubmed and Cochrane databases were searched systematically for HER2-positive or mBC trials published in English during January 1999–November 2017. Treatment arms with shorter PFS were considered as the “control” arm, whereas those with longer PFS as the “test” arm. The between-treatment drug differences were grouped into nine categories. Groups with or without anti-HER2 antibodies were pooled respectively for comparisons. The interrelationships between PFS and OS hazard ratios (HRs) and median survival time differences were investigated by conducting fixed-effects and mixed-effects linear meta-regression analyses. Results Twenty-eight trials (10,928 patients) from 438 articles were collected, and four with missing data were excluded in meta-regression analysis. Overall median PFS (HR = 0.73, 95% CI: 0.68–0.78) and median OS (HR = 0.82, 95% CI: 0.77–0.87) weakly favored the longer PFS arm with a weak correlation between the PFS and OS HRs. However, the between-treatment drug difference was anti-HER2 antibody, the absolute increment in median OS time was double that of median PFS time (p < 0.001) and linearly correlated, which was not found with any non-anti-HER2 antibody drug differences. Conclusions Anti-HER2 antibody in patients with HER2-positive mBC prolonged OS more than PFS and mandates further investigation. Our study dissected the overall survival benefit over progression-free survival in HER-2 positive metastatic breast cancer treated with anti-HER2 antibody by meta-analysis. Use of anti-HER2 antibodies, but not other anticancer drugs, to treat HER2-positive metastatic breast cancer independently predicted the OS compared with PFS. The design of future clinical trials should consider anti-HER2 antibodies carefully in the treatment armamentarium for HER2-positive metastatic breast cancer.
Collapse
|
13
|
Chen J, Wan R, Li Q, Rao Z, Wang Y, Zhang L, Teichmann AT. Utilizing the Hippo pathway as a therapeutic target for combating endocrine-resistant breast cancer. Cancer Cell Int 2021; 21:306. [PMID: 34112175 PMCID: PMC8194146 DOI: 10.1186/s12935-021-01999-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/27/2021] [Indexed: 12/14/2022] Open
Abstract
Drug resistance is always a great obstacle in any endocrine therapy of breast cancer. Although the combination of endocrine therapy and targeted therapy has been shown to significantly improve prognosis, refractory endocrine resistance is still common. Dysregulation of the Hippo pathway is often related to the occurrence and the development of many tumors. Targeted therapies of this pathway have played important roles in the study of triple negative breast cancer (TNBC). Targeting the Hippo pathway in combination with chemotherapy or other targeted therapies has been shown to significantly improve specific antitumor effects and reduce cancer antidrug resistance. Further exploration has shown that the Hippo pathway is closely related to endocrine resistance, and it plays a "co-correlation point" role in numerous pathways involving endocrine resistance, including related pathways in breast cancer stem cells (BCSCs). Agents and miRNAs targeting the components of the Hippo pathway are expected to significantly enhance the sensitivity of breast cancer cells to endocrine therapy. This review initially explains the possible mechanism of the Hippo pathway in combating endocrine resistance, and it concludes by recommending endocrine therapy in combination with therapies targeting the Hippo pathway in the study of endocrine-resistant breast cancers.
Collapse
Affiliation(s)
- Jing Chen
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China.,Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Runlan Wan
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China
| | - Qinqin Li
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China.,Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Zhenghuan Rao
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China.,Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yanlin Wang
- North Sichuan Medical College, Nanchong, 637000, China
| | - Lei Zhang
- Department of Gynaecology, The Second People's Hospital of Yibin, Yibin, 644000, China
| | - Alexander Tobias Teichmann
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China. .,Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
14
|
Punturi NB, Seker S, Devarakonda V, Mazumder A, Kalra R, Chen CH, Li S, Primeau T, Ellis MJ, Kavuri SM, Haricharan S. Mismatch repair deficiency predicts response to HER2 blockade in HER2-negative breast cancer. Nat Commun 2021; 12:2940. [PMID: 34011995 PMCID: PMC8134423 DOI: 10.1038/s41467-021-23271-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 04/22/2021] [Indexed: 01/02/2023] Open
Abstract
Resistance to endocrine treatment occurs in ~30% of ER+ breast cancer patients resulting in ~40,000 deaths/year in the USA. Preclinical studies strongly implicate activation of growth factor receptor, HER2 in endocrine treatment resistance. However, clinical trials of pan-HER inhibitors in ER+/HER2- patients have disappointed, likely due to a lack of predictive biomarkers. Here we demonstrate that loss of mismatch repair activates HER2 after endocrine treatment in ER+/HER2- breast cancer cells by protecting HER2 from protein trafficking. Additionally, HER2 activation is indispensable for endocrine treatment resistance in MutL- cells. Consequently, inhibiting HER2 restores sensitivity to endocrine treatment. Patient data from multiple clinical datasets supports an association between MutL loss, HER2 upregulation, and sensitivity to HER inhibitors in ER+/HER2- patients. These results provide strong rationale for MutL loss as a first-in-class predictive marker of sensitivity to combinatorial treatment with endocrine intervention and HER inhibitors in endocrine treatment-resistant ER+/HER2- breast cancer patients.
Collapse
MESH Headings
- Animals
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Cell Line, Tumor
- DNA Mismatch Repair/drug effects
- DNA Mismatch Repair/genetics
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Knockdown Techniques
- Humans
- MCF-7 Cells
- Mice
- Mice, Nude
- Mice, SCID
- MutL Protein Homolog 1/genetics
- MutL Protein Homolog 1/metabolism
- MutL Proteins/genetics
- MutL Proteins/metabolism
- Proteins/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptors, Estrogen/metabolism
- Signal Transduction
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Nindo B Punturi
- Tumor Microenvironment and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Sinem Seker
- Tumor Microenvironment and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Vaishnavi Devarakonda
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Aloran Mazumder
- Tumor Microenvironment and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Rashi Kalra
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Ching Hui Chen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Shunqiang Li
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Tina Primeau
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Shyam M Kavuri
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - Svasti Haricharan
- Tumor Microenvironment and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
15
|
Roßwag S, Cotarelo CL, Pantel K, Riethdorf S, Sleeman JP, Schmidt M, Thaler S. Functional Characterization of Circulating Tumor Cells (CTCs) from Metastatic ER+/HER2- Breast Cancer Reveals Dependence on HER2 and FOXM1 for Endocrine Therapy Resistance and Tumor Cell Survival: Implications for Treatment of ER+/HER2- Breast Cancer. Cancers (Basel) 2021; 13:cancers13081810. [PMID: 33920089 PMCID: PMC8070196 DOI: 10.3390/cancers13081810] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Acquired endocrine resistance and late recurrence in patients with ER+/HER2− breast cancer are complex and not fully understood. Here, we evaluated mechanisms of acquired resistance in circulating tumor cells (CTCs) from an ER+/HER2− breast cancer patient who initially responded but later progressed under endocrine treatment. We found a switch from ERα-dependent to HER2-dependent and ERα-independent expression of FOXM1, which may enable disseminated ER+/HER2− cells to re-initiate tumor cell growth and metastasis formation in the presence of endocrine treatment. We found that NFkB signaling sustains HER2 and FOXM1 expression in CTCs in the presence of ERα inhibitors suggesting that NFkB and FOXM1 might be an efficient therapeutic approach to prevent late recurrence and to treat endocrine resistance. Collectively our data show that CTCs from patients with endocrine resistance allow mechanisms of acquired endocrine resistance to be delineated, and can be used to test potential drug regimens for combatting resistance. Abstract Mechanisms of acquired endocrine resistance and late recurrence in patients with ER+/HER2− breast cancer are complex and not fully understood. Here, we evaluated mechanisms of acquired resistance in circulating tumor cells (CTCs) from an ER+/HER2− breast cancer patient who initially responded but later progressed under endocrine treatment. We found a switch from ERα-dependent to HER2-dependent and ERα-independent expression of FOXM1, which may enable disseminated ER+/HER2− cells to re-initiate tumor cell growth and metastasis formation in the presence of endocrine treatment. Our results also suggest a role for HER2 in resistance, even in ER+ breast cancer cells that have neither HER2 amplification nor activating HER2 mutations. We found that NFkB signaling sustains HER2 and FOXM1 expression in CTCs in the presence of ERα inhibitors. Inhibition of NFkB signaling blocked expression of HER2 and FOXM1 in the CTCs, and induced apoptosis. Thus, targeting of NFkB and FOXM1 might be an efficient therapeutic approach to prevent late recurrence and to treat endocrine resistance. Collectively our data show that CTCs from patients with endocrine resistance allow mechanisms of acquired endocrine resistance to be delineated, and can be used to test potential drug regimens for combatting resistance.
Collapse
Affiliation(s)
- Sven Roßwag
- European Center for Angioscience, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany or (S.R.); (J.P.S.)
| | - Cristina L. Cotarelo
- Institute of Pathology, University Medical Center of Heinrich-Heine University, 40225 Duesseldorf, Germany;
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (K.P.); (S.R.)
| | - Sabine Riethdorf
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (K.P.); (S.R.)
| | - Jonathan P. Sleeman
- European Center for Angioscience, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany or (S.R.); (J.P.S.)
- Karlsruhe Institute of Technology (KIT) Campus Nord, Institute of Biological and Chemical Systems—Biological Information Processing, 76344 Eggenstein-Leupoldshafen, Germany
| | - Marcus Schmidt
- Department of Gynecology and Obstetrics, University Medical Center of Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Sonja Thaler
- European Center for Angioscience, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany or (S.R.); (J.P.S.)
- Correspondence: ; Tel.: +49-621-3837-1599
| |
Collapse
|
16
|
Zhou S, Zheng F, Zhan CG. Clinical data mining reveals analgesic effects of lapatinib in cancer patients. Sci Rep 2021; 11:3528. [PMID: 33574423 PMCID: PMC7878815 DOI: 10.1038/s41598-021-82318-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 01/14/2021] [Indexed: 12/03/2022] Open
Abstract
Microsomal prostaglandin E2 synthase 1 (mPGES-1) is recognized as a promising target for a next generation of anti-inflammatory drugs that are not expected to have the side effects of currently available anti-inflammatory drugs. Lapatinib, an FDA-approved drug for cancer treatment, has recently been identified as an mPGES-1 inhibitor. But the efficacy of lapatinib as an analgesic remains to be evaluated. In the present clinical data mining (CDM) study, we have collected and analyzed all lapatinib-related clinical data retrieved from clinicaltrials.gov. Our CDM utilized a meta-analysis protocol, but the clinical data analyzed were not limited to the primary and secondary outcomes of clinical trials, unlike conventional meta-analyses. All the pain-related data were used to determine the numbers and odd ratios (ORs) of various forms of pain in cancer patients with lapatinib treatment. The ORs, 95% confidence intervals, and P values for the differences in pain were calculated and the heterogeneous data across the trials were evaluated. For all forms of pain analyzed, the patients received lapatinib treatment have a reduced occurrence (OR 0.79; CI 0.70–0.89; P = 0.0002 for the overall effect). According to our CDM results, available clinical data for 12,765 patients enrolled in 20 randomized clinical trials indicate that lapatinib therapy is associated with a significant reduction in various forms of pain, including musculoskeletal pain, bone pain, headache, arthralgia, and pain in extremity, in cancer patients. Our CDM results have demonstrated the significant analgesic effects of lapatinib, suggesting that lapatinib may be repurposed as a novel type of analgesic.
Collapse
Affiliation(s)
- Shuo Zhou
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA
| | - Fang Zheng
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA. .,Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA.
| | - Chang-Guo Zhan
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA. .,Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA.
| |
Collapse
|
17
|
Abstract
The functional diversity of the mammalian intestinal microbiome far exceeds that of the host organism, and microbial genes contribute substantially to the well-being of the host. However, beneficial gut organisms can also be pathogenic when present in the gut or other locations in the body. Among dominant beneficial bacteria are several species of Bacteroides, which metabolize polysaccharides and oligosaccharides, providing nutrition and vitamins to the host and other intestinal microbial residents. These topics and the specific organismal and molecular interactions that are known to be responsible for the beneficial and detrimental effects of Bacteroides species in humans comprise the focus of this review. The complexity of these interactions will be revealed.
Collapse
Affiliation(s)
- Hassan Zafar
- Department of Molecular Biology, Division of Biological Sciences, University of California at San Diego, USA
- Department of Microbiology and Molecular Genetics, Faculty of Life Sciences, University of Okara,Okara, PunjabPakistan
| | - Milton H. Saier
- Department of Molecular Biology, Division of Biological Sciences, University of California at San Diego, USA
| |
Collapse
|
18
|
Yusufov M, Nathan M, Wiley A, Russell J, Partridge A, Joffe H. Predictors of increased risk for early treatment non-adherence to oral anti-estrogen therapies in early-stage breast cancer patients. Breast Cancer Res Treat 2020; 185:53-62. [PMID: 32918659 DOI: 10.1007/s10549-020-05920-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Non-adherence to the oral anti-estrogen therapies (AET) tamoxifen and aromatase inhibitors in early-stage hormone receptor-positive breast cancer is associated with numerous negative clinical outcomes. Prior studies have identified that non-adherence is associated with psychological and menopause-related factors which are present during AET, but the presence of these characteristics prior to AET initiation has not been investigated. METHODS Psychological and menopause symptoms (depression, generalized anxiety, insomnia, somatosensory amplification, hot flash frequency, and hot flash-related interference) were assessed pre-AET initiation as predictors of subsequent non-adherence in 73 participants (Mage = 55.0, SD = 10.1 years). Participants self-reported treatment adherence after three and 6 weeks on AET. Participants who did not initiate treatment were excluded from the analysis. RESULTS Discriminant function analyses revealed that the hypothesized set of psychological and menopause symptoms at baseline (pre-AET) together statistically distinguished between those who were non-adherent (n = 19; 26.0%) from adherent (n = 54; 74.0%) at 6 weeks. Model classification accuracy was statistically significant (Wilks' ƛ = 0.782, χ2(6) = 15.50, p = 0.017) at the 6-week timepoint. Results were consistent at 3 weeks. Pre-AET psychological and menopause symptoms correctly classified 6-week treatment adherence 77.9% of the time. Depression contributed most to distinguishing between adherers and non-adherers. CONCLUSIONS The presence of a composite profile of psychological and menopause symptoms prior to AET initiation may help to identify early treatment non-adherence. Results can be used to identify patients at risk for non-adherence and to guide psychological and symptom management interventions.
Collapse
Affiliation(s)
- Miryam Yusufov
- Department of Psychosocial Oncology & Palliative Care, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA.,Department of Psychiatry, Harvard Medical School Brigham and Women's Hospital, 75 Francis Street, Thorn 1111, Boston, MA, 02215, USA
| | - Margo Nathan
- Women's Hormones and Aging Research Program, Department of Psychiatry, Brigham & Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA.,Department of Psychiatry, Harvard Medical School Brigham and Women's Hospital, 75 Francis Street, Thorn 1111, Boston, MA, 02215, USA
| | - Aleta Wiley
- Women's Hormones and Aging Research Program, Department of Psychiatry, Brigham & Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Julia Russell
- Women's Hormones and Aging Research Program, Department of Psychiatry, Brigham & Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Ann Partridge
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Hadine Joffe
- Department of Psychosocial Oncology & Palliative Care, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA. .,Women's Hormones and Aging Research Program, Department of Psychiatry, Brigham & Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA. .,Department of Psychiatry, Harvard Medical School Brigham and Women's Hospital, 75 Francis Street, Thorn 1111, Boston, MA, 02215, USA.
| |
Collapse
|
19
|
Brandão M, Maurer C, Ziegelmann PK, Pondé NF, Ferreira A, Martel S, Piccart M, de Azambuja E, Debiasi M, Lambertini M. Endocrine therapy-based treatments in hormone receptor-positive/HER2-negative advanced breast cancer: systematic review and network meta-analysis. ESMO Open 2020; 5:e000842. [PMID: 32847835 PMCID: PMC7451473 DOI: 10.1136/esmoopen-2020-000842] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/15/2020] [Accepted: 06/19/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Several endocrine therapy (ET)-based treatments are available for patients with advanced breast cancer. We assessed the efficacy of different ET-based treatments in patients with hormone receptor-positive/HER2-negative advanced breast cancer with endocrine-sensitive or endocrine-resistant disease. METHODS We searched Medline and Cochrane Central Register of Controlled Trials up to 15 October 2019 and abstracts from major conferences from 2016 to October 2019. We included phase II/III randomised trials, comparing ≥2 ET-based treatments. Progression-free survival (PFS) and overall survival (OS) were analysed by network meta-analyses using MTC Bayesian models based on both fixed-effect and random-effect models; relative treatment effects were measured as HRs and 95% credibility intervals (CrI). All statistical tests were two-sided. Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed and this systematic review is registered in the PROSPERO database. RESULTS 55 publications reporting on 32 trials (n=12 293 patients) were included. Regarding PFS in the endocrine sensitive setting (n=5200; 12 trials), the combination of cyclin-dependent kinases (CDK)4/6-inhibitors (CDK4/6i)+fulvestrant 500 mg (F500) was likely the most effective treatment (surface under the cumulative ranking curve (SUCRA)=97.3%), followed by CDK4/6i+aromatase inhibitor ±goserelin; there was no significant difference between them (HR 0.82; 95% CrI 0.54-1.25). Regarding OS (n=2157; five trials), the most effective treatment was probably CDK4/6i+F500 (SUCRA=97.3%); comparing CDK4/6i+F500 versus F500 held a HR of 0.77 (95% CrI 0.63-0.95). Regarding PFS in the endocrine-resistant setting (n=6635; 20 trials), CDK4/6i+F500 was likely the most effective treatment (SUCRA=95.7%), followed by capivasertib+F500, without significant difference between them (HR 0.91; 95% CrI 0.60-1.36). For OS (n=4377; 11 trials), the most effective treatments were capivasertib+F500 (SUCRA=84.7%) and CDK4/6i+F500 (SUCRA=69.9%). Comparing CDK4/6i+F500 versus F500 held a HR of 0.77 (95% CrI 0.67-0.89). CONCLUSIONS CDK4/6i+F500 is likely the best treatment option in both endocrine-sensitive and endocrine-resistant diseases for PFS, and in endocrine-sensitive patients for OS. Concerning OS in endocrine-resistant patients, capivasertib+F500 and CDK4/6i+F500 are likely the best treatments. PROSPERO REGISTRATION NUMBER CRD42018104628.
Collapse
Affiliation(s)
- Mariana Brandão
- Academic Trials Promoting Team, Institut Jules Bordet, Bruxelles, Belgium
| | - Christian Maurer
- Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn, University of Cologne, Cologne, Germany
| | - Patricia Klarmann Ziegelmann
- National Institute for Health Technology Assessment (IATS), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Postgraduate Program in Epidemiology and Department of Statistics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Noam F Pondé
- Medical Oncology, AC Camargo Cancer Center, Sao Paulo, Brazil
| | - Arlindo Ferreira
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | - Samuel Martel
- Institut Jules Bordet, Bruxelles, Belgium
- CISSS Montérégie-Centre, Hopital Charles-Lemoyne, Greenfield Park, Quebec, Canada
| | - Martine Piccart
- Université Libre de Bruxelles, Institut Jules Bordet, Bruxelles, Belgium
| | - Evandro de Azambuja
- Academic Trials Promoting Team, Institut Jules Bordet, Bruxelles, Belgium
- Department of Medical Oncology, Institut Jules Bordet, Brussels, Belgium
| | | | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O.C, Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
20
|
Tolaney SM, Wardley AM, Zambelli S, Hilton JF, Troso-Sandoval TA, Ricci F, Im SA, Kim SB, Johnston SRD, Chan A, Goel S, Catron K, Chapman SC, Price GL, Yang Z, Gainford MC, André F. Abemaciclib plus trastuzumab with or without fulvestrant versus trastuzumab plus standard-of-care chemotherapy in women with hormone receptor-positive, HER2-positive advanced breast cancer (monarcHER): a randomised, open-label, phase 2 trial. Lancet Oncol 2020; 21:763-775. [DOI: 10.1016/s1470-2045(20)30112-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/10/2020] [Accepted: 02/13/2020] [Indexed: 12/15/2022]
|
21
|
Hua YM. An evidence-based review of the outcome of fulvestrant plus a targeted agent versus fulvestrant alone in treating hormone receptor-positive endocrine therapy-resistant metastatic breast cancer. Arch Gynecol Obstet 2019; 300:1377-1382. [PMID: 31599350 DOI: 10.1007/s00404-019-05304-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/14/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Fulvestrant is approved for the hormone receptor-positive advanced metastatic breast cancer (MBC) patients during or after prior endocrine treatment. The adding of a targeted agent to fulvestrant has improved the outlook for these patients from recent studies. However, these results were still under investigation, the analysis was undertaken to assess the clinical outcomes of the fulvestrant-based combination therapy compared with fulvestrant monotherapy. METHODS I systematically searched electronic databases to identify eligible literatures till January 2019. Randomized-controlled trials (RCTs) assessing the efficacy of a targeted agent to fulvestrant with fulvestrant mono-therapy in MBC patients who are refractory to or intolerant of prior endocrine therapy were included. RESULTS Six RCTs were included in this analysis. The group of a targeted agent to fulvestrant was significantly improved overall survival (OR = 0.86, 95%CI = 0.76-0.97, P = 0.02), progression-free survival (OR = 0.66, 95%CI = 0.54-0.81, P < 0.0001), as well with the objective response rate (OR = 2.30, 95%CI = 1.67-3.18, P < 0.00001), respectively. However, there are more adverse effects with the combination group (OR = 6.71, 95%CI = 5.58-8.06, P < 0.00001). CONCLUSIONS Pooled results indicate that adding a targeted agent to fulvestrant prolonged OS, PFS and ORR in relapse or metastatic hormone receptor-positive breast cancer after prior endocrine therapy. Combination of fulvestrant with a targeted agent was associated with more frequent grade 3/4 toxicities. Further research is needed to develop a database of reliable biomarkers and their individual impact on the fulvestrant-based combination treatments.
Collapse
Affiliation(s)
- Yu-Ming Hua
- Department of General Surgery, Wuxi No. 3 People's Hospital, No. 585 Xingyuan Bei Road, Wuxi, 214002, Jiangsu, China.
| |
Collapse
|
22
|
Soleja M, Raj GV, Unni N. An evaluation of fulvestrant for the treatment of metastatic breast cancer. Expert Opin Pharmacother 2019; 20:1819-1829. [DOI: 10.1080/14656566.2019.1651293] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Mohsin Soleja
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ganesh V. Raj
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nisha Unni
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
23
|
Buchta Rosean C, Bostic RR, Ferey JCM, Feng TY, Azar FN, Tung KS, Dozmorov MG, Smirnova E, Bos PD, Rutkowski MR. Preexisting Commensal Dysbiosis Is a Host-Intrinsic Regulator of Tissue Inflammation and Tumor Cell Dissemination in Hormone Receptor-Positive Breast Cancer. Cancer Res 2019; 79:3662-3675. [PMID: 31064848 PMCID: PMC6983951 DOI: 10.1158/0008-5472.can-18-3464] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/12/2019] [Accepted: 05/03/2019] [Indexed: 02/07/2023]
Abstract
It is unknown why some patients with hormone receptor-positive (HR+) breast cancer present with more aggressive and invasive disease. Metastatic dissemination occurs early in disease and is facilitated by cross-talk between the tumor and tissue environment, suggesting that undefined host-intrinsic factors enhance early dissemination and the probability of developing metastatic disease. Here, we have identified commensal dysbiosis as a host-intrinsic factor associated with metastatic dissemination. Using a mouse model of HR+ mammary cancer, we demonstrate that a preestablished disruption of commensal homeostasis results in enhanced circulating tumor cells and subsequent dissemination to the tumor-draining lymph nodes and lungs. Commensal dysbiosis promoted early inflammation within the mammary gland that was sustained during HR+ mammary tumor progression. Furthermore, dysbiosis enhanced fibrosis and collagen deposition both systemically and locally within the tumor microenvironment and induced significant myeloid infiltration into the mammary gland and breast tumor. These effects were recapitulated both by directly targeting gut microbes using nonabsorbable antibiotics and by fecal microbiota transplantation of dysbiotic cecal contents, demonstrating the direct impact of gut dysbiosis on mammary tumor dissemination. This study identifies dysbiosis as a preexisting, host-intrinsic regulator of tissue inflammation, myeloid recruitment, fibrosis, and dissemination of tumor cells in HR+ breast cancer. SIGNIFICANCE: Identification of commensal dysbiosis as a host-intrinsic factor mediating evolution of metastatic breast cancer allows for development of interventions or diagnostic tools for patients at highest risk for developing metastatic disease.See related commentary by Ingman, p. 3539.
Collapse
Affiliation(s)
- Claire Buchta Rosean
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Raegan R Bostic
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Joshua C M Ferey
- University of Virginia School of Medicine, Charlottesville, Virginia
| | - Tzu-Yu Feng
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Francesca N Azar
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Kenneth S Tung
- Department of Pathology, University of Virginia, Charlottesville, Virginia
| | - Mikhail G Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia
| | - Ekaterina Smirnova
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia
| | - Paula D Bos
- Department of Pathology, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Melanie R Rutkowski
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
24
|
Lux MP, Böhme S, Hücherig S, Jeratsch U, Kürschner N, Lüftner D. Surrogate threshold effect based on a meta-analysis for the predictive value of progression-free survival for overall survival in hormone receptor-positive, HER2-negative metastatic breast cancer. Breast Cancer Res Treat 2019; 176:495-506. [PMID: 31065873 PMCID: PMC6586722 DOI: 10.1007/s10549-019-05262-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/26/2019] [Indexed: 01/13/2023]
Abstract
PURPOSE Clinical trials investigating therapies for metastatic breast cancer (mBC) generally use progression-free survival (PFS) as primary endpoint, which is not accepted as patient-relevant outcome within the German benefit assessment. Hence a validation of PFS as surrogate endpoint for overall survival (OS) is needed, e.g., in the indication of HR+, HER2-negative mBC. METHODS A systematic search was conducted. RCT were included if at least one study arm investigated fulvestrant, letrozole, tamoxifen, exemestane, or anastrozole. Additionally, hazard ratios reported for OS/PFS including confidence interval or standard error were mandatory. Pearson correlation coefficient was calculated to estimate the relation of surrogate endpoint PFS and patient-relevant outcome OS as well as the surrogate threshold effect (STE) which is used to determine thresholds for the estimate of the surrogate endpoint. RESULTS 16 studies with 5324 patients in total were included in the analyses. The correlation between hazard ratios of PFS and OS was statistically significant (r = 0.72, 95% CI 0.35-0.90) representing a positive linear relationship. STE analysis was applied. The meta-regression model revealed a STE for HRPFS of 0.60 and sensitivity analyses underlined robustness of the results. CONCLUSIONS Based on the derived STE, it is possible to draw conclusions on a significant effect in OS for a hypothetical trial demonstrating an upper confidence limit of HRPFS < 0.60 in PFS. However, only final OS results are able to confirm if a clinical relevant difference in survival time can be achieved.
Collapse
Affiliation(s)
- Michael Patrick Lux
- Kooperatives Brustzentrum Paderborn, Paderborn, Germany. .,Frauenklinik St. Louise, Paderborn, Germany. .,St. Josefs-Krankenhaus, Salzkotten, Germany. .,Frauen- und Kinderklinik St. Louise, Husener Str. 81, 33098, Paderborn, Germany.
| | - Sarah Böhme
- Pfizer Deutschland GmbH, Linkstraße 10, 10785, Berlin, Germany
| | - Stephanie Hücherig
- AMS Advanced Medical Services GmbH, Rosa-Bavarese-Str. 5, 80639, Munich, Germany
| | - Ulli Jeratsch
- AMS Advanced Medical Services GmbH, Rosa-Bavarese-Str. 5, 80639, Munich, Germany
| | | | - Diana Lüftner
- Klinik für Hämatologie, Onkologie und Tumorimmunologie, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany
| |
Collapse
|
25
|
Li J, Wang Z, Shao Z. Fulvestrant in the treatment of hormone receptor-positive/human epidermal growth factor receptor 2-negative advanced breast cancer: A review. Cancer Med 2019; 8:1943-1957. [PMID: 31004402 PMCID: PMC6536994 DOI: 10.1002/cam4.2095] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/30/2019] [Accepted: 02/26/2019] [Indexed: 12/13/2022] Open
Abstract
Nearly 75% of breast cancers are hormone receptor-positive (HR+) and human epidermal growth factor receptor type 2-negative (HER2-), making endocrine therapy the mainstay of treatment for HR+ and HER2- combination. Although endocrine therapy, such as therapy with fulvestrant, is widely used in the clinic, endocrine resistance (primary or secondary) is inevitable and poses a serious clinical concern. However, the therapeutic landscape of HR+/HER2- breast cancer is rapidly changing and evolving. In recent years, molecular insights into the genome of HR+/HER2- breast cancer have helped to identify promising targets, such as alterations in signaling pathways [phosphatidylinositide 3-kinase (PI3K/AKT/mammalian target of rapamycin (mTOR)], dysregulation of the cell cycle (CDK4/6), and identification of new ESR1 mutations. These insights have led to the development of newer targeted therapies, which aims at significantly improving survival in these patients. This review summarizes the role and rationale of fulvestrant when used as a monotherapy or in combination with targeted therapies in patients with HR+/HER2- advanced breast cancer. We also discuss other novel agents and potential future combination treatment options.
Collapse
Affiliation(s)
- Junjie Li
- Department of Surgery in Breast CancerFudan University Shanghai Cancer CenterShanghaiChina
| | - Zhonghua Wang
- Department of Surgery in Breast CancerFudan University Shanghai Cancer CenterShanghaiChina
| | - Zhimin Shao
- Department of Surgery in Breast CancerFudan University Shanghai Cancer CenterShanghaiChina
| |
Collapse
|
26
|
Li J. Diarrhea With HER2-Targeted Agents in Cancer Patients: A Systematic Review and Meta-Analysis. J Clin Pharmacol 2019; 59:935-946. [DOI: 10.1002/jcph.1382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/10/2019] [Indexed: 11/07/2022]
Affiliation(s)
- Jing Li
- College of Pharmacy; Southwest Minzu University; Chengdu Sichuan P. R. China
| |
Collapse
|
27
|
Huang HW, Huang LS, Xu QN, Wang HB, Li XY, Lin JZ. CDK4/6 inhibition versus mTOR blockade as second-line strategy in postmenopausal patients with hormone receptor-positive advanced breast cancer: A network meta-analysis. Medicine (Baltimore) 2019; 98:e13909. [PMID: 30608416 PMCID: PMC6344177 DOI: 10.1097/md.0000000000013909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Cyclin-dependent kinase 4/6 (CDK4/6) inhibitors (palbociclib and abemaciclib) and mammalian target of rapamycin (mTOR) inhibitors (everolimus) are effective agents for restoring endocrine sensitivity in patients with advanced breast cancer progression on prior aromatase inhibitors. We conducted a network meta-analysis to compare these treatments in terms of progression-free survival (PFS), objective response rate (ORR), and clinical benefit rate (CBR). METHODS The PubMed and Embase databases were searched for relevant publications between January 2000 and June 2018. Treatments were ranked based on a network meta-analysis. Ranking was determined by P-score. A random-effect model was used when heterogeneity was detected; otherwise, a fixed-effect model was used. RESULTS Six trials comprising 4063 patients formed the comparison network. Compared with everolimus plus exemestane, the combinations of palbociclib or abemaciclib with fulvestrant showed similar efficacies in PFS and no differences in ORR. For the CBR, palbociclib demonstrated improvement, while abemaciclib did not. Incidences of severe adverse events did not significantly differ. A total of 29%, 15.9%, and 4% of patients discontinued everolimus, abemaciclib, and palbociclib, respectively, due to toxicity. CONCLUSION These results suggest similar efficacies between CDK4/6 inhibition and mTOR blockade; however, CDK4/6 inhibitors were associated with favorable toxicity profiles.
Collapse
Affiliation(s)
- Hong-Wei Huang
- The Seventh Department of Surgery, Puning People's Hospital, Puning
| | | | - Qi-Ni Xu
- Department of Medical Oncology, Cancer Hospital of Shantou University Medical College
| | - Hong-Biao Wang
- Department of Medical Oncology, Cancer Hospital of Shantou University Medical College
| | - Xu-Yuan Li
- Department of Medical Oncology, Shantou Central Hospital
| | - Jia-Zhou Lin
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
28
|
Sen F, Aydiner A. Endocrine Therapy of Metastatic Breast Cancer. Breast Cancer 2019. [DOI: 10.1007/978-3-319-96947-3_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
29
|
Klinge CM. Non-Coding RNAs in Breast Cancer: Intracellular and Intercellular Communication. Noncoding RNA 2018; 4:E40. [PMID: 30545127 PMCID: PMC6316884 DOI: 10.3390/ncrna4040040] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/29/2018] [Accepted: 12/04/2018] [Indexed: 02/07/2023] Open
Abstract
Non-coding RNAs (ncRNAs) are regulators of intracellular and intercellular signaling in breast cancer. ncRNAs modulate intracellular signaling to control diverse cellular processes, including levels and activity of estrogen receptor α (ERα), proliferation, invasion, migration, apoptosis, and stemness. In addition, ncRNAs can be packaged into exosomes to provide intercellular communication by the transmission of microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) to cells locally or systemically. This review provides an overview of the biogenesis and roles of ncRNAs: small nucleolar RNA (snRNA), circular RNAs (circRNAs), PIWI-interacting RNAs (piRNAs), miRNAs, and lncRNAs in breast cancer. Since more is known about the miRNAs and lncRNAs that are expressed in breast tumors, their established targets as oncogenic drivers and tumor suppressors will be reviewed. The focus is on miRNAs and lncRNAs identified in breast tumors, since a number of ncRNAs identified in breast cancer cells are not dysregulated in breast tumors. The identity and putative function of selected lncRNAs increased: nuclear paraspeckle assembly transcript 1 (NEAT1), metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), steroid receptor RNA activator 1 (SRA1), colon cancer associated transcript 2 (CCAT2), colorectal neoplasia differentially expressed (CRNDE), myocardial infarction associated transcript (MIAT), and long intergenic non-protein coding RNA, Regulator of Reprogramming (LINC-ROR); and decreased levels of maternally-expressed 3 (MEG3) in breast tumors have been observed as well. miRNAs and lncRNAs are considered targets of therapeutic intervention in breast cancer, but further work is needed to bring the promise of regulating their activities to clinical use.
Collapse
Affiliation(s)
- Carolyn M Klinge
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40292, USA.
| |
Collapse
|
30
|
Xu L, Yan N, Li Z, Luo L, Wu X, Liu Q, Xu Y, Cao Y. A comparison of fulvestrant plus a targeted agent with fulvestrant alone in hormone receptor-positive advanced breast cancer that progressed on prior endocrine therapy: a meta-analysis. Onco Targets Ther 2018; 11:8389-8398. [PMID: 30568462 PMCID: PMC6267349 DOI: 10.2147/ott.s166653] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Fulvestrant is recommended for the hormone receptor-positive metastatic breast cancer (MBC) patients progressed during or after prior endocrine therapy. Notably, recent evidence has also demonstrated that adding a targeted agent to fulvestrant conferred a significantly clinical benefit in these patients. Since these results were inconsistent among the studies, this meta-analysis herein was conducted to compare the efficacy and toxicities of the fulvestrant-based combination therapy with fulvestrant monotherapy. Thus, a systemic research was performed in PubMed, Embase, and Cochrane library to identify relevant Phase II or Phase III randomized controlled trials. The progression-free survival (PFS), overall response rate (ORR), and toxicities were evaluated. And HR, risk ratio (RR), and their 95% CIs were employed to complete the pooled analyses. In total, 13 studies with 3,910-hour positive MBC patients progressed on prior endocrine therapy were included in our meta-analysis. Improvements of doublet-agents group were proven in terms of PFS (HR 0.73, 95% CI =0.63–0.86, P=0.000) and ORR (RR 2.07, 95% CI =1.67–2.58, P=0.000). And the further subgroup analysis also demonstrated that fulvestrant in combination with a cyclin-dependent kinase (CDK4/6) inhibitor or a PI3K/mTOR inhibitor was associated with a superior efficacy (RR 2.72, 95% CI =1.93–3.83, P=0.000 and RR 1.60, 95% CI =1.15–2.23, P=0.005, respectively). However, the efficacy was comparable between the other combination strategies and fulvestrant alone. With respect to the adverse effects, adding a targeted agent to fulvestrant also produced more frequent grade 3/4 toxicities (RR 3.86, 95% CI =2.66–5.61, P=0.000). Taken together, combination of fulvestrant with a targeted agent, especially inhibitors targeting CDK4/6 or PI3K/mTOR pathway, may open a new avenue for more effective therapies in relapse or metastatic hormone receptor-positive breast cancer after prior aromatase inhibitors or tamoxifen treatment. In addition, identifying reliable biomarkers to delineate which subgroup of patients will specially benefit from fulvestrant-based combination therapy is warranted.
Collapse
Affiliation(s)
- Liang Xu
- The First Department of Prevention and Cure Centre of Breast Disease, The Third Hospital of Nanchang City, Key Laboratory of Breast Disease in Jiangxi Province, Nanchang, JiangXi 330009, China,
| | - Ningning Yan
- Department of Oncology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Zhihua Li
- The First Department of Prevention and Cure Centre of Breast Disease, The Third Hospital of Nanchang City, Key Laboratory of Breast Disease in Jiangxi Province, Nanchang, JiangXi 330009, China,
| | - Lihua Luo
- Medical Department, Graduate School of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xiaobo Wu
- The First Department of Prevention and Cure Centre of Breast Disease, The Third Hospital of Nanchang City, Key Laboratory of Breast Disease in Jiangxi Province, Nanchang, JiangXi 330009, China,
| | - Qiuming Liu
- The First Department of Prevention and Cure Centre of Breast Disease, The Third Hospital of Nanchang City, Key Laboratory of Breast Disease in Jiangxi Province, Nanchang, JiangXi 330009, China,
| | - Yingchun Xu
- Department of Oncology, Shanghai Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China,
| | - Yali Cao
- The First Department of Prevention and Cure Centre of Breast Disease, The Third Hospital of Nanchang City, Key Laboratory of Breast Disease in Jiangxi Province, Nanchang, JiangXi 330009, China,
| |
Collapse
|
31
|
Chanchan G, Xiangyu S, Fangfang S, Yan C, Xiaoyi G. The efficacy and safety of targeted therapy plus fulvestrant in postmenopausal women with hormone-receptor positive advanced breast cancer: A meta-analysis of randomized-control trials. PLoS One 2018; 13:e0204202. [PMID: 30235292 PMCID: PMC6157814 DOI: 10.1371/journal.pone.0204202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 09/05/2018] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE To evaluate the efficacy and safety of targeted therapy plus fulvestrant for postmenopausal patients with hormone receptor-positive advanced breast cancer. METHODS Pubmed, Embase and Web of Science databases were systematically searched on February 26, 2018. Eligible studies were screened according to selection criteria, and two reviewers independently extracted outcome data which included progression-free survival, overall survival, objective response rate, clinical benefit rate and toxicities. RevMan 5.3 and STATA 11.0 software were used to conduct meta-analysis. RESULTS Thirteen articles including twelve randomized-control trials fulfilled selection criteria. There was no evidence regarding the existence of publication bias and high-risk bias of quality in the selected studies. In previously endocrine therapy-treated postmenopausal patients with hormone-receptor positive advanced breast cancer, the PFS (HR = 0.77, 95%CI: 0.66-0.91) and ORR (RR = 1.78, 95%CI: 1.35-2.34) of combination therapy group were significantly higher than that from fulvestrant monotherapy group. Besides, a statistically significant difference in PFS was found across the two arms in postmenopausal women with PIK3CA-mutant ctDNA tumor (HR = 0.52, 95% CI: 0.39-0.69). Moreover, the risk of adverse events (RR = 1.09, 95%CI: 1.05-1.13), CTCAE≥3 (RR = 1.97, 95%CI: 1.49-2.60) and discontinuation due to adverse events (RR = 4.91, 95%CI: 3.37-7.15) were also significantly different between two treatment groups. Sensitivity analysis showed PLOMA-3 trial was an important factor of heterogeneity. DISCUSSION Even though the combination of targeted therapy plus fulvestrant improved PFS and increased ORR in advanced breast cancer patients, the toxicities of combination therapy were also higher than fulvestrant monotherapy. Further studies related to inhibitors targeting the specific signaling pathway or receptors are urgently needed, and more efforts concerning precision medicine of targeted therapy plus endocrine therapy should be taken to improve the clinical benefits.
Collapse
Affiliation(s)
- Gao Chanchan
- Department of Oncology, Zhongda Hospital of Southeast University, Nanjing, China
| | - Su Xiangyu
- Department of Oncology, Zhongda Hospital of Southeast University, Nanjing, China
| | - Shi Fangfang
- Department of Oncology, Zhongda Hospital of Southeast University, Nanjing, China
| | - Chen Yan
- Department of Oncology, Zhongda Hospital of Southeast University, Nanjing, China
| | - Gu Xiaoyi
- Department of Oncology, Zhongda Hospital of Southeast University, Nanjing, China
| |
Collapse
|
32
|
Siersbæk R, Kumar S, Carroll JS. Signaling pathways and steroid receptors modulating estrogen receptor α function in breast cancer. Genes Dev 2018; 32:1141-1154. [PMID: 30181360 PMCID: PMC6120708 DOI: 10.1101/gad.316646.118] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Estrogen receptor α (ER) is the major driver of ∼75% of breast cancers, and multiple ER targeting drugs are routinely used clinically to treat patients with ER+ breast cancer. However, many patients relapse on these targeted therapies and ultimately develop metastatic and incurable disease, and understanding the mechanisms leading to drug resistance is consequently of utmost importance. It is now clear that, in addition to estrogens, ER function is modulated by other steroid receptors and multiple signaling pathways (e.g., growth factor and cytokine signaling), and many of these pathways affect drug resistance and patient outcome. Here, we review the mechanisms through which these pathways impact ER function and drug resistance as well as discuss the clinical implications.
Collapse
Affiliation(s)
- Rasmus Siersbæk
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| | - Sanjeev Kumar
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
- Addenbrookes Hospital, Cambridge CB2 0QQ, United Kingdom
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| |
Collapse
|
33
|
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: 14] [Impact Index Per Article: 2.3] [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
|
34
|
Abouelghar A, Hasnah R, Taouk G, Saad M, Karam M. Prognostic values of the mRNA expression of natural killer receptor ligands and their association with clinicopathological features in breast cancer patients. Oncotarget 2018; 9:27171-27196. [PMID: 29930758 PMCID: PMC6007477 DOI: 10.18632/oncotarget.25506] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/14/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Natural killer (NK) cells are lymphocytes of the innate immune system that have potent cytotoxic activity against tumor cells. NK cell recognition and activity towards cancer cells are regulated by an integrated interplay between numerous inhibitory and activating receptors acting in concert to eliminate tumor cells expressing cognate ligands. Despite strong evidence supporting the role of NK cells in breast cancer (BC) control, BC still develops and progresses to form large tumors and metastases. A major mechanism of BC escape from NK immunity is the alteration of the expression of NK receptor ligands. The aim of this study was to determine whether NK receptor ligands' mRNA expression might influence prognosis in BC patients and whether these effects differ by molecular subtypes and clinicopathological features. METHODS We used the KM plotter platform to analyze the correlation between mRNA expression of 32 NK receptor ligands and relapse-free survival (RFS) and overall survival (OS) in 3951 and 1402 BC patients, respectively. The association with tumor subtypes and clinicopathological features was determined. BC samples were split into high and low expression groups according to the best cutoff value and the two patient cohorts were compared by Kaplan-Meier survival plots. The hazard ratios with 95% confidence intervals and log rank P values were calculated and FDR-adjusted for multiple testing correction. The data was considered to be statistically significant when FDR-adjusted P value < 0.05. RESULTS High mRNA expression of around 80% of ligands for NK activating and inhibitory receptors associated with better RFS, which correlated with longer OS for only about half of the NK-activating ligands but for most NK-inhibitory ligands. Also, five NK-activating ligands correlated with worse prognosis. These prognostic values were differentially associated with the BC clinical criteria. In addition, the favorable prognostic influence of NK-activating ligands' upregulation, as a whole, was mainly significantly associated with HER2-positive and basal-like subtypes, lymph node positive phenotype, and high-grade tumors. CONCLUSIONS NK receptor ligands appear to play an important role in defining BC patient prognosis. Identification of a group of patients with worse prognosis expressing high levels of NK-activating ligands and low levels of NK-inhibitory ligands makes them ideal potential candidates for NK-based immunotherapy to eliminate residual tumor cells, prevent relapse and improve patient survival.
Collapse
Affiliation(s)
- Ali Abouelghar
- Cancer Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Reem Hasnah
- Department of Biological Sciences, Carnegie Mellon University in Qatar, Doha, Qatar
| | - Ghina Taouk
- Cancer Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Mohamad Saad
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Manale Karam
- Cancer Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| |
Collapse
|
35
|
Patient Case Lessons: Endocrine Management of Advanced Breast Cancer. Clin Breast Cancer 2018; 18:192-204. [DOI: 10.1016/j.clbc.2017.05.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 04/07/2017] [Accepted: 05/22/2017] [Indexed: 12/20/2022]
|
36
|
Patel HK, Bihani T. Selective estrogen receptor modulators (SERMs) and selective estrogen receptor degraders (SERDs) in cancer treatment. Pharmacol Ther 2018; 186:1-24. [DOI: 10.1016/j.pharmthera.2017.12.012] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
37
|
Hayashi T, Hikichi M, Yukitake J, Wakatsuki T, Nishio E, Utsumi T, Harada N. Forskolin increases the effect of everolimus on aromatase inhibitor-resistant breast cancer cells. Oncotarget 2018; 9:23451-23461. [PMID: 29805747 PMCID: PMC5955115 DOI: 10.18632/oncotarget.25217] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 04/06/2018] [Indexed: 01/31/2023] Open
Abstract
Aromatase inhibitor (AI) resistance is a major obstacle in the treatment of estrogen receptor-positive breast cancer. Everolimus (EVE) ameliorates AI-resistant breast cancer and is therefore used in cancer treatment. However, some patients show resistance to EVE. Here, we used 30 clones of long-term estrogen-deprived (LTED) MCF-7 cells as a model of AI-resistant breast cancer. We examined changes in protein phosphatase type 2A (PP2A) and cancerous inhibitor of PP2A (CIP2A), a negative regulator of PP2A, in LTED cells treated with EVE. In LTED cells with high sensitivity to EVE, CIP2A expression decreased at low EVE concentrations; however, in LTED cells poorly sensitive to EVE, CIP2A and PP2A did not change upon exposure to EVE. Therefore, we hypothesized that there is a relation between expression of CIP2A and sensitivity to EVE. Knockdown of CIP2A increased the sensitivity to EVE in three clones poorly sensitive to EVE. Additionally, we found that treatment with FSK, which activates PP2A, increased the sensitivity of the cells to EVE. Our data point to CIP2A and PP2A as novel therapeutic targets for AI-resistant breast cancer.
Collapse
Affiliation(s)
- Takanori Hayashi
- Department of Biochemistry, School of Medicine, Fujita Health University, Aichi, Japan
| | - Masahiro Hikichi
- Department of Breast Surgery, School of Medicine, Fujita Health University, Aichi, Japan
| | - Jun Yukitake
- Department of Clinical Immunology, School of Health Sciences, Fujita Health University, Aichi, Japan
| | - Toru Wakatsuki
- Department of Health Science, School of Medicine, Fujita Health University, Aichi, Japan
| | - Eiji Nishio
- Department of Obstetrics and Gynecology, School of Medicine, Fujita Health University, Aichi, Japan
| | - Toshiaki Utsumi
- Department of Breast Surgery, School of Medicine, Fujita Health University, Aichi, Japan
| | - Nobuhiro Harada
- Department of Biochemistry, School of Medicine, Fujita Health University, Aichi, Japan
| |
Collapse
|
38
|
Olson E. Combination Therapies in Advanced, Hormone Receptor-Positive Breast Cancer. J Adv Pract Oncol 2018; 9:43-54. [PMID: 30564467 PMCID: PMC6296414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
About 17% of women with breast cancer have locally advanced or metastatic disease at the time of diagnosis, and 30% to 40% of women diagnosed with early-stage disease will eventually have recurrence. The majority of breast cancers express estrogen or progesterone receptors, and hormonal therapies (HTs) remain the treatment of choice for these cancers after the resection of primary tumors. In addition to their effectiveness, HTs often have fewer severe side effects compared with chemotherapies. As breast cancer recurs or progresses, however, it becomes less responsive to successive HT, and the duration of response decreases. Recent advances have identified specific combinations of HTs that can extend the duration of response in appropriate patients with advanced breast cancer. Furthermore, research into signaling pathways has led to the availability of targeted agents that improve efficacy and duration of response when used in combination with specific HTs. Despite their effectiveness and advantages, these combination therapies increase the burden of side effects and the care required for proper management. In addition, practitioners must educate patients about the increasing complexity regarding treatment decisions, and provide care as part of a patient-centered team that optimizes both the medical outcomes and quality of life of patients with breast cancer.
Collapse
|
39
|
Martel S, Bruzzone M, Ceppi M, Maurer C, Ponde NF, Ferreira AR, Viglietti G, Del Mastro L, Prady C, de Azambuja E, Lambertini M. Risk of adverse events with the addition of targeted agents to endocrine therapy in patients with hormone receptor-positive metastatic breast cancer: A systematic review and meta-analysis. Cancer Treat Rev 2018; 62:123-132. [DOI: 10.1016/j.ctrv.2017.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 09/27/2017] [Indexed: 10/18/2022]
|
40
|
|
41
|
Liu CY, Wu CY, Petrossian K, Huang TT, Tseng LM, Chen S. Treatment for the endocrine resistant breast cancer: Current options and future perspectives. J Steroid Biochem Mol Biol 2017; 172:166-175. [PMID: 28684381 DOI: 10.1016/j.jsbmb.2017.07.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 05/31/2017] [Accepted: 07/01/2017] [Indexed: 02/07/2023]
Abstract
Endocrine resistance remains a challenge and an unmet need for managing hormone receptor-positive breast cancer. The mechanisms of endocrine resistance are multifaceted and are likely to evolve over time following various single or combination therapies. The purpose of this review article is to provide general understanding of molecular basis of endocrine resistance of breast cancer and to offer comprehensive review on current treatment options and potential new treatment strategies for endocrine resistant breast cancers. Last but not the least, we discuss current challenges and future directions for management of endocrine resistant breast cancers.
Collapse
Affiliation(s)
- Chun-Yu Liu
- Division of Medical Oncology, Department of Oncology, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chia-Yun Wu
- Division of Medical Oncology, Department of Oncology, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Karineh Petrossian
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, United States
| | - Tzu-Ting Huang
- Division of Medical Oncology, Department of Oncology, Taiwan
| | - Ling-Ming Tseng
- Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, United States.
| |
Collapse
|
42
|
Martel S, Maurer C, Lambertini M, Pondé N, De Azambuja E. Breast cancer treatment-induced cardiotoxicity. Expert Opin Drug Saf 2017; 16:1021-1038. [PMID: 28697311 DOI: 10.1080/14740338.2017.1351541] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Breast cancer is the most frequent cancer affecting women worldwide. In every setting, the majority of women are treated with an evergrowing arsenal of therapeutic agents that have greatly improved their outcomes. However, these therapies can also be associated with significant adverse events. Areas covered: This review aims to thoroughly describe the current state of the evidence regarding the potential cardiotoxicity of agents commonly used in the treatment of breast cancer. These include chemotherapeutic agents, anti-HER2 therapies and CDK4/6 and mTOR inhibitors. Furthermore, issues related to the risk stratification and monitoring tools are explored. Expert opinion: Anthracycline- and trastuzumab-related cardiac toxicities have been extensively studied. Substantial evidence is now available concerning additional anti-HER2 agents such as pertuzumab, T-DM1 and tyrosine kinase inhibitors; overall, the cardiotoxicity profile is reassuring. Cardiac events due to endocrine therapy are mostly ischemic and, in the context of prolonged therapy, need specific attention. Novel agents implicated in the treatment of hormone receptor-positive disease are potentially arrhythmogenic and the exact risk will need to be further refined. As for today, assessment of baseline risk factors prior to treatment initiation and cardiac imaging before and during treatment remains the optimal way to prevent cardiac dysfunction. Cardioprotective therapy in primary prevention is still a matter of debate.
Collapse
Affiliation(s)
- Samuel Martel
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium.,b Département d'hémato-oncologie , CISSS Montérégie centre/Hôpital Charles Lemoyne, centre affilié de l'Université de Sherbrooke , Greenfield Park , Qc , Canada
| | - Christian Maurer
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium.,c Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn , University of Cologne , Cologne , Germany
| | - Matteo Lambertini
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium.,d Breast Cancer Translational Research Laboratory, Institut Jules Bordet , Université Libre de Bruxelles (U.L.B) , Brussels , Belgium
| | - Noam Pondé
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium
| | - Evandro De Azambuja
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium
| |
Collapse
|
43
|
Boér K. Fulvestrant in advanced breast cancer: evidence to date and place in therapy. Ther Adv Med Oncol 2017; 9:465-479. [PMID: 28717399 PMCID: PMC5502950 DOI: 10.1177/1758834017711097] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/27/2017] [Indexed: 12/19/2022] Open
Abstract
Breast cancer is a classical hormone-dependent tumour; therefore, endocrine therapy is the mainstay of treatment for hormone receptor-positive, human epidermal growth factor 2-negative advanced breast cancer. Until recently, classical endocrine agents such as tamoxifen, steroidal and nonsteroidal aromatase inhibitors and fulvestrant have been widely used in postmenopausal patients to treat locally advanced or metastatic disease. However, for patients with this subtype of breast cancer, the landscape of endocrine therapy is rapidly changing. Therapies targeting oestrogen modulation have evolved in recent years following the introduction of targeted agents, mTOR and CDK 4/6 inhibitors that are administered in combination with hormone therapy. As a result, options for endocrine therapy have expanded in recent years, and a variety of single-agent or combinations of targeted drugs and endocrine therapies are accepted. Fulvestrant is a selective oestrogen receptor downregulator (SERD) which was introduced to clinical practice in 2002, initially with the indication to treat postmenopausal women with hormone-receptor-positive advanced breast cancer as second-line therapy postdisease progression after aromatase inhibitors or tamoxifen. Additionally, fulvestrant has also been shown to be active in patients previously untreated with endocrine therapy, either both in the neoadjuvant and the metastatic setting, alone or in combination with other targeted therapies. Currently, the standard dose is 500 mg, which is administered with a loading dose. Fulvestrant received a new FDA indication in December 2016, in combination with palbociclib, both in pre/peri/postmenopausal women with breast cancer progressing after endocrine therapy. This manuscript aims to give an overview of new efficacy data and the current role of fulvestrant in the systemic therapy of hormone-receptor-positive advanced breast cancer, in the context of other available therapeutic modalities.
Collapse
Affiliation(s)
- Katalin Boér
- Department of Medical Oncology, Szent Margit Hospital, 132 Bécsi Street, Budapest 1032, Hungary
| |
Collapse
|
44
|
Lin WZ, Xu QN, Wang HB, Li XY. Fulvestrant plus targeted agents versus fulvestrant alone for treatment of hormone-receptor positive advanced breast cancer progressed on previous endocrine therapy: a meta-analysis of randomized controlled trials. Breast Cancer 2017; 24:345-352. [PMID: 28324247 DOI: 10.1007/s12282-017-0770-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 03/14/2017] [Indexed: 02/05/2023]
Abstract
To compare the addition of targeted agents to fulvestrant with fulvestrant alone in hormone-receptor positive advanced breast cancer progressed on previous endocrine therapy; a meta-analysis of all relevant randomized controlled trials was performed. The PubMed, Embase databases and the Cochrane Central Register of Controlled Trials were searched for relevant publications reporting randomized controlled trials between January 2000 and June 2016. Progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), and toxicity were assessed. Eight trials with a total of 2,470 patients were included in this meta-analysis. Compared with fulvestrant alone, combination therapy improved PFS (HR = 0.79; 95% CI 0.72-0.87; P = 0.00), increased ORR (RR = 1.70; 95% CI 1.30-2.21; P = 0.00), and showed a trend of increase in DCR (RR = 1.27; 95% CI 0.96-1.69, P = 0.09). In network analysis, only CD4/6 and PI3K/m-TOR inhibitors showed significant treatment effects with a P-score of 0.9999 and 0.7615, respectively. Patients treated with combination therapy developed more grade 3 or greater toxic effects (RR = 1.24; 95% CI 1.13-1.36; P = 0.00). Combining targeted agents with fulvestrant showed benefit but with increased toxicity in patients with advanced breast cancer compared with fulvestrant alone. Biomarkers for treatment optimization are lacking. The CD4/6 and PI3K/m-TOR pathways merit further investigation.
Collapse
Affiliation(s)
- Wen-Zhao Lin
- Department of Medical Oncology, Shantou Central Hospital, Shantou, Guangdong, China
| | - Qi-Ni Xu
- Department of Respiratory Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Hong-Biao Wang
- Department of Respiratory Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xu-Yuan Li
- Department of Medical Oncology, Shantou Central Hospital, Shantou, Guangdong, China.
| |
Collapse
|
45
|
Chakraborty A, Hatzis C, DiGiovanna MP. Co-targeting the HER and IGF/insulin receptor axis in breast cancer, with triple targeting with endocrine therapy for hormone-sensitive disease. Breast Cancer Res Treat 2017; 163:37-50. [PMID: 28236033 DOI: 10.1007/s10549-017-4169-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/17/2017] [Indexed: 01/22/2023]
Abstract
PURPOSE Interactions between HER2, estrogen receptor (ER), and insulin-like growth factor I receptor (IGF1R) are implicated in resistance to monotherapies targeting these receptors. We have previously shown in pre-clinical studies synergistic anti-tumor effects for co-targeting each pairwise combination of HER2, IGF1R, and ER. Strikingly, synergy for HER2/IGF1R targeting occurred not only in a HER2+ model, but also in a HER2-normal model. The purpose of the current study was therefore to determine the generalizability of synergistic anti-tumor effects of co-targeting HER2/IGF1R, the anti-tumor activity of triple-targeting HER2/IGF1R/ER in hormone-dependent cell lines, and the effect of using the multi-targeting drugs neratinib (pan-HER) and BMS-754807 (dual IGF1R/insulin receptor). METHODS Proliferation and apoptosis assays were performed in a large panel of cell lines representing varying receptor expression levels. Mechanistic effects were studied using phospho-protein immunoblotting. Analyses of drug interaction effects were performed using linear mixed-effects regression models. RESULTS Enhanced anti-proliferative effects of HER/IGF-insulin co-targeting were seen in most, though not all, cell lines, including HER2-normal lines. For ER+ lines, triple targeting with inclusion of anti-estrogen generally resulted in the greatest anti-tumor effects. Double or triple targeting generally resulted in marked increases in apoptosis in the sensitive lines. Mechanistic studies demonstrated that the synergy between drugs was correlated with maximal inhibition of Akt and ERK pathway signaling. CONCLUSIONS Dual HER/IGF-insulin targeting, and triple targeting with inclusion of anti-estrogen drugs, shows striking anti-tumor activity across breast cancer types, and drugs with broader receptor specificity may be more effective than single receptor selective drugs, particularly for ER- cells.
Collapse
Affiliation(s)
- Ashok Chakraborty
- Section of Medical Oncology, Departments of Internal Medicine, Yale Cancer Center, Smilow Cancer Hospital, Yale University School of Medicine, 300 George Street, Suite 120, New Haven, CT, 06510, USA
| | - Christos Hatzis
- Section of Medical Oncology, Departments of Internal Medicine, Yale Cancer Center, Smilow Cancer Hospital, Yale University School of Medicine, 300 George Street, Suite 120, New Haven, CT, 06510, USA
| | - Michael P DiGiovanna
- Section of Medical Oncology, Departments of Internal Medicine, Yale Cancer Center, Smilow Cancer Hospital, Yale University School of Medicine, 300 George Street, Suite 120, New Haven, CT, 06510, USA.
| |
Collapse
|
46
|
Koleva-Kolarova RG, Oktora MP, Robijn AL, Greuter MJW, Reyners AKL, Buskens E, de Bock GH. Increased life expectancy as a result of non-hormonal targeted therapies for HER2 or hormone receptor positive metastatic breast cancer: A systematic review and meta-analysis. Cancer Treat Rev 2017; 55:16-25. [PMID: 28288388 DOI: 10.1016/j.ctrv.2017.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 01/02/2017] [Accepted: 01/03/2017] [Indexed: 11/28/2022]
Abstract
This article aimed to assess the clinical effectiveness of non-hormonal targeted therapies (TTs) in terms of increase of median progression-free survival (PFS) and overall survival (OS) in receptor-positive metastatic breast cancer (MBC) patients by performing a systematic review and meta-analysis. We systematically searched relevant randomized controlled trials and extracted data about number of patients on targeted and comparator therapy, receptor status, line of treatment, median PFS and OS, p values, hazard ratios (HRs) and 95% confidence intervals (CI). Inverse variance was used to estimate pooled HRs, chi-square test for heterogeneity and Jadad scale for quality were applied. Thirty-eight studies (n=17,192 patients) were eligible for inclusion. TTs added 3.3months to the median PFS [0.7-9.6; HRs 0.74, 95% CI 0.71-0.77] of receptor-positive MBC patients and prolonged their median OS with 3.5months [0-4.7; HRs 0.90, 95% CI 0.82-0.98]. The highest increase in median PFS of 3.6months was found in HER2-/hormone receptor(HR)+ patients, while the highest increase in median OS of 7.2months was observed in HER2+/HRmixed status patients. First-line TTs were most effective in increasing the median PFS in the HR+/HER2- group with 2.0months, and in the HER2+/HRmixed group by adding 4.7months to the median OS. Second-line TTs were most effective for HER2-/HR+ patients by adding 2.6months to their PFS, and for HER2+/HRmixed patients by adding 3.1months to their median OS. Albeit small, the gain in months of median PFS and median OS was significant. Importantly, the results reported show large variation, and thus routinely applying a personalized approach seems warranted.
Collapse
Affiliation(s)
- Rositsa G Koleva-Kolarova
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700RB Groningen, The Netherlands; Department of Primary Care and Public Health Sciences, Division of Health and Social Care Research, King's College London, Guy's, AH 3.2, SE1 1UL London, United Kingdom.
| | - Monika P Oktora
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700RB Groningen, The Netherlands.
| | - Annelies L Robijn
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700RB Groningen, The Netherlands.
| | - Marcel J W Greuter
- Department of Radiology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700RB Groningen, The Netherlands.
| | - Anna K L Reyners
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700RB Groningen, The Netherlands.
| | - Erik Buskens
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700RB Groningen, The Netherlands.
| | - Geertruida H de Bock
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700RB Groningen, The Netherlands.
| |
Collapse
|
47
|
Surrogacy of progression free survival for overall survival in metastatic breast cancer studies: Meta-analyses of published studies. Contemp Clin Trials 2017; 53:20-28. [DOI: 10.1016/j.cct.2016.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 11/30/2016] [Accepted: 12/03/2016] [Indexed: 11/19/2022]
|
48
|
Freedman RA, Foster JC, Seisler DK, Lafky JM, Muss HB, Cohen HJ, Mandelblatt J, Winer EP, Hudis CA, Partridge AH, Carey LA, Cirrincione C, Moreno-Aspitia A, Kimmick G, Jatoi A, Hurria A. Accrual of Older Patients With Breast Cancer to Alliance Systemic Therapy Trials Over Time: Protocol A151527. J Clin Oncol 2017; 35:421-431. [PMID: 27992272 PMCID: PMC5455700 DOI: 10.1200/jco.2016.69.4182] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose Despite increasing awareness of accrual challenges, it is unknown if accrual of older patients to breast cancer treatment trials is improving. Methods We examined accrual of older patients to Alliance for Clinical Trials in Oncology systemic therapy breast cancer trials during 1985-2012 and compared disease characteristics and reasons for therapy cessation for older (age ≥ 65 years and ≥ 70 years) versus younger (age < 65 years and < 70 years) participants. To examine accrual trends, we modeled age as a function of time, using logistic regression. Results Overall, 17% of study participants were ≥ 65 years of age. Approximately 15%, 24%, and 24% of participants in adjuvant, neoadjuvant, and metastatic trials were age ≥ 65 years, and 7%, 15%, and 13% were age ≥ 70 years, respectively. The odds of a patient age ≥ 65 years enrolling significantly increased over time for adjuvant trials (odds ratio [OR] per year, 1.04; 95% CI, 1.04 to 1.05) but decreased significantly for neoadjuvant and metastatic trials (OR, 0.62; 95% CI, 0.58 to 0.67 and OR, 0.98, 95% CI, 0.97 to 1.00). Similar trends were seen for those age ≥ 70 years but these were statistically significant for adjuvant and neoadjuvant trials only (OR, 1.05, 95% CI, 1.04 to 1.07; and OR, 0.57, 95% CI, 0.52 to 0.62). In general, those age ≥ 65 years ( v those < 65 years) in adjuvant studies had a higher mean number of lymph nodes involved and more hormone receptor-negative tumors, although tumor sizes were similar. Early protocol treatment cessation was also more frequent in those age ≥ 65 years (50%) versus < 65 years (35.9%) across trials. Conclusion Older patients with breast cancer remain largely underrepresented in cooperative group therapeutic trials. We observed some improvement in accrual to adjuvant trials but worsening of accrual for neoadjuvant/metastatic trials. Novel strategies to increase accrual of older patients are critical to meaningfully change the evidence base for this growing patient population.
Collapse
Affiliation(s)
- Rachel A. Freedman
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Jared C. Foster
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Drew K. Seisler
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Jacqueline M. Lafky
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Hyman B. Muss
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Harvey J. Cohen
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Jeanne Mandelblatt
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Eric P. Winer
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Clifford A. Hudis
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Ann H. Partridge
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Lisa A. Carey
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Constance Cirrincione
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Alvaro Moreno-Aspitia
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Gretchen Kimmick
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Aminah Jatoi
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| | - Arti Hurria
- Rachel A. Freedman, Eric P. Winer, and Ann H. Partridge, Dana-Farber Cancer Institute, Boston, MA; Jared C. Foster, Drew K. Seisler, Jacqueline M. Lafky, and Aminah Jatoi, Mayo Clinic; Jared C. Foster and Drew K. Seisler, Mayo Cancer Center, Rochester, MN; Hyman B. Muss and Lisa A. Carey, University of North Carolina, Chapel Hill, NC; Harvey J. Cohen, Constance Cirrincione, and Gretchen Kimmick, Duke University, Durham, NC; Jeanne Mandelblatt, Georgetown University, Washington, DC; Clifford A. Hudis, Memorial Sloan-Kettering Cancer Center, New York, NY; Alvaro Moreno-Aspitia, Mayo Clinic, Jacksonville, FL; and Arti Hurria, City of Hope, Duarte, CA
| |
Collapse
|
49
|
Chia SK, Ellard SL, Mates M, Welch S, Mihalcioiu C, Miller WH, Gelmon K, Lohrisch C, Kumar V, Taylor S, Hagerman L, Goodwin R, Wang T, Sakashita S, Tsao MS, Eisenhauer E, Bradbury P. A phase-I study of lapatinib in combination with foretinib, a c-MET, AXL and vascular endothelial growth factor receptor inhibitor, in human epidermal growth factor receptor 2 (HER-2)-positive metastatic breast cancer. Breast Cancer Res 2017; 19:54. [PMID: 28464908 PMCID: PMC5414192 DOI: 10.1186/s13058-017-0836-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 03/16/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The mechanisms of resistance to anti-human epidermal growth factor receptor 2 (HER 2) therapies are unclear but may include the tyrosine-protein kinase Met (c-Met), vascular endothelial growth factor (VEGF) and AXL pathways. Foretinib is an inhibitor of c-Met, VEGF receptor 2 (VEGFR-2), platelet-derived growth factor receptor beta (PDGFRB), AXL, Fms-like tyrosine kinase 3 (FLT3), angiopoiten receptor (TIE-2), RET and RON kinases. This phase Ib study sought to establish the associated toxicities, pharmacokinetics (PK) and recommended phase II doses (RP2D) of foretinib and lapatinib in a cohort of HER-2-positive patients with metastatic breast cancer (MBC). METHODS Women with HER-2 positive MBC, Performance status (PS 0-2), and no limit on number of prior chemotherapies or lines of anti-HER-2 therapies were enrolled. A 3 + 3 dose escalation design was utilized. Four dose levels were intended with starting doses of foretinib 30 mg and lapatinib 750 mg orally once a day (OD) on a 4-weekly cycle. Assessment of c-MET status from the primary archival tissue was performed. RESULTS We enrolled 19 patients, all evaluable for toxicity assessment and for response evaluation. Median age was 60 years (34-86 years), 95% were PS 0-1, 53% were estrogen receptor-positive and 95% had at least one prior anti-HER-2-based regimen. The fourth dose level was reached (foretinib 45 mg/lapatinib 1250 mg) with dose-limiting toxicities of grade-3 diarrhea and fatigue. There was only one grade-4 non-hematological toxicity across all dose levels. There were no PK interactions between the agents. A median of two cycles was delivered across the dose levels (range 1-20) with associated progression-free survival of 3.2 months (95% CI 1.61-4.34 months). By immunohistochemical assessment with a specified cutoff, none of the 17 samples tested were classified as positive for c-Met. CONCLUSIONS The RP2D of the combined foretinib and lapatinib is 45 mg and 1000 mg PO OD, respectively. Limited activity was seen with this combination in a predominantly unselected cohort of HER-2-positive patients with MBC.
Collapse
Affiliation(s)
- Stephen K. Chia
- 0000 0001 0702 3000grid.248762.dMedical Oncology, British Columbia Cancer Agency (BCCA), Vancouver, BC Canada
| | - Susan L. Ellard
- 0000 0001 0702 3000grid.248762.dMedical Oncology, BCCA, Kelowna, BC Canada
| | - Mihaela Mates
- 0000 0004 0633 727Xgrid.415354.2Queen’s University and Cancer Centre of South Eastern Ontario at Kingston General Hospital, Kingston, ON Canada
| | - Stephen Welch
- 0000 0000 9132 1600grid.412745.1London Regional Cancer Program, London, ON Canada
| | - Catalin Mihalcioiu
- 0000 0004 1936 8649grid.14709.3bJewish General Hospital and Rossy Cancer Network, McGill University, Montreal, QC Canada
| | - Wilson H. Miller
- 0000 0004 1936 8649grid.14709.3bJewish General Hospital and Rossy Cancer Network, McGill University, Montreal, QC Canada
| | - Karen Gelmon
- 0000 0001 0702 3000grid.248762.dMedical Oncology, British Columbia Cancer Agency (BCCA), Vancouver, BC Canada
| | - Caroline Lohrisch
- 0000 0001 0702 3000grid.248762.dMedical Oncology, British Columbia Cancer Agency (BCCA), Vancouver, BC Canada
| | - Vikaash Kumar
- 0000 0004 0633 727Xgrid.415354.2Queen’s University and Cancer Centre of South Eastern Ontario at Kingston General Hospital, Kingston, ON Canada
| | - Sara Taylor
- 0000 0001 0702 3000grid.248762.dMedical Oncology, BCCA, Kelowna, BC Canada
| | | | - Rachel Goodwin
- 0000 0000 9606 5108grid.412687.eThe Ottawa Hospital Cancer Centre, Ottawa, ON Canada
| | - Tao Wang
- 0000 0001 2157 2938grid.17063.33Princess Margaret Cancer Centre and University Health Network, University of Toronto, Toronto, ON Canada
| | - Shingo Sakashita
- 0000 0001 2157 2938grid.17063.33Princess Margaret Cancer Centre and University Health Network, University of Toronto, Toronto, ON Canada
| | - Ming S. Tsao
- 0000 0001 2157 2938grid.17063.33Princess Margaret Cancer Centre and University Health Network, University of Toronto, Toronto, ON Canada
| | - Elizabeth Eisenhauer
- 0000 0004 0633 727Xgrid.415354.2Queen’s University and Cancer Centre of South Eastern Ontario at Kingston General Hospital, Kingston, ON Canada
| | - Penelope Bradbury
- 0000 0001 2157 2938grid.17063.33Princess Margaret Cancer Centre and University Health Network, University of Toronto, Toronto, ON Canada
| |
Collapse
|
50
|
Pritchard KI, Chia SK, Simmons C, McLeod D, Paterson A, Provencher L, Rayson D. Enhancing Endocrine Therapy Combination Strategies for the Treatment of Postmenopausal HR+/HER2- Advanced Breast Cancer. Oncologist 2016; 22:12-24. [PMID: 27864574 DOI: 10.1634/theoncologist.2016-0185] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/01/2016] [Indexed: 01/16/2023] Open
Abstract
Breast cancer (BC) is the most common malignancy in women worldwide, with approximately two-thirds having hormone receptor-positive (HR+) tumors. New endocrine therapy (ET) strategies include combining ET agents as well as adding inhibitors targeting growth factors, angiogenesis, the mechanistic target of rapamycin, phosphoinositide 3-kinase (PI3K), or cyclin-dependent kinase 4/6 to ET. Level 1 evidence supports use of fulvestrant plus anastrozole or palbociclib plus letrozole as first-line therapy for HR+/HER- advanced BC with special consideration for the former in ET-naïve patients, as well as everolimus plus exemestane or palbociclib plus fulvestrant as second-line therapy with special consideration in select first-line patients. Although the safety profiles of these combinations are generally predictable and manageable, both everolimus and palbociclib are associated with an increased risk of potentially serious or early-onset toxicities requiring individualized a priori adverse event risk stratification, earlier and more rigorous agent-specific monitoring, and patient education. Although each of these combinations improves progression-free survival, none with the exception of anastrazole plus fulvestrant have demonstrated improved overall survival. PI3K catalytic-α mutations assessed from circulating tumor DNA represent the first potentially viable serum biomarker for the selection of ET combinations, and new data demonstrate the feasibility of this minimally invasive technique as an alternative to traditional tissue analysis. Therapeutic ratios of select ET combinations support their use in first- and second-line settings, but optimal sequencing has yet to be determined. THE ONCOLOGIST 2017;22:12-24 IMPLICATIONS FOR PRACTICE: Emerging data show that new endocrine therapy (ET) combinations can improve progression-free and overall survival outcomes in patients with hormone receptor-positive, HER2-negative (HR+/HER-) advanced breast cancer. Level 1 evidence supports consideration of dual ET regimens, particularly in ET-naïve patients, or palbociclib plus letrozole as first-line therapy, as well as the addition of mTOR or CDK4/6 inhibitors to established ET in the second-line setting and in select first-line patients. Some combinations are associated with increased risk of class-specific toxicities that will require individualized risk stratification, earlier and more rigorous agent-specific monitoring, and patient education. Recent data on a noninvasive biomarker assay that predicts response to a phosphoinositide 3-kinase inhibitor demonstrates the feasibility of this minimally invasive technique as an alternative to traditional tissue analysis.
Collapse
Affiliation(s)
- Kathleen I Pritchard
- Sunnybrook Odette Cancer Centre and University of Toronto, Toronto, Ontario, Canada
| | - Stephen K Chia
- British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | | | - Deanna McLeod
- Kaleidoscope Strategic, Inc., Toronto, Ontario, Canada
| | | | | | - Daniel Rayson
- Division of Medical Oncology, Dalhousie University, and Atlantic Clinical Cancer Research Unit, Halifax, Nova Scotia, Canada
| |
Collapse
|