51
|
HER2 intratumoral heterogeneity is independently associated with distal metastasis and overall survival in HER2-positive breast carcinomas. Breast Cancer Res Treat 2020; 181:519-527. [PMID: 32335801 DOI: 10.1007/s10549-020-05650-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 04/17/2020] [Indexed: 01/02/2023]
Abstract
PURPOSE Human epidermal growth factor receptor 2 (HER2) intratumoral heterogeneity (ITH) occurs in a subset of breast cancers. Our recent study revealed it as an independent predictive factor for the response to anti-HER2 neoadjuvant therapy. In this study, we aimed to investigate its association with distal metastasis. METHODS HER2 ITH was assessed using HER2 gene protein assay (GPA) on whole tissue sections of pretreatment biopsies from a cohort of 158 HER2-positive invasive breast carcinomas and correlated with patients' clinical follow-up outcomes along with other clinicopathologic characteristics. RESULTS Fifty-seven cases (36%) showed HER2 ITH including 19 with genetic, 8 with both genetic and non-genetic, and 30 with non-genetic ITH. Multivariate analysis demonstrated larger tumor size, positive resected lymph node(s), negative PR, and the presence of HER2 ITH were independently associated with distal metastasis. Additionally, multivariate analysis demonstrated larger tumor size and the presence of HER2 ITH were the only independent factors associated with decreased overall survival (death). CONCLUSION The presence of HER2 ITH is an independent factor associated with poor overall survival and increased distal metastasis in HER2-positive breast cancer patients.
Collapse
|
52
|
Montemurro F, Nuzzolese I, Ponzone R. Neoadjuvant or adjuvant chemotherapy in early breast cancer? Expert Opin Pharmacother 2020; 21:1071-1082. [PMID: 32237920 DOI: 10.1080/14656566.2020.1746273] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION The administration of chemotherapy before (neoadjuvant), rather than after surgery (adjuvant) in early breast cancer has been considered an optional strategy for patients with operable breast cancer. We reviewed this concept considering recent results in the field. AREAS COVERED Herein, the authors cover neoadjuvant chemotherapy with or without biologics in triple-negative and HER2-positive operable breast cancer with a focus on rates of complete pathological remission (pCR) in the breast and axilla. The impact of the CREATE X and KATERINE randomized clinical trials of post-surgical treatments in patients with residual disease after neoadjuvant chemotherapy is also discussed. EXPERT OPINION The CREATE X and KATERINE clinical trials show for the first time and with methodological strengths that, in TNBC and HER2-positive breast cancer patients, post-surgical capecitabine and T-DM1, respectively, can improve prognosis when the disease persists after neoadjuvant chemotherapy. Therefore, the role of pCR as a treatment endpoint and a guide for further treatment decisions is now demonstrated. On account of these results, neoadjuvant chemotherapy becomes not an option, but rather the preferred treatment strategy for more and more TNBC and HER2-positive breast cancer patients in clinical practice.
Collapse
Affiliation(s)
- Filippo Montemurro
- Multidisciplinary Oncology Outpatient Clinic, Istituto di Candiolo, FPO-IRCCS , Candiolo, Italy
| | | | - Riccardo Ponzone
- Division of Gynecological Surgery, Istituto di Candiolo, FPO-IRCCS , Candiolo, Italy
| |
Collapse
|
53
|
Testa U, Castelli G, Pelosi E. Breast Cancer: A Molecularly Heterogenous Disease Needing Subtype-Specific Treatments. Med Sci (Basel) 2020; 8:E18. [PMID: 32210163 PMCID: PMC7151639 DOI: 10.3390/medsci8010018] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/23/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most commonly occurring cancer in women. There were over two-million new cases in world in 2018. It is the second leading cause of death from cancer in western countries. At the molecular level, breast cancer is a heterogeneous disease, which is characterized by high genomic instability evidenced by somatic gene mutations, copy number alterations, and chromosome structural rearrangements. The genomic instability is caused by defects in DNA damage repair, transcription, DNA replication, telomere maintenance and mitotic chromosome segregation. According to molecular features, breast cancers are subdivided in subtypes, according to activation of hormone receptors (estrogen receptor and progesterone receptor), of human epidermal growth factors receptor 2 (HER2), and or BRCA mutations. In-depth analyses of the molecular features of primary and metastatic breast cancer have shown the great heterogeneity of genetic alterations and their clonal evolution during disease development. These studies have contributed to identify a repertoire of numerous disease-causing genes that are altered through different mutational processes. While early-stage breast cancer is a curable disease in about 70% of patients, advanced breast cancer is largely incurable. However, molecular studies have contributed to develop new therapeutic approaches targeting HER2, CDK4/6, PI3K, or involving poly(ADP-ribose) polymerase inhibitors for BRCA mutation carriers and immunotherapy.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Regina Elena 299, 00161 Rome, Italy; (G.C.); (E.P.)
| | | | | |
Collapse
|
54
|
Morganti S, Curigliano G. Moving beyond endocrine therapy for luminal metastatic breast cancer in the precision medicine era: looking for new targets. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020. [DOI: 10.1080/23808993.2020.1720508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Stefania Morganti
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
55
|
Godoy-Ortiz A, Sanchez-Muñoz A, Chica Parrado MR, Álvarez M, Ribelles N, Rueda Dominguez A, Alba E. Deciphering HER2 Breast Cancer Disease: Biological and Clinical Implications. Front Oncol 2019; 9:1124. [PMID: 31737566 PMCID: PMC6828840 DOI: 10.3389/fonc.2019.01124] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 10/09/2019] [Indexed: 12/31/2022] Open
Abstract
The main obstacle for designing effective treatment approaches in breast cancer is the extensive and the characteristic heterogeneity of this tumor. The vast majority of critical genomic changes occurs during breast cancer progression, creating a significant variability within primary tumors as well as between the primary breast cancer and their metastases, a hypothesis have already demonstrated in retrospective studies (1). A clear example of this is the HER2-positive breast cancer. In these tumors, we can find all of the transcriptional subtypes of breast cancer, even the basal like or luminal A subtypes. Although the HER2-enriched is the most representative transcriptional subtype in the HER2-positive breast cancer, we can find it too in breast cancers with HER2-negative status. This intrinsic subtype shows a high expression of the HER2 and is associated with proliferation-related genes clusters, among other features. Therefore, two hypotheses can be suggested. First, the HER2 amplification can be a well-defined driver event present in all of the intrinsic subtypes, and not a subtype marker isolated. Secondly, HER2-enriched subtype can have a distinctive transcriptional landscape independent of HER2 amplification. In this review, we present an extensive revision about the last highlights and advances in clinical and genomic settings of the HER2-positive breast cancer and the HER2-enriched subtype, in an attempt to improving the knowledge of the underlying biology of both entities and to explaining the intrinsic heterogeneity of HER2-positive breast cancers.
Collapse
Affiliation(s)
- Ana Godoy-Ortiz
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain
- Laboratorio de Biología Molecular del Centro de Investigaciones Médico-Sanitarias de Málaga (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, Spain
| | - Alfonso Sanchez-Muñoz
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain
- Laboratorio de Biología Molecular del Centro de Investigaciones Médico-Sanitarias de Málaga (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, Spain
| | - Maria Rosario Chica Parrado
- Laboratorio de Biología Molecular del Centro de Investigaciones Médico-Sanitarias de Málaga (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, Spain
| | - Martina Álvarez
- Laboratorio de Biología Molecular del Centro de Investigaciones Médico-Sanitarias de Málaga (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, Spain
| | - Nuria Ribelles
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain
- Laboratorio de Biología Molecular del Centro de Investigaciones Médico-Sanitarias de Málaga (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, Spain
| | - Antonio Rueda Dominguez
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain
- Laboratorio de Biología Molecular del Centro de Investigaciones Médico-Sanitarias de Málaga (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, Spain
| | - Emilio Alba
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain
- Laboratorio de Biología Molecular del Centro de Investigaciones Médico-Sanitarias de Málaga (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, Spain
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Madrid, Spain
| |
Collapse
|
56
|
Reagan M. CAUSES OF CANCER. Cancer 2019. [DOI: 10.1002/9781119645214.ch3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
57
|
Kaczmarek E, Saint-Martin C, Pierga JY, Brain E, Rouzier R, Savignoni A, Mouret-Fourme E, Dieras V, Piot I, Dubot C, Carton M, Lerebours F. Long-term survival in HER2-positive metastatic breast cancer treated with first-line trastuzumab: results from the french real-life curie database. Breast Cancer Res Treat 2019; 178:505-512. [DOI: 10.1007/s10549-019-05423-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/27/2019] [Indexed: 11/30/2022]
|
58
|
The 41-gene classifier TRAR predicts response of HER2 positive breast cancer patients in the NeoALTTO study. Eur J Cancer 2019; 118:1-9. [DOI: 10.1016/j.ejca.2019.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 12/29/2022]
|
59
|
Cullin7 enhances resistance to trastuzumab therapy in Her2 positive breast cancer via degrading IRS-1 and downregulating IGFBP-3 to activate the PI3K/AKT pathway. Cancer Lett 2019; 464:25-36. [PMID: 31461670 DOI: 10.1016/j.canlet.2019.08.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/17/2019] [Accepted: 08/20/2019] [Indexed: 12/15/2022]
Abstract
Patients with Her2-positive breast cancer exhibit de novo resistance or develop acquired resistance in less than one year after Her2 targeting treatment, but the mechanism is not fully elucidated. Compensatory pathways such as the IGF-1R/IRS-1 pathway, are activated, leading to aberrant enhanced PI3K/Akt/mTOR pathway activity to attenuate the efficacy of trastuzumab. Cullin7 could participate in the degradation of IRS-1 in a mTOR/S6K dependent manner. Whether Cullin7 participates in trastuzumab resistance needs to be further investigated. Here, we reveals that Cullin7 is overexpressed in trastuzumab-resistant Her2 positive breast cancer cells. Knockdown of Cullin7 reduces degradation of Ser phosphorylation of IRS-1, attenuates activation of the PI3K/AKT pathway, and partly restores trastuzumab sensitivity in trastuzumab-resistant Her2 positive breast cancer cells. IGFBP-3 expression is decreased in trastuzumab-resistant Her2 positive breast cancer cells, which leads to release of the Wnt signaling pathway inhibition and an increase in Cullin7 expression, as mediated by TCF7L2. Overexpression of Cullin7 in Her2-amplified breast cancer tissues has clinical implications because it positively correlates with shorter disease-free survival (DFS) and inadequate response to trastuzumab. Thus, our results suggest a critical role for Cullin7 in response to trastuzumab, which has significant implications for selection of the optimal therapeutic strategy for Her2 positive breast cancers.
Collapse
|
60
|
Abstract
Breast cancer is a heterogeneous disease, which over time acquires various adaptive changes leading to more aggressive biological characteristics and development of treatment resistance. Several mechanisms of resistance have been established; however, due to the complexity of oestrogen receptor (ER) signalling and its crosstalk with other signalling networks, various areas still need to be investigated. This article focusses on the role of nuclear factor kappa B (NF-KB) as a key link between inflammation and cancer and addresses its emerging role as a key player in endocrine therapy resistance. Understanding the precise mechanism of NF-KB-driven endocrine therapy resistance provides a possible opportunity for therapeutic intervention.
Collapse
Affiliation(s)
- Phungern Khongthong
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of MVLS, University of Glasgow, Glasgow, UK
| | - Antonia K Roseweir
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of MVLS, University of Glasgow, Glasgow, UK
| | - Joanne Edwards
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of MVLS, University of Glasgow, Glasgow, UK
| |
Collapse
|
61
|
Omarini C, Bettelli S, Caprera C, Manfredini S, Barbolini M, Moscetti L, Isca C, Toss A, Barbieri E, Cortesi L, Kaleci S, Maiorana A, Tazzioli G, Cascinu S, Piacentini F. Differential molecular pathways expression in HER2 positive early breast cancer according to hormone receptor status. J Cancer Res Clin Oncol 2019; 145:821-828. [PMID: 30603906 DOI: 10.1007/s00432-018-02833-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 12/21/2018] [Indexed: 01/19/2023]
Abstract
PURPOSE Hormone receptors (HR) status in HER2 + breast cancer (BC) is a recognized stratification factor with relevant clinical implication. According to HR expression, HER2 + BC show different clinical characteristics, treatment sensitivity and prognosis. The interaction between HR and HER2 pathways remains incompletely understood. METHODS Thirty-four HER2 + BC were included: 18 tumors with HER2+/HR + and 16 with HER2+/HR-. The expression of 770 genes and 13 molecular pathways were evaluated using Nanostring PanCancer Pathway panel performed on FFPE BC biopsies. RESULTS Gene expression analysis identified 127 genes with significantly different expression between the two cohorts. 83% of these genes were overexpressed in HER2+/HR- cohort. Globally, 23% of them belonged to PI3K pathway (41 genes), 15% to Trascriptional regulation (26 genes) and 12% to MAPK (22 genes). Regarding pathway expression, PI3K, MAPK and NOTCH were significantly differently expressed between the two groups (p = 0.003, p = 0.0018 and p = 0.02, respectively), all of them were overexpressed in HER2+/HR- tumors. CONCLUSIONS According to HR status, HER2 + tumors express different pathways profiles: the overexpression of PI3K, MAPK and NOTCH pathways in HER2+/HR- group could justify different survival outcomes and treatment sensitivity. The identification of tumor driver pathways may be a useful instrument for individualized pathway-directed therapies. Further clinical implications are warranted.
Collapse
Affiliation(s)
- Claudia Omarini
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Via del Pozzo 71, 41122, Modena, Italy.
| | - Stefania Bettelli
- Division of Pathological Anatomy, Department of Diagnostic, Clinical Medicine and Public Health, University Hospital of Modena, Modena, Italy
| | - Cecilia Caprera
- Division of Pathological Anatomy, Department of Diagnostic, Clinical Medicine and Public Health, University Hospital of Modena, Modena, Italy
| | - Samantha Manfredini
- Division of Pathological Anatomy, Department of Diagnostic, Clinical Medicine and Public Health, University Hospital of Modena, Modena, Italy
| | - Monica Barbolini
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Via del Pozzo 71, 41122, Modena, Italy
| | - Luca Moscetti
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Via del Pozzo 71, 41122, Modena, Italy
| | - Chrystel Isca
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Via del Pozzo 71, 41122, Modena, Italy
| | - Angela Toss
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Via del Pozzo 71, 41122, Modena, Italy
| | - Elena Barbieri
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Via del Pozzo 71, 41122, Modena, Italy
| | - Laura Cortesi
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Via del Pozzo 71, 41122, Modena, Italy
| | - Shaniko Kaleci
- Division of Pathological Anatomy, Department of Diagnostic, Clinical Medicine and Public Health, University Hospital of Modena, Modena, Italy
| | - Antonino Maiorana
- Division of Pathological Anatomy, Department of Diagnostic, Clinical Medicine and Public Health, University Hospital of Modena, Modena, Italy
| | - Giovanni Tazzioli
- Oncologic Breast Surgery Unit, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Modena, Italy
| | - Stefano Cascinu
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Via del Pozzo 71, 41122, Modena, Italy
| | - Federico Piacentini
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Via del Pozzo 71, 41122, Modena, Italy
| |
Collapse
|
62
|
Abstract
Neratinib (Nerlynx™) is an oral, irreversible inhibitor of the human epidermal growth factor receptors HER1 (EGFR), HER2 and HER4. The drug originally arose from research by Wyeth (now Pfizer) and is now being developed by Puma Biotechnology primarily for the treatment of HER2-positive (HER+) breast cancer. Neratinib is approved in the USA for the extended adjuvant treatment of patients with HER2+ early-stage breast cancer who have been previously treated with a trastuzumab-based adjuvant regimen, and is in the preregistration phase for this indication in the EU. Neratinib, as monotherapy and/or combination therapy, is also in phase 3 development for metastatic breast cancer and in phase 1/2 development for advanced breast cancer and other solid tumours, including non-small cell lung cancer, colorectal cancer and glioblastoma. This article summarizes the milestones in the development of neratinib leading to this first approval for breast cancer.
Collapse
Affiliation(s)
- Emma D Deeks
- Springer, Private Bag 65901, Mairangi Bay, 0754, Auckland, New Zealand.
| |
Collapse
|
63
|
Giannone G, Milani A, Geuna E, Galizia D, Biello F, Montemurro F. What is the best pharmacotherapeutic strategy for HER-2 positive breast cancer? Expert Opin Pharmacother 2018; 20:5-9. [DOI: 10.1080/14656566.2018.1543406] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Gaia Giannone
- Medical Oncology, Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Italy
- Medical School, University of Turin, Turin, Italy
| | - Andrea Milani
- Investigational and Clinical Oncology, Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Italy
| | - Elena Geuna
- Investigational and Clinical Oncology, Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Italy
| | - Danilo Galizia
- Investigational and Clinical Oncology, Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Italy
| | - Federica Biello
- Investigational and Clinical Oncology, Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Italy
| | - Filippo Montemurro
- Investigational and Clinical Oncology, Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Italy
| |
Collapse
|
64
|
Nicolini A, Ferrari P, Duffy MJ. Prognostic and predictive biomarkers in breast cancer: Past, present and future. Semin Cancer Biol 2018; 52:56-73. [DOI: 10.1016/j.semcancer.2017.08.010] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/14/2017] [Accepted: 08/24/2017] [Indexed: 12/19/2022]
|
65
|
Wang M, Hu Y, Yu T, Ma X, Wei X, Wei Y. Pan-HER-targeted approach for cancer therapy: Mechanisms, recent advances and clinical prospect. Cancer Lett 2018; 439:113-130. [PMID: 30218688 DOI: 10.1016/j.canlet.2018.07.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/08/2018] [Accepted: 07/09/2018] [Indexed: 02/05/2023]
Abstract
The Human Epidermal Growth Factor Receptor family is composed of 4 structurally related receptor tyrosine kinases that are involved in many human cancers. The efficacy and safety of HER inhibitors have been compared in a wide range of clinical trials, suggesting the superior inhibitory ability of multiple- HER-targeting blockade compared with single receptor antagonists. However, many patients are currently resistant to current therapeutic treatment and novel strategies are warranted to conquer the resistance. Thus, we performed a critical review to summarize the molecular involvement of HER family receptors in tumour progression, recent anti-HER drug development based on clinical trials, and the potential resistance mechanisms of anti-HER therapy.
Collapse
Affiliation(s)
- Manni Wang
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Yuzhu Hu
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Ting Yu
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Xuelei Ma
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Xiawei Wei
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China.
| | - Yuquan Wei
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| |
Collapse
|
66
|
Rusz O, Kószó R, Dobi Á, Csenki M, Valicsek E, Nikolényi A, Uhercsák G, Cserháti A, Kahán Z. Clinical benefit of fulvestrant monotherapy in the multimodal treatment of hormone receptor and HER2 positive advanced breast cancer: a case series. Onco Targets Ther 2018; 11:5459-5463. [PMID: 30233207 PMCID: PMC6129034 DOI: 10.2147/ott.s170736] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Fulvestrant is a pure estrogen receptor (ER) antagonist approved for the treatment of metastatic ER positive breast cancer in postmenopausal women with disease progression following antiestrogen therapy. The clinical results of fulvestrant demonstrated encouraging activity in tumors in spite of HER2 positivity, but data about its use after progression on anti-HER2 agents are limited. Partial responses and durations of response of 12, 25, and 38 months in three cases with multiple metastases of ER positive and HER2 positive breast cancer were observed; all patients had been treated with 1–4 regimens of an anti-HER2 agent in combination with chemotherapy or an aromatase inhibitor before the initiation of fulvestrant. Fulvestrant is a valuable option with limited toxicity and durable response in metastatic HER2 and ER positive breast cancer after progression on anti-HER2 agents as well. Therapeutic benefit even in extensive skin metastases and (irradiated) brain metastases may be expected. Further investigations are warranted to establish where it fits into the multimodal management of ER and HER positive breast cancer.
Collapse
Affiliation(s)
- Orsolya Rusz
- Department of Oncotherapy, University of Szeged, Szeged, Hungary,
| | - Renáta Kószó
- Department of Oncotherapy, University of Szeged, Szeged, Hungary,
| | - Ágnes Dobi
- Department of Oncotherapy, University of Szeged, Szeged, Hungary,
| | - Melinda Csenki
- Department of Oncotherapy, University of Szeged, Szeged, Hungary,
| | | | - Alíz Nikolényi
- Department of Oncotherapy, University of Szeged, Szeged, Hungary,
| | | | | | - Zsuzsanna Kahán
- Department of Oncotherapy, University of Szeged, Szeged, Hungary,
| |
Collapse
|
67
|
Zhang W, Wu M, Chong QY, Zhang M, Zhang X, Hu L, Zhong Y, Qian P, Kong X, Tan S, Li G, Ding K, Lobie PE, Zhu T. Loss of Estrogen-Regulated MIR135A1 at 3p21.1 Promotes Tamoxifen Resistance in Breast Cancer. Cancer Res 2018; 78:4915-4928. [PMID: 29945962 DOI: 10.1158/0008-5472.can-18-0069] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/11/2018] [Accepted: 06/20/2018] [Indexed: 11/16/2022]
Abstract
The dysregulation of miRNAs has been increasingly recognized as a critical mediator of cancer development and progression. Here, we show that frequent deletion of the MIR135A1 locus is associated with poor prognosis in primary breast cancer. Forced expression of miR-135a decreased breast cancer progression, while inhibition of miR-135a with a specific miRNA sponge elicited opposing effects, suggestive of a tumor suppressive role of miR-135a in breast cancer. Estrogen receptor alpha (ERα) bound the promoter of MIR135A1 for its transcriptional activation, whereas tamoxifen treatment inhibited expression of miR-135a in ERα+ breast cancer cells. miR-135a directly targeted ESR1, ESRRA, and NCOA1, forming a negative feedback loop to inhibit ERα signaling. This regulatory feedback between miR-135a and ERα demonstrated that miR-135a regulated the response to tamoxifen. The tamoxifen-mediated decrease in miR-135a expression increased the expression of miR-135a targets to reduce tamoxifen sensitivity. Consistently, miR-135a expression was downregulated in ERα+ breast cancer cells with acquired tamoxifen resistance, while forced expression of miR-135a partially resensitized these cells to tamoxifen. Tamoxifen resistance mediated by the loss of miR-135a was shown to be partially dependent on the activation of the ERK1/2 and AKT pathways by miR-135a-targeted genes. Taken together, these results indicate that deletion of the MIR135A1 locus and decreased miR-135a expression promote ERα+ breast cancer progression and tamoxifen resistance.Significance: Loss of miR-135a in breast cancer disrupts an estrogen receptor-induced negative feedback loop, perpetuating disease progression and resistance to therapy.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/17/4915/F1.large.jpg Cancer Res; 78(17); 4915-28. ©2018 AACR.
Collapse
Affiliation(s)
- Weijie Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Mingming Wu
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Qing-Yun Chong
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore
| | - Min Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiao Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Lan Hu
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Yanghao Zhong
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Pengxu Qian
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences and Institute of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiangjun Kong
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Sheng Tan
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Gaopeng Li
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Keshuo Ding
- Department of Pathology, Anhui Medical University, Hefei, Anhui, China
| | - Peter E Lobie
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore.
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, Guangdong, China
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
68
|
Ayad E, Soliman A, Anis SE, Salem AB, Hu P, Dong Y. Ki 67 assessment in breast cancer in an Egyptian population: a comparative study between manual assessment on optical microscopy and digital quantitative assessment. Diagn Pathol 2018; 13:63. [PMID: 30153851 PMCID: PMC6114705 DOI: 10.1186/s13000-018-0735-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 08/12/2018] [Indexed: 12/18/2022] Open
Abstract
Background Breast cancer is by far the most frequent cancer among women. The proliferative index, Ki-67, is more and more taken into consideration for treatment decisions. However, the reliability of the established Ki-67 scoring is limited. Digital pathology is currently suggested to be a potential solution to Ki 67 assessment problems. Methods This is a retrospective and prospective study including 100 patients diagnosed with invasive breast cancer. Three senior pathologists have been asked to estimate the Ki-67 proliferative index for each of the 100 cases by examining the whole glass slides on optical microscope and providing a continuous score then a categorical score (‘high’ and ‘low’ Ki 67 index) using once 14%, once 20% as threshold indicative of high Ki67 status. Finally, a digital quantitative assessment of Ki67 was performed. Results A high inter-observer agreement was found when using optical microscopy for Ki 67 assessment, with correlation coefficient (CC) estimated at 0.878 (p value < 0.01). The overall agreement between manual and automated evaluation of Ki 67 was only substantial (CC estimated at 0.745 (p value < 0.01)). When using categorical scores, the inter-observers concordance was substantial using both cutoff points with kappa value estimated at 0.796 ([0.696–0.925] while using 14% as a cut off point and at 0.766 ([0.672–0.938] while using 20% as a cutoff point (p value < 0). The inter-observers agreement was better while using 14% as cutoff point. Agreement between manual and automated assessment of Ki 67 indices using both cutoff points was only substantial (Kappa estimated at 0.623, p value < 0.01). In comparison to automated assessment of Ki 67 index, while using 14% as a cutoff point, the overall tendency of all observers was to overestimate the Ki 67 values but to underestimate the proliferation index while using 20% as a cutoff point. Conclusion Automated assessment of Ki 67 value would appear to be comparable to visual Ki 67 assessment on optical microscopy. Such study would help define the role of digital pathology as a potential easy-to use tool for a robust and standardized fully automated Ki 67 scoring.
Collapse
Affiliation(s)
- Essam Ayad
- Department of Pathology, Cairo University, Cairo, Egypt.
| | - Ahmed Soliman
- Department of Pathology, Cairo University, Cairo, Egypt
| | | | | | - Pengchao Hu
- Department of Oncology, XiangYang No.1 People's Hospital Affiliated to Hubei University of Medicine, Xiangyang, Hubei, 441000, People's Republic of China
| | - Youhong Dong
- Department of Oncology, XiangYang No.1 People's Hospital Affiliated to Hubei University of Medicine, Xiangyang, Hubei, 441000, People's Republic of China
| |
Collapse
|
69
|
Godone RLN, Leitão GM, Araújo NB, Castelletti CHM, Lima-Filho JL, Martins DBG. Clinical and molecular aspects of breast cancer: Targets and therapies. Biomed Pharmacother 2018; 106:14-34. [PMID: 29945114 DOI: 10.1016/j.biopha.2018.06.066] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 06/01/2018] [Accepted: 06/13/2018] [Indexed: 12/23/2022] Open
Abstract
Breast Cancer is a complex disease characterized by the occurrence of multiple molecular alterations. Currently, some molecular markers are in use for breast cancer diagnostic, prognostic, and predictive purposes. Thus, genetic signatures are available for improving the decision-making. The biomarkers are also essential as therapeutic approaches, but many questions remain due to the lack of efficacy on breast cancer treatment, mainly for triple-negative breast cancer subtype. Since the genetic profile of breast cancer can also be related to different ethnic groups and geographic areas, the reference populations of the genetic assays and clinical trials need to include a broader population beyond the European and North American patients. In this review, we analyzed the current and potential molecular markers that could help to improve the strategies for breast cancer therapy.
Collapse
Affiliation(s)
- R L N Godone
- Molecular Prospection and Bioinformatics Group, Laboratory Keizo Asami of Immunopathology (LIKA), Federal University of Pernambuco (UFPE), Brazil
| | - G M Leitão
- Molecular Prospection and Bioinformatics Group, Laboratory Keizo Asami of Immunopathology (LIKA), Federal University of Pernambuco (UFPE), Brazil; Clinical Hospital of Pernambuco - Professor Romero Marques, Federal University of Pernambuco (UFPE), Brazil
| | - N B Araújo
- Molecular Prospection and Bioinformatics Group, Laboratory Keizo Asami of Immunopathology (LIKA), Federal University of Pernambuco (UFPE), Brazil
| | - C H M Castelletti
- Molecular Prospection and Bioinformatics Group, Laboratory Keizo Asami of Immunopathology (LIKA), Federal University of Pernambuco (UFPE), Brazil; Agronomic Institute of Pernambuco (IPA), Recife, Pernambuco, Brazil
| | - J L Lima-Filho
- Laboratory Keizo Asami of Immunopathology (LIKA), Federal University of Pernambuco (UFPE), Brazil; Department of Biochemistry, Federal University of Pernambuco (UFPE), Brazil
| | - D B G Martins
- Molecular Prospection and Bioinformatics Group, Laboratory Keizo Asami of Immunopathology (LIKA), Federal University of Pernambuco (UFPE), Brazil; Department of Biochemistry, Federal University of Pernambuco (UFPE), Brazil.
| |
Collapse
|
70
|
Schedin TB, Borges VF, Shagisultanova E. Overcoming Therapeutic Resistance of Triple Positive Breast Cancer with CDK4/6 Inhibition. Int J Breast Cancer 2018; 2018:7835095. [PMID: 30018827 PMCID: PMC6029445 DOI: 10.1155/2018/7835095] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/20/2018] [Accepted: 05/08/2018] [Indexed: 01/30/2023] Open
Abstract
Triple positive breast cancers overexpress both the human epidermal growth factor receptor 2 (HER2) oncogene and the hormonal receptors (HR) to estrogen and progesterone. These cancers represent a unique therapeutic challenge because of a bidirectional cross-talk between the estrogen receptor alpha (ERα) and HER2 pathways leading to tumor progression and resistance to targeted therapy. Attempts to combine standard of care HER2-targeted drugs with antihormonal agents for the treatment of HR+/HER2+ breast cancer yielded encouraging results in preclinical experiments but did improve overall survival in clinical trial. In this review, we dissect multiple mechanisms of therapeutic resistance typical of HR+/HER2+ breast cancer, summarize prior clinical trials of targeted agents, and describe novel rational drug combinations that include antihormonal agents, HER2-targeted drugs, and CDK4/6 inhibitors for treatment of the HR+/HER2+ breast cancer subtype.
Collapse
Affiliation(s)
- Troy B. Schedin
- Young Women's Breast Cancer Translational Program, University of Colorado Denver, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Virginia F. Borges
- Young Women's Breast Cancer Translational Program, University of Colorado Denver, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Elena Shagisultanova
- Young Women's Breast Cancer Translational Program, University of Colorado Denver, 13001 E 17th Pl, Aurora, CO 80045, USA
| |
Collapse
|
71
|
Zhao B, Zhao H, Zhao J. Impact of hormone receptor status on the efficacy of HER2-targeted treatment. Endocr Relat Cancer 2018; 25:687-697. [PMID: 29739815 DOI: 10.1530/erc-18-0029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 11/08/2022]
Abstract
The introduction of human epidermal growth factor receptor 2 (HER2)-targeted drugs into routine clinical practice has a dramatic effect on the outlook for patients with HER2-positive breast cancer (BC). However, the association between efficacy of HER2-targeted therapy and hormone receptor (HR) status is still unclear. Here we conducted a meta-analysis of randomized controlled trials (RCTs) to address this issue in both neoadjuvant and adjuvant settings. PubMed and EMBASE were searched from inception to October 2017 for studies involving trastuzumab, lapatinib, pertuzumab, trastuzumab emtansine and neratinib. Efficacy endpoints were pathological complete response (pCR) for neoadjuvant therapy and disease-free survival (DFS) for adjuvant therapy. In neoadjuvant setting, pCR was reported in 7 trials with 2868 subjects. Hormone receptor (HR)-negative women derived substantially greater benefit from HER2-targeted agents than did HR-positive patients (odds ratio (OR), 2.34; 95% confidence interval (CI), 1.99-2.75). Additionally, the impact of HR status on pCR was independent of anti-HER2 agents. In adjuvant setting, DFS was investigated in 7 studies with 12,768 patients. HR-positive patients benefit more from anti-HER2 treatment than did HR-negative subjects (OR, 0.81; 95% CI, 0.74-0.89). Moreover, patients who did not receive any endocrine or anti-HER2 neoadjuvant treatment showed similar outcome but with a smaller effect (OR, 0.88; 95% CI, 0.78-0.99). In summary, compared with HER2-positive/HR-negative subjects, HER2-positive/HR-positive patients achieved greater benefit from HER2-targeted treatment although the efficacy from neoadjuvant therapy was relatively poor.
Collapse
Affiliation(s)
- Bin Zhao
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hong Zhao
- The Third Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiaxin Zhao
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
72
|
Parylo S, Vennepureddy A, Dhar V, Patibandla P, Sokoloff A. Role of cyclin-dependent kinase 4/6 inhibitors in the current and future eras of cancer treatment. J Oncol Pharm Pract 2018; 25:110-129. [PMID: 29726787 DOI: 10.1177/1078155218770904] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cyclin-dependent kinase 4/6 inhibitors, which act by inhibiting progression from the G1 to S phases of the cell cycle, include palbociclib, ribociclib, abemaciclib, and trilaciclib. Palbociclib and ribociclib are currently food and drug administration-approved for use in combination with aromatase inhibitors in postmenopausal women with metastatic hormone receptor-positive, human epidermal growth factor receptor 2-negative breast cancer. Palbociclib is also food and drug administration-approved for use in combination with fulvestrant in hormone receptor-positive, human epidermal growth factor receptor 2-negative breast cancer progressing after endocrine therapy. Abemaciclib is the newest cyclin-dependent kinase 4/6 inhibitor to gain Food and Drug Administration (FDA) approval, specifically as monotherapy for hormone receptor-positive, human epidermal growth factor receptor 2-negative metastatic breast cancer previously treated with chemotherapy and endocrine therapy. Abemaciclib also shares a similar indication with palbociclib for use in combination with fulvestrant in hormone receptor-positive, human epidermal growth factor receptor 2-negative breast cancer progressing after endocrine therapy. Trilaciclib use remains largely investigational at this time. However, despite FDA-approval for only metastatic hormone receptor-positive, human epidermal growth factor receptor 2-negative breast cancer, all four cyclin-dependent kinase 4/6 inhibitors have shown promise in hematologic malignancies and non-breast solid tumors. Although further research is needed, cyclin-dependent kinase 4/6 inhibitors represent intriguing developments in the treatment of various malignancies, including those with such poor prognoses as glioblastoma multiforme, mantle cell lymphoma, and metastatic melanoma. We discuss the approved indications, current research, and areas of future exploration for palbociclib, ribociclib, abemaciclib, and trilaciclib.
Collapse
Affiliation(s)
- S Parylo
- 1 Department of Internal Medicine, 7601 Staten Island University Hospital , New York, USA
| | - A Vennepureddy
- 2 Division of Hematology and Oncology, 7601 Staten Island University Hospital , New York, USA
| | - V Dhar
- 1 Department of Internal Medicine, 7601 Staten Island University Hospital , New York, USA
| | - P Patibandla
- 1 Department of Internal Medicine, 7601 Staten Island University Hospital , New York, USA
| | - A Sokoloff
- 2 Division of Hematology and Oncology, 7601 Staten Island University Hospital , New York, USA
| |
Collapse
|
73
|
Sakai H, Tsurutani J, Iwasa T, Komoike Y, Sakai K, Nishio K, Nakagawa K. HER2 genomic amplification in circulating tumor DNA and estrogen receptor positivity predict primary resistance to trastuzumab emtansine (T-DM1) in patients with HER2-positive metastatic breast cancer. Breast Cancer 2018; 25:605-613. [PMID: 29700710 PMCID: PMC6132843 DOI: 10.1007/s12282-018-0861-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 04/18/2018] [Indexed: 12/26/2022]
Abstract
Background Trastuzumab emtansine (T-DM1) is approved for the treatment of patients with human epidermal growth factor receptor 2 (HER2)-positive advanced breast cancer (ABC), and has high efficacy. However, some patients exhibit primary resistance to T-DM1, and thus methods that can predict resistance in clinical practice are needed. Genomic analysis of circulating tumor DNA (ctDNA) in plasma is a non-invasive and reproducible method. This study aimed to predict primary resistance to T-DM1 by combining genomic analysis of ctDNA and other clinicopathological features of patients with HER2-positive ABC. Methods The study population comprised 34 patients with HER2-positive ABC who had been treated with T-DM1. Correlations between clinicopathological characteristics of patients and primary resistance to T-DM1 were examined, and HER2 gene copy number and PIK3CA gene mutations were analyzed using plasma ctDNA samples obtained from 16 patients before T-DM1 administration. Results Among the 34 patients, nine (26.5%) had progressive disease at the first efficacy analysis; these patients were considered to have primary resistance to T-DM1. No significant difference was found in the rate of primary resistance to T-DM1 between groups. Among 16 patients whose ctDNA was analyzed, four showed primary resistance to T-DM1. These four patients showed negative HER2 gene amplification in ctDNA and were ER-positive and/or PR-positive by immunohistochemistry. Conclusions HER2 gene amplification in ctDNA and ER and PR status may predict primary resistance to T-DM1. A liquid biopsy before the initiation of T-DM1 treatment could be a non-invasive way to predict whether a patient would exhibit primary resistance to T-DM1.
Collapse
Affiliation(s)
- Hitomi Sakai
- Department of Medical Oncology, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka, Japan
| | - Junji Tsurutani
- Department of Medical Oncology, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka, Japan.
| | - Tsutomu Iwasa
- Department of Medical Oncology, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka, Japan
| | - Yoshifumi Komoike
- Department of Surgery, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-Sayama, Osaka, Japan
| |
Collapse
|
74
|
Keegan NM, Gleeson JP, Hennessy BT, Morris PG. PI3K inhibition to overcome endocrine resistance in breast cancer. Expert Opin Investig Drugs 2018; 27:1-15. [PMID: 29252036 DOI: 10.1080/13543784.2018.1417384] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Activation of the phosphatidylinositol-3 kinase (PI3K) pathway is a critical step in oncogenesis and plays a role in the development of treatment resistance for both estrogen receptor (ER) positive and human epidermal growth factor receptor 2 (HER2) positive breast cancers. Hence, there have been efforts to therapeutically inhibit this pathway. AREAS COVERED Several inhibitors of PI3K are now progressing through clinical trials with varying degrees of efficacy and toxicity to date. Numerous unresolved questions remain concerning the optimal isoform selectivity of PI3K inhibitors and use of predictive biomarkers. This review examines the most important PI3K inhibitors in ER positive breast cancer to date, with a particular focus on their role in overcoming endocrine therapy resistance and the possible use of PIK3CA mutations as a predictive biomarker. EXPERT OPINION We discuss some of the emerging challenges and questions encountered during the development of PI3K inhibitors from preclinical to phase III studies, including other novel biomarkers and future combinations to overcome endocrine resistance.
Collapse
Affiliation(s)
- Niamh M Keegan
- a Department of Medical Oncology , Cancer Clinical Trials and Research Unit, Beaumont Hospital , Dublin , Ireland.,b Department of Molecular Medicine , Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| | - Jack P Gleeson
- a Department of Medical Oncology , Cancer Clinical Trials and Research Unit, Beaumont Hospital , Dublin , Ireland
| | - Bryan T Hennessy
- a Department of Medical Oncology , Cancer Clinical Trials and Research Unit, Beaumont Hospital , Dublin , Ireland.,b Department of Molecular Medicine , Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| | - Patrick G Morris
- a Department of Medical Oncology , Cancer Clinical Trials and Research Unit, Beaumont Hospital , Dublin , Ireland.,b Department of Molecular Medicine , Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| |
Collapse
|
75
|
Johnston SRD, Hegg R, Im SA, Park IH, Burdaeva O, Kurteva G, Press MF, Tjulandin S, Iwata H, Simon SD, Kenny S, Sarp S, Izquierdo MA, Williams LS, Gradishar WJ. Phase III, Randomized Study of Dual Human Epidermal Growth Factor Receptor 2 (HER2) Blockade With Lapatinib Plus Trastuzumab in Combination With an Aromatase Inhibitor in Postmenopausal Women With HER2-Positive, Hormone Receptor-Positive Metastatic Breast Cancer: ALTERNATIVE. J Clin Oncol 2017; 36:741-748. [PMID: 29244528 DOI: 10.1200/jco.2017.74.7824] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose Human epidermal growth factor receptor 2 (HER2) targeting plus endocrine therapy (ET) improved clinical benefit in HER2-positive, hormone receptor (HR)-positive metastatic breast cancer (MBC) versus ET alone. Dual HER2 blockade enhances clinical benefit versus single HER2 blockade. The ALTERNATIVE study evaluated the efficacy and safety of dual HER2 blockade plus aromatase inhibitor (AI) in postmenopausal women with HER2-positive/HR-positive MBC who received prior ET and prior neo(adjuvant)/first-line trastuzumab (TRAS) plus chemotherapy. Methods Patients were randomly assigned (1:1:1) to receive lapatinib (LAP) + TRAS + AI, TRAS + AI, or LAP + AI. Patients for whom chemotherapy was intended were excluded. The primary end point was progression-free survival (PFS; investigator assessed) with LAP + TRAS + AI versus TRAS + AI. Secondary end points were PFS (comparison of other arms), overall survival, overall response rate, clinical benefit rate, and safety. Results Three hundred fifty-five patients were included in this analysis: LAP + TRAS + AI (n = 120), TRAS + AI (n = 117), and LAP + AI (n = 118). Baseline characteristics were balanced. The study met its primary end point; superior PFS was observed with LAP + TRAS + AI versus TRAS + AI (median PFS, 11 v 5.7 months; hazard ratio, 0.62; 95% CI, 0.45 to 0.88; P = .0064). Consistent PFS benefit was observed in predefined subgroups. Overall response rate, clinical benefit rate, and overall survival also favored LAP + TRAS + AI. The median PFS with LAP + AI versus TRAS + AI was 8.3 versus 5.7 months (hazard ratio, 0.71; 95% CI, 0.51 to 0.98; P = .0361). Common adverse events (AEs; ≥ 15%) with LAP + TRAS + AI, TRAS + AI, and LAP + AI were diarrhea (69%, 9%, and 51%, respectively), rash (36%, 2%, and 28%, respectively), nausea (22%, 9%, and 22%, respectively), and paronychia (30%, 0%, and 15%, respectively), mostly grade 1 or 2. Serious AEs were reported similarly across the three groups, and AEs leading to discontinuation were lower with LAP + TRAS + AI. Conclusion Dual HER2 blockade with LAP + TRAS + AI showed superior PFS benefit versus TRAS + AI in patients with HER2-positive/HR-positive MBC. This combination offers an effective and safe chemotherapy-sparing alternative treatment regimen for this patient population.
Collapse
Affiliation(s)
- Stephen R D Johnston
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Roberto Hegg
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Seock-Ah Im
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - In Hae Park
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Olga Burdaeva
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Galina Kurteva
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Michael F Press
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Sergei Tjulandin
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Hiroji Iwata
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Sergio D Simon
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Sarah Kenny
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Severine Sarp
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Miguel A Izquierdo
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Lisa S Williams
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - William J Gradishar
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| |
Collapse
|
76
|
Johnston SRD, Hegg R, Im SA, Park IH, Burdaeva O, Kurteva G, Press MF, Tjulandin S, Iwata H, Simon SD, Kenny S, Sarp S, Izquierdo MA, Williams LS, Gradishar WJ. Phase III, Randomized Study of Dual Human Epidermal Growth Factor Receptor 2 (HER2) Blockade With Lapatinib Plus Trastuzumab in Combination With an Aromatase Inhibitor in Postmenopausal Women With HER2-Positive, Hormone Receptor-Positive Metastatic Breast Cancer: ALTERNATIVE. J Clin Oncol 2017. [PMID: 29244528 DOI: 10.1200/jco.2017.74.7824.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Human epidermal growth factor receptor 2 (HER2) targeting plus endocrine therapy (ET) improved clinical benefit in HER2-positive, hormone receptor (HR)-positive metastatic breast cancer (MBC) versus ET alone. Dual HER2 blockade enhances clinical benefit versus single HER2 blockade. The ALTERNATIVE study evaluated the efficacy and safety of dual HER2 blockade plus aromatase inhibitor (AI) in postmenopausal women with HER2-positive/HR-positive MBC who received prior ET and prior neo(adjuvant)/first-line trastuzumab (TRAS) plus chemotherapy. Methods Patients were randomly assigned (1:1:1) to receive lapatinib (LAP) + TRAS + AI, TRAS + AI, or LAP + AI. Patients for whom chemotherapy was intended were excluded. The primary end point was progression-free survival (PFS; investigator assessed) with LAP + TRAS + AI versus TRAS + AI. Secondary end points were PFS (comparison of other arms), overall survival, overall response rate, clinical benefit rate, and safety. Results Three hundred fifty-five patients were included in this analysis: LAP + TRAS + AI (n = 120), TRAS + AI (n = 117), and LAP + AI (n = 118). Baseline characteristics were balanced. The study met its primary end point; superior PFS was observed with LAP + TRAS + AI versus TRAS + AI (median PFS, 11 v 5.7 months; hazard ratio, 0.62; 95% CI, 0.45 to 0.88; P = .0064). Consistent PFS benefit was observed in predefined subgroups. Overall response rate, clinical benefit rate, and overall survival also favored LAP + TRAS + AI. The median PFS with LAP + AI versus TRAS + AI was 8.3 versus 5.7 months (hazard ratio, 0.71; 95% CI, 0.51 to 0.98; P = .0361). Common adverse events (AEs; ≥ 15%) with LAP + TRAS + AI, TRAS + AI, and LAP + AI were diarrhea (69%, 9%, and 51%, respectively), rash (36%, 2%, and 28%, respectively), nausea (22%, 9%, and 22%, respectively), and paronychia (30%, 0%, and 15%, respectively), mostly grade 1 or 2. Serious AEs were reported similarly across the three groups, and AEs leading to discontinuation were lower with LAP + TRAS + AI. Conclusion Dual HER2 blockade with LAP + TRAS + AI showed superior PFS benefit versus TRAS + AI in patients with HER2-positive/HR-positive MBC. This combination offers an effective and safe chemotherapy-sparing alternative treatment regimen for this patient population.
Collapse
Affiliation(s)
- Stephen R D Johnston
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Roberto Hegg
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Seock-Ah Im
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - In Hae Park
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Olga Burdaeva
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Galina Kurteva
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Michael F Press
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Sergei Tjulandin
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Hiroji Iwata
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Sergio D Simon
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Sarah Kenny
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Severine Sarp
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Miguel A Izquierdo
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - Lisa S Williams
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| | - William J Gradishar
- Stephen R.D. Johnston, The Royal Marsden NHS Foundation Trust, London; Lisa S. Williams, Novartis Pharmaceuticals UK Limited, Frimley, United Kingdom; Roberto Hegg, Centro de Referência da Saúde da Mulher; Sergio D. Simon, Hospital Israelita Albert Einstein, São Paulo, Brazil; Seock-Ah Im, Seoul National University College of Medicine, Seoul; In Hae Park, National Cancer Center, Gyeonggi-do, Korea; Olga Burdaeva, Regional Oncology Dispensary, Arkhangelsk; Sergei Tjulandin, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; Galina Kurteva, University Cancer Center Hospital, Sofia, Bulgaria; Michael F. Press, University of Southern California, Los Angeles, CA; Hiroji Iwata, Aichi Cancer Center Hospital, Aichi, Japan; Sarah Kenny, Severine Sarp, and Miguel A. Izquierdo, Novartis Pharma AG, Basel, Switzerland; and William J. Gradishar, Northwestern University, Chicago, IL
| |
Collapse
|
77
|
Hsu PY, Wu VS, Kanaya N, Petrossian K, Hsu HK, Nguyen D, Schmolze D, Kai M, Liu CY, Lu H, Chu P, Vito CA, Kruper L, Mortimer J, Chen S. Dual mTOR Kinase Inhibitor MLN0128 Sensitizes HR +/HER2 + Breast Cancer Patient-Derived Xenografts to Trastuzumab or Fulvestrant. Clin Cancer Res 2017; 24:395-406. [PMID: 29079660 DOI: 10.1158/1078-0432.ccr-17-1983] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/24/2017] [Accepted: 10/23/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Therapeutic strategies against hormonal receptor-positive (HR+)/HER2+ breast cancers with poor response to trastuzumab need to be optimized.Experimental Design: Two HR+/HER2+ patient-derived xenograft (PDX) models named as COH-SC1 and COH-SC31 were established to explore targeted therapies for HER2+ breast cancers. RNA sequencing and RPPA (reverse phase protein array) analyses were conducted to decipher molecular features of the two PDXs and define the therapeutic strategy of interest, validated by in vivo drug efficacy examination and in vitro cell proliferation analysis.Results: Estrogen acted as a growth driver of trastuzumab-resistant COH-SC31 tumors but an accelerator in the trastuzumab-sensitive COH-SC1 model. In vivo trastuzumab efficacy examination further confirmed the consistent responses between PDXs and the corresponding tumors. Integrative omics analysis revealed that mammalian target of rapamycin (mTOR) and ERα signaling predominantly regulate tumor growth of the two HR+/HER2+ PDXs. Combination of the dual mTOR complex inhibitor MLN0128 and anti-HER2 trastuzumab strongly suppressed tumor growth of COH-SC1 PDX accompanied by increasing ER-positive cell population in vivo Instead, MLN0128 in combination with antiestrogen fulvestrant significantly halted the growth of HR+/HER2+ cancer cells in vitro and trastuzumab-resistant COH-SC31 as well as trastuzumab-sensitive COH-SC1 tumors in vivoConclusions: Compared with the standard trastuzumab treatment, this study demonstrates alternative therapeutic strategies against HR+/HER2+ tumors through establishment of two PDXs coupled with integrative omics analyses and in vivo drug efficacy examination. This work presents a prototype of future "co-clinical" trials to tailor personalized medicine in clinical practice. Clin Cancer Res; 24(2); 395-406. ©2017 AACR.
Collapse
Affiliation(s)
- Pei-Yin Hsu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Victoria Shang Wu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Noriko Kanaya
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Karineh Petrossian
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Hang-Kai Hsu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Duc Nguyen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Daniel Schmolze
- Department of Pathology, City of Hope Medical Center, Duarte, California
| | - Masaya Kai
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Chun-Yu Liu
- Department of Oncology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hannah Lu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Peiguo Chu
- Department of Pathology, City of Hope Medical Center, Duarte, California
| | - Courtney A Vito
- Department of Surgery, City of Hope Medical Center, Duarte, California
| | - Laura Kruper
- Department of Surgery, City of Hope Medical Center, Duarte, California
| | - Joanne Mortimer
- Department of Medical Oncology and Therapeutic Research, City of Hope Medical Center, Duarte, California
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California.
| |
Collapse
|
78
|
Zhao B, Zhao H. Impact of clinicopathological characteristics on the efficacy of neoadjuvant therapy in patients with human epidermal growth factor receptor-2-positive breast cancer. Int J Cancer 2017; 142:844-853. [PMID: 29023765 DOI: 10.1002/ijc.31097] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 10/02/2017] [Accepted: 10/05/2017] [Indexed: 12/25/2022]
Abstract
Neoadjuvant therapy has become increasingly common in human epidermal growth factor receptor-2 (HER2)-positive breast cancer. In this study, we examined the impact of different clinicopathological characteristics on pathological complete response (pCR) in patients treated with anti-HER2 agents. The PubMed and Embase databases were searched from inception through April 2017 to identify studies that met pre-specified criteria. The odds ratios (ORs) and 95% confidence intervals (CIs) were extracted directly or were calculated with other available information. Eleven randomized controlled trials (RCTs) that involved 3,269 HER2-positive women were included in this meta-analysis. Patients with hormone receptor (HR)-negative breast cancer benefited more from anti-HER2 therapy than did patients with HR-positive tumours (OR, 2.25; 95% CI, 1.93-2.62). Furthermore, this improvement in pCR was independent of anti-HER2 agents, phase, combined chemotherapy, neoadjuvant duration, year the trials started and region where the trials were conducted. Patients with small tumours achieved greater benefits than patients with large tumours (OR, 1.25; 95% CI, 1.00-1.55). Age did not predict an additional benefit from anti-HER2 neoadjuvant treatment (OR, 1.02; 95% CI, 0.73-1.45). The impact of nodal status on pCR was dependent on the anti-HER2 agents. In conclusion, for HER2-targeted neoadjuvant treatment in breast cancer, greater benefits were achieved in patients with small HR-negative tumours compared with patients with large HR-positive tumours. These results may improve drug development and treatment strategies, economic analyses and the design and interpretation of clinical trials.
Collapse
Affiliation(s)
- Bin Zhao
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hong Zhao
- Department of Medical Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
79
|
Annaratone L, Simonetti M, Wernersson E, Marchiò C, Garnerone S, Scalzo MS, Bienko M, Chiarle R, Sapino A, Crosetto N. Quantification of HER2 and estrogen receptor heterogeneity in breast cancer by single-molecule RNA fluorescence in situ hybridization. Oncotarget 2017; 8:18680-18698. [PMID: 28423635 PMCID: PMC5386639 DOI: 10.18632/oncotarget.15727] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 02/06/2017] [Indexed: 11/25/2022] Open
Abstract
Intra-tumor heterogeneity is a pervasive property of human cancers that poses a major clinical challenge. Here, we describe the characterization, at the transcriptional level, of the intra-tumor topography of two prominent breast cancer biomarkers and drug targets, epidermal growth factor receptor 2 (HER2) and estrogen receptor 1 (ER) in 49 archival breast cancer samples. We developed a protocol for single-molecule RNA FISH in formalin-fixed, paraffin-embedded tissue sections (FFPE-smFISH), which enabled us to simultaneously detect and perform absolute quantification of HER2 and ER mature transcripts in single cells and multiple tumor regions. We benchmarked our method with standard diagnostic techniques, demonstrating that FFPE-smFISH is able to correctly classify breast cancers into well-established molecular subgroups. By counting transcripts in thousands of single cells, we identified different expression modes and levels of inter-cellular variability. In samples expressing both HER2 and ER, many cells co-expressed both genes, although expression levels were typically uncorrelated. Finally, we applied diversity metrics from the field of ecology to assess the intra-tumor topography of HER2 and ER gene expression, revealing that the spatial distribution of these key biomarkers can vary substantially even among breast cancers of the same subtype. Our results demonstrate that FFPE-smFISH is a reliable diagnostic assay and a powerful method for quantification of intra-tumor transcriptional heterogeneity of selected biomarkers in clinical samples.
Collapse
Affiliation(s)
- Laura Annaratone
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Michele Simonetti
- Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Karolinska Institutet, Stockholm, Sweden
| | - Erik Wernersson
- Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Karolinska Institutet, Stockholm, Sweden
| | - Caterina Marchiò
- Department of Medical Sciences, University of Turin, Turin, Italy.,Department of Laboratory Medicine, Pathology Unit, Azienda Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
| | - Silvano Garnerone
- Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Karolinska Institutet, Stockholm, Sweden
| | | | - Magda Bienko
- Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Karolinska Institutet, Stockholm, Sweden
| | - Roberto Chiarle
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Anna Sapino
- Department of Medical Sciences, University of Turin, Turin, Italy.,Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Italy
| | - Nicola Crosetto
- Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
80
|
Chong QY, You ML, Pandey V, Banerjee A, Chen YJ, Poh HM, Zhang M, Ma L, Zhu T, Basappa S, Liu L, Lobie PE. Release of HER2 repression of trefoil factor 3 (TFF3) expression mediates trastuzumab resistance in HER2+/ER+ mammary carcinoma. Oncotarget 2017; 8:74188-74208. [PMID: 29088778 PMCID: PMC5650333 DOI: 10.18632/oncotarget.18431] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 05/10/2017] [Indexed: 12/15/2022] Open
Abstract
HER2+/ER+ breast cancer, a subset of the luminal B subtype, makes up approximately 10% of all breast cancers. The bidirectional crosstalk between HER2 and estrogen receptor (ER) in HER2+/ER+ breast cancer contributes to resistance towards both anti-estrogens and HER2-targeted therapies. TFF3 promotes breast cancer progression and has been implicated in anti-estrogen resistance in breast cancer. Herein, we investigated the cross-regulation between HER2 and estrogen-responsive TFF3, and the role of TFF3 in mediating trastuzumab resistance in HER2+/ER+ breast cancer. TFF3 expression was decreased by HER2 activation, and increased by inhibition of HER2 with trastuzumab in HER2+/ER+ breast cancer cells, partially in an ERα-independent manner. In contrast, the forced expression of TFF3 activated the entire HER family of receptor tyrosine kinases (HER1-4). Hence, HER2 negatively regulates its own signalling through the transcriptional repression of TFF3, while trastuzumab inhibition of HER2 results in increased TFF3 expression to compensate for the loss of HER2 signalling. In HER2+/ER+ breast cancer cells with acquired trastuzumab resistance, TFF3 expression was markedly upregulated and associated with a corresponding decrease in HER signalling. siRNA mediated depletion or small molecule inhibition of TFF3 decreased the survival and growth advantage of the trastuzumab resistant cells without re-sensitization to trastuzumab. Furthermore, TFF3 inhibition abrogated the enhanced cancer stem cell-like behaviour in trastuzumab resistant HER2+/ER+ breast cancer cells. Collectively, TFF3 may function as a potential biomarker and therapeutic target in trastuzumab resistant HER2+/ER+ breast cancer.
Collapse
Affiliation(s)
- Qing-Yun Chong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ming-Liang You
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Vijay Pandey
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Arindam Banerjee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Yi-Jun Chen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Han-Ming Poh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mengyi Zhang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lan Ma
- Tsinghua Berkeley Shenzhen Institute, Tsinghua University Graduate School at Shenzhen, Shenzhen, China
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Salundi Basappa
- Laboratory of Chemical Biology, Department of Chemistry, Bangalore University, Central College Campus, Bangalore, India
| | - Liang Liu
- Department of Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Radiology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Peter E. Lobie
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Tsinghua Berkeley Shenzhen Institute, Tsinghua University Graduate School at Shenzhen, Shenzhen, China
- National University Cancer Institute, Singapore
| |
Collapse
|
81
|
Buonomo OC, Caredda E, Portarena I, Vanni G, Orlandi A, Bagni C, Petrella G, Palombi L, Orsaria P. New insights into the metastatic behavior after breast cancer surgery, according to well-established clinicopathological variables and molecular subtypes. PLoS One 2017; 12:e0184680. [PMID: 28922402 PMCID: PMC5602519 DOI: 10.1371/journal.pone.0184680] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/29/2017] [Indexed: 02/03/2023] Open
Abstract
Despite advances in treatment, up to 30% of patients with early breast cancer (BC) experience distant disease relapse. However, a comprehensive understanding of tumor spread and site-specific recurrence patterns remains lacking. This retrospective case-control study included 103 consecutive patients with metastatic BC admitted to our institution (2000–2013). Cases were matched according to age, tumor biology, and clinicopathological features to 221 patients with non-metastatic BC (control group). The median follow-up period among the 324 eligible patients was 7.3 years. While relatively low values for sensitivity (71%) and specificity (56%) were found for axillary lymph node (ALN) involvement as an indicator of risk and pattern of distant relapse, nodal status remained the most powerful predictor of metastases (OR: 3.294; CL: 1.9–5.5). Rates of dissemination and metastatic efficiency differed according to molecular subtype. HER2-positive subtypes showed a stronger association with systemic spread (OR: 2.127; CL: 1.2–3.8) than other subgroups. Classification as Luminal or Non-Luminal showed an increased risk of lung and distant nodal recurrence, and a decreased risk in bone metastases in the Non-Luminal group (OR: 2.9, 3.345, and 0.2, respectively). Tumors with HER2 overexpression had a significantly high risk for distant relapse (OR: 2.127) compared with HER2-negative tumors and also showed higher central nervous system (CNS) and lung metastatic potential (OR: 5.6 and 2.65, respectively) and low risk of bone disease progression (OR: 0.294). Furthermore, we found significant associations between biological profiles and sites of recurrence. A new process of clinical/diagnostic staging, including molecular subtypes, could better predict the likelihood of distant relapses and their anatomical location. Recognition and appreciation of clinically distinct molecular subtypes may assist in evaluation of the probability of distant relapses and their sites. Our analysis provides new insights into management of metastatic disease behavior, to lead to an optimal disease-tailored approach and appropriate follow-up.
Collapse
Affiliation(s)
| | - Emanuele Caredda
- Department of Biomedicine and Prevention, Tor Vergata University Hospital, Rome, Italy
| | - Ilaria Portarena
- Department of Internal Medicine, Medical Oncology Unit, Tor Vergata University Hospital, Rome, Italy
| | - Gianluca Vanni
- Department of Surgery, Tor Vergata University Hospital, Rome, Italy
| | - Augusto Orlandi
- Anatomic Pathology, Department Biomedicine and Prevention, Tor Vergata University Hospital, Rome, Italy
| | - Claudia Bagni
- Department of Biomedicine and Prevention, Tor Vergata University Hospital, Rome, Italy
| | | | - Leonardo Palombi
- Department of Biomedicine and Prevention, Tor Vergata University Hospital, Rome, Italy
- * E-mail:
| | - Paolo Orsaria
- Department of Surgery, Tor Vergata University Hospital, Rome, Italy
| |
Collapse
|
82
|
Thaler S, Schmidt M, Roβwag S, Thiede G, Schad A, Sleeman JP. Proteasome inhibitors prevent bi-directional HER2/estrogen-receptor cross-talk leading to cell death in endocrine and lapatinib-resistant HER2+/ER+ breast cancer cells. Oncotarget 2017; 8:72281-72301. [PMID: 29069787 PMCID: PMC5641130 DOI: 10.18632/oncotarget.20261] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/04/2017] [Indexed: 12/26/2022] Open
Abstract
Amplification and/or overexpression of the human epidermal growth factor 2 (HER2) oncogene occurs in about 13–15% of invasive breast cancer and triggers breast cancer cell proliferation, survival and metastatic progression. Around half of all breast cancers with HER2 overexpression co-express hormone receptors (HR) such as those for estrogen and progesterone. Aberrant signaling through HER2 and other members of the HER-family mediates endocrine-resistance in estrogen receptor alpha (ERα) positive breast cancer. On the other hand, ERα co-expression has been shown to attenuate the efficiency of anti-HER2 therapies. These findings indicate that HER2 and ERα synergize to escape from both anti-ERα and anti-HER2-targeted therapies. Rationally designed clinical trials that combine endocrine therapy with anti-HER2 agents to interfere with HER2/ERα cross-talk have been conducted. However, the outcome of these trials suggests that novel therapeutic approaches are needed to further improve inhibition of HER2 and other HER-family members in conjunction with a more efficient ERα blockade. Here, we demonstrate that carfilzomib and bortezomib stabilize the HER2-specific protein tyrosine phosphatase BDP1 leading to decreased HER2 autophosphorylation, reduced HER2 activity and subsequently attenuated activation of the PI3K/Akt-pathway, together with blockade of ERα expression. We further observed that proteasome inhibitors (PIs) reverse autophosphorylation and thereby inhibit the activity of constitutively active mutant HER2. We also demonstrate that PIs cause cell death in lapatinib and endocrine-resistant HER2+/ER+ breast cancer cells. These findings suggest that PIs might have the potential to improve the management of HER2+/ER+ breast cancer patients by efficiently disrupting the bi-directional HER2/ERα cross-talk.
Collapse
Affiliation(s)
- Sonja Thaler
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Marcus Schmidt
- Department of Obstetrics and Gynecology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Sven Roβwag
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Gitta Thiede
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Arno Schad
- Institute of Pathology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Jonathan P Sleeman
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,KIT Campus Nord, Institute for Toxicology and Genetics, Karlsruhe, Germany
| |
Collapse
|
83
|
Zhang X, Harbeck N, Jeschke U, Doisneau-Sixou S. Influence of vitamin D signaling on hormone receptor status and HER2 expression in breast cancer. J Cancer Res Clin Oncol 2017; 143:1107-1122. [PMID: 28025696 DOI: 10.1007/s00432-016-2325-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/16/2016] [Indexed: 12/14/2022]
Abstract
PURPOSE Breast cancer is a significant global public health issue. It is the leading cause of death among women around the world, with an incidence increasing annually. In recent years, there has been more and more information in the literature regarding a protective role of vitamin D in cancer. Increasingly preclinical and clinical studies suggest that vitamin D optimal levels can reduce the risk of breast cancer development and regulate cancer-related pathways. METHOD In this review, we focus on the importance of vitamin D in breast cancers, discussing especially the influence of vitamin D signaling on estrogen receptor and human epidermal growth factor receptor 2 (HER2), two major biomarkers of breast cancer today. CONCLUSION We discuss the possibility of actual and future targeted therapeutic approaches for vitamin D signaling in breast cancer.
Collapse
Affiliation(s)
- Xi Zhang
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany
| | - Nadia Harbeck
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany
| | - Udo Jeschke
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany
| | - Sophie Doisneau-Sixou
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany.
- Faculté des Sciences Pharmaceutiques, Université Paul Sabatier Toulouse III, 31062, Toulouse Cedex 09, France.
| |
Collapse
|
84
|
Zhang X, Hofmann S, Rack B, Harbeck N, Jeschke U, Sixou S. Fluorescence Analysis of Vitamin D Receptor Status of Circulating Tumor Cells (CTCS) in Breast Cancer: From Cell Models to Metastatic Patients. Int J Mol Sci 2017. [PMID: 28632174 PMCID: PMC5486139 DOI: 10.3390/ijms18061318] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The Vitamin D receptor (VDR) expressed in normal breast tissue and breast tumors has been suggested as a new prognostic biomarker in breast cancer (BC). Besides, increasing evidence supports the view that the detection of circulating tumor cells (CTCs) predicts outcome in early and metastatic BC. Consequently, an evaluation of VDR expression in the CTCs of BC patients may allow optimization of their treatment. As an attempt to profile and subtype the CTCs of metastatic patients, we established an innovative fluorescence technique using nine BC cell lines to visualize, define, and compare their individual VDR status. Afterwards, we tested the CTC presence and VDR expression in blood samples (cytospins) collected from 23 metastatic BC patients. The results demonstrated major differences in the VDR levels among the nine cell lines, and VDR positive CTCs were detected in 46% of CTC-positive patients, with a total of 42 CTCs individually analyzed. Due to the limited number of patients in this study, no correlation between VDR expression and BC subtype classification (according to estrogen receptor (ER), progesterone receptor (PR) and HER2) could be determined, but our data support the view that VDR evaluation is a potential new prognostic biomarker to help in the optimization of therapy management for BC patients.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Obstetrics and Gynaecology, Breast Center, Ludwig-Maximilians University of Munich (LMU), Maistrasse 11, Munich 80337, Germany.
| | - Simone Hofmann
- Department of Obstetrics and Gynaecology, Breast Center, Ludwig-Maximilians University of Munich (LMU), Maistrasse 11, Munich 80337, Germany.
| | - Brigitte Rack
- Department of Obstetrics and Gynaecology, Breast Center, Ludwig-Maximilians University of Munich (LMU), Maistrasse 11, Munich 80337, Germany.
| | - Nadia Harbeck
- Department of Obstetrics and Gynaecology, Breast Center, Ludwig-Maximilians University of Munich (LMU), Maistrasse 11, Munich 80337, Germany.
| | - Udo Jeschke
- Department of Obstetrics and Gynaecology, Breast Center, Ludwig-Maximilians University of Munich (LMU), Maistrasse 11, Munich 80337, Germany.
| | - Sophie Sixou
- Department of Obstetrics and Gynaecology, Breast Center, Ludwig-Maximilians University of Munich (LMU), Maistrasse 11, Munich 80337, Germany.
- Faculty of Pharmacy, University Paul Sabatier Toulouse III, Toulouse cedex 09 31062, France.
| |
Collapse
|
85
|
Cocciolone V, Cannita K, Calandrella ML, Ricevuto E, Baldi PL, Sidoni T, Irelli A, Paradisi S, Pizzorno L, Resta V, Bafile A, Alesse E, Tessitore A, Ficorella C. Prognostic significance of clinicopathological factors in early breast cancer: 20 years of follow-up in a single-center analysis. Oncotarget 2017; 8:72031-72043. [PMID: 29069766 PMCID: PMC5641109 DOI: 10.18632/oncotarget.18526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 06/02/2017] [Indexed: 12/31/2022] Open
Abstract
Background To quantify the effect of traditional prognostic factors [nodal status, estrogen-receptor (ER), progesterone-receptor (PR), human epidermal growth factor receptor 2 (HER2)] on long-term outcome of patients with early breast cancer (EBC), treated in clinical practice over a period of about twenty years. Results 1198 consecutive patients were identified. Median DFS (disease-free survival): ER+/PR±/HER2−, 165 months (mo) if node-negative (N0) and 114mo if node-positive (N+) (p < 0.001); triple-negative (TN), 109mo if N0 and 65mo if N+ (p 0.144); ER+/PR±/HER2+ in patients not-treated with adjuvant trastuzumab (T−), not reached if N0 and 114mo if N+ (p 0.297); ER+/PR±/HER2+ in patients treated with trastuzumab (T+), 95mo if N0 and 85mo if N+ (p 0.615); ER−/PR−/HER2+ T−, not reached if N0 and 26mo if N+ (p 0.279); ER−/PR−/HER2+ T+, not reached if N0 and 66mo if N+ (p 0.014). Median OS (overall survival): ER+/ PR±/HER2−, 166mo if N0 and 144mo if N+ (p 0.028); TN, 158mo if N0 and 96mo if N+ (p 0.384); ER+/PR±/HER2+ T−, not reached if N0 and 157mo if N+ (p 0.475), ER+/PR±/HER2+ T+, not reached if N0 and 106mo if N+ (p 0.436); ER−/PR−/HER2+ T−, not reached if N0 and 34mo if N+ (p 0.273); ER−/PR−/HER2+ T+, not reached neither if N0 nor if N+ (p 0.094). Materials and Methods Disease-free survival (DFS) and overall survival (OS) were evaluated according to tumor characteristics, based on information retrospectively retrieved from patients’ medical records. Conclusions Pathological tumor characteristics and nodal status still represent useful tools in treatment selection and follow-up decision making of EBC patients in clinical practice.
Collapse
Affiliation(s)
- Valentina Cocciolone
- Medical Oncology, S. Salvatore Hospital, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Katia Cannita
- Medical Oncology, S. Salvatore Hospital, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | | | - Enrico Ricevuto
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy.,Oncology Network ASL1 Abruzzo, UOSD Oncology Territorial Care, S. Salvatore Hospital, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Paola Lanfiuti Baldi
- Medical Oncology, S. Salvatore Hospital, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Tina Sidoni
- Medical Oncology, S. Salvatore Hospital, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Azzurra Irelli
- Medical Oncology, S. Salvatore Hospital, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Stefania Paradisi
- Medical Oncology, S. Salvatore Hospital, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Laura Pizzorno
- Breast Unit, S. Salvatore Hospital, L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Valter Resta
- Breast Unit, S. Salvatore Hospital, L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Alberto Bafile
- Breast Unit, S. Salvatore Hospital, L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Edoardo Alesse
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Alessandra Tessitore
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Corrado Ficorella
- Medical Oncology, S. Salvatore Hospital, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| |
Collapse
|
86
|
Collins D, Jacob W, Cejalvo JM, Ceppi M, James I, Hasmann M, Crown J, Cervantes A, Weisser M, Bossenmaier B. Direct estrogen receptor (ER) / HER family crosstalk mediating sensitivity to lumretuzumab and pertuzumab in ER+ breast cancer. PLoS One 2017; 12:e0177331. [PMID: 28493933 PMCID: PMC5426757 DOI: 10.1371/journal.pone.0177331] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/26/2017] [Indexed: 11/24/2022] Open
Abstract
Bidirectional cross talk between members of the human epidermal growth factor family of receptors (HER) and the estrogen receptor (ER) is believed to underlie resistance mechanisms that develop in response to treatment with anti-HER agents and endocrine therapy. We investigated the interaction between HER2, HER3 and the ER in vitro using human embryonic kidney cells transfected with human HER2, HER3, and ERα. We also investigated the additive efficacy of combination regimens consisting of anti-HER3 (lumretuzumab), anti-HER2 (pertuzumab), and endocrine (fulvestrant) therapy in vivo. Our data show that both HER2 and HER3 can directly complex with the ER and can mediate phosphorylation of the ER. Phosphorylation of the ER was only observed in cells that expressed both HER2 and ERα or in heregulin-stimulated cells that expressed both HER3 and ERα. Using a mouse xenograft model of ER+/HER2-low (HER2 immunohistochemistry 1+ or 2+ without gene amplification) human breast cancer we show that the combination of lumretuzumab and pertuzumab is highly efficacious and induces long-lasting tumor regression in vivo and adding endocrine therapy (fulvestrant) to this combination further improved efficacy. In addition, a prolonged clinical response was observed with the combination of lumretuzumab and pertuzumab in a patient with ER+/HER2-low breast cancer who had failed endocrine therapy. These preclinical data confirm that direct cross talk exists between HER2/HER3 and ER which may explain the resistance mechanisms to endocrine therapy and monoclonal antibodies that target HER2 and HER3. Our data also indicate that the triplet of anti-HER2, anti-HER3, and endocrine therapy might be an efficacious combination for treating patients with ER+/HER2-low breast cancer, which is an area of significant unmet medical need.
Collapse
Affiliation(s)
- Denis Collins
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
- * E-mail: (DC); (MW)
| | | | - Juan Miguel Cejalvo
- Department of Medical Oncology, Institute of Health Research INCLIVA, University of Valencia, Valencia, Spain
| | | | - Ian James
- A4P Consulting Ltd, Sandwich, United Kingdom
| | - Max Hasmann
- Roche Innovation Center Munich, Penzberg, Germany
| | - John Crown
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
- Department of Medical Oncology, St. Vincent's University Hospital, Dublin, Ireland
| | - Andrés Cervantes
- Department of Medical Oncology, Institute of Health Research INCLIVA, University of Valencia, Valencia, Spain
| | - Martin Weisser
- Roche Innovation Center Munich, Penzberg, Germany
- * E-mail: (DC); (MW)
| | | |
Collapse
|
87
|
Yang HY, Ma D, Liu YR, Hu X, Zhang J, Wang ZH, Di GH, Hu XC, Shao ZM. Impact of hormone receptor status and distant recurrence-free interval on survival benefits from trastuzumab in HER2-positive metastatic breast cancer. Sci Rep 2017; 7:1134. [PMID: 28442763 PMCID: PMC5430907 DOI: 10.1038/s41598-017-00663-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 03/07/2017] [Indexed: 12/25/2022] Open
Abstract
We sought to investigate the impact of hormone receptor (HR) status and distant recurrence-free interval (DRFI) on the degree of overall survival (OS) benefit from palliative trastuzumab-containing treatment in HER2-positive metastatic breast cancer (MBC). Here, we retrospectively identified 588 eligible HER2-positive patients with postoperative distant recurrence. DRFI of HR+HER2+ MBC patients (median: 30.7 months, IQR: 18.5-45.9, P < 0.001) was significant longer compared with HR-HER2+ patients. Patients were categorized into four subgroups based on HR status and palliative trastuzumab (trast+) received. The most superior outcome was observed in the HR+HER2+trast+ subgroup, with a median OS of 48.3 months. Moreover, DRFI > 24 months is an independent favourable prognostic factor for both HR-HER2+ patients (Hazard Ratio (HzR) = 0.55, 95% CI: 0.39-0.76, P < 0.001) and HR+HER2+ patients (HzR = 0.45, 95% CI: 0.32-0.64, P < 0.001). Upon further analysis of the interaction between trastuzumab and DRFI, the degree of trastuzumab benefits in HR-HER2+ MBC patients remained basically unchanged regardless of DRFI length. Unlikely, the degree in HR+HER2+ MBC patients decreased gradually along with DRFI extending, indicating that trastuzumab failed to translate into an OS benefit for late recurrent (DRFI > 5years) HR+HER2+ MBC patients.
Collapse
Affiliation(s)
- Hai-Yuan Yang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ding Ma
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi-Rong Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xin Hu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jian Zhang
- Department of Medical oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhong-Hua Wang
- Department of Medical oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Gen-Hong Di
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Xi-Chun Hu
- Department of Medical oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| |
Collapse
|
88
|
Park HS, Sohn J, Kim SI, Park S, Park HS, Gho SG, Chung HC, Paik S, Kim GM. Effects of hormone receptor status on the durable response of trastuzumab-based therapy in metastatic breast cancer. Breast Cancer Res Treat 2017; 163:255-262. [PMID: 28243895 DOI: 10.1007/s10549-017-4175-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 02/22/2017] [Indexed: 12/28/2022]
Abstract
PURPOSE Trastuzumab-based treatment is the standard care for patients with HER2+ metastatic breast cancer (MBC). About 10% of HER2+ MBC showed a long-term durable response (progression-free survival, PFS > 3 years) to trastuzumab-based therapy. The aim of this study is to identify clinico-pathologic factors for a durable response to trastuzumab-based therapy in HER2-positive MBC. METHODS In the Yonsei Breast Cancer MBC Database, we identified 1218 MBC patients who were diagnosed from 2006 to 2015. Among them, 294 had HER2+ disease, and 153 received trastuzumab plus taxane chemotherapy as first-line treatment. Clinico-pathologic factors, such as hormone receptor (HR) status and metastatic sites, were reviewed. To evaluate a durable response, landmark analysis was performed. RESULTS The median follow-up time was 28 months (95% CI 4.4-83.0 months). Of 153 HER2+ patients, there were 73 HR- patients (47.7%), and bone was the most common metastatic site. The median PFS and overall survival (OS) were 12 and 39 months, respectively. HR- patients showed a tendency toward longer PFS (median, 13 vs. 11 months, P = 0.160) compared with HR+ patients. Patients with non-visceral metastases had longer median PFS and OS than those with visceral disease (median PFS, 15 vs. 11 months, P = 0.012; median OS, 75 vs. 34 months, P = 0.03). Landmark analysis at 9 months suggested that the PFS of HR- patients was significantly longer than that of HR+ patients (median, 19 vs. 9 months, P = 0.008). CONCLUSIONS Among patients with HER2+ MBC, HR status is a possible predictive biomarker of a durable response to trastuzumab-based therapy.
Collapse
Affiliation(s)
- Hyung Soon Park
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, 120-752, Seoul, Korea
| | - Joohyuk Sohn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, 120-752, Seoul, Korea
| | - Seung Il Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Seho Park
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Hyung Seok Park
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Seul Ghi Gho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, 120-752, Seoul, Korea
| | - Hyun Cheol Chung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, 120-752, Seoul, Korea
| | - Soonmyung Paik
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, 120-752, Seoul, Korea.,Department of Biomedical Science, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Gun Min Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, 120-752, Seoul, Korea.
| |
Collapse
|
89
|
Vici P, Pizzuti L, Sperduti I, Frassoldati A, Natoli C, Gamucci T, Tomao S, Michelotti A, Moscetti L, Gori S, Baldini E, Giotta F, Cassano A, Santini D, Giannarelli D, Di Lauro L, Corsi DC, Marchetti P, Sini V, Sergi D, Barba M, Maugeri-Saccà M, Russillo M, Mentuccia L, D'Onofrio L, Iezzi L, Scinto AF, Da Ros L, Bertolini I, Basile ML, Rossi V, De Maria R, Montemurro F. "Triple positive" early breast cancer: an observational multicenter retrospective analysis of outcome. Oncotarget 2017; 7:17932-44. [PMID: 26910921 PMCID: PMC4951261 DOI: 10.18632/oncotarget.7480] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/11/2016] [Indexed: 01/18/2023] Open
Abstract
We recently found that trastuzumab benefit may be lower in a small subset of early breast cancer (BC) patients (pts) with tumors expressing high levels of both hormonal receptors (HRs), i.e. triple positive (TP). To better investigate the role of HRs in HER2 positive BC, we retrospectively identified 872 TP BC pts treated with adjuvant chemotherapy alone (cohort A-366 pts), or plus trastuzumab (cohort B-506 pts). Relapse-free-survival (RFS) and breast-cancer-specific-survival (BCSS) were evaluated. Trastuzumab improved RFS and BCSS in all the subsets analyzed, but the effect on BCSS in tumors expressing both HRs in >30% of cells (TP30), and even on RFS in tumors with both HRs expressed in >50% of cells (TP50) was not significant. Distinct patterns of relapse were observed in TP50 and no-TP50 tumors, the former showing low and constant risk in the first 5 years, a late increase beyond 5 years and modest trastuzumab effect. Trastuzumab effect tended to disappear in pts whose tumors expressed ER in >50% of cells. Multivariate analysis of RFS confirmed a significant interaction between trastuzumab and ER expression, with benefit confined to pts whose tumors expressed ER in ≤50% of cells. Our data suggest that the pattern of relapse of TP tumors with high HRs is similar to that of "luminal", HER2 negative tumors, without clear benefit from adjuvant trastuzumab, which remains the standard treatment even in TP tumors. Confirmatory findings on the extent to which quantitative expression of HRs may impact clinical behavior of HER2 positive BC are warranted.
Collapse
Affiliation(s)
- Patrizia Vici
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Rome, Italy
| | - Laura Pizzuti
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Rome, Italy
| | - Isabella Sperduti
- Biostatistics Unit, "Regina Elena" National Cancer Institute, Rome, Italy
| | | | - Clara Natoli
- Department of Experimental and Clinical Sciences, University "G. d'Annunzio", Chieti, Italy
| | | | - Silverio Tomao
- Department of Medico-Surgical Sciences and Biotechnologies, "Sapienza" University of Rome, Oncology Unit, Istituto Chirurgico Ortopedico Traumatologico, Latina, Italy
| | - Andrea Michelotti
- Oncology Unit I, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Luca Moscetti
- Department of Oncology, Division of Medical Oncology, Belcolle Hospital, ASL Viterbo, Viterbo, Italy
| | - Stefania Gori
- Department of Oncology, Ospedale Sacro Cuore Don Calabria, Negrar, Verona, Italy
| | - Editta Baldini
- Department of Medical Oncology, S. Luca Hospital, Lucca, Italy
| | - Francesco Giotta
- Division of Medical Oncology, IRCCS, Giovanni Paolo II Hospital, Bari, Italy
| | | | - Daniele Santini
- Department of Medical Oncology, University Campus Bio-Medico, Rome, Italy
| | - Diana Giannarelli
- Biostatistics Unit, "Regina Elena" National Cancer Institute, Rome, Italy
| | - Luigi Di Lauro
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Rome, Italy
| | | | - Paolo Marchetti
- Oncology Unit, Sant'Andrea Hospital, "Sapienza" University of Rome, Rome, Italy
| | - Valentina Sini
- Oncology Unit, Sant'Andrea Hospital, "Sapienza" University of Rome, Rome, Italy.,Medical Oncology, S. Spirito Hospital, Rome, Italy
| | - Domenico Sergi
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Rome, Italy
| | - Maddalena Barba
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Rome, Italy.,Scientific Direction, "Regina Elena" National Cancer Institute, Rome, Italy
| | - Marcello Maugeri-Saccà
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Rome, Italy.,Scientific Direction, "Regina Elena" National Cancer Institute, Rome, Italy
| | | | | | - Loretta D'Onofrio
- Department of Medical Oncology, University Campus Bio-Medico, Rome, Italy
| | - Laura Iezzi
- Department of Experimental and Clinical Sciences, University "G. d'Annunzio", Chieti, Italy
| | | | - Lucia Da Ros
- Division of Oncology, S. Anna Hospital, Ferrara, Italy
| | - Ilaria Bertolini
- Oncology Unit I, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Maria Luisa Basile
- Department of Molecular Medicine, "Umberto I", "Sapienza" University of Rome, Roma, Italy
| | - Valentina Rossi
- Investigative Clinical Oncology, Fondazione del Piemonte per l'Oncologia-Candiolo Cancer Institute (IRCCs), Candiolo, Italy.,Division of Medical Oncology, Ospedale Civile di Saluzzo, Saluzzo, Italy
| | - Ruggero De Maria
- Scientific Direction, "Regina Elena" National Cancer Institute, Rome, Italy
| | - Filippo Montemurro
- Investigative Clinical Oncology, Fondazione del Piemonte per l'Oncologia-Candiolo Cancer Institute (IRCCs), Candiolo, Italy
| |
Collapse
|
90
|
Mancuso MR, Massarweh SA. Endocrine therapy and strategies to overcome therapeutic resistance in breast cancer. Curr Probl Cancer 2016; 40:95-105. [PMID: 27839747 DOI: 10.1016/j.currproblcancer.2016.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 09/01/2016] [Indexed: 01/04/2023]
Abstract
Despite the remarkable success of endocrine therapy in the treatment of patients with estrogen receptor (ER)- positive breast cancer, not all patients derive benefit from such therapy, or may benefit only temporarily before disease progression or relapse occurs. The value of endocrine therapy, which blocks ER signaling by a variety of strategies, lies in its simplicity, lower toxicity, and better alignment with preserved quality of life, particularly when compared to chemotherapy, which is more toxic and has only modest benefits for many patients with ER-positive breast cancer. It is therefore critical that we discover ways to extend endocrine therapy benefit in patients and prevent therapeutic resistance whenever possible. The tremendous evolution in our understanding of endocrine resistance mechanisms, coupled with the increasing availability of novel agents that target resistance pathways, has led to enhanced treatment approaches for patients with ER-positive breast cancer, primarily through combinations of endocrine agents with a variety of pathway inhibitors. Despite these treatment advances and our changing view of ER-positive breast cancer, there is much work that needs to be done. It remains a problem that we cannot reliably predict which subsets of patients will experience disease relapse or progression on endocrine therapy, and as such, combination strategies with targeted agents have largely been used in unselected patients with ER-positive breast cancer, including those who continue to have endocrine-sensitive disease. Patient selection is a significant issue since most of the targeted therapeutics that we use with endocrine therapy are expensive and can be toxic, and we may be inadvertently overtreating patients whose disease can still be controlled with endocrine therapy alone. In this article, we will review current and future strategies in the treatment of ER-positive breast cancer, as well as the evolving role of targeted therapy in the management of endocrine-resistance.
Collapse
Affiliation(s)
| | - Suleiman Alfred Massarweh
- Division of Oncology, Stanford University School of Medicine; Stanford Cancer Institute, Stanford, CA, 94305.
| |
Collapse
|
91
|
Ding J, Hu P, Chen J, Wu X, Cao Y. The importance of tissue confirmation of metastatic disease in patients with breast cancer: lesson from a brain metastasis case. Oncoscience 2016; 3:268-274. [PMID: 28050577 PMCID: PMC5116944 DOI: 10.18632/oncoscience.320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/12/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The discrepancy of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) statuses in breast cancers has been reported. Available systemic therapy for patients with breast cancer is based on the molecular subtypes as identified by IHC and/or FISH. However, these biomarkers may change throughout tumor progression. CASE PRESENTATION We report a relatively uncommon case of a 39-year-old Chinese woman with local advanced breast cancer (LABC) treated with 6 cycles of docetaxel, doxorubicin and cyclophosphamide (TAC) regimen neoadjuvant chemotherapy, and subsequently mastectomy, intensity-modulated radiation therapy (IMRT) and tamoxifen followed as regularly. Brain metastatic event appeared in 6 months after mastectomy. Treatment for brain metastasis was surgical resection and followed by whole brain radiotherapy (WBRT) approved by multidisciplinary team (MDT). Initial pathological diagnosis was IDC, cT4N1M0, luminal B (ER+ 90%, PR+90%, HER2 0, Ki67+ 70%) based on ultrasound-guided core needle biopsy. Surgical pathology revealed IDC, pT2N3M0 luminal B (ER+ 20%, PR+20%, HER2 0, Ki67+ 20%). Histological response to neoadjuvant chemotherapy is grade 3 according to the Miller/Payne grading system. Final pathology of brain metastasis showed a HER2 overexpression metastatic breast cancer luminal B (ER+ 70%, PR+ 70%, HER2 2+, Ki67+ 30%), FISH confirmed HER2 overexpression. Weekly paclitaxel plus trastuzumab was given for 12 weeks, then trastuzumab every 3 weeks for a whole year. Patient follow-up is still ongoing, no new events appear yet. CONCLUSIONS The determination of hormone receptors and HER2 status should be routinely performed in all involved tissues, if possible, and systemic therapy should be tailored following the latest finding.
Collapse
Affiliation(s)
- Jingxian Ding
- Department of Radiotherapy, Breast Cancer Institute, The Third Hospital of Nanchang, Nanchang 330009, China
| | - Pinghua Hu
- Department of Breast Surgery, Breast Cancer Institute, The Third Hospital of Nanchang, Nanchang 330009, China
| | - Jun Chen
- Department of Breast Surgery, Breast Cancer Institute, The Third Hospital of Nanchang, Nanchang 330009, China
| | - Xiaobo Wu
- Department of Breast Surgery, Breast Cancer Institute, The Third Hospital of Nanchang, Nanchang 330009, China
| | - Yali Cao
- Department of Breast Surgery, Breast Cancer Institute, The Third Hospital of Nanchang, Nanchang 330009, China
| |
Collapse
|
92
|
Harano K, Lei X, Gonzalez-Angulo AM, Murthy RK, Valero V, Mittendorf EA, Ueno NT, Hortobagyi GN, Chavez-MacGregor M. Clinicopathological and surgical factors associated with long-term survival in patients with HER2-positive metastatic breast cancer. Breast Cancer Res Treat 2016; 159:367-74. [PMID: 27522517 DOI: 10.1007/s10549-016-3933-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 07/29/2016] [Indexed: 10/21/2022]
Abstract
Trastuzumab-based treatment has dramatically improved the outcomes of HER2-positive (HER2+) metastatic breast cancer (MBC) patients, with some patients achieving prolonged survival times. In this study, we aim to identify factors that are associated with long-term survival. Patients with HER2+ MBC treated with anti-HER2 target therapy were identified. Patients were grouped according to overall survival (OS) and categorized as long-term survivors (LTS, OS ≥ 5 years), or non-long-term survivors (non-LTS, OS < 5 years). Descriptive statistics and multivariable logistic regression modeling were used. A sensitivity analysis was carried out, including only patients diagnosed before 2007; therefore, 5 years of potential follow-up was possible. 1063 patients with HER2+ MBC diagnosed between 1994 and 2012 and treated with anti-HER2 therapy were identified. Among them, 154 (14.5 %) patients were categorized as LTS (median OS 92.2 months). Among LTS, 63.4 % were HR-positive and 32 % had de novo stage IV disease. Hormone receptor positivity (OR) 1.69; 95 % CI 1.17-2.44), resection of metastases (OR 2.38; 95 % CI 1.53-3.69), and primary breast surgery in patients with de novo stage IV (OR 2.88; 95 % CI 1.47-5.66) were associated with improved long-term survival. Greater number of metastatic sites (≥3 vs. 1, OR 0.41; 95 % CI 0.23-0.72) and visceral metastases (OR 0.61; 95 % CI 0.4-0.91) were associated with poor survival. Hormone receptor positivity, low burden of disease, metastasis to soft and bone tissues, and surgical management with resection of the metastatic site and the primary tumor were associated with long-term survival in patients with MBC who received anti-HER2 treatment.
Collapse
Affiliation(s)
- K Harano
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - X Lei
- Division of Cancer Prevention, Department of Health Services Research, The University of Texas MD Anderson Cancer Center, FCT9.5024, 1515 Holcombe Blvd. Unit Number: 1444, Houston, TX, 77030, USA
| | | | - R K Murthy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - V Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - E A Mittendorf
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - N T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - G N Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Chavez-MacGregor
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Division of Cancer Prevention, Department of Health Services Research, The University of Texas MD Anderson Cancer Center, FCT9.5024, 1515 Holcombe Blvd. Unit Number: 1444, Houston, TX, 77030, USA.
| |
Collapse
|
93
|
Schettini F, Buono G, Cardalesi C, Desideri I, De Placido S, Del Mastro L. Hormone Receptor/Human Epidermal Growth Factor Receptor 2-positive breast cancer: Where we are now and where we are going. Cancer Treat Rev 2016; 46:20-6. [PMID: 27057657 DOI: 10.1016/j.ctrv.2016.03.012] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 11/19/2022]
Abstract
Near 75% of all breast cancers (BC) express estrogen receptors (ER) and/or progesterone receptors (PgR), while up to 20% of BC show an overexpression/amplification of Human Epidermal Growth Factor Receptor 2 (HER2). Around 50% of all HER2-overexpressing BC show the coexistence of both HER2 overexpression/amplification and ER and/or PgR overexpression. Numerous in vitro and in vivo studies suggest the existence of a cross-talk between their downstream pathways, which seem to affect the natural history, response to therapy and outcome of patients affected by this subset of BC. Meta-analyses or subgroup analysis of numerous neo-/adjuvant trials demonstrated significant clinical implications deriving from ER/HER2 co-existence, consisting in a different pattern of relapse and dissimilar outcome in response to anti-HER2 therapy. However, only two randomized trials in early disease and three in advanced disease specifically addressed the issue whether a combined approach with both hormonal and anti-HER2 therapy would have a better therapeutic impact in this subset of BC compared to the lone anti-HER2 or hormonal therapies (HT). None of these trials demonstrated improvements in overall survival, even though several efficacy end-points such as progression free survival, in advanced setting, or pCR rates in neoadjuvant setting, often favored the combined hormonal and anti-HER2 therapeutic approach. In the next few years, a certain number of ongoing randomized trials, both in neoadjuvant and advanced setting, will evaluate the efficacy of new anti-HER2 drugs, T-DM1 and pertuzumab, in combination with HT, helping to improve the therapeutic strategy for this specific subtype of breast tumors.
Collapse
Affiliation(s)
- Francesco Schettini
- Medical Oncology, Department of Clinical and Surgical Medicine, University of Naples "Federico II", Naples, Italy.
| | - Giuseppe Buono
- Medical Oncology, Department of Clinical and Surgical Medicine, University of Naples "Federico II", Naples, Italy
| | - Cinzia Cardalesi
- Medical Oncology, Department of Clinical and Surgical Medicine, University of Naples "Federico II", Naples, Italy
| | - Isacco Desideri
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Sabino De Placido
- Medical Oncology, Department of Clinical and Surgical Medicine, University of Naples "Federico II", Naples, Italy
| | - Lucia Del Mastro
- Department of Medical Oncology, IRCCS AOU San Martino-IST, Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| |
Collapse
|
94
|
Cardoso TC, Sakamoto SS, Stockmann D, Souza TFB, Ferreira HL, Gameiro R, Vieira FV, Louzada MJQ, Andrade AL, Flores EF. A three-dimensional cell culture system as an in vitro canine mammary carcinoma model for the expression of connective tissue modulators. Vet Comp Oncol 2016; 15:582-593. [PMID: 26991309 DOI: 10.1111/vco.12202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/22/2015] [Accepted: 11/03/2015] [Indexed: 11/27/2022]
Abstract
In this study, derived complex carcinoma (CC) and simple carcinoma (SC) cell lines were established and cultured under two-dimensional (2D) and three-dimensional (3D) conditions. The 3D was performed in six-well AlgiMatrix™ (LifeTechnologies®, Carlsbad, CA, USA) scaffolds, resulting in spheroids sized 50-125 µm for CC and 175-200 µm for SC. Cell viability was demonstrated up to 14 days for both models. Epidermal growth factor receptor (EGFR) was expressed in CC and SC in both systems. However, higher mRNA and protein levels were observed in SC 2D and 3D systems when compared with CC (P < 0.005). The connective tissue modulators, metalloproteinases-1, -2, -9 and -13 (MMPs), relaxin receptors 1 and 2 (RXR1 and RXR2) and E-cadherin (CDH1) were quantitated. All were upregulated similarly when canine mammary tumour (CMT)-derived cell lines were cultured under 3D AlgiMatrix, except CDH1 that was downregulated (P < 0.005). These results are promising towards the used of 3D system to increase a high throughput in vitro canine tumour model.
Collapse
Affiliation(s)
- T C Cardoso
- Veterinary Medicine School, Department of Support, Production and Animal Health, University of São Paulo State, Laboratory of Animal Virology and Cell Culture, Araçatuba, Brazil.,Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, Brazil
| | - S S Sakamoto
- Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, Brazil
| | - D Stockmann
- Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, Brazil
| | - T F B Souza
- Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, Brazil
| | - H L Ferreira
- Department of de Veterinary Medicine, Faculdade Zotecnia e Engenharia de Alimentos-USP, Pirassununga, SP, Brazil
| | - R Gameiro
- Veterinary Medicine School, Department of Support, Production and Animal Health, University of São Paulo State, Laboratory of Animal Virology and Cell Culture, Araçatuba, Brazil
| | - F V Vieira
- Veterinary Medicine School, Department of Support, Production and Animal Health, University of São Paulo State, Laboratory of Animal Virology and Cell Culture, Araçatuba, Brazil
| | - M J Q Louzada
- Veterinary Medicine School, Department of Support, Production and Animal Health, University of São Paulo State, Laboratory of Animal Virology and Cell Culture, Araçatuba, Brazil
| | - A L Andrade
- Veterinary Medicine School, Department of Clinical, Surgery and Animal Reproduction, University of São Paulo State, Veterinary Hospital Section, Araçatuba, Brazil
| | - E F Flores
- Department of Preventive Veterinary Medicine, Federal University of Santa Maria, Santa Maria, RS, Brazil
| |
Collapse
|
95
|
Martinello R, Milani A, Geuna E, Zucchini G, Aversa C, Nuzzo A, Montemurro F. Investigational ErbB-2 tyrosine kinase inhibitors for the treatment of breast cancer. Expert Opin Investig Drugs 2016; 25:393-403. [DOI: 10.1517/13543784.2016.1153063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
96
|
Clinical role of HER2 gene amplification and chromosome 17: a study on 154 IHC-equivocal cases of invasive breast carcinoma patients. Tumour Biol 2016; 37:8665-72. [PMID: 26738861 DOI: 10.1007/s13277-015-4657-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 08/25/2014] [Indexed: 01/21/2023] Open
Abstract
Accurate evaluation of human epidermal growth factor receptor 2 (HER2) status is quite crucial for invasive breast tumor patients in order to select anti-HER2 therapy for effective clinical outcomes. Immunohistochemistry (IHC) assay is routinely used to evaluate the HER2 oncoprotein overexpression but is unable to explain the chromosomal and genetic alterations and has been considered as a hot issue in IHC-equivocal cases. We investigated these molecular aberrations in correlation with prognostic factors. A cohort of 154 IHC-equivocal (+2) cases was selected and retrospectively analyzed by dual-probe fluorescence in situ hybridization (FISH) assay by using locus-specific HER2 and centromere enumeration probes (CEP17) for the identification of HER2 proto-oncogene amplification and chromosomal copy number per cell, respectively. The data were analyzed by SPSS 16.0 version using chi-square test (p < 0.05). We identified 36 out of 154 cases (23.4 %) showing HER2 gene amplification (average HER2 gene copies per cell >4 or <4 with HER2/CEP17 ratio >2) in concordance with HER2 oncoprotein overexpression, and significant correlation was observed with prognostic parameters including histological type, tumor grade II to III, histology and pathological type, lymphatic invasion, ductal carcinoma in situ (DCIS), and estrogen-positive and progesterone-negative receptors. Of the 154 cases, 18 cases (11.7 %) showed polysomy 17 with CEP17 probe signals per cell ≥3 and 22 cases (14.3 %) presented monosomy 17 (CEP17 probe signals per cell ≤1). Our data indicate that the use of anti-HER2 therapy should not be suggested unless true evaluation of HER2 protein expression is made regarding gene amplification essentially in IHC-ambiguous invasive breast tumors.
Collapse
|
97
|
Wang Y, Sun T, Wan D, Sheng L, Li W, Zhu H, Li Y, Lu J. Hormone receptor status predicts the clinical outcome of human epidermal growth factor 2-positive metastatic breast cancer patients receiving trastuzumab therapy: a multicenter retrospective study. Onco Targets Ther 2015; 8:3337-48. [PMID: 26648738 PMCID: PMC4648606 DOI: 10.2147/ott.s91166] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Objectives Trastuzumab, a humanized monoclonal antibody that binds human epidermal growth factor receptor 2 (HER2), dramatically improves the clinical outcomes of HER2-positive breast cancer. Emerging evidence implied that the clinical behavior and sensitivity to targeted agents in HER2-positive breast cancer differed by hormone receptor (HR) status. The objective of this study was to determine the effect of the HR status on survival benefit of HER2-positive metastatic breast cancer when treated with anti-HER2-targeted therapy in People’s Republic of China. Methods Metastatic breast cancer patients with HER2-positive diseases across six cancer centers in People’s Republic of China were retrospectively analyzed in our study. Patients were classified into four groups according to HR/HER2 status and trastuzumab treatment: HER2+/HR+ patients with first-line trastuzumab treatment, HER2+/HR+ patients with no trastuzumab treatment, HER2+/HR− patients with first-line trastuzumab treatment, and HER2+/HR− patients with no trastuzumab treatment. Kaplan–Meier analysis, log-rank test, and multivariate analysis were performed during analysis. Results A total of 295 patients were included in the final analysis. The median overall survival was 30 months (95% confidence interval: 27.521–32.479). Among patients with HER2+/HR− disease, significant survival benefit was observed when treated with trastuzumab (30 vs 21 months, P=0.000). However, in patients with HER2+/HR+ disease, trastuzumab administration had a survival improvement trend but no significant statistical differences (36 vs 30 months, P=0.258). In the multivariate analysis, HR status was an independent predictor of overall survival and trastuzumab treatment had significantly decreased risk of death in HER2+/HR− patients (hazard ratio =0.330). Conclusion HR status is an independent predictor of overall survival in HER2-positive metastatic breast cancer patients and patients with HER2+/HR− subtype might be associated with more survival benefits when treated with trastuzumab-based regimens.
Collapse
Affiliation(s)
- Yunchao Wang
- Department of Breast Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Tao Sun
- Department of Oncology, Liaoning Cancer Hospital and Institute, Shenyang, People's Republic of China
| | - Donggui Wan
- Department of Oncology, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Lijun Sheng
- Department of Oncology, Shandong Cancer Hospital, Jinan, People's Republic of China
| | - Wei Li
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Huayun Zhu
- Department of Oncology, Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Yanping Li
- Department of Breast Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Janice Lu
- Department of Breast Cancer, Beijing Shijitan Hospital, Capital Medical University, Beijing, People's Republic of China ; Division of Hematology and Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
98
|
Wu VS, Kanaya N, Lo C, Mortimer J, Chen S. From bench to bedside: What do we know about hormone receptor-positive and human epidermal growth factor receptor 2-positive breast cancer? J Steroid Biochem Mol Biol 2015; 153:45-53. [PMID: 25998416 PMCID: PMC4568143 DOI: 10.1016/j.jsbmb.2015.05.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/08/2015] [Accepted: 05/12/2015] [Indexed: 12/11/2022]
Abstract
Breast cancer is a heterogeneous disease. Thanks to extensive efforts from research scientists and clinicians, treatment for breast cancer has advanced into the era of targeted medicine. With the use of several well-established biomarkers, such as hormone receptors (HRs) (i.e., estrogen receptor [ER] and progesterone receptor [PgR]) and human epidermal growth factor receptor-2 (HER2), breast cancer patients can be categorized into multiple subgroups with specific targeted treatment strategies. Although therapeutic strategies for HR-positive (HR+) HER2-negative (HER2-) breast cancer and HR-negative (HR-) HER2-positive (HER2+) breast cancer are well-defined, HR+ HER2+ breast cancer is still an overlooked subgroup without tailored therapeutic options. In this review, we have summarized the molecular characteristics, etiology, preclinical tools and therapeutic options for HR+ HER2+ breast cancer. We hope to raise the attention of both the research and the medical community on HR+ HER2+ breast cancer, and to advance patient care for this subtype of disease.
Collapse
Affiliation(s)
- Victoria Shang Wu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Noriko Kanaya
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Chiao Lo
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Joanne Mortimer
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Medical Center Duarte, CA, United States
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States.
| |
Collapse
|
99
|
McBryan J, Fagan A, McCartan D, Bane FT, Varešlija D, Cocchiglia S, Byrne C, Bolger J, McIlroy M, Hudson L, Tibbitts P, Ó Gaora P, Hill AD, Young LS. Transcriptomic Profiling of Sequential Tumors from Breast Cancer Patients Provides a Global View of Metastatic Expression Changes Following Endocrine Therapy. Clin Cancer Res 2015; 21:5371-9. [DOI: 10.1158/1078-0432.ccr-14-2155] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 07/21/2015] [Indexed: 11/16/2022]
|
100
|
Affiliation(s)
- Filippo Montemurro
- Unit of Investigative Clinical Oncology (INCO), Fondazione del Piemonte per l'Oncologia, Candiolo Cancer Institute (IRCCs), Strada Provinciale 142, Km 3.95, Candiolo, 10060, Italy.
| |
Collapse
|