1
|
Pang Y, Chen Y, Lin M, Zhang Y, Zhang J, Wang L. MMSyn: A New Multimodal Deep Learning Framework for Enhanced Prediction of Synergistic Drug Combinations. J Chem Inf Model 2024; 64:3689-3705. [PMID: 38676916 DOI: 10.1021/acs.jcim.4c00165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
Combination therapy is a promising strategy for the successful treatment of cancer. The large number of possible combinations, however, mean that it is laborious and expensive to screen for synergistic drug combinations in vitro. Nevertheless, because of the availability of high-throughput screening data and advances in computational techniques, deep learning (DL) can be a useful tool for the prediction of synergistic drug combinations. In this study, we proposed a multimodal DL framework, MMSyn, for the prediction of synergistic drug combinations. First, features embedded in the drug molecules were extracted: structure, fingerprint, and string encoding. Then, gene expression data, DNA copy number, and pathway activity were used to describe cancer cell lines. Finally, these processed features were integrated using an attention mechanism and an interaction module and then input into a multilayer perceptron to predict drug synergy. Experimental results showed that our method outperformed five state-of-the-art DL methods and three traditional machine learning models for drug combination prediction. We verified that MMSyn achieved superior performance in stratified cross-validation settings using both the drug combination and cell line data. Moreover, we performed a set of ablation experiments to illustrate the effectiveness of each component and the efficacy of our model. In addition, our visual representation and case studies further confirmed the effectiveness of our model. All results showed that MMSyn can be used as a powerful tool for the prediction of synergistic drug combinations.
Collapse
Affiliation(s)
- Yu Pang
- Joint International Research Laboratory of Synthetic Biology and Medicine, Ministry of Education, Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yihao Chen
- Joint International Research Laboratory of Synthetic Biology and Medicine, Ministry of Education, Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Mujie Lin
- Joint International Research Laboratory of Synthetic Biology and Medicine, Ministry of Education, Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yanhong Zhang
- Joint International Research Laboratory of Synthetic Biology and Medicine, Ministry of Education, Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jiquan Zhang
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, College of Pharmacy, Guizhou Medical University, Guiyang 550025, P. R. China
| | - Ling Wang
- Joint International Research Laboratory of Synthetic Biology and Medicine, Ministry of Education, Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
2
|
Dragnev KH, Lubet RA, Miller MS, Sei S, Fox JT, You M. Primary Prevention and Interception Studies in RAS-Mutated Tumor Models Employing Small Molecules or Vaccines. Cancer Prev Res (Phila) 2023; 16:549-560. [PMID: 37468135 DOI: 10.1158/1940-6207.capr-23-0027] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/24/2023] [Accepted: 07/17/2023] [Indexed: 07/21/2023]
Abstract
Therapeutic targeting of RAS-mutated cancers is difficult, whereas prevention or interception (treatment before or in the presence of preinvasive lesions) preclinically has proven easier. In the A/J mouse lung model, where different carcinogens induce tumors with different KRAS mutations, glucocorticoids and retinoid X receptor (RXR) agonists are effective agents in prevention and interception studies, irrespective of specific KRAS mutations. In rat azoxymethane-induced colon tumors (45% KRAS mutations), cyclooxygenase 1/2 inhibitors and difluoromethylornithine are effective in preventing or intercepting KRAS-mutated or wild-type tumors. In two KRAS-mutant pancreatic models multiple COX 1/2 inhibitors are effective. Furthermore, combining a COX and an EGFR inhibitor prevented the development of virtually all pancreatic tumors in transgenic mice. In the N-nitroso-N-methylurea-induced estrogen receptor-positive rat breast model (50% HRAS mutations) various selective estrogen receptor modulators, aromatase inhibitors, EGFR inhibitors, and RXR agonists are profoundly effective in prevention and interception of tumors with wild-type or mutant HRAS, while the farnesyltransferase inhibitor tipifarnib preferentially inhibits HRAS-mutant breast tumors. Thus, many agents not known to specifically inhibit the RAS pathway, are effective in an organ specific manner in preventing or intercepting RAS-mutated tumors. Finally, we discuss an alternative prevention and interception approach, employing vaccines to target KRAS.
Collapse
Affiliation(s)
| | - Ronald A Lubet
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Mark Steven Miller
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Shizuko Sei
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Jennifer T Fox
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Ming You
- Houston Methodist Hospital, Houston, Texas
| |
Collapse
|
3
|
Chen JF, Wu SW, Shi ZM, Hu B. Traditional Chinese medicine for colorectal cancer treatment: potential targets and mechanisms of action. Chin Med 2023; 18:14. [PMID: 36782251 PMCID: PMC9923939 DOI: 10.1186/s13020-023-00719-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
Colorectal cancer (CRC) is a disease with complex pathogenesis, it is prone to metastasis, and its development involves abnormalities in multiple signaling pathways. Surgery, chemotherapy, radiotherapy, target therapy, and immunotherapy remain the main treatments for CRC, but improvement in the overall survival rate and quality of life is urgently needed. Traditional Chinese medicine (TCM) has a long history of preventing and treating CRC. It could affect CRC cell proliferation, apoptosis, cell cycle, migration, invasion, autophagy, epithelial-mesenchymal transition, angiogenesis, and chemoresistance by regulating multiple signaling pathways, such as PI3K/Akt, NF-κB, MAPK, Wnt/β-catenin, epidermal growth factor receptors, p53, TGF-β, mTOR, Hedgehog, and immunomodulatory signaling pathways. In this paper, the main signaling pathways and potential targets of TCM and its active ingredients in the treatment of CRC were systematically summarized, providing a theoretical basis for treating CRC with TCM and new ideas for further exploring the pathogenesis of CRC and developing new anti-CRC drugs.
Collapse
Affiliation(s)
- Jin-Fang Chen
- grid.412540.60000 0001 2372 7462Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032 Shanghai, People’s Republic of China ,grid.412540.60000 0001 2372 7462Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032 Shanghai, People’s Republic of China
| | - Shi-Wei Wu
- grid.412540.60000 0001 2372 7462Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032 Shanghai, People’s Republic of China ,grid.412540.60000 0001 2372 7462Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032 Shanghai, People’s Republic of China
| | - Zi-Man Shi
- grid.412540.60000 0001 2372 7462Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032 Shanghai, People’s Republic of China ,grid.412540.60000 0001 2372 7462Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032 Shanghai, People’s Republic of China
| | - Bing Hu
- Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, People's Republic of China. .,Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, People's Republic of China.
| |
Collapse
|
4
|
Xu QT, Wang ZW, Cai MY, Wei JF, Ding Q. A novel cuproptosis-related prognostic 2-lncRNAs signature in breast cancer. Front Pharmacol 2023; 13:1115608. [PMID: 36699089 PMCID: PMC9868634 DOI: 10.3389/fphar.2022.1115608] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
Background: Cuproptosis, a newly defined regulated form of cell death, is mediated by the accumulation of copper ions in cells and related to protein lipoacylation. Seven genes have been reported as key genes of cuproptosis phenotype. Cuproptosis may be developed by subsequent research as a target to treat cancer, such as breast cancer. Long-noncoding RNA (lncRNA) has been proved to play a vital role in regulating the biological process of breast cancer. However, the role of lncRNAs in cuproptosis is poorly studied. Methods: Based on TCGA (The Cancer Genome Atlas) database and integrated several R packages, we screened out 153 cuproptosis-related lncRNAs and constructed a novel cuproptosis-related prognostic 2-lncRNAs signature (BCCuS) in breast cancer and then verified. By using pRRophetic package and machine learning, 72 anticancer drugs, significantly related to the model, were screened out. qPCR was used to detect the differentially expression of two model lncRNAs and seven cuproptosis genes between 10 pairs of breast cancer tissue samples and adjacent samples. Results: We constructed a novel cuproptosis-related prognostic 2-lncRNAs (USP2-AS1, NIFK-AS1) signature (BCCuS) in breast cancer. Univariate COX analysis (p < .001) and multivariate COX analysis (p < .001) validated that BCCuS was an independent prognostic factor for breast cancer. Overall survival Kaplan Meier-plotter, ROC curve and Risk Plot validated the prognostic value of BCCuS both in test set and verification set. Nomogram and C-index proved that BCCuS has strong correlation with clinical decision-making. BCCuS still maintain inspection efficiency when patients were splitting into Stage I-II (p = .024) and Stage III-IV (p = .003) breast cancer. BCCuS-high group and BCCuS-low group showed significant differences in gene mutation frequency, immune function, TIDE (tumor immune dysfunction and exclusion) score and other phenotypes. TMB (tumor mutation burden)-high along with BCCuS-high group had the lowest Survival probability (p = .005). 36 anticancer drugs whose sensitivity (IC50) was significantly related to the model were screened out using pRRophetic package. qPCR results showed that two model lncRNAs (USP2-AS1, NIFK-AS1) and three Cuproptosis genes (FDX1, PDHA1, DLAT) expressed differently between 10 pairs of breast cancer tissue samples and adjacent samples. Conclusion: The current study reveals that cuproptosis-related prognostic 2-lncRNAs signature (BCCuS) may be useful in predicting the prognosis, biological characteristics, and appropriate treatment of breast cancer patients.
Collapse
Affiliation(s)
- Qi-Tong Xu
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Zi-Wen Wang
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Meng-Yuan Cai
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Ji-Fu Wei
- Department of Pharmacy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China,*Correspondence: Ji-Fu Wei, ; Qiang Ding,
| | - Qiang Ding
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China,*Correspondence: Ji-Fu Wei, ; Qiang Ding,
| |
Collapse
|
5
|
Liu J, Wang Q, Kang Y, Xu S, Pang D. Unconventional protein post-translational modifications: the helmsmen in breast cancer. Cell Biosci 2022; 12:22. [PMID: 35216622 PMCID: PMC8881842 DOI: 10.1186/s13578-022-00756-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/07/2022] [Indexed: 01/10/2023] Open
Abstract
AbstractBreast cancer is the most prevalent malignant tumor and a leading cause of mortality among females worldwide. The tumorigenesis and progression of breast cancer involve complex pathophysiological processes, which may be mediated by post-translational modifications (PTMs) of proteins, stimulated by various genes and signaling pathways. Studies into PTMs have long been dominated by the investigation of protein phosphorylation and histone epigenetic modifications. However, with great advances in proteomic techniques, several other PTMs, such as acetylation, glycosylation, sumoylation, methylation, ubiquitination, citrullination, and palmitoylation have been confirmed in breast cancer. Nevertheless, the mechanisms, effects, and inhibitors of these unconventional PTMs (particularly, the non-histone modifications other than phosphorylation) received comparatively little attention. Therefore, in this review, we illustrate the functions of these PTMs and highlight their impact on the oncogenesis and progression of breast cancer. Identification of novel potential therapeutic drugs targeting PTMs and development of biological markers for the detection of breast cancer would be significantly valuable for the efficient selection of therapeutic regimens and prediction of disease prognosis in patients with breast cancer.
Collapse
|
6
|
de Araújo RA, da Luz FAC, da Costa Marinho E, Nascimento CP, de Andrade Marques L, Delfino PFR, Antonioli RM, Araújo BJ, da Silva ACAL, Dos Reis Monteiro MLG, Neto MB, Silva MJB. Epidermal growth factor receptor (EGFR) expression in the serum of patients with triple-negative breast carcinoma: prognostic value of this biomarker. Ecancermedicalscience 2022; 16:1431. [PMID: 36158981 PMCID: PMC9458269 DOI: 10.3332/ecancer.2022.1431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Indexed: 11/15/2022] Open
Abstract
Background Epidermal growth factor receptor (EGFR) overexpression has been considered a poor prognostic factor in breast cancer. Methodology A prospective study of 206 women with breast cancer analysed by stages (I, II, III and IV) and by immunohistochemical subtype (Luminal A, Luminal B, HER2+ and triple-negative (TN)); 89 healthy controls with normal recent mammography were included. The EGFR measured in the serum (sEGFR) was detected by the Enzyme-Linked Immunosorbent Assay (ELISA) method (R&D Systems kit DY231) collected by blood before any treatment in patients. Kaplan-Meier method and Cox regression were carried out to obtain the prognostic value, considering significance if p < 0.05. Results With a median follow-up of 36.6 months, 47 deaths occurred. Multivariable Cox regression showed difference of overall survival (OS) associated with sEGFR levels (sEGFR ≤ or > 47.8 ng/mL) in patients with TN cancers, but not of Luminal A, Luminal B or HER2+ subtypes; adjusted by stage, the death risk increased by approximately 415% [hazard ratio (HR): 5.149 (1.900-13.955), p = 0.001] for patients with sEGFR > 47.8 ng/mL compared to patients with a lower sEGFR value. There was no significant correlation of sEGFR with staging, histological tumour grade (G1/G2/G3), Ki67 (< or ≥14%) or body mass index. Conclusions Increased sEGFR expression in patients with TN tumours is a significant predictor of lower OS and its quantification is inexpensive and straightforward.
Collapse
Affiliation(s)
- Rogério Agenor de Araújo
- Federal University of Uberlândia, Avenida Pará, Bloco 2U, 1720, Campus Umuarama, Uberlândia, MG, CEP 38400-902, Brazil
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0003-4653-6786
| | - Felipe Andrés Cordero da Luz
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-9381-4913
| | - Eduarda da Costa Marinho
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-1307-9104
| | - Camila Piqui Nascimento
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-0955-8559
| | - Lara de Andrade Marques
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-2734-8352
| | - Patrícia Ferreira Ribeiro Delfino
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-2196-9318
| | - Rafael Mathias Antonioli
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0003-3886-1562
| | - Breno Jeha Araújo
- São Paulo State Cancer Institute of the Medical School of the University of São Paulo, São Paulo, SP, CEP 38405-302, Brazil
- https://orcid.org/0000-0003-4892-9911
| | - Ana Cristina Araújo Lemos da Silva
- Federal University of Uberlândia, Avenida Pará, Bloco 2U, 1720, Campus Umuarama, Uberlândia, MG, CEP 38400-902, Brazil
- https://orcid.org/0000-0002-8220-938X
| | | | - Morun Bernardino Neto
- Department of Basic and Environmental Sciences, University of São Paulo, Lorena, SP, CEP 12602-810, Brazil
- https://orcid.org/0000-0003-4292-7800
| | - Marcelo José Barbosa Silva
- Laboratory of Tumor Biomarkers and Osteoimmunology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, MG, CEP 38405-320, Brazil
- https://orcid.org/0000-0002-5807-4286
| |
Collapse
|
7
|
Iancu G, Serban D, Badiu CD, Tanasescu C, Tudosie MS, Tudor C, Costea DO, Zgura A, Iancu R, Vasile D. Tyrosine kinase inhibitors in breast cancer (Review). Exp Ther Med 2022; 23:114. [PMID: 34970337 PMCID: PMC8713180 DOI: 10.3892/etm.2021.11037] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/20/2021] [Indexed: 12/23/2022] Open
Abstract
Anti-epidermal growth factor receptor (EGFR)-targeted therapy has been intensely researched in the last years, motivated by the favorable results obtained with monoclonal antibodies in HER2-enriched breast cancer (BC) patients. Most researched alternatives of anti-EGFR agents were tyrosine kinase inhibitors (TKIs) and monoclonal antibodies. However, excluding monoclonal antibodies trastuzumab and pertuzumab, the remaining anti-EGFR molecules have exhibited disappointing results, due to the lack of specificity and frequent adverse side effects. TKIs have several advantages, including reduced cardiotoxicity, oral administration and favorable penetration of blood-brain barrier for brain metastatic BC. Lapatinib and neratinib and recently pyrotinib (approved only in China) are the only TKIs from dozens of molecules researched over the years that were approved to be used in clinical practice with limited indications, in a subset of BC patients, single or in combination with other chemotherapy or hormonal therapeutic agents. Improved identification of BC subtypes and improved characterization of aggressive forms (triple negative BC or inflammatory BC) should lead to advancements in shaping of targeted agents to improve the outcome of patients.
Collapse
Affiliation(s)
- George Iancu
- Department of Obstetrics and Gynecology, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Gynecology, ‘Filantropia’ Clinical Hospital, 011132 Bucharest, Romania
| | - Dragos Serban
- Department of General Surgery, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Fourth Department of General Surgery, Emergency University Hospital, 050098 Bucharest, Romania
| | - Cristinel Dumitru Badiu
- Department of General Surgery, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of General Surgery, ‘Prof. Dr. Bagdasar Arseni’ Emergency Clinical Hospital, 041915 Bucharest, Romania
| | - Ciprian Tanasescu
- Third Clinico-Surgical Department, Faculty of Medicine, ‘Lucian Blaga’ University, 550169 Sibiu, Romania
| | - Mihai Silviu Tudosie
- Department of Orthopedia and Intensive care, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
- ICU II Toxicology, Clinical Emergency Hospital, 014461 Bucharest, Romania
| | - Corneliu Tudor
- Department of General Surgery, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Daniel Ovidiu Costea
- Department of General Surgery, Faculty of Medicine, ‘Ovidius’ University, 900470 Constanta, Romania
- First Surgery Department, Emergency County Hospital, 900591 Constanta, Romania
| | - Anca Zgura
- Department of Oncology Radiotherapy, Institute of Oncology ‘Prof. Dr. Trestioreanu’, 022328 Bucharest, Romania
| | - Raluca Iancu
- Department of ENT-Opthalmology, Faculty of Medicine, Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Ophthalmology, Emergency University Hospital, 050098 Bucharest, Romania
| | - Danut Vasile
- Department of General Surgery, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
- First Department of General Surgery, Emergency University Hospital, 050098 Bucharest, Romania
| |
Collapse
|
8
|
Ye J, Tian T, Chen X. The efficacy of gefitinib supplementation for breast cancer: A meta-analysis of randomized controlled studies. Medicine (Baltimore) 2020; 99:e22613. [PMID: 33120749 PMCID: PMC7581042 DOI: 10.1097/md.0000000000022613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION The efficacy of gefitinib supplementation for breast cancer remains controversial. We conduct a systematic review and meta-analysis to explore the influence of gefitinib supplementation vs placebo on the efficacy of breast cancer. METHODS We have searched PubMed, EMbase, Web of science, EBSCO, and Cochrane library databases through February 2019 and included randomized controlled trials assessing the effect of gefitinib supplementation vs placebo on overall response for breast cancer patients. This meta-analysis was performed using the random-effect model. RESULTS Seven randomized controlled trials involving 927 patients were included in the meta-analysis. Overall, compared with control group for breast cancer, gefitinib supplementation revealed no obvious impact on complete response (risk ration [RR] = 1.19; 95% confidence interval [CI] = 0.58 to 2.44; P = .63), progressive disease (RR = 0.81; 95% CI = 0.59-1.11; P = .18), partial response (RR = 0.67; 95% CI = 0.36-1.25; P = .21), stable disease (RR = 1.02; 95% CI = 0.65-1.60; P = .92), nausea or vomiting (RR = 0.99; 95% CI = 0.73-1.33; P = .93), but was associated with increased incidence of diarrhea (RR = 2.80; 95% CI = 2.23-3.52; P < .00001), decreased incidence of hot flash (RR = 0.53; 95% CI = 0.37-0.78; P = .001), and improved incidence of adverse events (RR = 1.12; 95% CI = 1.05-1.19; P = .0006). CONCLUSIONS Gefitinib supplementation may provide no positive effect on complete response, progressive disease, partial response or stable disease for breast cancer patients, but with the increase in adverse events.
Collapse
Affiliation(s)
- Jing Ye
- Department of Surgery, Chongqing Yongchuan Health Center for Women and Children
| | - Tian Tian
- Department of General Surgery, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaopin Chen
- Department of General Surgery, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Lubet RA, Heckman-Stoddard BM, Fox JT, Moeinpour F, Juliana MM, Shoemaker RH, Grubbs CJ. Use of Biomarker Modulation in Normal Mammary Epithelium as a Correlate for Efficacy of Chemopreventive Agents Against Chemically Induced Cancers. Cancer Prev Res (Phila) 2020; 13:283-290. [PMID: 31871222 PMCID: PMC7060128 DOI: 10.1158/1940-6207.capr-19-0318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/06/2019] [Accepted: 12/18/2019] [Indexed: 11/16/2022]
Abstract
In both estrogen receptor/progesterone receptor-positive (ER+/PR+) human breast cancer and in ER+/PR+ cancers in the methylnitrosourea (MNU)-induced rat model, short-term modulation of proliferation in early cancers predicts preventive/therapeutic efficacy. We determined the effects of known effective/ineffective chemopreventive agents on proliferative index (PI) in both rat mammary epithelium and small cancers. Female Sprague-Dawley rats were treated with MNU at 50 days of age. Five days later, the rats were treated with the individual compounds for a period of 14 days. At that time, normal mammary tissue from the inguinal gland area was surgically removed. After removal, the rats remained on the agents for an additional 5 months. This cancer prevention study confirmed our prior results of striking efficacy with tamoxifen, vorozole, Targretin, and gefitinib, and no efficacy with metformin, naproxen, and Lipitor. Employing a separate group of rats, the effects of short-term (7 days) drug exposure on small palpable cancers were examined. The PI in both small mammary cancers and in normal epithelium from control rats was >12%. In agreement with the cancer multiplicity data, tamoxifen, vorozole, gefitinib, and Targretin all strongly inhibited proliferation (>65%; P < 0.025) in the normal mammary epithelium. The ineffective agents metformin, naproxen, and Lipitor minimally affected PI. In the small cancers, tamoxifen, vorozole, and Targretin all reduced the PI, while metformin and Lipitor failed to do so. Thus, short-term changes in the PI in either normal mammary epithelium or small cancers correlated with long-term preventive efficacy in the MNU-induced rat model.
Collapse
Affiliation(s)
- Ronald A Lubet
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, NCI, Rockville, Maryland.
| | - Brandy M Heckman-Stoddard
- Breast and Gynecologic Cancer Research Group, Division of Cancer Prevention, NCI, Rockville, Maryland
| | - Jennifer T Fox
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, NCI, Rockville, Maryland
| | - Fariba Moeinpour
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - M Margaret Juliana
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Robert H Shoemaker
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, NCI, Rockville, Maryland
| | - Clinton J Grubbs
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
10
|
Abo-Zeid MAM, Abo-Elfadl MT, Gamal-Eldeen AM. Evaluation of lapatinib cytotoxicity and genotoxicity on MDA-MB-231 breast cancer cell line. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 71:103207. [PMID: 31234033 DOI: 10.1016/j.etap.2019.103207] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 05/20/2019] [Accepted: 06/06/2019] [Indexed: 06/09/2023]
Abstract
Lapatinib, one of the tyrosine kinase inhibitors (TKIs), is used to reduce epidermal growth factor family proteins overexpression. This study aims to assess the cytotoxic and genotoxic effects of lapatinib on the triple negative breast cancer cell line "MDA-MB-231". We investigated the cytotoxicity of lapatinib by MTT assay, mode of cell death using apoptosis-necrosis assay, DNA damage using micronucleus test, EGFR protein expression by immunocytochemistry, and assessed its effect on EGFR (7p11.2 locus) and TP53 (17p13 locus) genes using interphase-FISH technique. Lapatinib induced cytotoxicity on MDA-MB-231 cell line by elevating the concentration and its IC50 value was 32.5 μM after 24 h. Lapatinib increased apoptotic cells and micronuclei in binucleated cells gradually by increasing the concentration for 24 h. The EGFR protein expression was reduced by double fold that expressed in non-treated cells. Lapatinib enhanced deletion of EGFR gene signals highly significantly from the lowest concentration. Alternatively, lapatinib amplified signals of TP53 gene effectively by raising the concentration. In conclusion, lapatinib induced cytotoxic and genotoxic effects on MDA-MB-231 cell line. However, laptinib reduced the EGFR protein expression and EGFR signals, it raised the apoptotic cells and TP53 gene signals, which triggered extensive DNA damage. Therefore, lapatinib is an effective TKI in triple negative breast cancer cells as elucidated by its mode of cell death.
Collapse
Affiliation(s)
- Mona A M Abo-Zeid
- Genetics and Cytology Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Dokki, 12622, Cairo, Egypt; Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Dokki, 12622, Cairo, Egypt.
| | - Mahmoud T Abo-Elfadl
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Dokki, 12622, Cairo, Egypt; Biochemistry Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Dokki, 12622, Cairo, Egypt
| | - Amira M Gamal-Eldeen
- Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Dokki, 12622, Cairo, Egypt; Biochemistry Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Dokki, 12622, Cairo, Egypt; Clinical Laboratory Department, College of Applied Medical Sciences, Taif University, Al Mutamarat Rd, Al Mathnah, At Taif, 26521, Saudi Arabia
| |
Collapse
|
11
|
Zhang T, Feng F, Yao Y, Qi L, Tian J, Zhou C, Dong S, Wang X, Sun C. Efficacy and acceptability of neoadjuvant endocrine therapy in patients with hormone receptor-positive breast cancer: A network meta-analysis. J Cell Physiol 2019; 234:12393-12403. [PMID: 30652307 DOI: 10.1002/jcp.28068] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 12/18/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND The optimal sequence of endocrine therapy in a neoadjuvant setting for hormone receptor-positive (HR+) breast cancer is unclear. Our study evaluated the efficacy and acceptability of neoadjuvant endocrine therapy for HR+ breast cancer. METHODS We identified studies based on titles and abstracts that were published before 22 June 2018 in the following databases: PubMed, EMBASE, and the Cochrane Library. Eligible studies were randomised controlled trials with at least one arm that evaluated the effectiveness of one or a combination of anastrozole, letrozole, palbociclib, tamoxifen, fulvestrant, abemaciclib, everolimus, gefitinib, ribociclib, taselisib, and exemestane. We pooled effect sizes using the odds ratio (OR) and corresponding 95% credibility interval (95% CrI). The primary outcomes were response rate and treatment completion. RESULTS Our network meta-analysis included 3,306 participants and 16 eligible studies, which assessed 15 treatments. In terms of response rates, compared with letrozole combined therapy, tamoxifen was associated with a significant reduction in response rate (OR, 0.34; 95% CrI, 0.13-0.85; OR, 0.32; 95% CrI, 0.13-0.80; OR, 0.26; 95% CrI, 0.09-0.83; and OR, 0.30; 95% CrI, 0.09-0.96; for letrozole plus everolimus, letrozole plus taselisib, letrozole plus zoledronic acid, and letrozole plus lapatinib, respectively). Based on the surface under the cumulative ranking curves ranking, letrozole plus zoledronic acid was associated with the highest rate of response (87.6%), followed by letrozole plus lapatinib (85.2%), and letrozole plus taselisib (79.3%). CONCLUSIONS Ultimately, our study established that letrozole plus zoledronic acid may be an optimal treatment based on its current rank in a neoadjuvant setting for HR+ breast cancer.
Collapse
Affiliation(s)
- Tingting Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Fubin Feng
- Department of Oncology, Weifang Traditional Chinese Hospital, WeiFang, Shandong, People's Republic of China
| | - Yan Yao
- Clinical Medical Colleges, Weifang Medical University, WeiFang, Shandong, People's Republic of China
| | - Lingyu Qi
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Jinhui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Chao Zhou
- Department of Oncology, Weifang Traditional Chinese Hospital, WeiFang, Shandong, People's Republic of China
| | - Shengjie Dong
- Department of the Joint and Bone Surgery, Yantaishan Hospital, Yantai, Shandong, People's Republic of China
| | - Xue Wang
- Clinical Medical Colleges, Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, WeiFang, Shandong, People's Republic of China.,Department of Oncology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, People's Republic of China
| |
Collapse
|
12
|
Rani A, Stebbing J, Giamas G, Murphy J. Endocrine Resistance in Hormone Receptor Positive Breast Cancer-From Mechanism to Therapy. Front Endocrinol (Lausanne) 2019; 10:245. [PMID: 31178825 PMCID: PMC6543000 DOI: 10.3389/fendo.2019.00245] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/28/2019] [Indexed: 12/24/2022] Open
Abstract
The importance and role of the estrogen receptor (ER) pathway has been well-documented in both breast cancer (BC) development and progression. The treatment of choice in women with metastatic breast cancer (MBC) is classically divided into a variety of endocrine therapies, 3 of the most common being: selective estrogen receptor modulators (SERM), aromatase inhibitors (AI) and selective estrogen receptor down-regulators (SERD). In a proportion of patients, resistance develops to endocrine therapy due to a sophisticated and at times redundant interference, at the molecular level between the ER and growth factor. The progression to endocrine resistance is considered to be a gradual, step-wise process. Several mechanisms have been proposed but thus far none of them can be defined as the complete explanation behind the phenomenon of endocrine resistance. Although multiple cellular, molecular and immune mechanisms have been and are being extensively studied, their individual roles are often poorly understood. In this review, we summarize current progress in our understanding of ER biology and the molecular mechanisms that predispose and determine endocrine resistance in breast cancer patients.
Collapse
Affiliation(s)
- Aradhana Rani
- School of Life Sciences, University of Westminster, London, United Kingdom
- *Correspondence: Aradhana Rani
| | - Justin Stebbing
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Georgios Giamas
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - John Murphy
- School of Life Sciences, University of Westminster, London, United Kingdom
| |
Collapse
|
13
|
Tagliamento M, Genova C, Rijavec E, Rossi G, Biello F, Dal Bello MG, Alama A, Coco S, Boccardo S, Grossi F. Afatinib and Erlotinib in the treatment of squamous-cell lung cancer. Expert Opin Pharmacother 2018; 19:2055-2062. [DOI: 10.1080/14656566.2018.1540591] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
| | - Carlo Genova
- Lung Cancer Unit, Ospedale Policlinico San Martino , Genoa, Italy
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa , Genoa, Italy
| | - Erika Rijavec
- Lung Cancer Unit, Ospedale Policlinico San Martino , Genoa, Italy
| | - Giovanni Rossi
- Lung Cancer Unit, Ospedale Policlinico San Martino , Genoa, Italy
| | - Federica Biello
- Lung Cancer Unit, Ospedale Policlinico San Martino , Genoa, Italy
| | | | - Angela Alama
- Lung Cancer Unit, Ospedale Policlinico San Martino , Genoa, Italy
| | - Simona Coco
- Lung Cancer Unit, Ospedale Policlinico San Martino , Genoa, Italy
| | - Simona Boccardo
- Lung Cancer Unit, Ospedale Policlinico San Martino , Genoa, Italy
| | - Francesco Grossi
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Medical Oncology , Milan, Italy
| |
Collapse
|
14
|
Zhang Y, Bi J, Zhu H, Shi M, Zeng X. ANXA2 could act as a moderator of EGFR-directed therapy resistance in triple negative breast cancer. Biosci Biotechnol Biochem 2018; 82:1733-1741. [DOI: 10.1080/09168451.2018.1484275] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
ABSTRACT
Triple negative breast cancer (TNBC) patients cannot benefit from EGFR-targeted therapy even though the EGFR is highly expressed, because patients exhibit resistance to these drugs. Unfortunately, the molecular mechanisms remain relatively unknown. ANXA2, highly expressed in invasive breast cancer cells, is closely related with poor prognosis, and acts as a molecular switch to EGFR activation. In this study, MDA-MB-231 cells and MCF7 cells were used. Our results showed that ANXA2 expression is inversely correlated with cell sensitivity to gefitinib. Knockdown of ANXA2 expression in MDA-MB-231 cells increased the gefitinib induced cell death. When ANXA2 was overexpressed in MCF7 cells, the gefitinib induced cell death was decreased. Furthermore, we demonstrated that phosphorylation of ANXA2 at Tyr23 is negatively correlated with the sensitivity of TNBC to gefitinib. Altogether, our results suggest a new role of ANXA2 in regulating sensitivity of TNBC MDA-MB-231 cells to the EGFR inhibitor gefitinib.
Collapse
Affiliation(s)
- Yue Zhang
- School of Life Science, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Jiajia Bi
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, China
| | - Hongtao Zhu
- School of Life Science, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Mei Shi
- School of Life Science, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Xianlu Zeng
- School of Life Science, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| |
Collapse
|
15
|
Lubet RA, Steele VE, Shoemaker RH, Grubbs CJ. Screening of Chemopreventive Agents in Animal Models: Results on Reproducibility, Agents of a Given Class, and Agents Tested During Tumor Progression. Cancer Prev Res (Phila) 2018; 11:595-606. [PMID: 30045934 PMCID: PMC6186395 DOI: 10.1158/1940-6207.capr-18-0084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/08/2018] [Accepted: 07/17/2018] [Indexed: 12/22/2022]
Abstract
Because of the importance of testing reproducibility of results, we present our findings regarding screening agents in preclinical chemoprevention studies in rodent models performed by the Chemopreventive Agent Development Research Group (CADRG) of the Division of Cancer Prevention of the NCI. These studies were performed via contracts to various commercial and academic laboratories. Primarily, results with positive agents are reported because positive agents may progress to the clinics. In testing reproducibility, a limited number of direct repeats of our standard screening assays were performed; which entailed initiating treatment shortly after carcinogen administration or in young transgenic mice and continuing treatment until the end of the study. However, three additional protocols were employed relating to reproducibility: (i) testing agents at lower doses to determine efficacy and reduced toxicity; (ii) testing agents later in tumor progression when microscopic lesions existed and, (iii) testing multiple agents of the same mechanistic class. Data with six models that were routinely employed are presented: MNU-induced ER-positive mammary cancer in rats; MMTV-Neu ER-negative mammary cancers in transgenic mice; AOM-induced colon tumors in rats; intestinal adenomas in Min mice; OH-BBN-induced invasive rat urinary bladder cancers in rats; and UV-induced skin squamous carcinomas in mice. It was found that strongly positive results were highly reproducible in the preclinical models evaluated. Cancer Prev Res; 11(10); 595-606. ©2018 AACR.
Collapse
Affiliation(s)
- Ronald A Lubet
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Vernon E Steele
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Robert H Shoemaker
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Clinton J Grubbs
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
16
|
Lubet RA, Steele VE, Juliana MM, Bode A, Moeinpour F, Grubbs CJ. Daily or weekly dosing with EGFR inhibitors, gefitinib and lapatinib, and AKt inhibitor MK2206 in mammary cancer models. Oncol Rep 2018; 40:1545-1553. [PMID: 29565450 PMCID: PMC6072405 DOI: 10.3892/or.2018.6313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 01/12/2018] [Indexed: 12/26/2022] Open
Abstract
Daily vs. weekly dosing with EGFR inhibitors (gefitinib and lapatinib) and an AKT inhibitor (MK2206) were compared in two rodent breast cancer models. Female Sprague-Dawley rats were administered methylnitrosourea (MNU) at 50 days of age, and gefitinib (daily/weekly dosing at 10/70 mg/kg BW) or lapatinib (daily/weekly dosing at 75/525 mg/kg BW) were administered by gavage beginning 5 days after MNU. For the prevention studies, weekly or daily dosing with gefitinib or lapatinib reduced cancer multiplicity >75%, and all treatments reduced tumor weights by >90%. For the therapeutic studies, MNU-treated rats were followed until small palpable mammary cancers developed. The rats were then treated daily or weekly as above for 6 weeks. Either daily or weekly dosing with lapatinib or gefitinib caused regression in >50% of the tumors. Immunohistochemistry biomarker studies in palpable mammary cancers following a weekly dose of gefitinib showed that 1 day (but not 7 days) after treatment, the levels of phosphorylated EGFR1 were significantly decreased. In an ER-negative (ER−) Neu-overexpressing model employing MMTV-Neu/P53KO mice, daily (100 mg/kg BW/day, 5 days each week), or weekly dosing (500 or 250 mg/kg BW) with gefitinib reduced tumor multiplicity 65, 85 and 75%, respectively. In the MNU prevention model, daily dosing (100 mg/kg BW/day) with the allosteric AKT inhibitor MK2206 was ineffective, while weekly dosing (700 mg/kg BW) reduced the final tumor weight >70%. Combining weekly MK2206 with the aromatase inhibitor vorozole (0.12 mg/kg BW/day) showed that each compound alone reduced tumor multiplicity 40–50%. The combination reduced cancer multiplicity ~70%. These studies demonstrate the efficacy of weekly dosing with various protein kinase inhibitors; raising the possibility of employing these agents in a breast cancer preventive setting.
Collapse
Affiliation(s)
- Ronald A Lubet
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD 20852, USA
| | - Vernon E Steele
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD 20852, USA
| | - M M Juliana
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ann Bode
- Division of Cancer Biomarkers and Drug Resistance, University of Minnesota Hormel Institute, Austin, MN 55912, USA
| | - Fariba Moeinpour
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Clinton J Grubbs
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
17
|
Severson TM, Nevedomskaya E, Peeters J, Kuilman T, Krijgsman O, van Rossum A, Droog M, Kim Y, Koornstra R, Beumer I, Glas AM, Peeper D, Wesseling J, Simon IM, Wessels L, Linn SC, Zwart W. Neoadjuvant tamoxifen synchronizes ERα binding and gene expression profiles related to outcome and proliferation. Oncotarget 2017; 7:33901-18. [PMID: 27129152 PMCID: PMC5085127 DOI: 10.18632/oncotarget.8983] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/13/2016] [Indexed: 12/17/2022] Open
Abstract
Estrogen receptor alpha (ERα)-positive breast cancers are frequently treated with tamoxifen, but resistance is common. It remains elusive how tamoxifen resistance occurs and predictive biomarkers for treatment outcome are needed. Because most biomarker discovery studies are performed using pre-treatment surgical resections, the effects of tamoxifen therapy directly on the tumor cell in vivo remain unexamined. In this study, we assessed DNA copy number, gene expression profiles and ERα/chromatin binding landscapes on breast tumor specimens, both before and after neoadjuvant tamoxifen treatment. We observed neoadjuvant tamoxifen treatment synchronized ERα/chromatin interactions and downstream gene expression, indicating that hormonal therapy reduces inter-tumor molecular variability. ERα-synchronized sites are associated with dynamic FOXA1 action at these sites, which is under control of growth factor signaling. Genes associated with tamoxifen-synchronized sites are capable of differentiating patients for tamoxifen benefit. Due to the direct effects of therapeutics on ERα behavior and transcriptional output, our study highlights the added value of biomarker discovery studies after neoadjuvant drug exposure.
Collapse
Affiliation(s)
- Tesa M Severson
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Ekaterina Nevedomskaya
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands.,Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | | | - Thomas Kuilman
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Oscar Krijgsman
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Annelot van Rossum
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Marjolein Droog
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Yongsoo Kim
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands.,Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Rutger Koornstra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, GA, The Netherlands
| | | | | | - Daniel Peeper
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | | | - Lodewyk Wessels
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| | - Sabine C Linn
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands.,Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands.,Department of Pathology, University Medical Center Utrecht, CX, The Netherlands
| | - Wilbert Zwart
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, CX, The Netherlands
| |
Collapse
|
18
|
Matsuda N, Lim B, Wang X, Ueno NT. Early clinical development of epidermal growth factor receptor targeted therapy in breast cancer. Expert Opin Investig Drugs 2017; 26:463-479. [PMID: 28271910 PMCID: PMC5826640 DOI: 10.1080/13543784.2017.1299707] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 02/22/2017] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Epidermal growth factor receptor (EGFR) targeted treatment has been evaluated but has not shown a clear clinical benefit for breast cancer. This review article aims to consider the knowledge of the biological background of EGFR pathways in dissecting clinical studies of EGFR targeted treatment in breast cancer. Areas covered: This review focuses on the role of the EGFR pathway and the investigational drugs that target EGFR for breast cancer. Expert opinion: Recent studies have indicated that EGFR targeted therapy for breast cancer has some promising effects for patients with triple-negative breast cancer, basal-like breast cancer, and inflammatory breast cancer. However, predictive and prognostic biomarkers for EGFR targeted therapy have not been identified. The overexpression or amplification of EGFR itself may not be the true factor of induction of the canonical pathway as an oncogenic driver of breast cancer. Instead, downstream, non-canonical pathways related to EGFR may contribute to some aspects of the biological behavior of breast cancer; therefore, the blockade of the receptor could result in sufficient suppression of downstream pathways to inhibit the aggressive behavior of breast cancer. Mechanistic studies to investigate the dynamic interaction between the EGFR pathway and non-canonical pathways are warranted.
Collapse
Affiliation(s)
- Naoko Matsuda
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bora Lim
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoping Wang
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoto T. Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
19
|
Spring LM, Gupta A, Reynolds KL, Gadd MA, Ellisen LW, Isakoff SJ, Moy B, Bardia A. Neoadjuvant Endocrine Therapy for Estrogen Receptor-Positive Breast Cancer: A Systematic Review and Meta-analysis. JAMA Oncol 2017; 2:1477-1486. [PMID: 27367583 DOI: 10.1001/jamaoncol.2016.1897] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Importance Estrogen receptor-positive (ER+) tumors of the breast are generally highly responsive to endocrine treatment. Although endocrine therapy is the mainstay of adjuvant treatment for ER+ breast cancer, the role of endocrine therapy in the neoadjuvant setting is unclear. Objective To evaluate the effect of neoadjuvant endocrine therapy (NET) on the response rate and the rate of breast conservation surgery (BCS) for ER+ breast cancer. Data Sources Based on PRISMA guidelines, a librarian-led search of PubMed and Ovid MEDLINE was performed to identify eligible trials published from inception to May 15, 2015. The search was performed in May 2015. Study Selection Inclusion criteria were prospective, randomized, neoadjuvant clinical trials that reported response rates with at least 1 arm incorporating NET (n = 20). Two authors independently analyzed the studies for inclusion. Data Extraction and Synthesis Pooled odds ratios (ORs), 95% CIs, and P values were estimated for end points using the fixed- and random-effects statistical model. Results The analysis included 20 studies with 3490 unique patients. Compared with combination chemotherapy, NET as monotherapy with aromatase inhibitors had a similar clinical response rate (OR, 1.08; 95% CI, 0.50-2.35; P = .85; n = 378), radiological response rate (OR, 1.38; 95% CI, 0.92-2.07; P = .12; n = 378), and BCS rate (OR, 0.65; 95% CI, 0.41-1.03; P = .07; n = 334) but with lower toxicity. Aromatase inhibitors were associated with a significantly higher clinical response rate (OR, 1.69; 95% CI, 1.36-2.10; P < .001; n = 1352), radiological response rate (OR, 1.49; 95% CI, 1.18-1.89; P < .001; n = 1418), and BCS rate (OR, 1.62; 95% CI, 1.24-2.12; P < .001; n = 918) compared with tamoxifen. Dual combination therapy with growth factor pathway inhibitors was associated with a higher radiological response rate (OR, 1.59; 95% CI, 1.04-2.43; P = .03; n = 355), but not clinical response rate (OR, 0.76; 95% CI, 0.54-1.07; P = .11; n = 537), compared with endocrine monotherapy. The incidence of pathologic complete response was low (<10%). Conclusions and Relevance Neoadjuvant endocrine therapy, even as monotherapy, is associated with similar response rates as neoadjuvant combination chemotherapy but with significantly lower toxicity, suggesting that NET needs to be reconsidered as a potential option in the appropriate setting. Additional research is needed to develop rational NET combinations and predictive biomarkers to personalize the optimal neoadjuvant strategy for ER+ breast cancer.
Collapse
Affiliation(s)
- Laura M Spring
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston
| | - Arjun Gupta
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas
| | - Kerry L Reynolds
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston
| | - Michele A Gadd
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston
| | - Leif W Ellisen
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston
| | - Steven J Isakoff
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston
| | - Beverly Moy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston
| |
Collapse
|
20
|
Schmid P, Pinder SE, Purushotham A, Thompson AM. Reply to R.F. Sweis et al. J Clin Oncol 2017; 35:261-262. [PMID: 28056198 DOI: 10.1200/jco.2016.69.8019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Peter Schmid
- Peter Schmid, Queen Mary University London, London, United Kingdom; Sarah E. Pinder and Arnie Purushotham, King's College London; Guys & St Thomas NHS Trust, London, United Kingdom; and Alastair M. Thompson, MD Anderson Cancer Center, Houston, TX
| | - Sarah E Pinder
- Peter Schmid, Queen Mary University London, London, United Kingdom; Sarah E. Pinder and Arnie Purushotham, King's College London; Guys & St Thomas NHS Trust, London, United Kingdom; and Alastair M. Thompson, MD Anderson Cancer Center, Houston, TX
| | - Arnie Purushotham
- Peter Schmid, Queen Mary University London, London, United Kingdom; Sarah E. Pinder and Arnie Purushotham, King's College London; Guys & St Thomas NHS Trust, London, United Kingdom; and Alastair M. Thompson, MD Anderson Cancer Center, Houston, TX
| | - Alastair M Thompson
- Peter Schmid, Queen Mary University London, London, United Kingdom; Sarah E. Pinder and Arnie Purushotham, King's College London; Guys & St Thomas NHS Trust, London, United Kingdom; and Alastair M. Thompson, MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
21
|
Maugeri-Saccà M, Barba M, Vici P, Pizzuti L, Sergi D, Catenaro T, Di Lauro L, Mottolese M, Santini D, Milella M, De Maria R. Presurgical window of opportunity trial design as a platform for testing anticancer drugs: Pros, cons and a focus on breast cancer. Crit Rev Oncol Hematol 2016; 106:132-42. [PMID: 27637358 DOI: 10.1016/j.critrevonc.2016.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 06/17/2016] [Accepted: 08/16/2016] [Indexed: 12/24/2022] Open
Abstract
The high attrition rate is a major issue in anticancer drug development. Among the alternative trial designs, presurgical window of opportunity trials envision a short course treatment in the time window between diagnostic biopsy and surgery in a moderately-sized patient population. This approach allows testing therapeutics when pre- and post-treatment tumor tissues are available for comprehensive molecular analyses. The emerging evidence may help define the ability of a given agent to modulate its target(s) and help obtain a broader picture of the molecular changes operated by the treatment. The resulting gain may outweigh the potential harms for patients in the early disease setting. Window of opportunity trials have been extensively applied to breast cancer. Overall, a wider use of these trial designs might lead to the identification of potential responders, ineffective drugs or combinations, and ultimately contribute to enhance the efficiency of the clinical developmental process.
Collapse
Affiliation(s)
- Marcello Maugeri-Saccà
- Division of Medical Oncology 2, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy; Scientific Direction, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy.
| | - Maddalena Barba
- Division of Medical Oncology 2, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy; Scientific Direction, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy.
| | - Patrizia Vici
- Division of Medical Oncology 2, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
| | - Laura Pizzuti
- Division of Medical Oncology 2, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
| | - Domenico Sergi
- Division of Medical Oncology 2, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
| | - Teresa Catenaro
- Division of Medical Oncology 2, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
| | - Luigi Di Lauro
- Division of Medical Oncology 2, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
| | - Marcella Mottolese
- Department of Pathology, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
| | - Daniele Santini
- Department of Medical Oncology, Campus Bio-Medico, University of Rome, 00128 Rome, Italy
| | - Michele Milella
- Division of Medical Oncology A, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
| | - Ruggero De Maria
- Scientific Direction, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
| |
Collapse
|
22
|
Phase II trial of neoadjuvant letrozole and lapatinib in Asian postmenopausal women with estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2)-positive breast cancer [Neo-ALL-IN]: Highlighting the TILs, ER expressional change after neoadjuvant treatment, and FES-PET as potential significant biomarkers. Cancer Chemother Pharmacol 2016; 78:685-95. [DOI: 10.1007/s00280-016-3107-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 07/13/2016] [Indexed: 01/25/2023]
|
23
|
Klintman M, Dowsett M. Early Surrogate Markers of Treatment Activity: Where Are We Now? J Natl Cancer Inst Monogr 2016; 2015:24-8. [PMID: 26063881 DOI: 10.1093/jncimonographs/lgv002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The assessment of new therapies in the adjuvant setting in early breast cancer requires large numbers of patients and many years of follow-up for results to be presented. Therefore, the neoadjuvant study setting, which allows for early prediction of treatment response in smaller patient sets, has become increasingly popular. Ki67 is the most commonly used and extensively studied intermediate biomarker of treatment activity and residual risk in neoadjuvant trials on endocrine therapy, new biological therapies, and chemotherapy. It is increasingly being used as a primary endpoint for new therapies particularly those added to endocrine therapy. The PeriOperative Endocrine Therapy for Individualizing Care (POETIC) trial, including more than 4000 postmenopausal, estrogen receptor (ER)-positive patients randomly assigned to receive 2 weeks of presurgical treatment with an aromatase inhibitor or no further treatment, is the largest window-of-opportunity trial conducted and is assessing the clinical utility of on-treatment Ki67 as a predictor of long-term outcome. For generalizability, Ki67 measurements in the POETIC and other trials need to use standard methodology. The International Working Group on Ki67 in Breast Cancer is conducting a series of studies to bring this to reality.
Collapse
Affiliation(s)
- Marie Klintman
- Academic Department of Biochemistry, Royal Marsden Hospital, London, UK (MK, MD).
| | - Mitchell Dowsett
- Academic Department of Biochemistry, Royal Marsden Hospital, London, UK (MK, MD)
| |
Collapse
|
24
|
Abstract
Nowadays, neoadjuvant endocrine therapy is a clinically acceptable (and sometimes preferred) strategy in patients with operable estrogen receptor-positive (ER+) breast cancer. Despite the overall effectiveness of endocrine therapy in breast cancer in all settings, de novo (primary) and acquired (secondary) endocrine therapy resistance remains a major clinical problem. Neoadjuvant endocrine therapy trials for breast cancer are not only a great opportunity to determine which ER+ breast cancers can be treated without chemotherapy, but also a great strategy to develop insights into the biologic basis for the efficacy of estrogen-receptor-targeting agents, alone or in combination, in an effort to counteract resistance to endocrine therapy and discover actionable molecular targets that can be the focus of future drug discovery efforts and/or translational/clinical investigation in ER+ breast cancers.
Collapse
Affiliation(s)
- Laila S Agrawal
- Department of Medicine, Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | |
Collapse
|
25
|
Schmid P, Pinder SE, Wheatley D, Macaskill J, Zammit C, Hu J, Price R, Bundred N, Hadad S, Shia A, Sarker SJ, Lim L, Gazinska P, Woodman N, Korbie D, Trau M, Mainwaring P, Gendreau S, Lackner MR, Derynck M, Wilson TR, Butler H, Earl G, Parker P, Purushotham A, Thompson A. Phase II Randomized Preoperative Window-of-Opportunity Study of the PI3K Inhibitor Pictilisib Plus Anastrozole Compared With Anastrozole Alone in Patients With Estrogen Receptor-Positive Breast Cancer. J Clin Oncol 2016; 34:1987-94. [PMID: 26976426 DOI: 10.1200/jco.2015.63.9179] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Preclinical data support a key role for the PI3K pathway in estrogen receptor-positive breast cancer and suggest that combining PI3K inhibitors with endocrine therapy may overcome resistance. This preoperative window study assessed whether adding the PI3K inhibitor pictilisib (GDC-0941) can increase the antitumor effects of anastrozole in primary breast cancer and aimed to identify the most appropriate patient population for combination therapy. PATIENTS AND METHODS In this randomized, open-label phase II trial, postmenopausal women with newly diagnosed operable estrogen receptor-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancers were recruited. Participants were randomly allocated (2:1, favoring the combination) to 2 weeks of preoperative treatment with anastrozole 1 mg once per day (n = 26) or the combination of anastrozole 1 mg with pictilisib 260 mg once per day (n = 49). The primary end point was inhibition of tumor cell proliferation as measured by change in Ki-67 protein expression between tumor samples taken before and at the end of treatment. RESULTS There was significantly greater geometric mean Ki-67 suppression of 83.8% (one-sided 95% CI, ≥ 79.0%) for the combination and 66.0% (95% CI, ≤ 75.4%) for anastrozole (geometric mean ratio [combination:anastrozole], 0.48; 95% CI, ≤ 0.72; P = .004). PIK3CA mutations were not predictive of response to pictilisib, but there was significant interaction between response to treatment and molecular subtype (P = .03); for patients with luminal B tumors, the combination:anastrozole geometric mean ratio of Ki-67 suppression was 0.37 (95% CI, ≤ 0.67; P = .008), whereas no significant Ki-67 response was observed for pictilisib in luminal A tumors (1.01; P = .98). Multivariable analysis confirmed Ki-67 response to the combination treatment of patients with luminal B tumors irrespective of progesterone receptor status or baseline Ki-67 expression. CONCLUSION Adding pictilisib to anastrozole significantly increases suppression of tumor cell proliferation in luminal B primary breast cancer.
Collapse
Affiliation(s)
- Peter Schmid
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX.
| | - Sarah E Pinder
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Duncan Wheatley
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Jane Macaskill
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Charles Zammit
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Jennifer Hu
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Robert Price
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Nigel Bundred
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Sirwan Hadad
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Alice Shia
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Shah-Jalal Sarker
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Louise Lim
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Patrycja Gazinska
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Natalie Woodman
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Darren Korbie
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Matt Trau
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Paul Mainwaring
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Steven Gendreau
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Mark R Lackner
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Mika Derynck
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Timothy R Wilson
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Hannah Butler
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Gemma Earl
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Peter Parker
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Arnie Purushotham
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Alastair Thompson
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| |
Collapse
|
26
|
Abstract
Seventy five percent of all breast cancer (BC) patients express estrogen receptor (ER) but a quarter to half of patients with ER positive BC relapse on ET (endocrine therapy), tamoxifen, aromatase inhibitors (AIs), surgical castration, amongst other treatment strategies. ER positive BC at relapse loses ER expression in 20 % of cases and reduces quantitative ER expression most of the time. ER is not the only survival pathway driving ER positive BC and escape pathways intrinsic or acquired are activated during ET. This overview gives an account of ligand-independent ER activation, namely by receptor networks cross talk, and by the various genomic factors and mechanisms leading to ET response failure. Also the mechanisms of Her1 and Her2 inhibition resistance are dealt within this overview, along with the therapeutic indications and limitations of tyrosine kinase inhibitors, PARP inhibitors, PI3K/AKT/mTOR inhibitors, RAS/RAF/MEK/ERK/MAPK inhibitors, and antiangiogenic drugs. In spite of the many advances in controlling the division of BC cells and the progression of BC tumors these still remain the main cause of death among women in age range of 20-50 years requiring even more efforts in new therapeutic approaches besides the drugs within the scope of the overview.
Collapse
Affiliation(s)
- Sofia Braga
- José de Mello Saúde, Avenida Do Forte Edifício Suécia III, Piso 2, Carnaxide, Lisbon, Portugal.
- Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Algarve, Portugal.
| |
Collapse
|
27
|
Tryfonidis K, Basaran G, Bogaerts J, Debled M, Dirix L, Thery JC, Tjan-Heijnen VCG, Van den Weyngaert D, Cufer T, Piccart M, Cameron D. A European Organisation for Research and Treatment of Cancer randomized, double-blind, placebo-controlled, multicentre phase II trial of anastrozole in combination with gefitinib or placebo in hormone receptor-positive advanced breast cancer (NCT00066378). Eur J Cancer 2015; 53:144-54. [PMID: 26724641 DOI: 10.1016/j.ejca.2015.10.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 10/02/2015] [Accepted: 10/15/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND Preclinical data suggest that epidermal growth factor receptor (EGFR) inhibitors (e.g. gefitinib) can delay endocrine resistance in breast cancer. A double-blind, placebo-controlled, phase II trial investigated whether adding gefitinib (G) to anastrozole (A) would improve outcome in advanced breast cancer (ABC). METHODS Postmenopausal pre-treated hormone receptor-positive ABC patients (locally recurrent or metastatic) were 1:1 randomized to A (1 mg/d) plus G 250 mg/d or plus placebo (P). Patients who had prior treatment with an aromatase inhibitor in metastatic setting or with trastuzumab, anti-EGFR or anti-VEGF agents were excluded. Treatment was given until disease progression, unacceptable toxicity or patient withdrawal. Progression-free survival (PFS) rate at 1 year was assessed according to Response Evaluation Criteria in Solid Tumours, version 1.0. RESULTS Of 108 planned patients, 71 were recruited (36 in A/G and 35 in A/P). The trial closed prematurely due to slow recruitment; 31 patients had prior chemotherapy and 53 prior endocrine therapy (all except one received tamoxifen); 60% in adjuvant and 16% in metastatic setting received tamoxifen; 59 patients had visceral disease. Median follow-up was 18 months. PFS rate at 1 year was 35% for A/G and 32% for A/P arm. Objective responses were six (22%) in the A/G and nine (28%) in the A/P arm. Median duration of response was 13.8 and 18.6 months in the A/G and A/P arms, respectively. Fatigue (35%), diarrhoea (31%), rash (32%), dry skin (27%), and arthralgia/myalgia (27%) were the commonest adverse events in the A/G arm. CONCLUSIONS This phase II study, although prematurely closed, did not show a signal that adding G to A improves PFS at 1 year and its use is not supported. Gastrointestinal and skin toxicities were more pronounced with G resulting in premature therapy interruption in almost 1 in 3 patients (ClinicalTrials.gov number, NCT00066378).
Collapse
Affiliation(s)
| | - Gul Basaran
- Acıbadem Üniversitesi İç Hastalıkları/Tıbbi Onkoloji, Turkey.
| | - Jan Bogaerts
- EORTC-Headquarters, Statistical Department, Belgium.
| | | | - Luc Dirix
- GZA Sint Augustinus, Antwerp, Belgium.
| | | | | | | | | | - Martine Piccart
- Institute Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium.
| | - David Cameron
- NHS-Lothian, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
28
|
Marous M, Bièche I, Paoletti X, Alt M, Razak A, Stathis A, Kamal M, Le Tourneau C. Designs of preoperative biomarkers trials in oncology: a systematic review of the literature. Ann Oncol 2015; 26:2419-28. [DOI: 10.1093/annonc/mdv378] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 08/19/2015] [Indexed: 01/06/2023] Open
|
29
|
van Dam PA, van Dam VCN, Altintas S, Papadimitriou K, Rolfo C, Trinh XB. Neoadjuvant endocrine treatment in early breast cancer: An overlooked alternative? Eur J Surg Oncol 2015; 42:333-42. [PMID: 26776766 DOI: 10.1016/j.ejso.2015.10.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 10/16/2015] [Indexed: 12/21/2022] Open
Abstract
During the last decade neoadjuvant endocrine therapy (NET) has moved from being reserved for elderly and frail non-chemotherapy candidates to a primary systemic modality in selected patients with hormone sensitive breast cancer. Neoadjuvant hormonal treatment in patients with hormone receptor positive, HER-2 negative early breast cancer is proven to be an effective and safe option; it is associated with a higher rate of breast conserving surgery (BCS), may reduce the need for adjuvant chemotherapy and enables a delay of surgery for medical or practical reasons. Clinical responses range from 13% to 100% with at least 3 months of NET. Methods of assessing response should include MRI of the breast, particularly in lobular tumours. In studies comparing tamoxifen with aromatase inhibitors (AI), AI proved to be superior in terms of tumour response and rates of BCS. Change in Ki67 is accepted as a validated endpoint for comparing endocrine neoadjuvant agents. Levels of Ki67 during treatment are more closely related to long-term prognosis than pretreatment Ki67. Neoadjuvant endocrine therapy provides a unique opportunity for studies of endocrine responsiveness and the development of new experimental drugs combined with systemic hormonal treatment.
Collapse
Affiliation(s)
- P A van Dam
- Breast Unit of the Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Belgium.
| | - V C N van Dam
- Centre of Oncologic Research (CORE), Antwerp University, Edegem, Belgium
| | - S Altintas
- Breast Unit of the Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Belgium
| | - K Papadimitriou
- Breast Unit of the Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Belgium
| | - C Rolfo
- Breast Unit of the Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Belgium; Fase 1 Unit for Experimental Oncology, Antwerp University Hospital, Belgium
| | - X B Trinh
- Breast Unit of the Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Belgium
| |
Collapse
|
30
|
Peterson EA, Jenkins EC, Lofgren KA, Chandiramani N, Liu H, Aranda E, Barnett M, Kenny PA. Amphiregulin Is a Critical Downstream Effector of Estrogen Signaling in ERα-Positive Breast Cancer. Cancer Res 2015; 75:4830-8. [PMID: 26527289 DOI: 10.1158/0008-5472.can-15-0709] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/31/2015] [Indexed: 02/01/2023]
Abstract
Estrogen stimulation promotes epithelial cell proliferation in estrogen receptor (ERα)-positive breast cancer. Many ERα target genes have been enumerated, but the identities of the key effectors mediating the estrogen signal remain obscure. During mouse mammary gland development, the estrogen growth factor receptor (EGFR) ligand amphiregulin acts as an important stage-specific effector of estrogen signaling. In this study, we investigated the role of amphiregulin in breast cancer cell proliferation using human tissue samples and tumor xenografts in mice. Amphiregulin was enriched in ERα-positive human breast tumor cells and required for estrogen-dependent growth of MCF7 tumor xenografts. Furthermore, amphiregulin levels were suppressed in patients treated with endocrine therapy. Suppression of EGF receptor signaling appeared necessary for the therapeutic response in this setting. Our findings implicate amphiregulin as a critical mediator of the estrogen response in ERα-positive breast cancer, emphasizing the importance of EGF receptor signaling in breast tumor pathogenesis and therapeutic response.
Collapse
Affiliation(s)
- Esther A Peterson
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Edmund C Jenkins
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Kristopher A Lofgren
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York. Oncology Research Laboratory, Kabara Cancer Research Institute, Gundersen Medical Foundation, La Crosse, Wisconsin
| | - Natasha Chandiramani
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Hui Liu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Evelyn Aranda
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Maryia Barnett
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Paraic A Kenny
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York. Oncology Research Laboratory, Kabara Cancer Research Institute, Gundersen Medical Foundation, La Crosse, Wisconsin.
| |
Collapse
|
31
|
Gee JMW, Nicholson RI, Barrow D, Dutkowski CM, Goddard L, Jordan NJ, McClelland RA, Knowlden JM, Francies HE, Hiscox SE, Hutcheson IR. Antihormone induced compensatory signalling in breast cancer: an adverse event in the development of endocrine resistance. Horm Mol Biol Clin Investig 2015; 5:67-77. [PMID: 25961242 DOI: 10.1515/hmbci.2011.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 01/21/2011] [Indexed: 12/16/2022]
Abstract
Using MCF7 breast cancer cells, it has been shown that antihormones promote expression/activity of oestrogen-repressed tyrosine kinases, notably EGFR, HER2 and Src. These inductive events confer responsiveness to targeted inhibitors (e.g., gefitinib, trastuzumab, saracatinib). We observed that these antihormone-induced phenomena are common to ER+HER2- and ER+HER2+ breast cancer models in vitro, where targeting of EGFR, HER2 or Src alongside antihormone improves antitumour response and delays/prevents endocrine resistance. Such targeted inhibitors also subvert acquired endocrine resistant cells which retain increased EGFR, HER2 and Src (e.g., TAMR and FASR models derived after 6-12 months of tamoxifen or Faslodex treatment). Thus, antihormone-induced tyrosine kinases comprise "compensatory signalling" crucial in limiting maximal initial antihormone response and subsequently driving acquired resistance in vitro. However, despite such convincing preclinical findings from our group and others, clinical trials examining equivalent antigrowth factor strategies have proved relatively disappointing. Our new studies deciphering underlying causes reveal that further antihormone-promoted events could be pivotal in vivo. Firstly, Faslodex induces HER3 and HER4 which sensitise ER+ cells to heregulin, a paracrine growth factor that overcomes endocrine response and diminishes antitumour effect of agents targeting EGFR, HER2 or Src alongside antihormone. Secondly, extended antihormone exposure (experienced by ER+ cells prior to adjuvant clinical relapse) can "reprogramme" the compensatory kinase profile in vitro, hindering candidate antigrowth factor targeting of endocrine resistance. Faslodex resistant cells maintained with this antihormone for 3 years in vitro lose EGFR/HER2 dependency, gaining alternative mitogenic/invasion kinases. Deciphering these previously unrecognised antihormone-induced events could provide superior treatments to control endocrine relapse in the clinic.
Collapse
|
32
|
Grassadonia A, Caporale M, Tinari N, Zilli M, DeTursi M, Gamucci T, Vici P, Natoli C. Effect of targeted agents on the endocrine response of breast cancer in the neoadjuvant setting: a systematic review. J Cancer 2015; 6:575-82. [PMID: 26000050 PMCID: PMC4439944 DOI: 10.7150/jca.11566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 03/02/2015] [Indexed: 12/22/2022] Open
Abstract
Inhibition of aberrantly activated pathways cross-talking with hormone receptor (HR) improves response to endocrine therapy in patients with HR-positive advanced breast cancer. We performed a Pubmed database systematic review to ascertain the existence of a better clinical response when combining endocrine therapy with targeted agents in the neoadjuvant setting. Preclinical studies or trials evaluating toxicity were excluded. We found nine phase II trials that fulfilled the research criteria. The endocrine agents used were third generation aromatase inhibitors (AIs), anastrozole, letrozole or exemestane. The investigated targeted agents were inhibitors of tyrosine kinase receptors such as gefitinib, imatinib or trastuzumab/lapatinib, inhibitors of mTOR, such as everolimus, inhibitors of COX-2, such as celecoxib, and inhibitors of angiogenesis, such as bevacizumab. The response rate (RR) observed combining endocrine and targeted agents ranged between 36% and 90%. Overall the studies failed to show a remarkable advantage in RR in the combination group compared to historical control subjects receiving AIs alone.
Collapse
Affiliation(s)
- Antonino Grassadonia
- 1. Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio", Chieti, Italy ; 2. Medical Oncology Unit, "SS. Annunziata" Hospital, Chieti, Italy
| | - Marta Caporale
- 2. Medical Oncology Unit, "SS. Annunziata" Hospital, Chieti, Italy
| | - Nicola Tinari
- 1. Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio", Chieti, Italy ; 2. Medical Oncology Unit, "SS. Annunziata" Hospital, Chieti, Italy
| | - Marinella Zilli
- 2. Medical Oncology Unit, "SS. Annunziata" Hospital, Chieti, Italy
| | - Michele DeTursi
- 1. Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio", Chieti, Italy ; 2. Medical Oncology Unit, "SS. Annunziata" Hospital, Chieti, Italy
| | - Teresa Gamucci
- 3. Department of Oncology, "S.S. Trinita'" Hospital, Sora, Italy
| | - Patrizia Vici
- 4. Division of Medical Oncology B, Regina Elena National Cancer Institute, Rome, Italy
| | - Clara Natoli
- 1. Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio", Chieti, Italy ; 2. Medical Oncology Unit, "SS. Annunziata" Hospital, Chieti, Italy
| |
Collapse
|
33
|
Valentine J, Belum VR, Duran J, Ciccolini K, Schindler K, Wu S, Lacouture ME. Incidence and risk of xerosis with targeted anticancer therapies. J Am Acad Dermatol 2015; 72:656-67. [PMID: 25637330 DOI: 10.1016/j.jaad.2014.12.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 12/03/2014] [Accepted: 12/08/2014] [Indexed: 12/17/2022]
Abstract
BACKGROUND Many targeted therapies used in the treatment of cancer can lead to the development of xerosis, but the incidence and relative risk of xerosis have not been ascertained. OBJECTIVE We conducted a systematic review and metaanalysis of clinical trials, to ascertain the incidence and risk of developing xerosis after taking anticancer drugs. METHODS The PubMed (1966-October 2013), Web of Science (January 1998-October 2013), and American Society of Clinical Oncology abstracts (2004-2013) databases were searched for clinical trials of 58 targeted agents. Results were calculated using random or fixed effects models. RESULTS The incidences of all- and high-grade xerosis were 17.9% (95% confidence interval [CI]: 15.6-20.4%) and 1.0% (95% CI: 0.9-1.5%), respectively. The risk of developing all-grade xerosis was 2.99 (95% CI: 2.0-4.3), and it varied across different drugs (P < .001). LIMITATIONS The reporting of xerosis may vary among clinicians and institutions, and the incidence may be affected by age, concomitant medications, comorbidities, and underlying malignancies or skin conditions. CONCLUSION Patients receiving targeted therapies have a significant risk of developing xerosis. Patients should be counseled and treated early for this symptom to prevent suboptimal dosing and quality of life impairment.
Collapse
Affiliation(s)
- Johannah Valentine
- Department of Dermatology, Naval Medical Center San Diego, San Diego, California
| | | | - Juanita Duran
- Department of Dermatology, Universidad del Rosario, Bogota, Colombia
| | - Kathryn Ciccolini
- Dermatology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katja Schindler
- Dermatology Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Shenhong Wu
- Division of Hematology and Oncology, Stony Brook University Cancer Center, Stony Brook, New York; Division of Hematology and Oncology, Department of Medicine, Northport Veterans Affairs Medical Center, Northport, New York
| | - Mario E Lacouture
- Dermatology Service, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
34
|
BELIZZI ANTONIA, GRECO MARIARAFFAELLA, RUBINO ROSA, PARADISO ANGELO, FORCINITI STEFANIA, ZEEBERG KATRINE, CARDONE ROSAANGELA, RESHKIN STEPHANJOEL. The scaffolding protein NHERF1 sensitizes EGFR-dependent tumor growth, motility and invadopodia function to gefitinib treatment in breast cancer cells. Int J Oncol 2014; 46:1214-24. [DOI: 10.3892/ijo.2014.2805] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 11/26/2014] [Indexed: 11/06/2022] Open
|
35
|
Vici P, Pizzuti L, Natoli C, Gamucci T, Di Lauro L, Barba M, Sergi D, Botti C, Michelotti A, Moscetti L, Mariani L, Izzo F, D'Onofrio L, Sperduti I, Conti F, Rossi V, Cassano A, Maugeri-Saccà M, Mottolese M, Marchetti P. Triple positive breast cancer: a distinct subtype? Cancer Treat Rev 2014; 41:69-76. [PMID: 25554445 DOI: 10.1016/j.ctrv.2014.12.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/11/2014] [Accepted: 12/11/2014] [Indexed: 01/23/2023]
Abstract
Breast cancer is a heterogeneous disease, and within the HER-2 positive subtype this is highly exemplified by the presence of substantial phenotypical and clinical heterogeneity, mostly related to hormonal receptor (HR) expression. It is well known how HER-2 positivity is commonly associated with a more aggressive tumor phenotype and decreased overall survival and, moreover, with a reduced benefit from endocrine treatment. Preclinical studies corroborate the role played by functional crosstalks between HER-2 and estrogen receptor (ER) signaling in endocrine resistance and, more recently, the activation of ER signaling is emerging as a possible mechanism of resistance to HER-2 blocking agents. Indeed, HER-2 positive breast cancer heterogeneity has been suggested to underlie the variability of response not only to endocrine treatments, but also to HER-2 blocking agents. Among HER-2 positive tumors, HR status probably defines two distinct subtypes, with dissimilar clinical behavior and different sensitivity to anticancer agents. The triple positive subtype, namely, ER/PgR/Her-2 positive tumors, could be considered the subset which most closely resembles the HER-2 negative/HR positive tumors, with substantial differences in biology and clinical outcome. We argue on whether in this subgroup the "standard" treatment may be considered, in selected cases, i.e., small tumors, low tumor burden, high expression of both hormonal receptors, an overtreatment. This article review the existing literature on biologic and clinical data concerning the HER-2/ER/PgR positive tumors, in an attempt to better define the HER-2 subtypes and to optimize the use of HER-2 targeted agents, chemotherapy and endocrine treatments in the various subsets.
Collapse
Affiliation(s)
- Patrizia Vici
- Division of Medical Oncology B, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy.
| | - Laura Pizzuti
- Division of Medical Oncology B, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy.
| | - Clara Natoli
- Department of Experimental and Clinical Sciences, University "G. d'Annunzio", V dei Vestini, 29, 66100 Chieti, Italy.
| | - Teresa Gamucci
- Medical Oncology Unit ASL Frosinone, V Armando Fabi, 03100 Frosinone, Italy.
| | - Luigi Di Lauro
- Division of Medical Oncology B, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy.
| | - Maddalena Barba
- Division of Medical Oncology B, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy; Scientific Direction, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy.
| | - Domenico Sergi
- Division of Medical Oncology B, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy.
| | - Claudio Botti
- Department of Surgery, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy.
| | - Andrea Michelotti
- Oncology Unit I, Azienda Ospedaliera Universitaria Pisana, V Roma 67, 56126 Pisa, Italy.
| | - Luca Moscetti
- Division of Medical Oncology, Department of Oncology, Belcolle Hospital, ASL Viterbo, Strada S. Martinese, 01100 Viterbo, Italy.
| | - Luciano Mariani
- Department of Gynecologic Oncology, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy; HPV Unit, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy.
| | - Fiorentino Izzo
- Division of Medical Oncology B, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy.
| | - Loretta D'Onofrio
- Department of Medical Oncology, University Campus Bio-Medico, V Álvaro del Portillo 21, 00128 Rome, Italy.
| | - Isabella Sperduti
- Biostatistics Unit, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy.
| | - Francesca Conti
- Division of Medical Oncology B, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy.
| | - Valentina Rossi
- Division of Medical Oncology, Ospedale Civile di Saluzzo, V Spielberg 58, 12100 Saluzzo (CN), Italy.
| | - Alessandra Cassano
- Division of Medical Oncology, Catholic University of Sacred Heart, Largo Agostino Gemelli 8, 00168 Rome, Italy.
| | - Marcello Maugeri-Saccà
- Division of Medical Oncology B, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy; Scientific Direction, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy.
| | - Marcella Mottolese
- Department of Pathology, "Regina Elena" National Cancer Institute, V Elio Chianesi 53, 00144 Rome, Italy.
| | - Paolo Marchetti
- Oncology Unit, Sant'Andrea Hospital, "Sapienza" University of Rome, V Grottarossa 1035/1039, 00189 Rome, Italy.
| |
Collapse
|
36
|
Milani A, Geuna E, Mittica G, Valabrega G. Overcoming endocrine resistance in metastatic breast cancer: Current evidence and future directions. World J Clin Oncol 2014; 5:990-1001. [PMID: 25493235 PMCID: PMC4259959 DOI: 10.5306/wjco.v5.i5.990] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 05/12/2014] [Accepted: 07/18/2014] [Indexed: 02/06/2023] Open
Abstract
About 75% of all breast cancers are estrogen receptor (ER)-positive. They generally have a more favorable clinical behavior, prognosis, and pattern of recurrence, and endocrine therapy forms the backbone of treatment. Anti-estrogens (such as tamoxifen and fulvestrant) and aromatase inhibitors (such as anastrozole, letrozole, and exemestane) can effectively control the disease and induce tumor responses in a large proportion of patients. However, the majority of patients progress during endocrine therapy (acquired resistance) and a proportion of patients may fail to respond to initial therapy (de novo resistance). Endocrine resistance is therefore of clinical concern and there is great interest in strategies that delay or circumvent it. A deeper knowledge of the molecular mechanisms that drive endocrine resistance has recently led to development of new strategies that have the promise to effectively overcome it. Many resistance mechanisms have been described, and the crosstalk between ER and growth factor receptor signaling pathways seems to represent one of the most relevant. Compounds that are able to inhibit key elements of these pathways and restore endocrine sensitivity have been studied and more are currently under development. The aim of this review is to summarize the molecular pathophysiology of endocrine resistance in breast cancer and its impact on current clinical management.
Collapse
|
37
|
Knox AJ, Scaling AL, Pinto MP, Bliesner BS, Haughian JM, Abdel-Hafiz HA, Horwitz KB. Modeling luminal breast cancer heterogeneity: combination therapy to suppress a hormone receptor-negative, cytokeratin 5-positive subpopulation in luminal disease. Breast Cancer Res 2014; 16:418. [PMID: 25116921 PMCID: PMC4187339 DOI: 10.1186/s13058-014-0418-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 07/22/2014] [Indexed: 12/27/2022] Open
Abstract
Introduction Many Luminal breast cancers are heterogeneous, containing substantial numbers of estrogen (ER) and progesterone (PR) receptor-negative cells among the ER+ PR+ ones. One such subpopulation we call “Luminobasal” is ER-, PR- and cytokeratin 5 (CK5)-positive. It is not targeted for treatment. Methods To address the relationships between ER+PR+CK5– and ER–PR–CK5+ cells in Luminal cancers and tightly control their ratios we generated isogenic pure Luminal (pLUM) and pure Luminobasal (pLB) cells from the same parental Luminal human breast cancer cell line. We used high-throughput screening to identify pLB-specific drugs and examined their efficacy alone and in combination with hormone therapy in mixed-cell tumor models. Results We show that pLUM and MCF7 cells suppress proliferation of pLB cells in mixed-cell 3D colonies in vitro and that pLUM cells suppress growth of pLB cells in mixed-cell xenografts in vivo. High-throughput screening of 89 FDA-approved oncology drugs shows that pLB cells are sensitive to monotherapy with the epidermal growth factor receptor (EGFR) inhibitors gefitinib and erlotinib. By exploiting mixed-cell 3D colonies and mixed-cell solid mouse tumors models we demonstrate that combination therapy with gefitinib plus the anti-estrogen fulvestrant constitutes a robust treatment strategy. Conclusions We propose that response to combination endocrine/EGFR inhibitor therapies in heterogeneous Luminal cancers may improve long-term survival in patients whose primary tumors have been preselected for appropriate biomarkers, including ER, PR, CK5 and EGFR. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0418-6) contains supplementary material, which is available to authorized users.
Collapse
|
38
|
Zhao M, Ramaswamy B. Mechanisms and therapeutic advances in the management of endocrine-resistant breast cancer. World J Clin Oncol 2014; 5:248-262. [PMID: 25114842 PMCID: PMC4127598 DOI: 10.5306/wjco.v5.i3.248] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/11/2014] [Accepted: 06/20/2014] [Indexed: 02/06/2023] Open
Abstract
The estrogen receptor (ER) pathway plays a critical role in breast cancer development and progression. Endocrine therapy targeting estrogen action is the most important systemic therapy for ER positive breast cancer. However its efficacy is limited by intrinsic and acquired resistance. Mechanisms responsible for endocrine resistance include deregulation of the ER pathway itself, including loss of ER expression, post-translational modification of ER, deregulation of ER co-activators; increased receptor tyrosine kinase signaling leading to activation of various intracellular pathways involved in signal transduction, proliferation and cell survival, including growth factor receptor tyrosine kinases human epidermal growth factor receptor-2, epidermal growth factor receptor, PI3K/AKT/mammalian target of rapamycin (mTOR), Mitogen activated kinase (MAPK)/ERK, fibroblast growth factor receptor, insulin-like growth factor-1 receptor; alterations in cell cycle and apoptotic machinery; Epigenetic modification including dysregulation of DNA methylation, histone modification, and nucleosome remodeling; and altered expression of specific microRNAs. Functional genomics has helped us identify a catalog of genetic and epigenetic alterations that may be exploited as potential therapeutic targets and biomarkers of response. New treatment combinations targeting ER and such oncogenic signaling pathways which block the crosstalk between these pathways have been proven effective in preclinical models. Results of recent clinical studies suggest that subsets of patients benefit from the combination of inhibitor targeting certain oncogenic signaling pathway with endocrine therapy. Especially, inhibition of the mTOR signaling pathway, a key component implicated in mediating multiple signaling cascades, offers a promising approach to restore sensitivity to endocrine therapy in breast cancer. We systematically reviewed important publications cited in PubMed, recent abstracts from ASCO annual meetings and San Antonio Breast Cancer Symposium, and relevant trials registered at ClinicalTrials.gov. We present the molecular mechanisms contributing to endocrine resistance, in particular focusing on the biological rationale for the clinical development of novel targeted agents in endocrine resistant breast cancer. We summarize clinical trials utilizing novel strategies to overcome therapeutic resistance, highlighting the need to better identify the appropriate patients whose diseases are most likely to benefit from these specific strategies.
Collapse
|
39
|
Abstract
INTRODUCTION The EGFR has been associated with the pathogenesis and progression of breast cancer. Treatment based on an EGFR target is emerging as a promising option, especially in combination with conventional therapies. Unfortunately, there are no validated predictor biomarkers, and combinatorial treatments are meeting new resistance. AREAS COVERED The purpose of this review is to summarize the existing treatments and the current research based on targeting the EGFR pathway. EXPERT OPINION The existing EGFR treatments in breast cancer have shown limited benefit. The combination of the monoclonal antibody cetuximab and platinum salts achieves a 15 - 20% response rate. The effectiveness of tyrosine kinase inhibitors is not completely clear, showing modest or no benefits. Gefitinib treatment has offered some promising results in estrogen receptor + breast cancer. However, it has not been identified as a predictive factor for the appropriate selection of patients. Radioimmunotherapy with anti-EGFR radiolabeled antibodies is a promising strategy in BRCA-mutated breast cancer, but it still requires clinical confirmation. Nevertheless, the crosstalk between pathways frequently leads to treatment resistance. Current research is focused on increasing knowledge about the mechanisms of response and the discovery of predictive markers. Targeting several pathways simultaneously and a correct selection of patients seem essential.
Collapse
Affiliation(s)
- Ana Lluch
- Hospital Clinico Universitario, INCLIVA Biomedical Research Institute, Department of Oncology and Hematology , Valencia , Spain +0034 963987659 ;
| | | | | |
Collapse
|
40
|
Guarneri V, Generali DG, Frassoldati A, Artioli F, Boni C, Cavanna L, Tagliafico E, Maiorana A, Bottini A, Cagossi K, Bisagni G, Piacentini F, Ficarra G, Bettelli S, Roncaglia E, Nuzzo S, Swaby R, Ellis C, Holford C, Conte P. Double-blind, placebo-controlled, multicenter, randomized, phase IIb neoadjuvant study of letrozole-lapatinib in postmenopausal hormone receptor-positive, human epidermal growth factor receptor 2-negative, operable breast cancer. J Clin Oncol 2014; 32:1050-7. [PMID: 24590635 DOI: 10.1200/jco.2013.51.4737] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE This is a randomized, double-blind, placebo-controlled study aimed to evaluate the clinical and biologic effects of letrozole plus lapatinib or placebo as neoadjuvant therapy in hormone receptor (HR) -positive/human epidermal growth factor receptor 2 (HER2) -negative operable breast cancer. METHODS Ninety-two postmenopausal women with stage II to IIIA primary breast cancer were randomly assigned to preoperative therapy consisting of 6 months of letrozole 2.5 mg orally daily plus lapatinib 1,500 mg orally daily or placebo. Surgery was performed within 2 weeks from the last study medication. Clinical response was assessed by ultrasonography. Pre- and post-treatment samples were evaluated for selected biomarkers. Fresh-frozen tissue samples were collected for genomic analyses. RESULTS Numerically similar clinical response rates (partial + complete response) were observed (70% for letrozole-lapatinib and 63% for letrozole-placebo). Toxicities were generally mild and manageable. A significant decrease in Ki-67 and pAKT expression from baseline to surgery was observed in both arms. Overall, 34 patients (37%) had a mutation in PIK3CA exon 9 or 20. In the letrozole-lapatinib arm, the probability of achieving a clinical response was significantly higher in the presence of PIK3CA mutation (objective response rate, 93% v 63% in PIK3CA wild type; P = .040). CONCLUSION The combination of letrozole-lapatinib in early breast cancer was feasible, with expected and manageable toxicities. In unselected estrogen receptor-positive/HER2-negative patients, letrozole-lapatinib and letrozole-placebo resulted in a similar overall clinical response rate and similar effect on Ki-67 and pAKT. Our secondary end point findings of a significant correlation between PIK3CA mutation and response to letrozole-lapatinib in HR-positive/HER2-negative early breast cancer must now be independently confirmed.
Collapse
Affiliation(s)
- Valentina Guarneri
- Valentina Guarneri and PierFranco Conte, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), University of Padova, Padova; Daniele Giulio Generali and Alberto Bottini, U.O. Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona; Antonio Frassoldati, University Hospital, Ferrara; Fabrizio Artioli and Katia Cagossi, Ramazzini Hospital, Carpi; Corrado Boni and Giancarlo Bisagni, Azienda Ospedaliera Arcispedale S. Maria Nuova, IRCCS, Reggio Emilia; Luigi Cavanna, Hospital of Piacenza, Piacenza; Enrico Tagliafico, Enrica Roncaglia, and Simona Nuzzo, Center for Genome Research, University of Modena and Reggio Emilia; Antonino Maiorana, Federico Piacentini, Guido Ficarra, and Stefania Bettelli, Modena University Hospital, Modena, Italy; Ramona Swaby and Catherine Ellis, GlaxoSmithKline, Upper Providence, PA; and Clare Holford, GlaxoSmithKline, Stockley Park, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Palmieri C, Patten DK, Januszewski A, Zucchini G, Howell SJ. Breast cancer: current and future endocrine therapies. Mol Cell Endocrinol 2014; 382:695-723. [PMID: 23933149 DOI: 10.1016/j.mce.2013.08.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Revised: 07/31/2013] [Accepted: 08/01/2013] [Indexed: 12/29/2022]
Abstract
Endocrine therapy forms a central modality in the treatment of estrogen receptor positive breast cancer. The routine use of 5 years of adjuvant tamoxifen has improved survival rates for early breast cancer, and more recently has evolved in the postmenopausal setting to include aromatase inhibitors. The optimal duration of adjuvant endocrine therapy remains an active area of clinical study with recent data supporting 10 years rather than 5 years of adjuvant tamoxifen. However, endocrine therapy is limited by the development of resistance, this can occur by a number of possible mechanisms and numerous studies have been performed which combine endocrine therapy with agents that modulate these mechanisms with the aim of preventing or delaying the emergence of resistance. Recent trial data regarding the combination of the mammalian target of rapamycin (mTOR) inhibitor, everolimus with endocrine therapy have resulted in a redefinition of the clinical treatment pathway in the metastatic setting. This review details the current endocrine therapy utilized in both early and advanced disease, as well as exploring potential new targets which modulate pathways of resistance, as well as agents which aim to modulate adrenal derived steroidogenic hormones.
Collapse
Affiliation(s)
- Carlo Palmieri
- The University of Liverpool, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, Liverpool L69 3GA, UK; Liverpool & Merseyside Breast Academic Unit, The Linda McCartney Centre, Royal Liverpool University Hospital, Liverpool L7 8XP, UK; Academic Department of Medical Oncology, Clatterbridge Cancer Centre NHS Foundation Trust, Wiral CH63 4JY, UK.
| | - Darren K Patten
- Department of Surgery, Imperial College Healthcare NHS Trust, Fulham Palace Road, London W6 8RF, UK
| | - Adam Januszewski
- Department of Medical Oncology, Imperial College Healthcare NHS Trust, Fulham Palace Road, London W6 8RF, UK
| | - Giorgia Zucchini
- The University of Manchester, Institute of Cancer Studies, Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK
| | - Sacha J Howell
- The University of Manchester, Institute of Cancer Studies, Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
42
|
Azim Jr HA, Piccart MJ. Simultaneous targeting of estrogen receptor and HER2 in breast cancer. Expert Rev Anticancer Ther 2014; 10:1255-63. [DOI: 10.1586/era.10.99] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
43
|
Montemurro F, Scaltriti M. Biomarkers of drugs targeting HER-family signalling in cancer. J Pathol 2013; 232:219-29. [DOI: 10.1002/path.4269] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/15/2013] [Accepted: 09/16/2013] [Indexed: 12/12/2022]
Affiliation(s)
- Filippo Montemurro
- Unit of Investigative Clinical Oncology (INCO) and Division of Medical Oncology, Fondazione del Piemonte per l'Oncologia; Institute of Candiolo (IRCCs); Str Provinciale 142 10060 Candiolo Italy
| | - Maurizio Scaltriti
- Human Oncology & Pathogenesis Program (HOPP) and Memorial Sloan Kettering Cancer Center; 1275 York Avenue, Box 20 New York NY 10065 USA
| |
Collapse
|
44
|
Ensslin CJ, Rosen AC, Wu S, Lacouture ME. Pruritus in patients treated with targeted cancer therapies: systematic review and meta-analysis. J Am Acad Dermatol 2013; 69:708-720. [PMID: 23981682 DOI: 10.1016/j.jaad.2013.06.038] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 06/24/2013] [Accepted: 06/26/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND Pruritus has been anecdotally described in association with targeted cancer therapies. The risk of pruritus has not been systematically ascertained. OBJECTIVE A systematic review and meta-analysis of the literature was conducted for axitinib, cetuximab, dasatinib, erlotinib, everolimus, gefitinib, imatinib, ipilimumab, lapatinib, nilotinib, panitumumab, pazopanib, rituximab, sorafenib, temsirolimus, tositumomab, vandetanib, and vemurafenib. METHODS Databases from PubMed, Web of Science (January 1998 through July 2012), and American Society of Clinical Oncology abstracts (2004 through 2012) were searched. Incidence and relative risk of pruritus were calculated using random- or fixed-effects model. RESULTS The incidences of all-grade and high-grade pruritus were 17.4% (95% confidence interval 16.0%-19.0%) and 1.4% (95% confidence interval 1.2%-1.6%), respectively. There was an increased risk of all-grade pruritus (relative risk 2.90 [95% confidence interval 1.76-4.77, P < .001]) and variation among different drugs (P < .001). LIMITATIONS The reporting of pruritus may vary, resulting from concomitant medications, comorbidities, and underlying malignancies. We found a higher incidence of pruritus in patients with solid tumors, concordant with those targeted therapies with the highest pruritus incidences. CONCLUSION There is a significant risk of developing pruritus in patients receiving targeted therapies. To prevent suboptimal dosing and decreased quality of life, patients should be counseled and treated against this untoward symptom.
Collapse
Affiliation(s)
- Courtney J Ensslin
- Dermatology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Alyx C Rosen
- Dermatology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Shenhong Wu
- Division of Medical Oncology, Department of Medicine, State University of New York at Stony Brook, Stony Brook, New York
| | - Mario E Lacouture
- Dermatology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York.
| |
Collapse
|
45
|
Tyrosine kinase inhibitors (TKIs) in human and pet tumours with special reference to breast cancer: a comparative review. Crit Rev Oncol Hematol 2013; 88:293-308. [PMID: 23768779 DOI: 10.1016/j.critrevonc.2013.05.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 04/27/2013] [Accepted: 05/17/2013] [Indexed: 12/13/2022] Open
Abstract
Tyrosine kinase receptors (TKRs) play a key role in tumour cell proliferation and survival since they are involved in endothelial cell activation leading to tumour neoangiogenesis. In particular, vascular endothelial growth factor receptors (VEGFRs), platelet-derived growth factor receptor (PDGFR), stem cell factor receptor (c-KitR), and colony-stimulating factor 1 (CSF-1) are overexpressed or constitutively activated in human and pet malignancies. A variety of small molecule inhibitors targeting specific tyrosine kinases (known as tyrosine kinase inhibitors or TKIs) have recently been approved, or are under investigation, for the treatment of human cancer. TKI application in animal cancer is however relatively recent. This review aims to illustrate the major aspects of tyrosine kinase dysfunctions, with special regard to human and animal cancer of the mammary gland, providing an update on the background of the anti-angiogenic and anti-neoplastic properties of TKIs in human and veterinary cancer.
Collapse
|
46
|
Cellular functions regulated by phosphorylation of EGFR on Tyr845. Int J Mol Sci 2013; 14:10761-90. [PMID: 23702846 PMCID: PMC3709701 DOI: 10.3390/ijms140610761] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/06/2013] [Accepted: 05/13/2013] [Indexed: 11/17/2022] Open
Abstract
The Src gene product (Src) and the epidermal growth factor receptor (EGFR) are prototypes of oncogene products and function primarily as a cytoplasmic non-receptor tyrosine kinase and a transmembrane receptor tyrosine kinase, respectively. The identification of Src and EGFR, and the subsequent extensive investigations of these proteins have long provided cutting edge research in cancer and other molecular and cellular biological studies. In 1995, we reported that the human epidermoid carcinoma cells, A431, contain a small fraction of Src and EGFR in which these two kinase were in physical association with each other, and that Src phosphorylates EGFR on tyrosine 845 (Y845) in the Src-EGFR complex. Y845 of EGFR is located in the activation segment of the kinase domain, where many protein kinases contain kinase-activating autophosphorylation sites (e.g., cAMP-dependent protein kinase, Src family kinases, transmembrane receptor type tyrosine kinases) or trans-phosphorylation sites (e.g., cyclin-dependent protein kinase, mitogen-activated protein kinase, Akt protein kinase). A number of studies have demonstrated that Y845 phosphorylation serves an important role in cancer as well as normal cells. Here we compile the experimental facts involving Src phosphorylation of EGFR on Y845, by which cell proliferation, cell cycle control, mitochondrial regulation of cell metabolism, gamete activation and other cellular functions are regulated. We also discuss the physiological relevance, as well as structural insights of the Y845 phosphorylation.
Collapse
|
47
|
Masuda M, Miki Y, Hata S, Takagi K, Sakurai M, Ono K, Suzuki K, Yang Y, Abe E, Hirakawa H, Ishida T, Suzuki T, Ohuchi N, Sasano H. An induction of microRNA, miR-7 through estrogen treatment in breast carcinoma. J Transl Med 2013; 10 Suppl 1:S2. [PMID: 23227519 PMCID: PMC3445861 DOI: 10.1186/1479-5876-10-s1-s2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Background Estrogen plays an important role in the development of estrogen-dependent breast carcinoma. Recently, several studies demonstrated a possible involvement of several micro RNAs (miRNAs) in the development of resistance to endocrine therapy in breast cancer patients, but the correlation between estrogen actions and miRNA expression in breast carcinoma still remains largely unknown. Therefore, in this study, we examined the in vitro effects of estrogen upon miRNA expression profiles in breast carcinoma. Methods We first screened the miRNA expression profiles induced by 17β-Estradiol (E2) using RT2 miRNA PCR Array in the ER-positive breast carcinoma cell line MCF-7. We identified miR-7 as the important miRNA associated with estrogen actions in these cells and further examined the changes of estrogen-dependent EGFR expression by miR-7 in ER-positive or -negative breast carcinoma cell lines including MCF-7. We also evaluated the correlation between miR-7 and EGFR expression in breast carcinoma cells derived from 21 patients using laser capture microdissection combined with quantitative reverse transcriptase-PCR. Results Seventeen miRNAs were significantly induced by E2 treatment in the MCF-7 cell line. Among 17 miRNAs induced by estradiol treatment, only miR-7 expression was significantly decreased by subsequent ICI treatment. The expression of miR-7 was up-regulated 2.94-fold by E2 treatment. miR-7 was reported to suppress epidermal growth factor receptor (EGFR) expression in several human malignancies. Transfection of miR-7 significantly suppressed EGFR mRNA levels in MCF-7 cells. Depletion of E2 from cell culture media also increased the expression level of EGFR mRNA in MCF-7 and T-47D cells but not in ER-negative, MDA-MB-231 and SK-BR-3 cells. We also evaluated the status of miR-7 in breast carcinoma tissues, but the correlation between the status of miR-7 and EGFR in carcinoma cells isolated by laser capture microscopy was not detected. Conclusions These results suggest that miR-7 may play a role in the development of resistance to endocrine therapy in breast cancer patients through regulating EGFR expression of carcinoma cells.
Collapse
Affiliation(s)
- Mariko Masuda
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Stebbing J, Payne R, Reise J, Frampton AE, Avery M, Woodley L, Di Leo A, Pestrin M, Krell J, Coombes RC. The efficacy of lapatinib in metastatic breast cancer with HER2 non-amplified primary tumors and EGFR positive circulating tumor cells: a proof-of-concept study. PLoS One 2013; 8:e62543. [PMID: 23667487 PMCID: PMC3647066 DOI: 10.1371/journal.pone.0062543] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 03/18/2013] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Analysis of circulating tumor cells (CTCs) provides real-time measures of cancer sub-populations with potential for CTC-directed therapeutics. We examined whether lapatinib which binds both HER2 and EGFR could induce depletion of the EGFR-positive pool of CTCs, which may in turn lead to clinical benefits. PATIENTS AND METHODS Patients with metastatic breast cancer and HER2 non-amplified primary tumors with EGFR-positive CTCs were recruited and lapatinib 1500 mg daily was administered, in a standard two step phase 2 trial. RESULTS There were no responses leading to termination at the first analysis with 16 patients recruited out of 43 screened. In 6 out of 14 (43%) individuals eligible for the efficacy analysis, a decrease in CTCs was observed with most of these having a greater decrease in their EGFR-positive CTC pool. CONCLUSIONS This is one of the first studies of CTC-directed therapeutics and suggests that lapatinib monotherapy is not having any demonstrable clinical effects by reducing the EGFR-positive pool of CTCs in HER2 non-amplified primary tumors. Our attempt to expand the pool of patients eligible for a targeted therapy was unsuccessful; the role of clonal populations in cancer biology and therapeutic strategies to control them will require extensive evaluation in years to come. TRIAL REGISTRATION Clinical trials.gov NCT00820924.
Collapse
Affiliation(s)
- Justin Stebbing
- Department of Oncology, Imperial College Healthcare National Health Service Trust, Charing Cross Hospital, London, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Coombes RC, Tat T, Miller ML, Reise JA, Mansi JL, Hadjiminas DJ, Shousha S, Elsheikh SE, Lam EWF, Horimoto Y, El-Bahrawy M, Aboagye EO, Contractor KB, Shaw JA, Walker RA, Marconell MH, Palmieri C, Stebbing J. An open-label study of lapatinib in women with HER-2-negative early breast cancer: the lapatinib pre-surgical study (LPS study). Ann Oncol 2013; 24:924-30. [PMID: 23233650 DOI: 10.1093/annonc/mds594] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND This phase II, open-label, multicentre study aimed to evaluate changes in cell proliferation and biomarkers, as well as efficacy of lapatinib in treatment-naïve patients with HER-2-negative primary breast cancer. PATIENTS AND METHODS Patients received 1500 mg lapatinib for 28-42 days before surgery with repeat biopsies and measurements. The primary end point was inhibition of cell proliferation measured by Ki67; the secondary end points included clinical response, adverse events and changes in FOXO3a, FOXM1, p-AKT and HER-3. RESULTS Overall, there was no significant reduction in Ki67 with treatment (assessment carried out in 28 of 31 subjects enrolled). However, four patients (14%) showed a reduction in Ki67 ≥50%. Four of 25 patients (16%) had a partial response to treatment judged by sequential ultrasound measurements. Response, in terms of either Ki67 or ultrasound, did not relate to changes in any biomarker assessed at baseline, including the estrogen receptor (ER) and epidermal growth factor receptor (EGFR). However, all four clinical responders were HER-3 positive, as were three of four Ki67 responders. CONCLUSIONS Overall, a pre-surgical course of lapatinib monotherapy had little effect on this group of patients; however, in subsets of patients, especially those with HER-3-positive tumors, we observed either reduction in proliferation (Ki67) or tumor size; EGFR/ER status had no impact.
Collapse
Affiliation(s)
- R C Coombes
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, 1st Floor ICTEM, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lubet RA, Szabo E, Iwata KK, Gill SC, Tucker C, Bode A, Steele VE, Juliana MM, Nicastro HL, Grubbs CJ. Effect of intermittent dosing regimens of erlotinib on methylnitrosourea-induced mammary carcinogenesis. Cancer Prev Res (Phila) 2013; 6:448-54. [PMID: 23531447 DOI: 10.1158/1940-6207.capr-12-0322] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
EGF receptor (EGFR) inhibitors are used in the therapy of lung and pancreatic cancers and effectively prevent cancers in multiple animal models. Although daily dosing with erlotinib is effective, weekly dosing may reduce toxicity and have advantages, particularly for prevention. We tested alternative dosing regimens for preventive/therapeutic efficacy in a rat mammary cancer model. For prevention, erlotinib was administered by gavage beginning 5 days after methylnitrosourea (MNU). For therapy and biomarker studies, rats with palpable mammary cancers were treated for six weeks or for six days, respectively. Experiment A, erlotinib (6 mg/kg body weight/day, intragastric): daily (7 times/week); one day on/one day off; and two days on/two days off. All regimens decreased tumor incidence, increased tumor latency, and decreased cancer multiplicity versus controls (P < 0.01). However, intermittent dosing was less effective than daily dosing (P < 0.05). Experiment B, erlotinib (6 mg/kg body weight/day) daily or two days on/two days off or one time per week at 42 mg/kg body weight. All regimens reduced cancer incidence and multiplicity versus controls (P < 0.01). Interestingly, daily and weekly dosing were equally effective (P > 0.5). Experiment C, erlotinib administered at 42 or 21 mg/kg body weight 1 time per week, decreased tumor incidence and multiplicity (P < 0.01). Erlotinib had a serum half-life of ≤ 8 hours and weekly treatment yielded effective serum levels for ≤ 48 hours. Daily or weekly treatment of cancer bearing rats reduced mammary tumor size 25% to 35%, whereas control cancers increased >250%. Levels of phosphorylated extracellular signal-regulated kinase (ERK) were strongly decreased in rats treated daily/weekly with erlotinib. Thus, altering the dose of erlotinib retained most of its preventive and therapeutic efficacy, and based on prior clinical studies, is likely to reduce its toxicity.
Collapse
Affiliation(s)
- Ronald A Lubet
- National Cancer Institute, Executive Plaza North, Suite 2110, 6130 Executive Boulevard, Bethesda, MD 20852, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|