201
|
HER2-positive breast cancer: Current and new therapeutic strategies. Breast 2018; 39:80-88. [PMID: 29631097 DOI: 10.1016/j.breast.2018.03.006] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/11/2018] [Accepted: 03/19/2018] [Indexed: 11/20/2022] Open
Abstract
Since the identification of the HER2 receptor amplification as an adverse prognostic factor that defined a special subtype of metastatic breast cancer, there has been a substantial improvement in survival of patients affected with this disease due to the development of anti-HER2 targeted therapies. The approval of trastuzumab and pertuzumab associated to a taxane in first line and subsequent treatment with the antibody-drug conjugate T-DM1 has certainly contributed to achieve these outcomes. The Tyrosine Kinase Inhibitor lapatinib was also approved in the basis of an improvement in progression free survival, becoming another commonly used treatment in combination with capecitabine. Inevitably, despite these therapeutic advances most patients progress on therapy due to primary or acquired resistance or because of an incorrect HER2 positivity assessment. Hence, it is crucial to correctly categorize HER2 amplified tumors and define mechanisms of resistance to design effective new treatment approaches. In addition, identifying biomarkers of response or resistance permits to tailor the therapeutic options for each patient sparing them from unnecessary toxicity as well as improving their outcomes. The aim of this review is to examine new strategies in development to treat HER2-positive metastatic breast cancer referring to the mechanisms of action of new drugs and new combinations including results reported so far.
Collapse
|
202
|
Larionov AA. Current Therapies for Human Epidermal Growth Factor Receptor 2-Positive Metastatic Breast Cancer Patients. Front Oncol 2018; 8:89. [PMID: 29670855 PMCID: PMC5894159 DOI: 10.3389/fonc.2018.00089] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 03/14/2018] [Indexed: 01/01/2023] Open
Abstract
The median survival of patients with human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer (MBC) has more than doubled, since the discovery of HER2-targeted treatments: it rose from less than 2 years in 2001 (prior introduction of trastuzumab) to more than 4 years in 2017. The initial generation of HER2-targeted therapies included trastuzumab with taxanes in the first line, followed by the addition of lapatinib and by a switch to another cytotoxic agent after progression. Results of CLEOPATRA, EMILIA, and TH3RESA trials have changed this clinical practice. The current consensus includes horizontal dual blockade (trastuzumab + pertuzumab) with taxanes or vinorelbine in the first line, followed by trastuzumab-emtansine (T-DM1) in the second line, with addition of lapatinib in the later lines of treatment. However, the fast and simultaneous development of new drugs led to a relative shortage of clinical evidence to support this sequence. Triple-positive breast cancers (TPBC), which express both hormonal receptors and HER2, constitute nearly half of HER2-positive cases. For these tumors, the current consensus is to add endocrine therapy after completion of cytotoxic treatment. Again, this consensus is not fully evidence-based. In view of the recent progress in treatment of estrogen-receptor positive breast cancers, a series of trials is evaluating addition of CDK4/6 inhibitors, aromatase inhibitors or fulvestrant to HER2-targeted and cytotoxic chemotherapy in TPBC patients. Despite the remarkable progress in treatment of HER2-positive breast cancer, metastatic disease is still incurable in the majority of patients. A wide range of novel therapies are under development to prevent and overcome resistance to current HER2-targeted agents. This review discusses pivotal clinical trials that have shaped current clinical practices, the current consensus recommendations, and the new experimental treatments in metastatic HER2-positive breast cancer.
Collapse
Affiliation(s)
- Alexey A Larionov
- Department of Medical Genetics, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
203
|
Auclin E, Zaanan A, Vernerey D, Douard R, Gallois C, Laurent-Puig P, Bonnetain F, Taieb J. Subgroups and prognostication in stage III colon cancer: future perspectives for adjuvant therapy. Ann Oncol 2018; 28:958-968. [PMID: 28453690 DOI: 10.1093/annonc/mdx030] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Since the MOSAIC study, oxaliplatin-based adjuvant chemotherapy has been the standard treatment of stage III colon cancer. Combination therapy with fluoropyrimidines and oxaliplatin has improved overall survival (OS) and reduced the risk of recurrence in patients with resected stage III colon cancer. However, only 20% of patients really benefit from adjuvant chemotherapy, exposing 80% of patients to unnecessary toxicity. Recent analyses of large multicenter adjuvant studies have focused on the prognostication of OS and disease-free survival in stage III colon cancer in order to reduce over-treatment and to find more accurate prognostic tools than those used for adjuvant treatment decision-making in stage II disease. Indeed, clinical and pathological prognostic factors, although important, are not sufficient to decide which stage III patients will benefit from adjuvant therapy, and biomarkers will help select patient that need adjuvant treatment. Molecular markers such as microsatellite status and BRAF and KRAS mutations have recently been explored, and molecular signatures have been identified as promising prognostic factor for OS. Furthermore, recent studies have highlighted the prognostic value of immune infiltration. This review focuses on pathologic, immunologic and molecular prognostic markers for stage III colon cancer that could help clinicians tailor adjuvant treatment in a comprehensive transversal approach.
Collapse
Affiliation(s)
- E Auclin
- Department of Digestive Oncology, European Georges Pompidou Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France.,Methodological and Quality of Life in Oncology Unit, Besançon, France
| | - A Zaanan
- Department of Digestive Oncology, European Georges Pompidou Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France.,Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - D Vernerey
- Methodological and Quality of Life in Oncology Unit, Besançon, France
| | - R Douard
- Department of Digestive Surgery, European Georges Pompidou Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - C Gallois
- Department of Digestive Oncology, European Georges Pompidou Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - P Laurent-Puig
- Paris Descartes University, Sorbonne Paris Cité, Paris, France.,Department of Biology, European Georges Pompidou Hospital, Assistance Publique des Hôpitaux de Paris, INSERM-UMR-S1147, Paris, France
| | - F Bonnetain
- Methodological and Quality of Life in Oncology Unit, Besançon, France
| | - J Taieb
- Department of Digestive Oncology, European Georges Pompidou Hospital, Assistance Publique des Hôpitaux de Paris, Paris, France.,Paris Descartes University, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
204
|
Collovà E, Ferzi A, Scandurra G, Aurilio G, Torri V, Porcu L, Sanò MV, Taibi E, Foglietta J, Generali D, Andreis D, Dazzani MC, Bramati A, Marcon I, Atzori F, Cinieri S, Tondulli L, Grasso D, Nolè F, Petrella MC, Gori S, La Verde N. Efficacy of Trastuzumab in Unselected Patients with HER2-Positive Metastatic Breast Cancer: A Retrospective Analysis. TUMORI JOURNAL 2018. [DOI: 10.1177/1636.17902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Elena Collovà
- Ospedale Civile di Legnano, Department of Oncology, Legnano, Milan
| | - Antonella Ferzi
- Ospedale Civile di Legnano, Department of Oncology, Legnano, Milan
| | | | - Gaetano Aurilio
- European Institute of Oncology, Department of Oncology, Milan
| | - Valter Torri
- “Mario Negri” Institute, Laboratory of Methodology for Biomedical Research, Milan
| | - Luca Porcu
- “Mario Negri” Institute, Laboratory of Methodology for Biomedical Research, Milan
| | - Maria Vita Sanò
- Humanitas Centro Catanese di Oncologia, Department of Oncology, Catania
| | - Eleonora Taibi
- Humanitas Centro Catanese di Oncologia, Department of Oncology, Catania
| | - Jennifer Foglietta
- Azienda Ospedaliera Perugia, Department of Oncology, S. Andrea delle Fratte, Perugia
| | - Daniele Generali
- Istituti Ospitalieri di Cremona, Department of Mammarian Pathology, Cremona
| | - Daniele Andreis
- Istituti Ospitalieri di Cremona, Department of Mammarian Pathology, Cremona
| | | | - Annalisa Bramati
- Azienda Ospedaliera Fatebenefratelli e Oftalmico, Department of Oncology, Milan
| | - Ilaria Marcon
- Azienda Ospedaliera di Circolo e Fondazione Macchi, Department of Oncology, Varese
| | - Francesco Atzori
- University Hospital, Department of Oncology, bivio Sestu, Monserrato, Cagliari
| | - Saverio Cinieri
- Ospedale “A Perrino”, Department of Medical Oncology, Division & Breast Unit, Brindisi
| | | | - Donatella Grasso
- IRCC Policlinico San Matteo, Department of Hematology and Oncology, Pavia
| | - Franco Nolè
- European Institute of Oncology, Department of Oncology, Milan
| | | | - Stefania Gori
- Ospedale Sacro Cuore Don Calabria, Department of Oncology, Negrar (VR), Italy
| | - Nicla La Verde
- Azienda Ospedaliera Fatebenefratelli e Oftalmico, Department of Oncology, Milan
| |
Collapse
|
205
|
Lu Q, Wang L, Zhang Y, Yu X, Wang C, Wang H, Yang Y, Chong X, Xia T, Meng Y, Wang Y, Lu C, Zhou L, Li B. An anti-ErbB2 fully human antibody circumvents trastuzumab resistance. Oncotarget 2018; 7:67129-67141. [PMID: 27564098 PMCID: PMC5341862 DOI: 10.18632/oncotarget.11562] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/11/2016] [Indexed: 02/04/2023] Open
Abstract
Trastuzumab, an anti-HER2/ErbB2 humanized antibody, has shown great clinical benefits in ErbB2-positive breast cancer treatment. Despite of its effectiveness, response rate to trastuzumab is limited and resistance is common. Here, we developed a new anti-ErbB2 antibody, denoted as H2-18, which was isolated from a phage display human antibody library. Previous studies have demonstrated that trastuzumab recognizes the juxtamembrane region of domain IV, and pertuzumab, another humanized ErbB2-specific antibody, binds to ErbB2 near the center of domain II. Our crystallographic analysis showed that the epitope recognized by H2-18 is within domain I of the ErbB2 molecule. H2-18 potently induced programmed cell death (PCD) in both trastuzumab-sensitive and -resistant breast cancer cell lines, while trastuzumab and pertuzumab, either used alone or in combination, only exhibits very weak PCD-inducing activity. More importantly, H2-18 could inhibit the growth of trastuzumab-resistant breast cancer cells far more effectively than trastuzumab plus pertuzumab, both in vitro and in vivo. In conclusion, H2-18 shows a unique ability to overcome trastuzumab resistance, suggesting that it has the great potential to be translated to the clinic.
Collapse
Affiliation(s)
- Qiong Lu
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Lingfei Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Yajun Zhang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Xiaojie Yu
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Chao Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Huajing Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Yang Yang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Xiaodan Chong
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Tian Xia
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Yanchun Meng
- School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Yuxiao Wang
- Central Laboratory, Navy General Hospital, Beijing People's Republic of China
| | - Cuihua Lu
- Department of Gastroenterology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Lijun Zhou
- Central Laboratory, Navy General Hospital, Beijing People's Republic of China
| | - Bohua Li
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| |
Collapse
|
206
|
Butti R, Das S, Gunasekaran VP, Yadav AS, Kumar D, Kundu GC. Receptor tyrosine kinases (RTKs) in breast cancer: signaling, therapeutic implications and challenges. Mol Cancer 2018; 17:34. [PMID: 29455658 PMCID: PMC5817867 DOI: 10.1186/s12943-018-0797-x] [Citation(s) in RCA: 239] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 02/01/2018] [Indexed: 12/19/2022] Open
Abstract
Breast cancer is a multifactorial disease and driven by aberrant regulation of cell signaling pathways due to the acquisition of genetic and epigenetic changes. An array of growth factors and their receptors is involved in cancer development and metastasis. Receptor Tyrosine Kinases (RTKs) constitute a class of receptors that play important role in cancer progression. RTKs are cell surface receptors with specialized structural and biological features which respond to environmental cues by initiating appropriate signaling cascades in tumor cells. RTKs are known to regulate various downstream signaling pathways such as MAPK, PI3K/Akt and JAK/STAT. These pathways have a pivotal role in the regulation of cancer stemness, angiogenesis and metastasis. These pathways are also imperative for a reciprocal interaction of tumor and stromal cells. Multi-faceted role of RTKs renders them amenable to therapy in breast cancer. However, structural mutations, gene amplification and alternate pathway activation pose challenges to anti-RTK therapy.
Collapse
Affiliation(s)
- Ramesh Butti
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, SP Pune University Campus, Pune, 411007, India
| | - Sumit Das
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, SP Pune University Campus, Pune, 411007, India
| | - Vinoth Prasanna Gunasekaran
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, SP Pune University Campus, Pune, 411007, India
| | - Amit Singh Yadav
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, SP Pune University Campus, Pune, 411007, India
| | - Dhiraj Kumar
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77054, USA
| | - Gopal C Kundu
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science, SP Pune University Campus, Pune, 411007, India.
| |
Collapse
|
207
|
Hyman DM, Piha-Paul SA, Won H, Rodon J, Saura C, Shapiro GI, Juric D, Quinn DI, Moreno V, Doger B, Mayer IA, Boni V, Calvo E, Loi S, Lockhart AC, Erinjeri JP, Scaltriti M, Ulaner GA, Patel J, Tang J, Beer H, Selcuklu SD, Hanrahan AJ, Bouvier N, Melcer M, Murali R, Schram AM, Smyth LM, Jhaveri K, Li BT, Drilon A, Harding JJ, Iyer G, Taylor BS, Berger MF, Cutler RE, Xu F, Butturini A, Eli LD, Mann G, Farrell C, Lalani AS, Bryce RP, Arteaga CL, Meric-Bernstam F, Baselga J, Solit DB. HER kinase inhibition in patients with HER2- and HER3-mutant cancers. Nature 2018; 554:189-194. [PMID: 29420467 PMCID: PMC5808581 DOI: 10.1038/nature25475] [Citation(s) in RCA: 593] [Impact Index Per Article: 84.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 12/22/2017] [Indexed: 12/11/2022]
Abstract
Somatic mutations of ERBB2 and ERBB3 (which encode HER2 and HER3, respectively) are found in a wide range of cancers. Preclinical modelling suggests that a subset of these mutations lead to constitutive HER2 activation, but most remain biologically uncharacterized. Here we define the biological and therapeutic importance of known oncogenic HER2 and HER3 mutations and variants of unknown biological importance by conducting a multi-histology, genomically selected, 'basket' trial using the pan-HER kinase inhibitor neratinib (SUMMIT; clinicaltrials.gov identifier NCT01953926). Efficacy in HER2-mutant cancers varied as a function of both tumour type and mutant allele to a degree not predicted by preclinical models, with the greatest activity seen in breast, cervical and biliary cancers and with tumours that contain kinase domain missense mutations. This study demonstrates how a molecularly driven clinical trial can be used to refine our biological understanding of both characterized and new genomic alterations with potential broad applicability for advancing the paradigm of genome-driven oncology.
Collapse
Affiliation(s)
- David M. Hyman
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Helen Won
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jordi Rodon
- Vall d’Hebron University Hospital, Vall d’Hebron
Institute of Oncology (VHIO), Barcelona, Spain
| | - Cristina Saura
- Vall d’Hebron University Hospital, Vall d’Hebron
Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Dejan Juric
- Massachusetts Hospital Cancer Center, Boston, MA, USA
| | - David I. Quinn
- USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | | | | | | | - Valentina Boni
- START Madrid, Centro Integral Oncológico Clara Campal
(CIOCC), Madrid, Spain
| | - Emiliano Calvo
- START Madrid, Centro Integral Oncológico Clara Campal
(CIOCC), Madrid, Spain
| | - Sherene Loi
- Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Albert C. Lockhart
- Washington University in St. Louis School of Medicine, St. Louis,
MO, USA
| | | | | | - Gary A. Ulaner
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juber Patel
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jiabin Tang
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hannah Beer
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Nancy Bouvier
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Myra Melcer
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | - Komal Jhaveri
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bob T. Li
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Gopa Iyer
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | - Feng Xu
- Puma Biotechnology Inc., Los Angeles, CA, USA
| | | | - Lisa D. Eli
- Puma Biotechnology Inc., Los Angeles, CA, USA
| | - Grace Mann
- Puma Biotechnology Inc., Los Angeles, CA, USA
| | | | | | | | | | | | - José Baselga
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David B. Solit
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
208
|
Abstract
Somatic mutations of ERBB2 and ERBB3 (which encode HER2 and HER3, respectively) are found in a wide range of cancers. Preclinical modelling suggests that a subset of these mutations lead to constitutive HER2 activation, but most remain biologically uncharacterized. Here we define the biological and therapeutic importance of known oncogenic HER2 and HER3 mutations and variants of unknown biological importance by conducting a multi-histology, genomically selected, 'basket' trial using the pan-HER kinase inhibitor neratinib (SUMMIT; clinicaltrials.gov identifier NCT01953926). Efficacy in HER2-mutant cancers varied as a function of both tumour type and mutant allele to a degree not predicted by preclinical models, with the greatest activity seen in breast, cervical and biliary cancers and with tumours that contain kinase domain missense mutations. This study demonstrates how a molecularly driven clinical trial can be used to refine our biological understanding of both characterized and new genomic alterations with potential broad applicability for advancing the paradigm of genome-driven oncology.
Collapse
|
209
|
Metastatic Breast Cancer Treated with Lapatinib with a Prolonged Benefit: A Case Report and a Review of Therapeutic Options Available. TUMORI JOURNAL 2018. [DOI: 10.1177/030089161309900617] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A 53-year-old woman was treated, after trastuzumab progression of HER2-positive metastatic disease, with capecitabine plus lapatinib and subsequently with lapatinib alone. To date, she has had persistent remission of disease since 2009.
Collapse
|
210
|
Choi JH, Kim KH, Roh KH, Jung H, Lee A, Lee JY, Song JY, Park SJ, Kim I, Lee WS, Seo SK, Choi IW, Fu YX, Yea SS, Park S. A PI3K p110α-selective inhibitor enhances the efficacy of anti-HER2/neu antibody therapy against breast cancer in mice. Oncoimmunology 2018; 7:e1421890. [PMID: 29721370 DOI: 10.1080/2162402x.2017.1421890] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 10/18/2022] Open
Abstract
Combination therapies with phosphoinositide 3-kinase (PI3K) inhibitors and trastuzumab (anti-human epidermal growth factor receptor [HER]2/neu antibody) are effective against HER2+ breast cancer. Isoform-selective PI3K inhibitors elicit anti-tumor immune responses that are distinct from those induced by inhibitors of class I PI3K isoforms (pan-PI3K inhibitors). The present study investigated the therapeutic effect and potential for stimulating anti-tumor immunity of combined therapy with an anti-HER2/neu antibody and pan-PI3K inhibitor (GDC-0941) or a PI3K p110α isoform-selective inhibitor (A66) in mouse models of breast cancer. The anti-neu antibody inhibited tumor growth and enhanced anti-tumor immunity in HER2/neu+ breast cancer TUBO models, whereas GDC-0941 or A66 alone did not. Anti-neu antibody and PI3K inhibitor synergistically promoted anti-tumor immunity by increasing functional T cell production. In the presence of the anti-neu antibody, A66 was more effective than GDC-0941 at increasing the fraction of CD4+, CD8+, and IFN-γ+CD8+ T cells in the tumor-infiltrating lymphocyte population. Detection of IFN-γ levels by enzyme-linked immunospot assay showed that the numbers of tumor-specific T cells against neu and non-neu tumor antigens were increased by combined PI3K inhibitor plus anti-neu antibody treatment, with A66 exhibiting more potent effects than GDC-0941. In a TUBO (neu+) and TUBO-P2J (neu-) mixed tumor model representing immunohistochemistry 2+ tumors, A66 suppressed tumor growth and prolonged survival to a greater extent than GDC-0941 when combined with anti-neu antibody. These results demonstrate that a PI3K p110α-isoform-selective inhibitor is an effective adjunct to trastuzumab in the treatment of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Jae-Hyeog Choi
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan, Republic of Korea
| | - Ki Hyang Kim
- Department of Internal Medicine, Inje University College of Medicine, Busan, Republic of Korea
| | - Kug-Hwan Roh
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan, Republic of Korea
| | - Hana Jung
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan, Republic of Korea
| | - Anbok Lee
- Department of Surgery, Inje University College of Medicine, Busan, Republic of Korea
| | - Ji-Young Lee
- Department of Internal Medicine, Inje University College of Medicine, Busan, Republic of Korea
| | - Joo Yeon Song
- Department of Pathology, Dongnam Institute of Radiological and Medical Sciences, Busan, Republic of Korea
| | - Seung Jae Park
- Department of Internal Medicine, Inje University College of Medicine, Busan, Republic of Korea
| | - Ilhwan Kim
- Department of Internal Medicine, Inje University College of Medicine, Busan, Republic of Korea
| | - Won-Sik Lee
- Department of Internal Medicine, Inje University College of Medicine, Busan, Republic of Korea
| | - Su-Kil Seo
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan, Republic of Korea
| | - Il-Whan Choi
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan, Republic of Korea
| | - Yang-Xin Fu
- The Department of Pathology and Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sung Su Yea
- Department of Biochemistry, Inje University College of Medicine, Busan, Republic of Korea
| | - SaeGwang Park
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan, Republic of Korea
| |
Collapse
|
211
|
Insertional mutagenesis in a HER2-positive breast cancer model reveals ERAS as a driver of cancer and therapy resistance. Oncogene 2018; 37:1594-1609. [PMID: 29326437 PMCID: PMC6168451 DOI: 10.1038/s41388-017-0031-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 10/29/2017] [Accepted: 10/31/2017] [Indexed: 12/21/2022]
Abstract
Personalized medicine for cancer patients requires a deep understanding of the underlying genetics that drive cancer and the subsequent identification of predictive biomarkers. To discover new genes and pathways contributing to oncogenesis and therapy resistance in HER2+ breast cancer, we performed Mouse Mammary Tumor Virus (MMTV)-induced insertional mutagenesis screens in ErbB2/cNeu-transgenic mouse models. The screens revealed 34 common integration sites (CIS) in mammary tumors of MMTV-infected mice, highlighting loci with multiple independent MMTV integrations in which potential oncogenes are activated, most of which had never been reported as MMTV CIS. The CIS most strongly associated with the ErbB2-transgenic genotype was the locus containing Eras (ES cell-expressed Ras), a constitutively active RAS-family GTPase. We show that upon expression, Eras acts as a potent oncogenic driver through hyperactivation of the PI3K/AKT pathway, in contrast to other RAS proteins that signal primarily via the MAPK/ERK pathway and require upstream activation or activating mutations to induce signaling. We additionally show that ERAS synergistically enhances HER2-induced tumorigenesis and, in this role, can functionally replace ERBB3/HER3 by acting as a more powerful activator of PI3K/AKT signaling. Although previously reported as pseudogene in humans, we observed ERAS RNA and protein expression in a substantial subset of human primary breast carcinomas. Importantly, we show that ERAS induces primary resistance to the widely used HER2-targeting drugs Trastuzumab (Herceptin) and Lapatinib (Tykerb/Tyverb) in vivo, and is involved in acquired resistance via selective upregulation during treatment in vitro, indicating that ERAS may serve as a novel clinical biomarker for PI3K/AKT pathway hyperactivation and HER2-targeted therapy resistance.
Collapse
|
212
|
Angelbello AJ, Chen JL, Childs-Disney JL, Zhang P, Wang ZF, Disney MD. Using Genome Sequence to Enable the Design of Medicines and Chemical Probes. Chem Rev 2018; 118:1599-1663. [PMID: 29322778 DOI: 10.1021/acs.chemrev.7b00504] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Rapid progress in genome sequencing technology has put us firmly into a postgenomic era. A key challenge in biomedical research is harnessing genome sequence to fulfill the promise of personalized medicine. This Review describes how genome sequencing has enabled the identification of disease-causing biomolecules and how these data have been converted into chemical probes of function, preclinical lead modalities, and ultimately U.S. Food and Drug Administration (FDA)-approved drugs. In particular, we focus on the use of oligonucleotide-based modalities to target disease-causing RNAs; small molecules that target DNA, RNA, or protein; the rational repurposing of known therapeutic modalities; and the advantages of pharmacogenetics. Lastly, we discuss the remaining challenges and opportunities in the direct utilization of genome sequence to enable design of medicines.
Collapse
Affiliation(s)
- Alicia J Angelbello
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Jonathan L Chen
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Jessica L Childs-Disney
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Peiyuan Zhang
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Zi-Fu Wang
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Matthew D Disney
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
213
|
Kim YJ, Sung D, Oh E, Cho Y, Cho TM, Farrand L, Seo JH, Kim JY. Flubendazole overcomes trastuzumab resistance by targeting cancer stem-like properties and HER2 signaling in HER2-positive breast cancer. Cancer Lett 2018; 412:118-130. [DOI: 10.1016/j.canlet.2017.10.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/16/2017] [Accepted: 10/16/2017] [Indexed: 12/15/2022]
|
214
|
Abstract
The human epidermal growth factor receptor (HER) family of receptor tyrosine kinases plays an important role in the biology of many cancers. In breast and gastrointestinal cancer, and at lower rates also in additional tumor types, HER2 and its homo- or heterodimerization with HER1 or HER3 are essential for cancer cell growth and survival. Breast cancer patients overexpressing HER2 have a more aggressive course of their disease. The poor prognosis associated with HER2 overexpression can be substantially improved by adding HER2-targeted therapy to standard of care using the monoclonal antibody trastuzumab. Lapatinib, an oral dual tyrosine kinase inhibitor, blocks HER1 and HER2 tyrosine kinase activity by binding to the ATP-binding site of the receptor's intracellular domain, resulting in inhibition of tumor cell growth. Lapatinib is generally well tolerated with diarrhea being the most common adverse effect. However, although being mainly of mild to moderate severity, interruption or discontinuation of treatment has been reported in a substantial proportion of patients in clinical trials. In 2007, lapatinib has been approved in combination with capecitabine in patients with advanced HER2-positive breast cancer upon progressive disease following standard therapy with anthracyclines, taxanes, and trastuzumab. In 2013, the approval was extended to a chemotherapy-free combination with trastuzumab for patients with metastatic HER2-positive, hormone receptor-negative breast cancer progressing on prior trastuzumab and chemotherapy. Since 2010, lapatinib is approved in combination with letrozole in the treatment of postmenopausal women with advanced HER2- and hormone receptor-positive breast cancer. In contrast, in first-line cytotoxic-based therapy of both early and advanced HER2-positive breast cancer, data from clinical trials did not provide evidence of additional benefit of lapatinib compared to trastuzumab. Moreover, over the past few years, novel HER2-targeted drugs, either alone or as a combined anti-HER2 approach, have been extensively evaluated, demonstrating a more favorable outcome. Also, neither in first- nor second-line treatment of advanced gastric cancer, lapatinib has been proven to be superior compared to trastuzumab as hitherto standard of care HER2 blockade. Therefore, lapatinib has become somewhat less important in patients with HER2-positive breast cancer during the past 10 years since its first introduction. Nevertheless, consideration of treatment with lapatinib appears to be reasonable in selected patients not only in the approved applications but also beyond, and further indications such as HER2-positive refractory metastatic colorectal cancer may arise in future. Also, lapatinib may have distinct advantages over antibodies in targeting truncated HER2 and crossing the blood-brain barrier. Finally, the favorable cardiac toxicity profile of lapatinib makes it an attractive alternative to trastuzumab-based regimens in patients at risk for cardiac events.
Collapse
Affiliation(s)
- Minna Voigtlaender
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tanja Schneider-Merck
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Glaxo Smith Kline, Hamburg, Germany
| | - Martin Trepel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Department of Hematology and Oncology, Interdisciplinary Cancer Center Augsburg, Augsburg Medical Center, Stenglinstr. 2, 86156, Augsburg, Germany.
| |
Collapse
|
215
|
Abstract
Concurrent with an expansion in the number of agents available for the treatment of advanced CRC, there has been an increase in our understanding of selection biomarkers to optimize the management of patients with this disease. For CRC patients being considered for anti-EGFR therapy, expanded RAS testing is the standard of care to determine the subset of patients who can benefit from cetuximab or panitumumab in conjunction with chemotherapy. A small fraction of patients have HER2 amplification where emerging data suggest treatment with drugs targeting this alteration. Although advanced CRC patients who harbor the BRAF V600E mutation have a poorer prognosis, they are eligible for combinatorial therapy targeting EGFR/BRAF or BRAF/MEK within the MAP kinase signaling pathway. Once primarily thought to be a negative prognostic marker, BRAF V600E mutation is now considered as a positive predictive factor with an opportunity for clinical intervention. A growing body of evidence also supports MSI testing as clinical benefits with immune checkpoint blockade by cancer immunotherapy have been demonstrated in MSI-high patients whose tumors exhibit high mutational burden. It has been established that UGT1A1*28 polymorphism is associated with irinotecan toxicity, but this test is rarely performed as the management strategy has not been identified. No established predictive biomarker for anti-VEGF therapy has yet to be discovered.It is becoming increasingly apparent that our growing understanding of biomarkers is revolutionizing and improving our strategies in the treatment of advanced CRC. Traditional nonselective cytotoxic chemotherapy is gradually being augmented and even in some cases supplanted by selective targeted agents based on our increasing understanding of tumor signaling and mechanism at the molecular level. The prospect of personalized medicine in directing treatment approaches that are optimally beneficial for patients brings tremendous excitement to the growing field of cancer therapeutics. As discussed in this chapter, the concurrent development of molecular biomarkers with new treatment strategies holds great promise of precision medicine in improving outcomes for patients with advanced CRC.
Collapse
Affiliation(s)
- Patrick S Lin
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Thomas J Semrad
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA.
- Gene Upshaw Memorial Tahoe Forest Cancer Center, Truckee, CA, USA.
| |
Collapse
|
216
|
Cardiac biomarkers for early detection and prediction of trastuzumab and/or lapatinib-induced cardiotoxicity in patients with HER2-positive early-stage breast cancer: a NeoALTTO sub-study (BIG 1-06). Breast Cancer Res Treat 2017; 168:631-638. [PMID: 29280043 DOI: 10.1007/s10549-017-4628-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 12/18/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Biomarkers of cardiac damages, such as troponin T (TnT) and the amino-terminal fragment of brain natriuretic peptide (NT-proBNP), may be useful as early predictors of cardiac dysfunction. The role of these biomarkers in patients receiving lapatinib and/or trastuzumab before anthracyclines is unknown. This study explores TnT and NT-proBNP as predictors of early cardiac toxicity in neoadjuvant breast cancer patients. METHODS This sub-study of the NEOALTTO trial tested if changes in the levels of TnT and NT-proBNP occurred after 2 weeks of anti-HER2 therapy (lapatinib, trastuzumab or their combination) alone and/or after 18 weeks of anti-HER2 therapy plus weekly paclitaxel. RESULTS 173 and 172 were tested at all three timepoints for NT-proBNP and TnT, respectively. The incidence of biomarker elevation was overall low at all timepoints for all the three treatment arms. A total of 13 CEs in 11 patients occurred. Biomarker elevations in patients with CEs were very rare; only one patient with subsequent CE had a NT-proBNP elevation at baseline and at week 2. CONCLUSION These results suggest that TnT and proBNP may not be useful as early predictors of cardiac toxicity in anthracycline-naïve patients receiving trastuzumab and/or lapatinib.
Collapse
|
217
|
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
|
218
|
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
|
219
|
Schwartz AD, Barney LE, Jansen LE, Nguyen TV, Hall CL, Meyer AS, Peyton SR. A biomaterial screening approach reveals microenvironmental mechanisms of drug resistance. Integr Biol (Camb) 2017; 9:912-924. [PMID: 29159332 PMCID: PMC5725273 DOI: 10.1039/c7ib00128b] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Traditional drug screening methods lack features of the tumor microenvironment that contribute to resistance. Most studies examine cell response in a single biomaterial platform in depth, leaving a gap in understanding how extracellular signals such as stiffness, dimensionality, and cell-cell contacts act independently or are integrated within a cell to affect either drug sensitivity or resistance. This is critically important, as adaptive resistance is mediated, at least in part, by the extracellular matrix (ECM) of the tumor microenvironment. We developed an approach to screen drug responses in cells cultured on 2D and in 3D biomaterial environments to explore how key features of ECM mediate drug response. This approach uncovered that cells on 2D hydrogels and spheroids encapsulated in 3D hydrogels were less responsive to receptor tyrosine kinase (RTK)-targeting drugs sorafenib and lapatinib, but not cytotoxic drugs, compared to single cells in hydrogels and cells on plastic. We found that transcriptomic differences between these in vitro models and tumor xenografts did not reveal mechanisms of ECM-mediated resistance to sorafenib. However, a systems biology analysis of phospho-kinome data uncovered that variation in MEK phosphorylation was associated with RTK-targeted drug resistance. Using sorafenib as a model drug, we found that co-administration with a MEK inhibitor decreased ECM-mediated resistance in vitro and reduced in vivo tumor burden compared to sorafenib alone. In sum, we provide a novel strategy for identifying and overcoming ECM-mediated resistance mechanisms by performing drug screening, phospho-kinome analysis, and systems biology across multiple biomaterial environments.
Collapse
Affiliation(s)
- Alyssa D Schwartz
- Department of Chemical Engineering, University of Massachusetts Amherst, 686 N Pleasant St. 159 Goessmann Laboratory, Amherst, MA 01003, USA.
| | | | | | | | | | | | | |
Collapse
|
220
|
|
221
|
Takegawa N, Yonesaka K. HER2 as an Emerging Oncotarget for Colorectal Cancer Treatment After Failure of Anti-Epidermal Growth Factor Receptor Therapy. Clin Colorectal Cancer 2017; 16:247-251. [DOI: 10.1016/j.clcc.2017.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/10/2017] [Accepted: 03/01/2017] [Indexed: 12/15/2022]
|
222
|
Yu S, Liu Q, Han X, Qin S, Zhao W, Li A, Wu K. Development and clinical application of anti-HER2 monoclonal and bispecific antibodies for cancer treatment. Exp Hematol Oncol 2017; 6:31. [PMID: 29209558 PMCID: PMC5704598 DOI: 10.1186/s40164-017-0091-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 11/20/2017] [Indexed: 12/15/2022] Open
Abstract
HER2-targeted immunotherapy consists of monoclonal antibodies (e.g. trastuzumab, pertuzumab), bispecific antibodies (e.g. MM-111, ertumaxomab) and activated T cells armed with anti-HER2 bispecific antibody (HER2Bi-aATC). Trastuzumab is a classic drug for the treatment of HER2 positive metastatic breast cancer. The combined application of pertuzumab, trastuzumab and paclitaxel has been suggested as a standard therapy for HER2 positive advanced breast cancer. The resistance to anti-HER2 antibody has resulted in disease progression. HER2-directed bispecific antibody may be a promising therapeutic approach for these patients. Ertumaxomab enhanced the interaction of immune effector cells and tumor cells. MM-111 simultaneously binds to HER2 and HER3 and blocks downstream signaling. Besides, HER2Bi-aATC is also an alternative therapeutic approach for HER2 positive cancers. In this review, we summarized the recent advancement of HER2-targeted monoclonal antibodies (trastuzumab, pertuzumab and T-DM1) and bispecific antibodies (MM-111, ertumaxomab and HER2Bi-aATC), especially focus on clinical trial results.
Collapse
Affiliation(s)
- Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Xinwei Han
- Department of Interventional Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Anping Li
- Department of Interventional Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| |
Collapse
|
223
|
Nitz UA, Gluz O, Christgen M, Grischke EM, Augustin D, Kuemmel S, Braun M, Potenberg J, Kohls A, Krauss K, Stefek A, Schumacher C, Forstbauer H, Reimer T, Fischer H, Liedtke C, Wuerstlein R, Schumacher J, Kates R, Kreipe H, Harbeck N. De-escalation strategies in HER2-positive early breast cancer (EBC): final analysis of the WSG-ADAPT HER2+/HR- phase II trial: efficacy, safety, and predictive markers for 12 weeks of neoadjuvant dual blockade with trastuzumab and pertuzumab ± weekly paclitaxel. Ann Oncol 2017; 28:2768-2772. [PMID: 28945833 DOI: 10.1093/annonc/mdx494] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Response rates in HER2-overexpressing EBC treated with neoadjuvant chemotherapy and trastuzumab (T) have been improved by addition of pertuzumab (P). The prospective, phase II, neoadjuvant WSG-ADAPT HER2+/HR- trial assessed whether patients with strong early response to dual blockade alone might achieve pathological complete response (pCR) comparable to that of patients receiving dual blockade and chemotherapy. PATIENTS AND METHODS Female patients with HER2+/HR- EBC (M0) were randomized (5:2) to 12 weeks of T + P ± weekly paclitaxel (pac) at 80 mg/m2. Early response was defined as proliferation decrease ≥30% of Ki-67 (versus baseline) or low cellularity (<500 invasive tumor cells) in the 3-week biopsy. The trial was designed to test non-inferiority for pCR in early responding patients of the T + P arm versus all chemotherapy-treated patients. RESULTS From February 2014 to December 2015, 160 patients were screened, 92 were randomized to T + P and 42 to T + P+pac. Baseline characteristics were well balanced (median age 54 versus 51.5 years, cT2 51.1 versus 52.4%, cN0 54.3 versus 61.9%); 91.3% of patients completed T + P per protocol and 92.9% T + P+pac. The pCR rate in the T + P+pac arm was 90.5%, compared with 36.3% in the T + P arm as a whole. In the T + P arm, 24/92 were classified as non-responders, and their pCR rate was only 8.3% compared with 44.7% in responders (38/92) and 42.9% in patients with unclassified early response (30/92). No new safety signals were observed in the study population. CONCLUSION Addition of taxane monotherapy to dual HER2 blockade in a 12-week neoadjuvant setting substantially increases pCR rates in HER2+/HR- EBC compared with dual blockade alone, even within early responders to dual blockade. Early non-response under dual blockade strongly predicts failure to achieve pCR.
Collapse
Affiliation(s)
- U A Nitz
- West German Study Group GmbH, Moenchengladabach; Evangelical Hospital Johanniter Bethesda, Breast Center Niederrhein, Moenchengladbach
| | - O Gluz
- West German Study Group GmbH, Moenchengladabach; Evangelical Hospital Johanniter Bethesda, Breast Center Niederrhein, Moenchengladbach; University of Cologne, Cologne.
| | - M Christgen
- Institute of Pathology, Medical School Hannover, Hannover
| | - E-M Grischke
- Department of Gynecology and Obstetrics, University Clinics Tuebingen, Tuebingen
| | | | - S Kuemmel
- Breast Center, Clinics Essen-Mitte, Essen
| | - M Braun
- Rotkreuz Clinics Munich Breast Center, Munich
| | - J Potenberg
- Department of Oncology Evangelical Waldkrankenhaus Berlin, Berlin
| | - A Kohls
- Department of Gynecology and Obstetrics, Evangelical Hospital, Ludwigsfelde
| | - K Krauss
- Department of Gynecology and Obstetrics, University Clinics RWTH, Aachen
| | - A Stefek
- Breast Center, Evangelical Hospital Johanniter, Stendal
| | - C Schumacher
- Breast Center, St. Elisabeth Hospital Cologne, Cologne
| | | | - T Reimer
- Department of Gynecology and Obstetrics, University Clinics Rostock, Suedstadt
| | - H Fischer
- Breast Center, Evangelical Hospital Gelsenkirchen, Gelsenkirchen
| | - C Liedtke
- Department of Gynecology and Obstetrics, University Clinics Schleswig-Holstein/Campus Luebeck, Luebeck; Charite Berlin
| | - R Wuerstlein
- Breast Center, Department of Gynecology and Obstetrics, University of Munich (LMU) and CCCLMU, Munich
| | - J Schumacher
- Palleos Healthcare, Statistics, Wiesbaden, Germany
| | - R Kates
- West German Study Group GmbH, Moenchengladabach
| | | | - N Harbeck
- West German Study Group GmbH, Moenchengladabach; Breast Center, Department of Gynecology and Obstetrics, University of Munich (LMU) and CCCLMU, Munich
| |
Collapse
|
224
|
Abstract
Metastasis is one of the most characteristic yet problematic behaviors of cancer cells. Stage IV breast cancer accounts for a large portion of breast cancer-related morbidity and mortality. Despite early detection and improvement in survival owing to advancements in biomedical research and overall improvement of the health system, 6-10% of patients present with stage IV disease in the developed world, with a higher incidence noted elsewhere. Despite advances in biomedical research into cancer, up to 70-80% of patients with stage IV breast cancer die of cancer in 5 years, a disproportionally higher mortality compared with non-metastatic breast cancer. In this article, we review the incidence, survival, heterogeneity, current practice, and challenges in stage IV breast cancer, and we finish by noting new research initiatives to improve poor survival and suggesting future directions. By doing so, we hope to set the basis of future directions for both treating physicians and translational researchers to relieve the suffering of patients with stage IV breast cancer and improve the survival of patients with this dismal disease.
Collapse
Affiliation(s)
- Bora Lim
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Gabriel N Hortobagyi
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
225
|
Guerin M, Rezai K, Isambert N, Campone M, Autret A, Pakradouni J, Provansal M, Camerlo J, Sabatier R, Bertucci F, Charafe-Jauffret E, Hervieu A, Extra JM, Viens P, Lokiec F, Boher JM, Gonçalves A. PIKHER2: A phase IB study evaluating buparlisib in combination with lapatinib in trastuzumab-resistant HER2-positive advanced breast cancer. Eur J Cancer 2017; 86:28-36. [PMID: 28950146 DOI: 10.1016/j.ejca.2017.08.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND Phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin pathway is frequently activated in HER2-positive breast cancer and may play a major role in resistance to trastuzumab. Buparlisib is a pan-class-I PI3K inhibitor with potent and selective activity against wild-type and mutant PI3K p110 isoforms. PATIENTS AND METHODS PIKHER2 phase IB study aimed primarily to determine a maximum tolerated dose (MTD) and propose a recommended phase II dose (RP2D) for buparlisib in combination with lapatinib in HER2-positive, trastuzumab-resistant, advanced breast cancer. Oral buparlisib (40, 60 or 80 mg) and lapatinib (750, 1000 or 1250 mg) were administered daily. A modified continuous reassessment method using an adaptive Bayesian model guided the dose escalation of both agents. Secondary end-points included antitumour activity and pharmacokinetic (PK) assessments. RESULTS A total of 24 patients were treated across five dose levels. Dose-limiting toxicities included transaminases elevation, vomiting, stomatitis, hyperglycemia and diarrhoea. MTD was declared at buparlisib 80 mg/d + lapatinib 1250 mg/d, but toxicities and early treatment discontinuation rate beyond cycle 1 led to select buparlisib 80 mg + lapatinib 1000 mg/d as the RP2D. Main drug-related adverse events included diarrhoea, nausea, skin rash, asthenia, depression, anxiety and transaminases increase. There was no significant evidence for drug-drug PK interaction. Disease control rate was 79% [95% confidence interval [CI] 57-92%], one patient obtained a complete remission, and six additional patients experienced stable disease for ≥ 24 weeks (clinical benefit rate of 29% [95% CI 12-51%]). CONCLUSION Combining buparlisib and lapatinib in HER2-positive trastuzumab-resistant advanced breast cancer was feasible. Preliminary evidence of antitumour activity was observed in this heavily pre-treated population. TRIAL REGISTRATION ID NCT01589861.
Collapse
Affiliation(s)
- Mathilde Guerin
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Keyvan Rezai
- Institut Curie - Hôpital René Huguenin, Saint-Cloud, France
| | | | | | - Aurélie Autret
- Institut Paoli-Calmettes, Department of Clinical Research and Innovations, Marseille, France
| | - Jihane Pakradouni
- Institut Paoli-Calmettes, Department of Clinical Research and Innovations, Marseille, France
| | - Magali Provansal
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Jacques Camerlo
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Renaud Sabatier
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - François Bertucci
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Emmanuelle Charafe-Jauffret
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Biopathology, CRCM, Marseille, France
| | | | - Jean-Marc Extra
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Patrice Viens
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | | | - Jean-Marie Boher
- Institut Paoli-Calmettes, Department of Clinical Research and Innovations, Marseille, France; Aix Marseille University, INSERM, IRD, SESSTIM, Sciences Economiques & Sociales de La Santé & Traitement de L'Information Médicale, Marseille, France
| | - Anthony Gonçalves
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France.
| |
Collapse
|
226
|
Florido R, Smith KL, Cuomo KK, Russell SD. Cardiotoxicity From Human Epidermal Growth Factor Receptor-2 (HER2) Targeted Therapies. J Am Heart Assoc 2017; 6:JAHA.117.006915. [PMID: 28939718 PMCID: PMC5634312 DOI: 10.1161/jaha.117.006915] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Roberta Florido
- Division of Cardiology, Johns Hopkins University, Baltimore, MD
| | - Karen L Smith
- Breast and Ovarian Cancer Program, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | |
Collapse
|
227
|
Torres-García D, Pérez-Torres A, Manoutcharian K, Orbe U, Servín-Blanco R, Fragoso G, Sciutto E. GK-1 peptide reduces tumor growth, decreases metastatic burden, and increases survival in a murine breast cancer model. Vaccine 2017; 35:5653-5661. [PMID: 28890195 DOI: 10.1016/j.vaccine.2017.08.060] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/10/2017] [Accepted: 08/19/2017] [Indexed: 12/31/2022]
Abstract
GK-1 is a parasite-derived peptide adjuvant of 18 amino acid-length that enhances T-cell function and increases survival in B16-F10 melanoma tumor-bearing mice. This study was designed to evaluate in vivo the antitumor efficacy of GK-1 on 4T1 mouse mammary carcinoma. BALB/c mice with palpable primary tumors were weekly intravenously injected three times with saline solution or three different concentrations (10, 50, or 100μg per mouse) of GK-1. GK-1 significantly increased lifespan (p<0.0001) and reduced the primary tumor weight (p=0.014) and volume (p<0.0001) with respect to control mice, with no statistically significant differences among GK-1 doses. At the primary tumor, we found increased necrotic areas associated with a reduction in tumor mass, as well as an increase in the antitumor cytokine IL-12. Especially encouraging is the ability of GK-1 to reduce the number of lung metastasis (p=0.006) disregarding the dose used. The participation of IL-6 in metastasis development and the decreased levels of CCL-2, CCL-3, TNF-α, CXCL-9, GM-CSF, and b-FGF found in lungs of GK-1-treated mice is discussed. Our study supports the effectiveness of GK-1 as an antineoplastic agent that merits further exploration in combination with other therapeutic approaches in future translational studies.
Collapse
Affiliation(s)
- D Torres-García
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, 04510 Mexico City, Mexico
| | - A Pérez-Torres
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, 04510 Mexico City, Mexico
| | - K Manoutcharian
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, 04510 Mexico City, Mexico
| | - U Orbe
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, 04510 Mexico City, Mexico
| | - R Servín-Blanco
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, 04510 Mexico City, Mexico
| | - G Fragoso
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, 04510 Mexico City, Mexico
| | - E Sciutto
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, 04510 Mexico City, Mexico.
| |
Collapse
|
228
|
Jain D, Ahmad T, Cairo M, Aronow W. Cardiotoxicity of cancer chemotherapy: identification, prevention and treatment. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:348. [PMID: 28936442 PMCID: PMC5599271 DOI: 10.21037/atm.2017.06.35] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 06/07/2017] [Indexed: 12/21/2022]
Abstract
Cardiotoxicity is an important complication of several cancer therapeutic agents. Several well established and newer anticancer therapies such as anthracyclines, trastuzumab and other HER2 receptor blockers, antimetabolites, alkylating agents, tyrosine kinase inhibitors (TKIs), angiogenesis inhibitors, and checkpoint inhibitors are associated with significant cardiotoxicity. Cardiovascular imaging employing radionuclide imaging, echocardiography and magnetic resonance imaging are helpful in early detection and prevention of overt heart failure secondary to cardiotoxicity of cancer therapy. An understanding of the mechanism of the cardiotoxicity of cancer therapies can help prevent and treat their adverse cardiovascular consequences. Clinical implementation of algorithms based upon cardiac imaging and several non-imaging biomarkers can prevent cardiac morbidity and mortality associated with the use of cardiotoxic cancer therapies.
Collapse
Affiliation(s)
- Diwakar Jain
- Section of Cardiovascular Medicine, Department of Pediatrics, New York Medical College and Westchester Medical Center, Valhalla, NY, USA
| | - Tauseef Ahmad
- Section of Oncology and Hematology, Department of Pediatrics, New York Medical College and Westchester Medical Center, Valhalla, NY, USA
| | - Mitchel Cairo
- Section of Pediatric Hematology and Oncology, Department of Pediatrics, New York Medical College and Westchester Medical Center, Valhalla, NY, USA
| | - Wilbert Aronow
- Section of Cardiovascular Medicine, Department of Pediatrics, New York Medical College and Westchester Medical Center, Valhalla, NY, USA
| |
Collapse
|
229
|
Sadek I, Keaton M, Maihle NJ, Tang SC. Anti-HER-2 therapy following severe trastuzumab-induced cardiac toxicity. Genes Dis 2017; 4:159-162. [PMID: 30258918 PMCID: PMC6147109 DOI: 10.1016/j.gendis.2017.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 07/25/2017] [Indexed: 01/05/2023] Open
Abstract
The human epidermal growth factor receptor 2 (HER2) is overexpressed in 25%-30% of breast cancer patients. Anti-HER2 therapies have changed the aggressive course of HER2+ breast cancer. In spite of the therapeutic benefits, their cardiotoxicities are major concerns, especially when used concurrently with anthracyclines. Here we present an elderly patient with relapsed HER2+ breast cancer. Her presentation for relapsed disease was unusual for the physical finding as well as the history of trastuzumab-induced severe cardiotoxicity while requiring additional anti-HER2 therapy. She received neoadjuvant anti-HER2 treatment for stage III breast caner. Due to severe reduction of cardiac ejection fraction (EF), she only received five doses of adjuvant transtuzumab. Unfortunately her disease relapsed one year later with chest wall lesions and a persistent low EF. We treated the patient with lapatinib combined with capecitabine which resulted rapid resolution of her chest wall lesion. More importantly, the patient had one year of disease control without deterioration in her ejection fraction. We discussed the management of recurrent HER2+ breast cancer with chest wall disease and the choice of anti-HER2 therapy in patients with a history of transtuzumab-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Ibrahim Sadek
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Mark Keaton
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Nita J. Maihle
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Shou-Ching Tang
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Tianjin Medical University Cancer Institute and Hospital, Huanhuxi Road, Hexi District, Tianjin 300060, China
| |
Collapse
|
230
|
Does dual HER-2 blockade treatment increase the risk of severe toxicities of special interests in breast cancer patients: A meta-analysis of randomized controlled trials. Oncotarget 2017; 8:19923-19933. [PMID: 28199966 PMCID: PMC5386734 DOI: 10.18632/oncotarget.15252] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/19/2016] [Indexed: 11/25/2022] Open
Abstract
Although dual HER-2 blockade treatment could offer greater clinical efficacy in breast cancer, the risk of severe toxicities of special interest related to this combined regimen in breast cancer remained unknown. We systematically searched public databases (MEDLINE, EMBASE, Cochrane library) to identify relevant studies that comparing anti-HER2 monotherapy (lapatinib or trastuzumab or pertuzumab) with dual HER-2 blockade treatment (pertuzumab plus trastuzumab or trastuzumab plus lapatinib) in breast cancer. A total of 11,941 breast cancer patients from 9 trials were included for analysis. Meta-analysis showed that dual HER2 blockade treatment significantly increased the risk of severe diarrhea (OR 2.52, p<0.001) and treatment discontinuation (OR 1.52, p=0.014), but not for severe rash (OR 1.06, p=0.81), liver toxicities (OR 1.16, p=0.28), CHF (OR 1.46, p=0.09), LVEF decline (OR 1.09, p=0.40) and FAEs (OR 0.97, p=0.91). Similar results were observed in sub-group analysis according to anti-HER2 regimens in terms of severe diarrhea and treatment discontinuation. Additionally, trastuzumab plus lapatinib significantly increased the risk of LVEF decline in comparison with lapatinib alone (OR 1.48, p=0.002). Our analysis indicated that dual anti-HER2 blockade treatment significantly increased the risk of developing severe diarrhea and treatment discontinuation in comparison with anti-HER2 monotherapy. These were no evidence of an increased risk of fatal adverse events with dual-HER2 blockade treatment.
Collapse
|
231
|
Abstract
Breast cancer is one of the most common cancers affecting women worldwide. The controlled release of drugs to the precise site of the disease using a nanocarrier vehicle increases the therapeutic efficiency of the drugs. Nanotechnology-based approaches used to endorse clinical improvement from a disease also help to understand the interaction of malignant cells with their microenvironment. Receptor-based targeting is another approach for drug delivery which is undergoing clinical trials. Nanoparticles (NPs) delivery has been proven to promise high loading capacity, less toxicity, and stability of the drugs or biomolecules compared to traditional chemotherapeutic drugs. The goal of this review is to present the current problems of breast cancer therapy and discuss the NP-based targeting to overcome the hurdles of conventional drug therapy approach.
Collapse
Affiliation(s)
- Santosh Kumar Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Shriti Singh
- Department of Kriya Sharir, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - James W Lillard
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Rajesh Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
232
|
Economic evaluation of sequencing strategies in HER2-positive metastatic breast cancer in Mexico: a contrast between public and private payer perspectives. Breast Cancer Res Treat 2017; 166:951-963. [PMID: 28840424 DOI: 10.1007/s10549-017-4473-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 08/17/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE Breast cancer is the most common malignancy among women in Mexico. A large proportion of Mexican patients present with advanced disease, and 25% have HER2-positive tumors. We performed a cost-effectiveness analysis of different sequencing strategies of HER2-targeted agents in Mexico according to various payer perspectives. METHODS A Markov model was constructed to evaluate the cost-effectiveness of four different HER2-targeted treatment sequences among patients with HER2-positive metastatic breast cancer treated in Mexico according to three public and one private payer perspectives. Patients were followed weekly over their remaining life expectancies within the model. Health states considered were progression-free survival (PFS) 1st-3rd lines, and death. Transition probabilities between states were based on published trials. Cost data were obtained from official publications from Mexican healthcare institutions. The evaluated outcomes were PFS, OS, costs, QALYs, and incremental cost effectiveness ratio (ICER). RESULTS In the public payer perspective, sequences containing pertuzumab or T-DM1 were not cost-effective when compared with a sequence including the combination of trastuzumab/docetaxel as first line without subsequent T-DM1 or pertuzumab, even when utilizing alternate definitions for willingness to pay thresholds. In the private payer perspective, a sequence containing T-DM1 but not pertuzumab proved cost-effective at a lower clinical effectiveness. CONCLUSIONS In Mexico, the use of at least three lines of trastuzumab in combination with other therapies, but not with pertuzumab or TDM-1, represents the most cost-effective option for patients covered by the public healthcare system, and this sequence should be made available for all patients.
Collapse
|
233
|
Choi HD, Chang MJ. Cardiac toxicities of lapatinib in patients with breast cancer and other HER2-positive cancers: a meta-analysis. Breast Cancer Res Treat 2017; 166:927-936. [PMID: 28825152 DOI: 10.1007/s10549-017-4460-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 08/09/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE Lapatinib is a tyrosine kinase inhibitor that targets the human epidermal growth factor receptor 2 (HER2) and the epidermal growth factor receptor (EGFR/HER1), and there are concerns about its cardiac toxicity. Recent studies of lapatinib have reported cardiac adverse events; however, the results have been inconsistent among the studies. The aim of our study was to estimate the cardiac toxicity of lapatinib in patients with breast cancer and other HER2-positive cancers. METHODS To evaluate the cardiotoxicity of lapatinib, the results of previous studies were quantitatively integrated using meta-analysis. Forty-five articles regarding cardiac adverse events, including left ventricular dysfunction, left ventricular ejection fraction (LVEF) decrease, arrhythmia, and other cardiac adverse events, were assessed. As a subgroup analysis in patients with breast cancer, 26 studies of lapatinib-induced cardiac adverse events were assessed. RESULTS The overall incidence of cardiac adverse events was 2.70% (95% confidence interval [CI] 1.60-4.50%). The incidences of left ventricular dysfunction and LVEF decrease were 1.60% (95% CI 1.30-2.00%) and 2.20% (95% CI 1.30-3.60%), respectively. The overall incidence of cardiac adverse events was 3.00% (95% CI 1.50-6.10%) in patients with breast cancer, which was marginally higher than the rate in patients with all type of cancers. CONCLUSION The overall incidence of lapatinib-induced cardiac toxicity was relatively low based on an indirect comparison with trastuzumab. However, careful monitoring of cardiac toxicity is still needed when patients are treated with lapatinib because the related risk factors have not been clearly identified.
Collapse
Affiliation(s)
- Hye Duck Choi
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan, Gyeongsangbuk-do, 38541, Republic of Korea.
| | - Min Jung Chang
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Republic of Korea.,Department of Pharmaceutical Medicine and Regulatory Sciences, Colleges of Medicine and Pharmacy, Yonsei University, Incheon, Republic of Korea
| |
Collapse
|
234
|
Zhao M, Howard EW, Parris AB, Guo Z, Zhao Q, Ma Z, Xing Y, Liu B, Edgerton SM, Thor AD, Yang X. Activation of cancerous inhibitor of PP2A (CIP2A) contributes to lapatinib resistance through induction of CIP2A-Akt feedback loop in ErbB2-positive breast cancer cells. Oncotarget 2017; 8:58847-58864. [PMID: 28938602 PMCID: PMC5601698 DOI: 10.18632/oncotarget.19375] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 07/11/2017] [Indexed: 11/25/2022] Open
Abstract
Lapatinib, a small molecule ErbB2/EGFR inhibitor, is FDA-approved for the treatment of metastatic ErbB2-overexpressing breast cancer; however, lapatinib resistance is an emerging clinical challenge. Understanding the molecular mechanisms of lapatinib-mediated anti-cancer activities and identifying relevant resistance factors are of pivotal significance. Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a recently identified oncoprotein that is overexpressed in breast cancer. Our study investigated the role of CIP2A in the anti-cancer efficacy of lapatinib in ErbB2-overexpressing breast cancer cells. We found that lapatinib concurrently downregulated CIP2A and receptor tyrosine kinase signaling in ErbB2-overexpressing SKBR3 and 78617 cells; however, these effects were attenuated in lapatinib-resistant (LR) cells. CIP2A overexpression rendered SKBR3 and 78617 cells resistant to lapatinib-induced apoptosis and growth inhibition. Conversely, CIP2A knockdown via lentiviral shRNA enhanced cell sensitivity to lapatinib-induced growth inhibition and apoptosis. Results also suggested that lapatinib downregulated CIP2A through regulation of protein stability. We further demonstrated that lapatinib-induced CIP2A downregulation can be recapitulated by LY294002, suggesting that Akt mediates CIP2A upregulation. Importantly, lapatinib induced differential CIP2A downregulation between parental BT474 and BT474/LR cell lines. Moreover, CIP2A shRNA knockdown significantly sensitized the BT474/LR cells to lapatinib. Collectively, our results demonstrate that CIP2A is a molecular target and resistance factor of lapatinib with a critical role in lapatinib-induced cellular responses, including the inhibition of the CIP2A-Akt feedback loop. Further investigation of lapatinib-mediated CIP2A regulation will advance our understanding of lapatinib-associated anti-tumor activities and drug resistance.
Collapse
Affiliation(s)
- Ming Zhao
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA
| | - Erin W Howard
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA
| | - Amanda B Parris
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA
| | - Zhiying Guo
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA
| | - Qingxia Zhao
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA.,Basic Medical College of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Zhikun Ma
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA
| | - Ying Xing
- Basic Medical College of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Bolin Liu
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Susan M Edgerton
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ann D Thor
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Xiaohe Yang
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA.,College of Medicine, Henan University of Sciences and Technology, Luoyang, Henan, P.R. China
| |
Collapse
|
235
|
Martel S, Maurer C, Lambertini M, Pondé N, De Azambuja E. Breast cancer treatment-induced cardiotoxicity. Expert Opin Drug Saf 2017; 16:1021-1038. [PMID: 28697311 DOI: 10.1080/14740338.2017.1351541] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Breast cancer is the most frequent cancer affecting women worldwide. In every setting, the majority of women are treated with an evergrowing arsenal of therapeutic agents that have greatly improved their outcomes. However, these therapies can also be associated with significant adverse events. Areas covered: This review aims to thoroughly describe the current state of the evidence regarding the potential cardiotoxicity of agents commonly used in the treatment of breast cancer. These include chemotherapeutic agents, anti-HER2 therapies and CDK4/6 and mTOR inhibitors. Furthermore, issues related to the risk stratification and monitoring tools are explored. Expert opinion: Anthracycline- and trastuzumab-related cardiac toxicities have been extensively studied. Substantial evidence is now available concerning additional anti-HER2 agents such as pertuzumab, T-DM1 and tyrosine kinase inhibitors; overall, the cardiotoxicity profile is reassuring. Cardiac events due to endocrine therapy are mostly ischemic and, in the context of prolonged therapy, need specific attention. Novel agents implicated in the treatment of hormone receptor-positive disease are potentially arrhythmogenic and the exact risk will need to be further refined. As for today, assessment of baseline risk factors prior to treatment initiation and cardiac imaging before and during treatment remains the optimal way to prevent cardiac dysfunction. Cardioprotective therapy in primary prevention is still a matter of debate.
Collapse
Affiliation(s)
- Samuel Martel
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium.,b Département d'hémato-oncologie , CISSS Montérégie centre/Hôpital Charles Lemoyne, centre affilié de l'Université de Sherbrooke , Greenfield Park , Qc , Canada
| | - Christian Maurer
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium.,c Department I of Internal Medicine and Center of Integrated Oncology Cologne Bonn , University of Cologne , Cologne , Germany
| | - Matteo Lambertini
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium.,d Breast Cancer Translational Research Laboratory, Institut Jules Bordet , Université Libre de Bruxelles (U.L.B) , Brussels , Belgium
| | - Noam Pondé
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium
| | - Evandro De Azambuja
- a Clinique d'Oncologie Médicale, Institut Jules Bordet , Université Libre de Bruxelles (U.LB) , Brussels , Belgium
| |
Collapse
|
236
|
Lee J, Park HY, Kim WW, Lee SJ, Jeong JH, Kang SH, Jung JH, Chae YS. Biological function of long noncoding RNA snaR in HER2-positive breast cancer cells. Tumour Biol 2017; 39:1010428317707374. [PMID: 28653903 DOI: 10.1177/1010428317707374] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Long noncoding RNA, snaR (small NF90-associated RNA), has been reported to be upregulated in various cancer cell lines. We evaluated the additional role of snaR in HER2-positive breast cancer cell lines. METHODS We explored changes of expression of snaR among the selected long noncoding RNAs which have a potential in cancer proliferation or progression. The proliferation, migration, and invasion of HER2-positive breast cancer cells (SK-BR3) were evaluated by snaR with RNA interruption in 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyltetrazolium bromide, wound-healing assay, and Transwell assay. RESULTS The expression of snaR was remarkably upregulated in SK-BR3 cell lines together with ANRIL, while the SFMBT2 was downregulated in SK-BR3 cell lines. Although Nespas, 7SK, PSF inhibiting RNA, mascRNA, Hoxa11as, NRON, AK023948, MER11C, p53 mRNA, CAR Intergenic 10, HUC 1 and 2, ZFAS1, SCA8, and SNHG5 were also upregulated and UCA1 was downregulated, the differences were not dominent. Based on the expression result, we explored the functional role of snaR in HER2-positive breast cancer. Downregulation of snaR with small interfering RNA was identified to significanlty inhibit migration as well as proliferation of SK-BR3 cells. CONCLUSION In this study, snaR was identified as upregulated and to play a role in cancer progression of HER2-positive breast cancer cells. These results suggest snaR as a potential biomarker for HER2-positive breast cancer.
Collapse
Affiliation(s)
- Jeeyeon Lee
- 1 Department of Surgery, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Ho Yong Park
- 1 Department of Surgery, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Wan Wook Kim
- 1 Department of Surgery, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Soo Jung Lee
- 2 Department of Hemato-Oncology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Jae-Hwan Jeong
- 3 Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Seung Hee Kang
- 3 Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Jin Hyang Jung
- 1 Department of Surgery, Kyungpook National University School of Medicine, Daegu, Republic of Korea.,4 Breast Cancer Center, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Yee Soo Chae
- 2 Department of Hemato-Oncology, Kyungpook National University School of Medicine, Daegu, Republic of Korea.,4 Breast Cancer Center, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| |
Collapse
|
237
|
Evolution of anti-HER2 therapies for cancer treatment. Cancer Treat Rev 2017; 59:1-21. [PMID: 28715775 DOI: 10.1016/j.ctrv.2017.06.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 01/13/2023]
Abstract
The development of HER2-directed monoclonal antibodies and tyrosine kinase inhibitors have provided benefits to cancer patients, as well as produced many insights into the biology of the ErbB receptor family. Current therapies based on ErbB family members have resulted in improved overall survival with associated improvements in quality of life for the cancer patients that respond to treatment. Compared to monotherapy using either two antibodies to block the HER2 receptor blockade or combinatorial approaches with HER2 antibodies and standard therapies has provided additional benefits. Despite the therapeutic success of existing HER2 therapies, personalising treatment and overcoming resistance to these therapies remains a significant challenge. The heterogeneous intra-tumoural HER2 expression and lack of fully predictive and prognostic biomarkers remain significant barriers to improving the use of HER2 antibodies. Imaging modalities using radiolabelled pertuzumab and trastuzumab allow quantitative assessment of intra-tumoural HER2 expression, HER2 antibody saturation and the success of different drug delivery systems to be assessed. Molecular imaging with HER2 antibodies has the potential to be a non-invasive, predictive and prognostic technique capable of influencing therapeutic decisions, predicting response and failure of treatments as well as providing insights into receptor recycling and signalling. Similarly, conjugating HER2 antibodies with novel toxic payloads or combining HER2 antibodies with cellular immunotherapy provide exciting new opportunities for the management of tumours overexpressing HER2. Future research will lead to higher therapeutic responses, lower toxicities and providing insight into the mechanisms of resistance to HER2-targeted treatments.
Collapse
|
238
|
Dey N, De P, Leyland-Jones B. PI3K-AKT-mTOR inhibitors in breast cancers: From tumor cell signaling to clinical trials. Pharmacol Ther 2017; 175:91-106. [DOI: 10.1016/j.pharmthera.2017.02.037] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
239
|
Echavarria I, López-Tarruella S, Márquez-Rodas I, Jerez Y, Martin M. Neratinib for the treatment of HER2-positive early stage breast cancer. Expert Rev Anticancer Ther 2017. [PMID: 28649882 DOI: 10.1080/14737140.2017.1338954] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Despite the advances in the treatment of HER2-positive breast cancer, resistance to actual chemotherapeutic regimens eventually occurs. Neratinib, an orally available pan-inhibitor of the ERBB family, represents an interesting new option for early-stage HER2-positive breast cancer. Areas covered: In this article, the development of neratinib, with a special focus on its potential value in the treatment of early-stage HER2-positive breast cancer, has been reviewed. For this purpose, a literature search was conducted, including preclinical studies, early-phase trials in advanced cancer with neratinib in monotherapy and in combination, and phase II and large phase III trials in the early setting. Management of neratinib-induced toxicity, future perspectives for the drug, and ongoing trials are also discussed in this review. Expert commentary: Neratinib is emerging as a promising oral drug for the treatment of HER2-positive breast cancer. Although FDA and EMA approval is derived from the extended adjuvant treatment, this setting may not be the ideal scenario to obtain the beneficial effects of neratinib. Confirmatory data in the neoadjuvant setting and subgroup analysis from the ExTENET trial might bring some light into the best setting for neratinib therapy. Data from confirmatory trials in the metastatic setting are also required.
Collapse
Affiliation(s)
- Isabel Echavarria
- a Medical Oncology Department , Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM) , Madrid , Spain
| | - Sara López-Tarruella
- b Medical Oncology Department , Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CiberOnc , Madrid , Spain
| | - Iván Márquez-Rodas
- b Medical Oncology Department , Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CiberOnc , Madrid , Spain
| | - Yolanda Jerez
- b Medical Oncology Department , Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), CiberOnc , Madrid , Spain
| | - Miguel Martin
- c Instituto de Investigación Sanitaria Gregorio Maranon (IiSGM), Universidad Complutense, CiberOnc, GEICAM , Madrid , Spain
| |
Collapse
|
240
|
Trastuzumab emtansine versus treatment of physician's choice in patients with previously treated HER2-positive metastatic breast cancer (TH3RESA): final overall survival results from a randomised open-label phase 3 trial. Lancet Oncol 2017; 18:743-754. [DOI: 10.1016/s1470-2045(17)30313-3] [Citation(s) in RCA: 381] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/24/2017] [Accepted: 02/27/2017] [Indexed: 11/19/2022]
|
241
|
Nasir A, Holzer TR, Chen M, Man MZ, Schade AE. Differential expression of VEGFR2 protein in HER2 positive primary human breast cancer: potential relevance to anti-angiogenic therapies. Cancer Cell Int 2017; 17:56. [PMID: 28533703 PMCID: PMC5438568 DOI: 10.1186/s12935-017-0427-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/12/2017] [Indexed: 12/28/2022] Open
Abstract
Background Clinically relevant predictive biomarkers to tailor anti-angiogenic therapies to breast cancer (BRC) patient subpopulations are an unmet need. Methods We analyzed tumor vascular density and VEGFR2 protein expression in various subsets of primary human BRCs (186 females; Mean age: 59 years; range 33–88 years), using a tissue microarray. Discrete VEGFR2+ and CD34+ tumor vessels were manually scored in invasive ductal, lobular, mixed ductal-lobular and colloid (N = 139, 22, 18, 7) BRC cores. Results The observed CD34+ and VEGFR2+ tumor vascular counts in individual cases were heterogeneous. Mean CD34+ and VEGFR2+ tumor vessel counts were 11 and 3.4 per tumor TMA core respectively. Eighty-nine of 186 (48%) cases had >10 CD34+ tumor vessels, while 97/186 (52%) had fewer CD34+ vessels in each TMA core. Of 169 analyzable cores in the VEGFR2 stained TMA, 90 (53%) showed 1–5 VEGFR2+ tumor vessels/TMA core, while 42/169 (25%) cores had no detectable VEGFR2+ tumor vessels. Thirteen of 169 (8%) cases also showed tumor cell (cytoplasmic/membrane) expression of VEGFR2. Triple-negative breast cancers (TNBCs) appeared to be less vascular (Mean VD = 9.8, range 0–34) than other breast cancer subtypes. Overall, VEGFR2+ tumor vessel counts were significantly higher in HER2+ as compared to HR+ (p = 0.04) and TNBC (p = 0.02) tissues. Compared to HER2− cases, HER2+ breast cancers had higher VEGFR2+ tumor vessel counts (p = 0.007). Conclusion Characterization of pathologic angiogenesis in HER2+ breast cancer provides scientific rationale for future investigation of clinical activity of agents targeting the VEGF/VEGFR2 axis in this clinically aggressive breast cancer subtype.
Collapse
Affiliation(s)
- Aejaz Nasir
- Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, IN USA.,Eli Lilly and Company, Lilly Corporate Center, DC0424, Indianapolis, IN 46285 USA
| | - Timothy R Holzer
- Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, IN USA
| | - Mia Chen
- Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, IN USA
| | - Michael Z Man
- Oncology Statistics, Eli Lilly and Company, Indianapolis, IN USA
| | - Andrew E Schade
- Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, IN USA
| |
Collapse
|
242
|
Santra T, Roche S, Conlon N, O’Donovan N, Crown J, O’Connor R, Kolch W. Identification of potential new treatment response markers and therapeutic targets using a Gaussian process-based method in lapatinib insensitive breast cancer models. PLoS One 2017; 12:e0177058. [PMID: 28481952 PMCID: PMC5421758 DOI: 10.1371/journal.pone.0177058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 04/23/2017] [Indexed: 12/15/2022] Open
Abstract
Molecularly targeted therapeutics hold promise of revolutionizing treatments of advanced malignancies. However, a large number of patients do not respond to these treatments. Here, we take a systems biology approach to understand the molecular mechanisms that prevent breast cancer (BC) cells from responding to lapatinib, a dual kinase inhibitor that targets human epidermal growth factor receptor 2 (HER2) and epidermal growth factor receptor (EGFR). To this end, we analysed temporal gene expression profiles of four BC cell lines, two of which respond and the remaining two do not respond to lapatinib. For this analysis, we developed a Gaussian process based algorithm which can accurately find differentially expressed genes by analysing time course gene expression profiles at a fraction of the computational cost of other state-of-the-art algorithms. Our analysis identified 519 potential genes which are characteristic of lapatinib non-responsiveness in the tested cell lines. Data from the Genomics of Drug Sensitivity in Cancer (GDSC) database suggested that the basal expressions 120 of the above genes correlate with the response of BC cells to HER2 and/or EGFR targeted therapies. We selected 27 genes from the larger panel of 519 genes for experimental verification and 16 of these were successfully validated. Further bioinformatics analysis identified vitamin D receptor (VDR) as a potential target of interest for lapatinib non-responsive BC cells. Experimentally, calcitriol, a commonly used reagent for VDR targeted therapy, in combination with lapatinib additively inhibited proliferation in two HER2 positive cell lines, lapatinib insensitive MDA-MB-453 and lapatinib resistant HCC 1954-L cells.
Collapse
Affiliation(s)
- Tapesh Santra
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
- * E-mail:
| | - Sandra Roche
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Neil Conlon
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Norma O’Donovan
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - John Crown
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
- Department of Medical Oncology, St Vincent’s University Hospital, Dublin, Elm Park, Ireland
| | - Robert O’Connor
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Walter Kolch
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
243
|
Jain D, Russell RR, Schwartz RG, Panjrath GS, Aronow W. Cardiac Complications of Cancer Therapy: Pathophysiology, Identification, Prevention, Treatment, and Future Directions. Curr Cardiol Rep 2017; 19:36. [PMID: 28374177 DOI: 10.1007/s11886-017-0846-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW Cardiotoxicity is an important complication of cancer therapy. With a significant improvement in the overall survival and prognosis of patients undergoing cancer therapy, cardiovascular toxicity of cancer therapy has become an important public health issue. Several well-established as well as newer anticancer therapies such as anthracyclines, trastuzumab, and other HER2 receptor blockers, antimetabolites, alkylating agents, tyrosine kinase inhibitors, angiogenesis inhibitors, checkpoint inhibitors, and thoracic irradiation are associated with significant cardiotoxicity. RECENT FINDINGS Cardiovascular imaging employing radionuclide imaging, echocardiography, and magnetic resonance imaging is helpful in early detection of the cardiotoxicity and prevention of overt heart failure. These techniques also provide important tools for understanding the mechanism of cardiotoxicity of these modalities, which would help develop strategies for the prevention of cardiac morbidity and mortality related to the use of these agents. An understanding of the mechanism of the cardiotoxicity of cancer therapies can help prevent and treat their adverse cardiovascular consequences. Clinical implementation of algorithms based upon cardiac imaging and several non-imaging biomarkers can prevent cardiac morbidity and mortality associated with the use of cardiotoxic cancer therapies.
Collapse
Affiliation(s)
- Diwakar Jain
- Section of Cardiovascular Medicine, New York Medical College and Westchester Medical Center, 100 Woods Road, Valhalla, NY, USA.
| | - Raymond R Russell
- Rhode Island Cardiovascular Institute, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Ronald G Schwartz
- Cardiology Division, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Nuclear Medicine Division, Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, USA
| | - Gurusher S Panjrath
- Heart and Vascular Institute, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Wilbert Aronow
- Section of Cardiovascular Medicine, New York Medical College and Westchester Medical Center, 100 Woods Road, Valhalla, NY, USA
| |
Collapse
|
244
|
Tyrosine kinase-targeting drugs-associated heart failure. Br J Cancer 2017; 116:1366-1373. [PMID: 28399109 PMCID: PMC5482733 DOI: 10.1038/bjc.2017.88] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 01/17/2017] [Accepted: 02/23/2017] [Indexed: 02/02/2023] Open
Abstract
Background: The impact of cancer therapies on cardiac disease in the general adult cancer survivor population is largely unknown. Our objective was to evaluate which tyrosine kinase-targeting drugs are associated with greater risk for new-onset heart failure (HF). Methods: A nested case–control analysis was conducted within a cohort of 27 992 patients of Clalit Health Services, newly treated with a tyrosine kinase-targeting, and/or chemotherapeutic drug, for a malignant disease, between 1 January 2005 and 31 December 2012. Each new case of HF was matched to up to 30 controls from the cohort on calendar year of cohort entry, age, gender, and duration of follow-up. Main outcome measure was odds ratio (OR) with 95% confidence interval (CI) of new-onset HF. Results: There were 936 incident cases of HF during 71 742 person-years of follow-up. Trastuzumab (OR 1.90, 95% CI 1.46–2.49), cetuximab (OR 1.72, 1.10–2.69), panitumumab (OR 3.01, 1.02–8.85), and sunitinib (OR 3.39, 1.78–6.47) were associated with increased HF risk. Comorbidity independently associated with higher risk in a multivariable conditional regression model was diabetes mellitus, hypertension, chronic renal failure, ischaemic heart disease, valvular heart disease, arrhythmia, and smoking. Conclusions: Trastuzumab, cetuximab, panitumumab, and sunitinib are associated with increased risk for new-onset HF.
Collapse
|
245
|
Jackisch C, Lammers P, Jacobs I. Evolving landscape of human epidermal growth factor receptor 2-positive breast cancer treatment and the future of biosimilars. Breast 2017; 32:199-216. [PMID: 28236776 PMCID: PMC10187060 DOI: 10.1016/j.breast.2017.01.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/17/2017] [Accepted: 01/19/2017] [Indexed: 01/06/2023] Open
Abstract
Human epidermal growth factor receptor 2-positive (HER2+) breast cancer comprises approximately 15%-20% of all breast cancers and is associated with a poor prognosis. The introduction of anti-HER2 therapy has significantly improved clinical outcomes for patients with HER2+ breast cancer, and multiple HER2-directed agents (ie, trastuzumab, pertuzumab, lapatinib, and ado-trastuzumab emtansine [T-DM1]) are approved for clinical use in various settings. The treatment landscape for patients with HER2+ breast cancer is continuing to evolve. While novel agents and therapeutic strategies are emerging, biologic therapies, particularly trastuzumab, are likely to remain a mainstay of treatment. However, access issues create barriers to the use of biologics, and there is evidence for underuse of trastuzumab worldwide. A biosimilar is a biologic product that is highly similar to a licensed biologic in terms of product safety and effectiveness. Biosimilars of trastuzumab are in development and may soon become available. The introduction of biosimilars may improve access to anti-HER2 therapies by providing additional treatment options and lower-cost alternatives. Because HER2-targeted drugs may be administered for extended periods of time and in combination with other systemic therapies, biosimilars have the potential to result in significant savings for healthcare systems. Herein we review current and emerging treatment options for, and discuss the possible role of biosimilars in, treating patients with HER2+ breast cancer.
Collapse
Affiliation(s)
- Christian Jackisch
- Sana Klinikum Offenbach, Starkenburgring 66, D-63069 Offenbach, Germany.
| | - Philip Lammers
- Meharry Medical College, 1005 Dr. D.B. Todd Jr. Blvd., Nashville, TN 37208-3501, USA.
| | - Ira Jacobs
- Pfizer Inc., 235 East 42nd Street, New York, NY 10017-5755, USA.
| |
Collapse
|
246
|
O’Sullivan CC, Davarpanah NN, Abraham J, Bates SE. Current challenges in the management of breast cancer brain metastases. Semin Oncol 2017; 44:85-100. [DOI: 10.1053/j.seminoncol.2017.06.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 06/29/2017] [Indexed: 02/06/2023]
|
247
|
Villanueva C, Malvestiti J, Chaigneau L, Cals L, Pivot X. Nouvelles approches thérapeutiques dans le cancer du sein HER2 positif. ONCOLOGIE 2017. [DOI: 10.1007/s10269-015-2534-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
248
|
Chang YW, Singh KP. Long-term exposure to estrogen enhances chemotherapeutic efficacy potentially through epigenetic mechanism in human breast cancer cells. PLoS One 2017; 12:e0174227. [PMID: 28323900 PMCID: PMC5360320 DOI: 10.1371/journal.pone.0174227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/05/2017] [Indexed: 01/10/2023] Open
Abstract
Chemotherapy is the most common clinical option for treatment of breast cancer. However, the efficacy of chemotherapy depends on the age of breast cancer patients. Breast tissues are estrogen responsive and the levels of ovarian estrogen vary among the breast cancer patients primarily between pre- and post-menopausal age. Whether this age-dependent variation in estrogen levels influences the chemotherapeutic efficacy in breast cancer patients is not known. Therefore, the objective of this study was to evaluate the effects of natural estrogen 17 beta-estradiol (E2) on the efficacy of chemotherapeutic drugs in breast cancer cells. Estrogen responsive MCF-7 and T47D breast cancer cells were long-term exposed to 100 pg/ml estrogen, and using these cells the efficacy of chemotherapeutic drugs doxorubicin and cisplatin were determined. The result of cell viability and cell cycle analysis revealed increased sensitivities of doxorubicin and cisplatin in estrogen-exposed MCF-7 and T47D cells as compared to their respective control cells. Gene expression analysis of cell cycle, anti-apoptosis, DNA repair, and drug transporter genes further confirmed the increased efficacy of chemotherapeutic drugs in estrogen-exposed cells at molecular level. To further understand the role of epigenetic mechanism in enhanced chemotherapeutic efficacy by estrogen, cells were pre-treated with epigenetic drugs, 5-aza-2-deoxycytidine and Trichostatin A prior to doxorubicin and cisplatin treatments. The 5-aza-2 deoxycytidine pre-treatment significantly decreased the estrogen-induced efficacy of doxorubicin and cisplatin, suggesting the role of estrogen-induced hypermethylation in enhanced sensitivity of these drugs in estrogen-exposed cells. In summary, the results of this study revealed that sensitivity to chemotherapy depends on the levels of estrogen in breast cancer cells. Findings of this study will have clinical implications in selecting the chemotherapy strategies for treatment of breast cancer patients depending on the serum estrogen levels that varies among pre- and post-menopausal age of the patients.
Collapse
Affiliation(s)
- Yu-Wei Chang
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, Texas, United States of America
| | - Kamaleshwar P. Singh
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, Texas, United States of America
- * E-mail:
| |
Collapse
|
249
|
Xu ZQ, Zhang Y, Li N, Liu PJ, Gao L, Gao X, Tie XJ. Efficacy and safety of lapatinib and trastuzumab for HER2-positive breast cancer: a systematic review and meta-analysis of randomised controlled trials. BMJ Open 2017; 7:e013053. [PMID: 28289045 PMCID: PMC5353336 DOI: 10.1136/bmjopen-2016-013053] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES The anti-HER2 monoclonal antibody trastuzumab and the tyrosine kinase inhibitor lapatinib have complementary mechanisms of action and synergistic antitumour actively in models of HER2-positive breast cancer. However, the efficacy of trastuzumab in combination with lapatinib remains controversial. Therefore, we conducted this meta-analysis to compare combination treatment with lapatinib and trastuzumab to trastuzumab or lapatinib alone in the treatment of HER2-positive breast cancer. METHODS Randomised controlled trials (RCTs), published in PubMed, Embase and Web of Science, were systematically reviewed to assess the survival benefits and toxicity profile of HER2-positive patients with breast cancer who were treated with lapatinib and trastuzumab. Outcomes included pathological complete response (pCR), event-free survival (EFS), overall survival (OS) and toxicities. Results were expressed as the risk ratio (RR) or HR with 95% CIs. Pooled estimates were calculated by using a fixed-effects model or a randomised-effects model. RESULTS A total of 7 RCTs involving 2084 patients met the inclusion criteria and were included in this meta-analysis. The combination of lapatinib and trastuzumab significantly improved pCR (RR=1.43, 95% CI 1.23 to 1.67; p<0.001), EFS (HR=0.75, 95% CI 0.60 to 0.93; p=0.009) and OS (HR=0.72, 95% CI 0.56 to 0.93; p=0.011) in the treatment of HER2-positive breast cancer compared with trastuzumab or lapatinib alone. The combination treatment also increased the pCR irrespective of hormone receptor status and tumour size. More frequent grade 3 or 4 adverse events, including diarrhoea, rash or erythema, neutropenia and hepatic adverse events, were found in the combination group than in the trastuzumab or lapatinib group. CONCLUSIONS On the basis of the current evidence, our results reveal that the addition of lapatinib to trastuzumab can significantly improve pCR, EFS and OS with a tolerated toxicity in patients with HER2-positive breast cancer. Further well-conducted, large-scale trials are needed to validate these findings.
Collapse
Affiliation(s)
- Zhi-Qiao Xu
- Tumor Center, Kaifeng Central Hospital, Kaifeng, China
| | - Yan Zhang
- Tumor Center, Kaifeng Central Hospital, Kaifeng, China
| | - Ning Li
- Tumor Center, Kaifeng Central Hospital, Kaifeng, China
| | - Pei-Jie Liu
- Tumor Center, Kaifeng Central Hospital, Kaifeng, China
| | - Ling Gao
- Tumor Center, Kaifeng Central Hospital, Kaifeng, China
| | - Xin Gao
- Tumor Center, Kaifeng Central Hospital, Kaifeng, China
| | - Xiao-Jing Tie
- Tumor Center, Kaifeng Central Hospital, Kaifeng, China
| |
Collapse
|
250
|
Surrogacy of progression free survival for overall survival in metastatic breast cancer studies: Meta-analyses of published studies. Contemp Clin Trials 2017; 53:20-28. [DOI: 10.1016/j.cct.2016.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 11/30/2016] [Accepted: 12/03/2016] [Indexed: 11/19/2022]
|