1
|
Wang H, Zhang G, Gao W, Li S, Yang S, Zhang X, Wang J, Li C, Gao S, Geng C. Dalpiciclib plus aromatase inhibitor versus neoadjuvant chemotherapy for ER-positive, HER2-negative breast cancer. Front Oncol 2025; 15:1566146. [PMID: 40371225 PMCID: PMC12074949 DOI: 10.3389/fonc.2025.1566146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
Background The combination of cyclin-dependent kinases 4 and 6 inhibitors (CDK4/6i) with endocrine therapy (ET) has emerged as an effective alternative to neoadjuvant chemotherapy (NCT) for patients with hormone receptor-positive (HR+) and human epidermal growth factor receptor 2 (HER2) -negative breast cancer (BC). This single-center study retrospectively evaluated the efficacy and safety of dalpiciclib combined with an aromatase inhibitor (AI) compared to NCT. Methods The clinicopathological data and treatment details of patients with HR+ HER2 negative BC who underwent either neoadjuvant endocrine therapy (NET) or NCT were collected from the Fourth Hospital of Hebei Medical University. The primary endpoint of the study was the Residual Cancer Burden (RCB), while secondary endpoints included breast pathological complete response (bpCR), clinical response rates (ORR), proliferation markers, and safety profiles. Results Between May 2022 and June 2023, a total of 36 patients were treated with dalpiciclib plus AI, while 137 patients received NCT for the final analysis. Prior to propensity score matching (PSM), the rates of RCB 0 were 0% in the NET group and 7.3% in the NCT group (p=0.205). The rates of bpCR were 2.8% for the NET group and 13.1% for the NCT group (p=0.142). The ORR was comparable between the two groups (p=0.397), as were the rates of BCS (p=0.608). Both treatment groups demonstrated significant reductions in Ki-67 levels, although the extent of reduction was similar (p=0.174). Notably, a Ki-67 level of ≤ 10% post-treatment was more prevalent in the NET group (p<0.0001). Only two patients in the NET group achieved a Preoperative Endocrine Prognostic Index (PEPI) 0 score. The incidence of grade 3-4 toxicities was significantly higher in the NCT group compared to the NET group (p<0.05). Following PSM, patients treated with dalpiciclib plus AI exhibited comparable clinical responses and a safety advantage relative to those receiving NCT. Conclusion This study indicates that the combination of dalpiciclib and AI elicits comparable responses and demonstrates improved safety profiles when compared to NCT in patients with HR+ HER2 negative breast tumors. Furthermore, RCB and pCR may not serve as optimal endpoints for evaluating the efficacy of CDK4/6i-based NET.
Collapse
Affiliation(s)
- Haoqi Wang
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Breast Cancer Molecular Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Guoyu Zhang
- Department of Breast Surgery, Handan Central Hospital, Handan, Hebei, China
| | - Wei Gao
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sainan Li
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Breast Cancer Molecular Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shan Yang
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Breast Cancer Molecular Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xi Zhang
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Breast Cancer Molecular Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiaxing Wang
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Breast Cancer Molecular Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chunxiao Li
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shan Gao
- Gland Surgery, The Hebei Province People’s Hospital, Shijiazhuang, Hebei, China
| | - Cuizhi Geng
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Breast Cancer Molecular Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
2
|
Chen A, Kim BJ, Mitra A, Vollert CT, Lei JT, Fandino D, Anurag M, Holt MV, Gou X, Pilcher JB, Goetz MP, Northfelt DW, Hilsenbeck SG, Marshall CG, Hyer ML, Papp R, Yin SY, De Angelis C, Schiff R, Fuqua SAW, Ma CX, Foulds CE, Ellis MJ. PKMYT1 Is a Marker of Treatment Response and a Therapeutic Target for CDK4/6 Inhibitor-Resistance in ER+ Breast Cancer. Mol Cancer Ther 2024; 23:1494-1510. [PMID: 38781103 PMCID: PMC11443213 DOI: 10.1158/1535-7163.mct-23-0564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 03/25/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
Endocrine therapies (ET) with cyclin-dependent kinase 4/6 (CDK4/6) inhibition are the standard treatment for estrogen receptor-α-positive (ER+) breast cancer, however drug resistance is common. In this study, proteogenomic analyses of patient-derived xenografts (PDXs) from patients with 22 ER+ breast cancer demonstrated that protein kinase, membrane-associated tyrosine/threonine one (PKMYT1), a WEE1 homolog, is estradiol (E2) regulated in E2-dependent PDXs and constitutively expressed when growth is E2-independent. In clinical samples, high PKMYT1 mRNA levels associated with resistance to both ET and CDK4/6 inhibition. The PKMYT1 inhibitor lunresertib (RP-6306) with gemcitabine selectively and synergistically reduced the viability of ET and palbociclib-resistant ER+ breast cancer cells without functional p53. In vitro the combination increased DNA damage and apoptosis. In palbociclib-resistant, TP53 mutant PDX-derived organoids and PDXs, RP-6306 with low-dose gemcitabine induced greater tumor volume reduction compared to treatment with either single agent. Our study demonstrates the clinical potential of RP-6306 in combination with gemcitabine for ET and CDK4/6 inhibitor resistant TP53 mutant ER+ breast cancer.
Collapse
Affiliation(s)
- Anran Chen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, Texas
- Repare Therapeutics, Cambridge, Massachusetts
| | - Beom-Jun Kim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Aparna Mitra
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Craig T Vollert
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Employee of Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana
| | - Jonathan T Lei
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas
| | - Diana Fandino
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Meenakshi Anurag
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Matthew V Holt
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Xuxu Gou
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Jacob B Pilcher
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | | | - Donald W Northfelt
- Division of Hematology and Medical Oncology at Mayo Clinic, Phoenix, Arizona
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | | | - Marc L Hyer
- Repare Therapeutics, Cambridge, Massachusetts
| | - Robert Papp
- Repare Therapeutics, Saint-Laurent, Quebec, Canada
| | - Shou-Yun Yin
- Repare Therapeutics, Saint-Laurent, Quebec, Canada
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Suzanne A W Fuqua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Cynthia X Ma
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Charles E Foulds
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
3
|
Jeong H, Kim SB. Neoadjuvant endocrine therapy in ER-positive breast cancer: evolution, indication, and tailored treatment strategy. Ther Adv Med Oncol 2023; 15:17588359231200457. [PMID: 37786536 PMCID: PMC10541763 DOI: 10.1177/17588359231200457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 08/25/2023] [Indexed: 10/04/2023] Open
Abstract
In recent years, endocrine therapy (ET), an effective systemic treatment for the management of estrogen receptor (ER)-positive breast cancers, has regained interest as a neoadjuvant therapy based on evidence that ET can fulfill the aim of neoadjuvant systemic treatment for tumor shrinkage as well as elucidate important clinical information on endocrine sensitivity that enables the prognostication of patients. Moreover, neoadjuvant endocrine therapy (NET) potentially provides an opportunity for early assessment of the clinical efficacy of novel agents. Furthermore, recently reported trials have generated evidence for a more tailored approach for perioperative management of ER-positive breast cancer using clinical and molecular biomarkers, and this has provided a rationale that enables the broadening of clinical indications for NET. This review discusses the current evidence for NET, the evolution of NET trials, clinical indications, and NET-based treatment strategies.
Collapse
Affiliation(s)
- Hyehyun Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Republic of Korea
| |
Collapse
|
4
|
Li S, Yu X, Xu Y. Breast cancer gene expression signatures: development and clinical significance-a narrative review. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2022; 4:7. [PMID: 38751471 PMCID: PMC11093086 DOI: 10.21037/tbcr-22-39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/18/2022] [Indexed: 05/18/2024]
Abstract
Background and Objective Breast cancer gene expression signatures are developing rapidly and are expected to better understand the intrinsic features of the tumor, and also to optimize the treatment strategy in clinical practice. This review is to summarize the controversy and consensus in clinical practice of gene expression signatures, and to provide our perspective on these issues as well as recommendation for future direction. Methods We reviewed English publications in PubMed related to breast cancer gene expression signatures from 2002 to 2022. Key Content and Findings Five mature commercial gene expression signatures: Oncotype, MammaPrint, Prosigna/PAM50, EndoPredict and Breast Cancer Index (BCI) are available to provide the prognostic and predictive assessment. Although they could help to evaluate the risk of recurrence and to predict the benefits of certain treatments, their applications remain challenging. Treatment decisions should be determined by a combination of related clinical pathological factors in clinical practice. Conclusions Gene expression signatures could assist in the determination of the adjuvant therapy of early-stage breast cancer. The prospective randomized clinical trials showed that chemotherapy may be exempted in low-risk patients. More sufficient data are expected for the application in radiotherapy, extended endocrine therapy, and neoadjuvant treatment. The treatment cannot be determined by a single factor but by comprehensive assessments of clinicopathological factors, test purpose, and cost-effectiveness. Patients will benefit from personalized treatments with the publication of further evidence.
Collapse
Affiliation(s)
- Siyan Li
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Xinmiao Yu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yingying Xu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Gouveia MC, Amorim de Araújo Lima Santos C, Impieri Souza A. Study protocol: Randomized, open-label, non-inferiority clinical trial for evaluating the clinical and pathological response rates to neoadjuvant hormone therapy and chemotherapy in patients with luminal-subtype breast tumors. Contemp Clin Trials Commun 2022; 30:101013. [PMID: 36262803 PMCID: PMC9574413 DOI: 10.1016/j.conctc.2022.101013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/19/2022] [Accepted: 10/01/2022] [Indexed: 11/04/2022] Open
Abstract
Background Despite neoadjuvant hormone therapy (NHT) is being underused, it is an effective treatment for luminal tumors at a lower cost and with fewer side effects compared to those associated with neoadjuvant chemotherapy (NCT). The lack of robust comparative data between NHT and NCT is a factor that limits its use in clinical practice. Methods This study will be a randomized, open-label, non-inferiority clinical trial. Patients diagnosed with HER2-negative luminal-subtype breast cancer will be identified at the time of diagnosis. Menopausal patients randomized for NHT should receive anastrozole for at least six months. Premenopausal women should receive anastrozole associated with subcutaneous goserelin acetate every 12 weeks for at least six months. Patients randomized for NCT will receive a standard institutional regimen based on anthracyclines and taxanes. Sample size was calculated considering the CPS + EG as a method for evaluating response and prognosis, where a score <3 was defined as good. The non-inferiority margin for NHT was set at 15%. The study considered a power of 80%, a significance level of 5%, and an outcome proportion in each group of 69%, resulting in 118 patients in each group. We estimated at 10% of losses, resulting in a sample of 130 patients in each group. Conclusion The non-inferiority of NHT in relation to NCT will provide further evidence that replacing NCT with NHT is safe and effective in eligible patients, which is particularly relevant for populations with limited access to health services and for institutions with few available resources.
Collapse
Affiliation(s)
- Maria Carolina Gouveia
- Corresponding author.. Research department, Rua do Coelhos, 300, Boa Vista, Recife, PE, 50070-550, Brazil.
| | | | | |
Collapse
|
6
|
Türkmen NB, Çiftçi O, Taşlıdere A, Aydın M, Eke BC. The effect of aromatase inhibitors against possible testis toxicity in pembrolizumab treated rats. Andrologia 2022; 54:e14557. [DOI: 10.1111/and.14557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/17/2022] [Accepted: 08/01/2022] [Indexed: 11/28/2022] Open
Affiliation(s)
- Neşe Başak Türkmen
- Faculty of Pharmacy, Department of Pharmaceutical Toxicology Inonu University Malatya Turkey
| | - Osman Çiftçi
- Faculty of Medicine, Department of Medicinal Pharmacology Pamukkale University Denizli Turkey
| | - Aslı Taşlıdere
- Faculty of Medicine, Histology and Embryology Department Inonu University Malatya Turkey
| | - Muhterem Aydın
- Faculty of Veterinary Medicine, Department of Obstetrics and Gynecology Firat University Elazig Turkey
| | - Binay Can Eke
- Faculty of Pharmacy, Department of Pharmaceutical Toxicology Ankara University Ankara Turkey
| |
Collapse
|
7
|
Wang X, Fan Z, Wang X, He Y, Liu Y, Wang X, Zhang B, Jiang Z, Wang T, Yu Z, Wang F, Liu Y, Li Y, Zhang J, Luo B, Jiang H, Wang T, Xie Y, Li J, Ouyang T. Neoadjuvant endocrine therapy for strongly hormone receptor-positive and HER2-negative early breast cancer: results of a prospective multi-center study. Breast Cancer Res Treat 2022; 195:301-310. [PMID: 35917052 DOI: 10.1007/s10549-022-06686-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/08/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE For estrogen receptor (ER)-positive breast cancer, neoadjuvant endocrine therapy (NET) has been shown to be as effective as neoadjuvant chemotherapy (NACT). We evaluated the prognostic significance of Preoperative Endocrine Prognostic Index (PEPI). METHODS We conducted a prospective, multi-center, non-randomized, controlled trial that enrolled postmenopausal early-stage strongly ER-positive (≥ 50%) and HER2-negative breast cancer patients. All patients were given 4-month NET before surgery. The primary objective was to investigate the 5-year recurrence-free survival (RFS) in patients who had PEPI 0-1 or pathological complete response (pCR) without chemotherapy. Patients who had PEPI 0-1 or pCR were recommended to receive adjuvant endocrine therapy only and patients had PEPI ≥ 2 may receive adjuvant chemotherapy at the discretion of the treating physician. RESULTS A total of 410 patients were included and 352 patients constituted the per-protocol population. Overall, 9 patients (2.5%) had pCR (ypT0/is ypN0), 128 patients (36.4%) had PEPI = 0, and 56 patients (15.9%) had PEPI = 1. After a median follow-up of 60 months (4-104 months), patients who had PEPI 0-1 or pCR showed an improved 5-year RFS [99.5% (95% CI 98.5-99.9%) for PEPI 0-1 or pCR group vs. 93.7% (95% CI 89.6-97.8%) for PEPI ≥ 2 group, P = 0.028]. No survival difference was detected between patients received adjuvant chemotherapy vs. no chemotherapy among PEPI ≥ 2 cases. CONCLUSION PEPI 0-1 or pCR may be used to define a group of ER-positive and HER2-negative postmenopausal early breast cancer patients with low relapse risk for whom adjuvant chemotherapy can be safely withheld. Studies on the identification and alternative treatment options for endocrine-resistant tumors are warranted. CLINICAL TRIAL REGISTRATION NCT01613560.
Collapse
Affiliation(s)
- Xinguang Wang
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zhaoqing Fan
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xing Wang
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yingjian He
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yiqiang Liu
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiang Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bailin Zhang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zefei Jiang
- Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Tao Wang
- Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Zhigang Yu
- Department of Breast Surgery, Cheeloo College of Medicine, The Second Hospital, Shandong University, Shandong, China
| | - Fei Wang
- Department of Breast Surgery, Cheeloo College of Medicine, The Second Hospital, Shandong University, Shandong, China
| | - Yinhua Liu
- Breast Disease Center, Peking University First Hospital, Beijing, China
| | - Yanping Li
- Department of Breast Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jianguo Zhang
- Department of Breast Surgery, The Second Hospital of Harbin Medical University, Heilongjiang, China
| | - Bin Luo
- Department of General Surgery, Clinical School of Medicine, Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Hongchuan Jiang
- Department of Breast Surgery, Beijing Chao Yang Hospital, Capital Medical University, Beijing, China
| | - Tianfeng Wang
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yuntao Xie
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jinfeng Li
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Tao Ouyang
- Breast Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
8
|
Wang Y, Wang T, Fang H, Huang H, Fang L, Zhang X. Evaluation of Pharmacokinetics and Safety With Bioequivalence of Anastrozole in Healthy Chinese Volunteers: Bioequivalence Study Findings. Clin Pharmacol Drug Dev 2022; 11:687-694. [PMID: 35485182 DOI: 10.1002/cpdd.1099] [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: 06/15/2021] [Accepted: 03/20/2022] [Indexed: 11/06/2022]
Abstract
Anastrozole is a third-generation aromatase inhibitor that exerts potent anti-breast cancer effects. This trial aimed to explore the pharmacokinetics (PK) and safety with bioequivalence of orally administered anastrozole provided by 2 sponsors in healthy volunteers.Two separate open-label, randomized, single-dose, crossover-design studies consisting of a fasting study (n = 23) and a fed study (n = 23, 1 participant withdrew before taking medication) were conducted. In each study, healthy volunteers were randomized to receive the test product (Haizheng Pharmaceutical Group) followed by the reference drug (AstraZeneca Pharmaceuticals LP), or vice versa. Each study subject received a 1-mg anastrozole tablet with a 21-day washout. The plasma concentrations of anastrozole were measured with liquid chromatography-tandem mass spectrometry, and PK parameters were determined by noncompartmental analysis. Forty-six healthy female volunteers were enrolled. For patients enrolled in the fasting study, the mean age was 55.0 years, mean weight was 57.1 kg, mean body mass index was 23.6 kg/m2 , and mean height was 155.5 cm. For patients enrolled in the fed study, the mean age was 54.2 years, mean weight was 55.9 kg, mean body mass index was 23.9 kg/m2 , and mean height was 152.8 cm. All PK end points met the predefined criteria for PK equivalence. In fasting subjects, the median maximum plasma concentration was 23.4 and 22.6 at 1 hour for test and reference formulations, respectively. The maximum plasma concentration in fed subjects was 18.7 and 18.5 at 4 hours for test and reference formulations, respectively. Both fasting and fed studies achieved plausible bioequivalence. Anastrozole was well tolerated and exhibited a favorable safety profile at the prescribed doses. The severity of observed adverse events assessed according to the Common Terminology Criteria for Adverse Events (version CTCAE4.03) was mild, and some of the adverse events were not caused by anastrozole. Furthermore, the results of our study under fasting and fed conditions demonstrated bioequivalence of the test and reference products.
Collapse
Affiliation(s)
- Ying Wang
- Phase I Clinical Research Center, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Tongtong Wang
- Intensive Care Unit, Shaanxi Traditional Chinese Medicine Hospital, Xi'an, China
| | - Han Fang
- Department of Laboratory Medicine, Taizhou First People's Hospital, Taizhou, Zhejiang, China
| | - Hui Huang
- Department of Laboratory Medicine, Quzhou People's Hospital, Quzhou, Zhejiang, China
| | - Lijuan Fang
- Department of Laboratory Medicine, Hangzhou Ninth People's Hospital, Hangzhou, Zhejiang, China
| | - XuanGuo Zhang
- Intensive Care Unit, Shaanxi Traditional Chinese Medicine Hospital, Xi'an, China
| |
Collapse
|
9
|
Martí C, Yébenes L, Oliver JM, Moreno E, Frías L, Berjón A, Loayza A, Meléndez M, Roca MJ, Córdoba V, Hardisson D, Rodríguez MÁ, Sánchez-Méndez JI. The Clinical Impact of Neoadjuvant Endocrine Treatment on Luminal-like Breast Cancers and Its Prognostic Significance: Results from a Single-Institution Prospective Cohort Study. Curr Oncol 2022; 29:2199-2210. [PMID: 35448153 PMCID: PMC9026529 DOI: 10.3390/curroncol29040179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/15/2022] [Accepted: 03/21/2022] [Indexed: 01/02/2023] Open
Abstract
Purpose: Neoadjuvant endocrine treatment (NET) has become a useful tool for the downstaging of luminal-like breast cancers in postmenopausal patients. It enables us to increase breast- conserving surgery (BCS) rates, provides an opportunity for us to assess in vivo NET effectiveness, and allows us to study any biological changes that may act as valid biomarkers. The purpose of this study was to evaluate the safety and effectiveness of NET, and to assess the role of Ki67 proliferation rate changes as an indicator of endocrine responsiveness. Methods: From 2016 to 2020, a single-institution cohort of patients, treated with NET and further surgery, was evaluated. In patients with Ki67 ≥ 10%, a second core biopsy was performed after four weeks. Information regarding histopathological and clinical changes was gathered. Results: A total of 115 estrogen receptor-positive (ER+)/HER2-negative patients were included. The median treatment duration was 5.0 months (IQR: 2.0−6.0). The median maximum size in the surgical sample was 40% smaller than the pretreatment size measured by ultrasound (p < 0.0001). The median pretreatment Ki67 expression was 20.0% (IQR: 12.0−30.0), and was reduced to 5.0% (IQR: 1.8−10.0) after four weeks, and to 2.0% (IQR: 1.0−8.0) in the surgical sample (p < 0.0001). BCS was performed on 98 patients (85.2%). No pathological complete responses were recorded. A larger Ki67 fold change after four weeks was significantly related to a PEPI score of zero (p < 0.002). No differences were observed between luminal A- and B-like tumors, with regard to fold change and PEPI score. Conclusions: In our cohort, NET was proven to be effective for tumor size and Ki67 downstaging. This resulted in a higher rate of conservative surgery, aided in therapeutic decision making, provided prognostic information, and constituted a safe and well-tolerated approach.
Collapse
Affiliation(s)
- Covadonga Martí
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.Y.); (J.M.O.); (E.M.); (L.F.); (A.B.); (A.L.); (M.M.); (M.J.R.); (V.C.); (D.H.); (M.Á.R.); (J.I.S.-M.)
- Department of Gynecology, Hospital Universitario La Paz, 28046 Madrid, Spain
- Correspondence:
| | - Laura Yébenes
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.Y.); (J.M.O.); (E.M.); (L.F.); (A.B.); (A.L.); (M.M.); (M.J.R.); (V.C.); (D.H.); (M.Á.R.); (J.I.S.-M.)
- Department of Pathology, Hospital Universitario La Paz, 28046 Madrid, Spain
- IdiPaz—Instituto de Investigación La Paz, 28046 Madrid, Spain
| | - José María Oliver
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.Y.); (J.M.O.); (E.M.); (L.F.); (A.B.); (A.L.); (M.M.); (M.J.R.); (V.C.); (D.H.); (M.Á.R.); (J.I.S.-M.)
- Department of Radiology, Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Elisa Moreno
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.Y.); (J.M.O.); (E.M.); (L.F.); (A.B.); (A.L.); (M.M.); (M.J.R.); (V.C.); (D.H.); (M.Á.R.); (J.I.S.-M.)
- Department of Gynecology, Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Laura Frías
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.Y.); (J.M.O.); (E.M.); (L.F.); (A.B.); (A.L.); (M.M.); (M.J.R.); (V.C.); (D.H.); (M.Á.R.); (J.I.S.-M.)
- Department of Gynecology, Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Alberto Berjón
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.Y.); (J.M.O.); (E.M.); (L.F.); (A.B.); (A.L.); (M.M.); (M.J.R.); (V.C.); (D.H.); (M.Á.R.); (J.I.S.-M.)
- Department of Pathology, Hospital Universitario La Paz, 28046 Madrid, Spain
- IdiPaz—Instituto de Investigación La Paz, 28046 Madrid, Spain
| | - Adolfo Loayza
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.Y.); (J.M.O.); (E.M.); (L.F.); (A.B.); (A.L.); (M.M.); (M.J.R.); (V.C.); (D.H.); (M.Á.R.); (J.I.S.-M.)
- Department of Gynecology, Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Marcos Meléndez
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.Y.); (J.M.O.); (E.M.); (L.F.); (A.B.); (A.L.); (M.M.); (M.J.R.); (V.C.); (D.H.); (M.Á.R.); (J.I.S.-M.)
- Department of Gynecology, Hospital Universitario La Paz, 28046 Madrid, Spain
| | - María José Roca
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.Y.); (J.M.O.); (E.M.); (L.F.); (A.B.); (A.L.); (M.M.); (M.J.R.); (V.C.); (D.H.); (M.Á.R.); (J.I.S.-M.)
- Department of Radiology, Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Vicenta Córdoba
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.Y.); (J.M.O.); (E.M.); (L.F.); (A.B.); (A.L.); (M.M.); (M.J.R.); (V.C.); (D.H.); (M.Á.R.); (J.I.S.-M.)
- Department of Radiology, Hospital Universitario La Paz, 28046 Madrid, Spain
| | - David Hardisson
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.Y.); (J.M.O.); (E.M.); (L.F.); (A.B.); (A.L.); (M.M.); (M.J.R.); (V.C.); (D.H.); (M.Á.R.); (J.I.S.-M.)
- Department of Pathology, Hospital Universitario La Paz, 28046 Madrid, Spain
- IdiPaz—Instituto de Investigación La Paz, 28046 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid, 28046 Madrid, Spain
- Center for Biomedical Research in the Cancer Network (CIBERONC), 28029 Madrid, Spain
| | - María Ángeles Rodríguez
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.Y.); (J.M.O.); (E.M.); (L.F.); (A.B.); (A.L.); (M.M.); (M.J.R.); (V.C.); (D.H.); (M.Á.R.); (J.I.S.-M.)
| | - José Ignacio Sánchez-Méndez
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain; (L.Y.); (J.M.O.); (E.M.); (L.F.); (A.B.); (A.L.); (M.M.); (M.J.R.); (V.C.); (D.H.); (M.Á.R.); (J.I.S.-M.)
- Department of Gynecology, Hospital Universitario La Paz, 28046 Madrid, Spain
- IdiPaz—Instituto de Investigación La Paz, 28046 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid, 28046 Madrid, Spain
| |
Collapse
|
10
|
Cyclin-dependent kinase 4 and 6 inhibitors in combination with neoadjuvant endocrine therapy in estrogen receptor-positive early breast cancer: a systematic review and meta-analysis. Clin Exp Med 2022; 23:245-254. [PMID: 35304677 DOI: 10.1007/s10238-022-00814-3] [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: 01/08/2022] [Accepted: 02/22/2022] [Indexed: 11/03/2022]
Abstract
The combination of cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors and endocrine treatment has benefited patients with estrogen receptor-positive/human epidermal growth factor receptor 2-negative (ER + /HER2-) metastatic breast cancer; however, its effects in the neoadjuvant setting for ER + /HER2- early breast cancer (EBC) are unclear. Systematic searches were performed in PubMed, Embase, Cochrane Library, and major oncological meetings for trials of CDK4/6 inhibitors plus neoadjuvant endocrine treatment (NET) vs. NET/neoadjuvant chemotherapy (NACT) alone up to January 30, 2021. We assessed the efficacy of CDK4/6 inhibitors plus NET vs. NET/NACT alone in ER + /HER2- EBC. Six studies that included 803 patients treated with CDK4/6 inhibitors plus NET vs. NET/NACT alone were used. Compared with NET/NACT alone, CDK4/6 inhibitors plus NET increased the complete cell cycle arrest (CCCA) rate (OR, 9.00; 95% CI, 5.42-14.96; P < 0.001). Nonsignificant differences between CDK4/6 inhibitors and NET/NACT alone occurred in the preoperative endocrine prognostic index (PEPI)-0 rate (OR, 1.13; 95% CI, 0.59-2.18; P = 0.71), pathological complete response (pCR) rate (OR, 0.75; 95% CI, 0.13-4.29; P = 0.74), objective response rate (ORR) (OR, 0.70; 95% CI, 0.21-2.29; P = 0.55), and disease control rate (DCR) (OR, 1.16; 95% CI, 0.47-2.89; P = 0.74). CDK4/6 inhibitors plus NET indicated a high risk of neutropenia (OR, 56.43; 95% CI, 15.76-202.11; P < 0.001) as an adverse effect (AE) and elevated alanine aminotransferase (ALT) level (OR, 15.30; 95% CI, 2.02-115.98; P = 0.008) as grade 3/4 AEs. Compared with NET/NACT alone, CDK4/6 inhibitors plus NET increased CCCA rate in ER + /HER2- EBC patients. CDK4/6 inhibitors plus NET did not substantially improve the PEPI-0 rate, pCR rate, ORR, or DCR. The combination increased the risk of neutropenia and elevated ALT levels. In the neoadjuvant setting, addition of CDK4/6 inhibitors to NET may be an option for treating ER + /HER2- EBC.
Collapse
|
11
|
The Present and Future of Neoadjuvant Endocrine Therapy for Breast Cancer Treatment. Cancers (Basel) 2021; 13:cancers13112538. [PMID: 34064183 PMCID: PMC8196711 DOI: 10.3390/cancers13112538] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/16/2021] [Accepted: 05/19/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The treatment of breast cancer has evolved considerably over the last two decades, leading toward individualized disease management. Hormone-sensitive breast cancers constitute the vast majority of cases and endocrine therapy is the mainstay of their treatment. On the other hand, neoadjuvant or pre-surgical treatments provide a number of advantages for tumor management. In this review we will discuss the existing evidence on neoadjuvant endocrine therapy, as well as its possible future indications. Abstract Endocrine therapy (ET) has established itself as an efficacious treatment for estrogen receptor-positive (ER+) breast cancers, with a reduction in recurrence rates and increased survival rates. The pre-surgical approach with chemotherapy (NCT) has become a common form of management for large, locally advanced, or high-risk tumors. However, a good response to NCT is not usually expected in ER+ tumors. Good results with primary ET, mainly in elderly women, have encouraged studies in other stages of life, and nowadays neoadjuvant endocrine treatment (NET) has become a useful approach to many ER+ breast cancers. The aim of this review is to provide an update on the current state of art regarding the present and the future role of NET.
Collapse
|
12
|
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: 1.5] [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
|
13
|
Martí C, Sánchez-Méndez JI. Neoadjuvant endocrine therapy for luminal breast cancer treatment: a first-choice alternative in times of crisis such as the COVID-19 pandemic. Ecancermedicalscience 2020; 14:1027. [PMID: 32368252 PMCID: PMC7192402 DOI: 10.3332/ecancer.2020.1027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Indexed: 01/09/2023] Open
Abstract
The epidemiological emergency caused by CoV-2 (COVID-19) has changed priorities in breast cancer management. In those places where the pandemic has had the greatest effect, it is of paramount importance for most patients to be at home, reducing or postponing their attendance at clinics, as well as avoiding surgeries. In this scenario, neoadjuvant endocrine treatment could be an appropriate alternative treatment for hormone receptor positive breast cancer (luminal-like tumours) in order to minimise hospital admissions and to delay elective surgeries. Accordingly, we present a simple protocol that can be applied to most cases of luminal-like breast cancer and is appropriate for the majority of secondary or tertiary medical centres, or even primary care.
Collapse
Affiliation(s)
- Covadonga Martí
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain
- Gynaecology Department, Hospital Universitario La Paz, 28046 Madrid, Spain
| | - José I Sánchez-Méndez
- Breast Cancer Unit, Hospital Universitario La Paz, 28046 Madrid, Spain
- Gynaecology Department, Hospital Universitario La Paz, 28046 Madrid, Spain
| |
Collapse
|
14
|
Kurozumi S, Yamaguchi Y, Matsumoto H, Kurosumi M, Hayashi SI, Fujii T, Horiguchi J, Shirabe K, Inoue K. Utility of Ki67 labeling index, cyclin D1 expression, and ER-activity level in postmenopausal ER-positive and HER2-negative breast cancer with neoadjuvant chemo-endocrine therapy. PLoS One 2019; 14:e0217279. [PMID: 31112577 PMCID: PMC6528995 DOI: 10.1371/journal.pone.0217279] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/08/2019] [Indexed: 12/19/2022] Open
Abstract
In this study, we investigated the relationships of pathological response after neoadjuvant chemo-endocrine therapy with alterations in the Ki67 labeling index (LI), expression of cyclin D1 (CCND1) and progesterone receptor (PgR), and estrogen receptor (ER) activity in breast cancer. A total of 43 Japanese post-menopausal ER-positive and human epidermal growth factor receptor 2-negative invasive breast cancer patients with tumors >2 cm or positive lymph nodes were enrolled. Exemestane alone was administered for 2 months. Neoadjuvant chemo-endocrine therapy included four cycles of doxorubicin plus paclitaxel followed by weekly paclitaxel. The immunohistochemical expression of Ki67 LI, CCND1, and PgR, and ER activity were evaluated using core needle biopsy samples at the pretreatment and post-exemestane alone stages. ER activity was evaluated through transfection of an adenovirus vector carrying an estrogen-response element-green fluorescent protein gene. In current study, marked pathological responses (including 4.7% with pathological complete response) were obtained in 34.9% of patients. Ki67 LI and PgR expression were significantly decreased after treatment. High Ki67 LI at pretreatment was a significant predictive factor of marked pathological response. At the stage of post-exemestane alone, Ki67 LI was not significantly associated with pathological response; however, high CCND1 expression was significantly correlated with high Ki67 LI. Moreover, low-level ER activity at the post-exemestane alone stage was significantly associated with marked pathological response. In conclusions, pretreatment Ki67 LI was a predictor of response to neoadjuvant chemo-endocrine therapy. CCND1 expression and ER activity at the post-endocrine therapy alone stage may be useful in determining further treatments.
Collapse
Affiliation(s)
- Sasagu Kurozumi
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Gunma, Japan
- Division of Breast Surgery, Saitama Cancer Center, Saitama, Japan
- * E-mail:
| | - Yuri Yamaguchi
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | | | | | - Shin-ichi Hayashi
- Department of Molecular and Functional Dynamics, Tohoku University, Miyagi, Japan
| | - Takaaki Fujii
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Jun Horiguchi
- Department of Breast Surgery, International University of Health and Welfare, Chiba, Japan
| | - Ken Shirabe
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Kenichi Inoue
- Division of Breast Oncology, Saitama Cancer Center, Saitama, Japan
| |
Collapse
|
15
|
Ruffalo M, Thomas R, Chen J, Lee AV, Oesterreich S, Bar-Joseph Z. Network-guided prediction of aromatase inhibitor response in breast cancer. PLoS Comput Biol 2019; 15:e1006730. [PMID: 30742607 PMCID: PMC6386390 DOI: 10.1371/journal.pcbi.1006730] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 02/22/2019] [Accepted: 12/19/2018] [Indexed: 01/07/2023] Open
Abstract
Prediction of response to specific cancer treatments is complicated by significant heterogeneity between tumors in terms of mutational profiles, gene expression, and clinical measures. Here we focus on the response of Estrogen Receptor (ER)+ post-menopausal breast cancer tumors to aromatase inhibitors (AI). We use a network smoothing algorithm to learn novel features that integrate several types of high throughput data and new cell line experiments. These features greatly improve the ability to predict response to AI when compared to prior methods. For a subset of the patients, for which we obtained more detailed clinical information, we can further predict response to a specific AI drug. Breast cancer is the second most common type of cancer in women, with an incidence rate of over 250,000 cases per year, and breast cancer cases show significant heterogeneity in clinical and omic measures. Estrogen receptor positive (ER+) tumors typically grow in response to estrogen, and in post menopausal women, estrogen is only produced in peripheral tissues via the aromatase enzyme. Inhibition of aromatase is often an effective treatment for ER+ tumors, but aromatase inhibitor therapy is not effective for all tumors, and causes of this heterogeneity in response are largely not known. In this work, we present a feature construction and classification method to predict response to aromatase inhibitor therapy. We use network smoothing techniques to combine tumor omic data into predictive features, which we use as input to standard machine learning algorithms. We train predictive models using clinical data, including high-quality clinical data from UPMC patients, and show that our method outperforms previous approaches in predicting response to aromatase inhibitor therapy.
Collapse
Affiliation(s)
- Matthew Ruffalo
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Roby Thomas
- Women’s Cancer Research Center, Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, Magee Womens Research Institute, Pittsburgh, Pennsylvania, United States of America
| | - Jian Chen
- Women’s Cancer Research Center, Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, Magee Womens Research Institute, Pittsburgh, Pennsylvania, United States of America
| | - Adrian V. Lee
- Women’s Cancer Research Center, Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, Magee Womens Research Institute, Pittsburgh, Pennsylvania, United States of America
| | - Steffi Oesterreich
- Women’s Cancer Research Center, Department of Pharmacology and Chemical Biology, UPMC Hillman Cancer Center, Magee Womens Research Institute, Pittsburgh, Pennsylvania, United States of America
| | - Ziv Bar-Joseph
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
16
|
Post-neoadjuvant strategies in breast cancer: From risk assessment to treatment escalation. Cancer Treat Rev 2019; 72:7-14. [DOI: 10.1016/j.ctrv.2018.10.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/16/2018] [Accepted: 10/29/2018] [Indexed: 01/25/2023]
|
17
|
Zhang Y, Wu J, Zhou Y, Yin Y, Chen H. Effects of psoralen on the pharmacokinetics of anastrozole in rats. PHARMACEUTICAL BIOLOGY 2018; 56:433-439. [PMID: 30345900 PMCID: PMC6201813 DOI: 10.1080/13880209.2018.1501584] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
CONTEXT Psoralen and anastrozole are always used together for breast cancer patients in Chinese clinics. OBJECTIVE This study investigates the effects of psoralen on the pharmacokinetics of anastrozole in rats and its potential mechanism. MATERIALS AND METHODS The pharmacokinetics of orally administered anastrozole (0.5 mg/kg) with (test group) or without (Control group) psoralen pretreatment (20 mg/kg/day for 10 days) in male Sprague-Dawley rats (six rats in each group) were investigated. The plasma concentration of anastrozole was determined using a sensitive and reliable LC-MS/MS method. Additionally, the effects of psoralen on the intestine transport and metabolic stability of anastrozole (1 μM) were investigated using a Caco-2 cell transwell model and rat liver microsome incubation systems. RESULTS The results indicated that psoralen could significantly increase the Cmax (from 56.74 ± 3.17 ng/mL to 83.26 ± 6.87 ng/mL), and t1/2 (from 10.80 ± 1.05 to 14.29 ± 1.38 h) of anastrozole (p < 0.05). Psoralen could also significantly decrease the efflux ratio of anastrozole from 1.88 to 1.32 (p < 0.05). Additionally, the intrinsic clearance rates of anastrozole decreased significantly (from 62.83 to 43.97 μL/min/mg protein) (p < 0.05) with psoralen pretreatment in rat liver microsome incubation systems. DISCUSSION AND CONCLUSIONS This study indicates that when the rats were pretreated with psoralen, the system exposure of anastrozole would be increased significantly. The results showed that the herb-drug interaction between psoralen and anastrozole might occur when they were co-administered, and future studies in humans also need to investigate its herb-drug interaction potential.
Collapse
Affiliation(s)
- Yuzhu Zhang
- TCM Department of Breast, Longhua Hospital Affiliated to Shanghai University of TCM, Shanghai, China
| | - Jingjing Wu
- TCM Department of Breast, Longhua Hospital Affiliated to Shanghai University of TCM, Shanghai, China
| | - Yue Zhou
- TCM Department of Breast, Longhua Hospital Affiliated to Shanghai University of TCM, Shanghai, China
| | - Yulian Yin
- TCM Department of Breast, Longhua Hospital Affiliated to Shanghai University of TCM, Shanghai, China
| | - Hongfeng Chen
- TCM Department of Breast, Longhua Hospital Affiliated to Shanghai University of TCM, Shanghai, China
- CONTACT Hongfeng Chen Department of Breast, Longhua Hospital Affiliated to Shanghai University of TCM, Shanghai, 200032, China
| |
Collapse
|
18
|
Mosly D, Turnbull A, Sims A, Ward C, Langdon S. Predictive markers of endocrine response in breast cancer. World J Exp Med 2018; 8:1-7. [PMID: 30191138 PMCID: PMC6125140 DOI: 10.5493/wjem.v8.i1.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/26/2018] [Accepted: 08/04/2018] [Indexed: 02/07/2023] Open
Abstract
Ongoing clinical and research efforts seek to optimise the use of endocrine therapy in the treatment of breast cancer. Accurate biomarkers are needed that predict response for individual patients. The presence of the estrogen receptor (ER) as the direct (for tamoxifen and fulvestrant) or indirect (for aromatase inhibitors) target molecule for endocrine therapy remains the foremost biomarker and determinant of response. However, ER expression only poorly predicts outcome and further indicators of response or resistance are required. The development and application of molecular signature assays such as Oncotype Dx, Prosigna, Mammaprint and Endopredict have provided valuable information on prognosis and these are being used to support clinical decision making on whether endocrine therapy alone alongside surgery is sufficient for ER-positive early stage breast cancers or whether combination of endocrine with chemotherapy are also warranted. Ki67, the proliferation marker, has been widely used in the neo-adjuvant (pre-operative) setting to help predict response and long term outcome. Gene expression studies within the same setting have allowed monitoring of changes of potential predictive markers. These have identified frequent changes in estrogen-regulated and proliferation genes. Specific molecules such as mutant ER may also prove helpful biomarkers in predicting outcome and monitoring response to treatment.
Collapse
Affiliation(s)
- Duniya Mosly
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh EH4 2XR, United Kingdom
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Arran Turnbull
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh EH4 2XR, United Kingdom
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Andrew Sims
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh EH4 2XR, United Kingdom
| | - Carol Ward
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
- the Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh EH25 9RG, United Kingdom
| | - Simon Langdon
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| |
Collapse
|
19
|
Baxter DE, Kim B, Hanby AM, Verghese ET, Sims AH, Hughes TA. Neoadjuvant Endocrine Therapy in Breast Cancer Upregulates the Cytotoxic Drug Pump ABCG2/BCRP, and May Lead to Resistance to Subsequent Chemotherapy. Clin Breast Cancer 2018; 18:481-488. [PMID: 30055962 DOI: 10.1016/j.clbc.2018.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 07/03/2018] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Neoadjuvant treatments for primary breast cancer are becoming more common; however, little is known about how these impact on response to subsequent adjuvant therapies. Conveniently, neoadjuvant therapy provides opportunities to consider this question, by studying therapy-induced expression changes using comparisons between pre- and posttreatment samples. These data are relatively lacking in the context of neoadjuvant endocrine therapy, as opposed to the more common neoadjuvant chemotherapy. Here, we investigate the relevance of expression of the xenobiotic transporter ABCG2/BCRP, a gene/protein associated with chemoresistance, in the context of neoadjuvant endocrine therapy and particularly with reference to subsequent chemotherapy treatment. MATERIALS AND METHODS ABCG2/BCRP expression was assessed by immunohistochemistry or by expression arrays in matched patient samples pre- and post-neoadjuvant endocrine therapy. Cell culture was used to model the impact of endocrine therapy-induced changes in ABCG2/BCRP on subsequent chemotherapy response, using Western blots, quantitative polymerase chain reaction, survival assays, and cell cycle analyses. RESULTS ABCG2/BCRP was commonly and significantly upregulated in breast cancers after treatment with neoadjuvant endocrine therapy in 3 separate cohorts encompassing a total of 200 patients. Treatment with the endocrine therapeutic tamoxifen similarly induced ABCG2/BCRP upregulation in a relevant model cell line, the estrogen receptor-positive line T47D. Critically, this upregulation was associated with significantly increased chemoresistance to subsequent treatment with epirubicin, an anthracycline commonly used in breast cancer adjuvant chemotherapy. CONCLUSION Our data suggest that neoadjuvant endocrine therapy may induce poor responses to adjuvant chemotherapy, and therefore, that clinical outcomes following this treatment sequence warrant further study.
Collapse
Affiliation(s)
- Diana E Baxter
- School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Baek Kim
- Department of Breast Surgery, St. James's University Hospital, Leeds, United Kingdom
| | - Andrew M Hanby
- School of Medicine, University of Leeds, Leeds, United Kingdom; Department of Histopathology, St. James's University Hospital, Leeds, United Kingdom
| | - Eldo T Verghese
- Department of Histopathology, St. James's University Hospital, Leeds, United Kingdom
| | - Andrew H Sims
- Applied Bioinformatics of Cancer Group, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, United Kingdom
| | - Thomas A Hughes
- School of Medicine, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
20
|
Fingleton B, Lange K, Caldwell B, Bankaitis KV. Perspective on the interpretation of research and translation to clinical care with therapy-associated metastatic breast cancer progression as an example. Clin Exp Metastasis 2018; 34:443-447. [PMID: 29484519 DOI: 10.1007/s10585-017-9872-8] [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: 11/10/2017] [Accepted: 12/29/2017] [Indexed: 11/29/2022]
Abstract
This commentary was written as a collaboration between the Board of the Metastasis Research Society and two patients with metastatic breast cancer. It was conceived in response to how preclinical scientific research is sometimes presented to non-scientists in a way that can cause stress and confusion. Translation of preclinical findings to the clinic requires overcoming multiple barriers. This is irrespective of whether the findings relate to exciting responses to new therapies or problematic effects of currently used therapies. It is important that these barriers are understood and acknowledged when research findings are summarized for mainstream reporting. To minimize confusion, patients should continue to rely on their oncology care team to help them interpret whether research findings presented in mainstream media have relevance for their individual care. Researchers, both bench and clinical, should work together where possible to increase options for patients with metastatic disease, which is still in desperate need of effective therapeutic approaches.
Collapse
Affiliation(s)
- Barbara Fingleton
- Department of Pharmacology, Vanderbilt University, 771 PRB, 2220 Pierce Ave, Nashville, TN, 37233-6480, USA.
| | - Kelly Lange
- METAvivor Research and Support Inc., Annapolis, MD, USA
| | | | | |
Collapse
|
21
|
Mueller C, Haymond A, Davis JB, Williams A, Espina V. Protein biomarkers for subtyping breast cancer and implications for future research. Expert Rev Proteomics 2018; 15:131-152. [PMID: 29271260 PMCID: PMC6104835 DOI: 10.1080/14789450.2018.1421071] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Breast cancer subtypes are currently defined by a combination of morphologic, genomic, and proteomic characteristics. These subtypes provide a molecular portrait of the tumor that aids diagnosis, prognosis, and treatment escalation/de-escalation options. Gene expression signatures describing intrinsic breast cancer subtypes for predicting risk of recurrence have been rapidly adopted in the clinic. Despite the use of subtype classifications, many patients develop drug resistance, breast cancer recurrence, or therapy failure. Areas covered: This review provides a summary of immunohistochemistry, reverse phase protein array, mass spectrometry, and integrative studies that are revealing differences in biological functions within and between breast cancer subtypes. We conclude with a discussion of rigor and reproducibility for proteomic-based biomarker discovery. Expert commentary: Innovations in proteomics, including implementation of assay guidelines and standards, are facilitating refinement of breast cancer subtypes. Proteomic and phosphoproteomic information distinguish biologically functional subtypes, are predictive of recurrence, and indicate likelihood of drug resistance. Actionable, activated signal transduction pathways can now be quantified and characterized. Proteomic biomarker validation in large, well-designed studies should become a public health priority to capitalize on the wealth of information gleaned from the proteome.
Collapse
Affiliation(s)
- Claudius Mueller
- a Center for Applied Proteomics and Molecular Medicine , George Mason University , Manassas , VA , USA
| | - Amanda Haymond
- a Center for Applied Proteomics and Molecular Medicine , George Mason University , Manassas , VA , USA
| | - Justin B Davis
- a Center for Applied Proteomics and Molecular Medicine , George Mason University , Manassas , VA , USA
| | - Alexa Williams
- a Center for Applied Proteomics and Molecular Medicine , George Mason University , Manassas , VA , USA
| | - Virginia Espina
- a Center for Applied Proteomics and Molecular Medicine , George Mason University , Manassas , VA , USA
| |
Collapse
|