1
|
Hosonaga M, Habano E, Arakawa H, Kaneko K, Nakajima T, Hayashi N, Fukada I, Nakamura A, Haruyama Y, Maeda T, Inari H, Kobayashi T, Nakashima E, Ueno T, Takano T, Takahashi S, Ohno S, Ueki A. Case series of Li-Fraumeni syndrome: carcinogenic mechanisms in breast cancer with TP53 pathogenic variant carriers. Breast Cancer 2024; 31:988-996. [PMID: 39017822 PMCID: PMC11341599 DOI: 10.1007/s12282-024-01612-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 06/30/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND Li-Fraumeni syndrome (LFS), a hereditary condition attributed to TP53 pathogenic variants,(PV), is associated with high risks for various malignant tumors, including breast cancer. Notably, individuals harboring TP53 PVs are more likely (67-83%) to develop HER2 + breast cancer than noncarriers (16-25%). In this retrospective study, we evaluated the associations between TP53 variants and breast cancer phenotype. METHODS We conducted a retrospective review of the medical records of patients with LFS treated at a single institution and reviewed the literature on TP53 functions and the mechanisms underlying HER2 + breast cancer development in LFS. RESULTS We analyzed data for 10 patients with LFS from 8 families. The median age at the onset of the first tumor was 35.5 years. Only case 2 met the classic criteria; this patient harbored a nonsense variant, whereas the other patients carried missense variants. We observed that 9 of 10 patients developed breast cancer. Immunohistochemical analyses revealed that 40% of breast cancers in patients with LFS were HR - /HER2 + . The median age at the onset of breast cancer was slightly younger in HR - /HER2 + tumors than in HR + /HER2 - tumors (31 years and 35.5 years, respectively). CONCLUSIONS The occurrence of HER2 + breast cancer subtype was 40% in our LFS case series, which is greater than that in the general population (16-25%). Some TP53 PVs may facilitate HER2-derived oncogenesis in breast cancer. However, further studies with larger sample sizes are warranted to clarify the oncogenic mechanisms underlying each subtype of breast cancer in TP53 PV carriers.
Collapse
Affiliation(s)
- Mari Hosonaga
- Department of Breast Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan.
| | - Eri Habano
- Department of Clinical Genetic Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Hiromi Arakawa
- Department of Clinical Genetic Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Keika Kaneko
- Department of Clinical Genetic Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Takeshi Nakajima
- Medical Ethics and Medical Genetics, Kyoto University, Graduate of School of Medicine, School of Public Health, Kyoto, 606-8501, Japan
| | - Naomi Hayashi
- Division of Genomic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Ippei Fukada
- Department of Breast Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
- Division of Genomic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Akira Nakamura
- Department of Breast Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Yurie Haruyama
- Department of Breast Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Tetsuyo Maeda
- Department of Breast Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Hitoshi Inari
- Department of Breast Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Takayuki Kobayashi
- Department of Breast Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Eri Nakashima
- Department of Breast Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Takayuki Ueno
- Department of Breast Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Toshimi Takano
- Department of Breast Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Shunji Takahashi
- Division of Genomic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Shinji Ohno
- Department of Breast Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
- Department of Breast Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Arisa Ueki
- Department of Clinical Genetic Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| |
Collapse
|
2
|
Glen C, Morrow A, Roditi G, Hopkins T, Macpherson I, Stewart P, Petrie MC, Berry C, Epstein F, Lang NN, Mangion K. Cardiovascular sequelae of trastuzumab and anthracycline in long-term survivors of breast cancer. Heart 2024; 110:650-656. [PMID: 38103912 DOI: 10.1136/heartjnl-2023-323437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023] Open
Abstract
OBJECTIVES Long-term follow-up of patients treated with trastuzumab largely focuses on those with reduced left ventricular ejection fraction (LVEF) on treatment completion. This study sought to evaluate the prevalence of cardiovascular risk factors, overt cardiovascular disease and cardiac imaging abnormalities using cardiac magnetic resonance (CMR), in participants with normal LVEF on completion of trastuzumab±anthracycline therapy at least 5 years previously. METHODS Participants with human epidermal growth factor receptor 2-positive breast cancer treated with trastuzumab±anthracycline ≥5 years previously were identified from a clinical database. All participants had normal LVEF prior to, and on completion of, treatment. Participants underwent clinical cardiovascular evaluation, ECG, cardiac biomarker evaluation and CMR. Left ventricular systolic dysfunction (LVSD) was defined as LVEF <50%. RESULTS Forty participants were recruited between 15 March 2021 and 19 July 2022. Median time since completion of trastuzumab was 7.8 years (range 5.9-10.8 years) and 90% received prior anthracycline. 25% of participants had LVSD; median LVEF was 55.2% (Q1-Q3, 51.3-61.2). 30% of participants had N-terminal pro-B-type natriuretic peptide >125 pg/mL and 8% had high-sensitivity cardiac troponin T >14 ng/L. 33% of participants had a new finding of hypertension. 58% had total cholesterol >5.0 mmol/L, 43% had triglycerides >1.7 mmol/L and 5% had a new diagnosis of diabetes. CONCLUSIONS The presence of asymptomatic LVSD, abnormal cardiac biomarkers and cardiac risk factors in participants treated with trastuzumab and anthracycline at least 5 years previously is common, even in those with normal LVEF on completion of treatment. Our findings reinforce the relevance of comprehensive evaluation of cardiovascular risk factors following completion of cancer therapy, in addition to LVEF assessment.
Collapse
Affiliation(s)
- Claire Glen
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Andrew Morrow
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Giles Roditi
- Clinical Research Imaging Facility, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Tracey Hopkins
- Clinical Research Imaging Facility, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Iain Macpherson
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Philip Stewart
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mark C Petrie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
- Regional Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, UK
| | - Colin Berry
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
- Cardiology, Golden Jubilee National Hospital, Clydebank, UK
| | - Fred Epstein
- School of Engineering and Applied Science, University of Virginia, Charlottesville, Virginia, USA
| | - Ninian N Lang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Kenneth Mangion
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
- Regional Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, UK
| |
Collapse
|
3
|
Basu D, Pal R, Sarkar M, Barma S, Halder S, Roy H, Nandi S, Samadder A. To Investigate Growth Factor Receptor Targets and Generate Cancer Targeting Inhibitors. Curr Top Med Chem 2023; 23:2877-2972. [PMID: 38164722 DOI: 10.2174/0115680266261150231110053650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/20/2023] [Accepted: 10/02/2023] [Indexed: 01/03/2024]
Abstract
Receptor tyrosine kinase (RTK) regulates multiple pathways, including Mitogenactivated protein kinases (MAPKs), PI3/AKT, JAK/STAT pathway, etc. which has a significant role in the progression and metastasis of tumor. As RTK activation regulates numerous essential bodily processes, including cell proliferation and division, RTK dysregulation has been identified in many types of cancers. Targeting RTK is a significant challenge in cancer due to the abnormal upregulation and downregulation of RTK receptors subfamily EGFR, FGFR, PDGFR, VEGFR, and HGFR in the progression of cancer, which is governed by multiple RTK receptor signalling pathways and impacts treatment response and disease progression. In this review, an extensive focus has been carried out on the normal and abnormal signalling pathways of EGFR, FGFR, PDGFR, VEGFR, and HGFR and their association with cancer initiation and progression. These are explored as potential therapeutic cancer targets and therefore, the inhibitors were evaluated alone and merged with additional therapies in clinical trials aimed at combating global cancer.
Collapse
Affiliation(s)
- Debroop Basu
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Riya Pal
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, IndiaIndia
| | - Maitrayee Sarkar
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Soubhik Barma
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Sumit Halder
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Harekrishna Roy
- Nirmala College of Pharmacy, Vijayawada, Guntur, Andhra Pradesh, India
| | - Sisir Nandi
- Global Institute of Pharmaceutical Education and Research (Affiliated to Uttarakhand Technical University), Kashipur, 244713, India
| | - Asmita Samadder
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
- Cytogenetics and Molecular Biology Lab., Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| |
Collapse
|
4
|
El-Gamal MI, Mewafi NH, Abdelmotteleb NE, Emara MA, Tarazi H, Sbenati RM, Madkour MM, Zaraei SO, Shahin AI, Anbar HS. A Review of HER4 (ErbB4) Kinase, Its Impact on Cancer, and Its Inhibitors. Molecules 2021; 26:7376. [PMID: 34885957 PMCID: PMC8659013 DOI: 10.3390/molecules26237376] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 11/16/2022] Open
Abstract
HER4 is a receptor tyrosine kinase that is required for the evolution of normal body systems such as cardiovascular, nervous, and endocrine systems, especially the mammary glands. It is activated through ligand binding and activates MAPKs and PI3K/AKT pathways. HER4 is commonly expressed in many human tissues, both adult and fetal. It is important to understand the role of HER4 in the treatment of many disorders. Many studies were also conducted on the role of HER4 in tumors and its tumor suppressor function. Mostly, overexpression of HER4 kinase results in cancer development. In the present article, we reviewed the structure, location, ligands, physiological functions of HER4, and its relationship to different cancer types. HER4 inhibitors reported mainly from 2016 to the present were reviewed as well.
Collapse
Affiliation(s)
- Mohammed I. El-Gamal
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; (N.H.M.); (N.E.A.); (M.A.E.); (H.T.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.M.S.); (M.M.M.); (S.-O.Z.); (A.I.S.)
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Nada H. Mewafi
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; (N.H.M.); (N.E.A.); (M.A.E.); (H.T.)
| | - Nada E. Abdelmotteleb
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; (N.H.M.); (N.E.A.); (M.A.E.); (H.T.)
| | - Minnatullah A. Emara
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; (N.H.M.); (N.E.A.); (M.A.E.); (H.T.)
| | - Hamadeh Tarazi
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; (N.H.M.); (N.E.A.); (M.A.E.); (H.T.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.M.S.); (M.M.M.); (S.-O.Z.); (A.I.S.)
| | - Rawan M. Sbenati
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.M.S.); (M.M.M.); (S.-O.Z.); (A.I.S.)
| | - Moustafa M. Madkour
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.M.S.); (M.M.M.); (S.-O.Z.); (A.I.S.)
| | - Seyed-Omar Zaraei
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.M.S.); (M.M.M.); (S.-O.Z.); (A.I.S.)
| | - Afnan I. Shahin
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.M.S.); (M.M.M.); (S.-O.Z.); (A.I.S.)
| | - Hanan S. Anbar
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai 19099, United Arab Emirates
| |
Collapse
|
5
|
Kulukian A, Lee P, Taylor J, Rosler R, de Vries P, Watson D, Forero-Torres A, Peterson S. Preclinical Activity of HER2-Selective Tyrosine Kinase Inhibitor Tucatinib as a Single Agent or in Combination with Trastuzumab or Docetaxel in Solid Tumor Models. Mol Cancer Ther 2021; 19:976-987. [PMID: 32241871 DOI: 10.1158/1535-7163.mct-19-0873] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/26/2019] [Accepted: 02/18/2020] [Indexed: 11/16/2022]
Abstract
HER2 is a transmembrane tyrosine kinase receptor that mediates cell growth, differentiation, and survival. HER2 is overexpressed in approximately 20% of breast cancers and in subsets of gastric, colorectal, and esophageal cancers. Both antibody and small-molecule drugs that target HER2 and block its tyrosine kinase activity are effective in treating HER2-driven cancers. In this article, we describe the preclinical properties of tucatinib, an orally available, reversible HER2-targeted small-molecule tyrosine kinase inhibitor. In both biochemical and cell signaling experiments, tucatinib inhibits HER2 kinase activity with single-digit nanomolar potency and provides exceptional selectivity for HER2 compared with the related receptor tyrosine kinase EGFR, with a >1,000-fold enhancement in potency for HER2 in cell signaling assays. Tucatinib potently inhibits signal transduction downstream of HER2 and HER3 through the MAPK and PI3K/AKT pathways and is selectively cytotoxic in HER2-amplified breast cancer cell lines in vitro. In vivo, tucatinib is active in multiple HER2+ tumor models as a single agent and shows enhanced antitumor activity in combination with trastuzumab or docetaxel, resulting in improved rates of partial and complete tumor regression. These preclinical data, taken together with the phase-I tucatinib clinical trial results demonstrating preliminary safety and activity, establish the unique pharmacologic properties of tucatinib and underscore the rationale for investigating its utility in HER2+ cancers. GRAPHICAL ABSTRACT: http://mct.aacrjournals.org/content/molcanther/19/4/976/F1.large.jpg.
Collapse
|
6
|
Kong X, Zhang K, Wang X, Yang X, Li Y, Zhai J, Xing Z, Qi Y, Gao R, Feng X, Wang J, Fang Y. Mechanism of trastuzumab resistance caused by HER-2 mutation in breast carcinomas. Cancer Manag Res 2019; 11:5971-5982. [PMID: 31308740 PMCID: PMC6618040 DOI: 10.2147/cmar.s194137] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 04/23/2019] [Indexed: 11/23/2022] Open
Abstract
Background: Trastuzumab is an effective drug for the treatment of Her2-positive breast cancer. But, primary or secondary resistances to trastuzumab have become an important factor influencing the curative effect. The mechanisms of trastuzumab resistance are somewhat complex. The present work aims to explore the mechanism of trastuzumab resistance caused by HER-2 mutation in breast carcinomas. Methods: Firstly, the HER2 wild type (WT) and HER2 mutant (HER2 Q429R, HER2 Q429H and HER2 T798M are the commonest 3 types of mutations) MCF7 cell lines were established. Cell proliferation inhibition was then assessed by the Cell Counting Kit-8 assay and BrdU assay. Transwell invasion assays were also conducted to investigate the metastatic potential influenced by the HER2 mutation. Furthermore, Western blotting and co-immunoprecipitation were conducted to detect protein levels and the physical interaction of HER2 and trastuzumab. Results: The results showed that the mutant MCF7 cells were less sensitive to trastuzumab than the WTMCF7 cells. The mutation of HER2 almost had no influence on the expression of HER2 and the interaction of HER2 and trastuzumab. Finally, the mutation of HER2 weakened the inhibition of trastuzumab in the PI3K/AKT pathways. In addition, the inhibition of PI3K/AKT signaling-pathway increased the trastuzumab-sensitivity of HER2-mutant MCF7 cells. Conclusions: Dysregulation of the PI3K-AKT signaling-pathway was a key mechanism inducing the trastuzumab-resistance to HER2 mutant breast cancer cells.
Collapse
Affiliation(s)
- Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China.,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Harvard University, Boston, MA 02114-3117, USA
| | - Kai Zhang
- Department of Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Xiangyu Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China.,Department of Laboratory Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Xue Yang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Yalun Li
- Department of Breast Surgery, Yantai Yuhuangding Hospital, Yantai 264000, People's Republic of China
| | - Jie Zhai
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Zeyu Xing
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Yihang Qi
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Ran Gao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Xiaoli Feng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, People's Republic of China
| |
Collapse
|
7
|
Ong G, Brezden-Masley C, Dhir V, Deva DP, Chan KKW, Chow CM, Thavendiranathan D, Haq R, Barfett JJ, Petrella TM, Connelly KA, Yan AT. Myocardial strain imaging by cardiac magnetic resonance for detection of subclinical myocardial dysfunction in breast cancer patients receiving trastuzumab and chemotherapy. Int J Cardiol 2018; 261:228-233. [PMID: 29555336 DOI: 10.1016/j.ijcard.2018.03.041] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/19/2018] [Accepted: 03/09/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND Our objectives were to evaluate the temporal changes in CMR-based strain imaging, and examine their relationship with left ventricular ejection fraction (LVEF), in patients treated with trastuzumab. PATIENTS AND METHODS In this prospective longitudinal observational study, 41 women with HER2+ breast cancer treated with chemotherapy underwent serial CMR (baseline, 6, 12, and 18 months) after initiation of trastuzumab (treatment duration 12 months). LVEF and LV strain (global longitudinal[GLS] and circumferential[GCS]) measurements were independently measured by 2 blinded readers. RESULTS Of the 41 patients, 56% received anthracycline-based chemotherapy. Compared to baseline (60.4%, 95%CI 59.2-61.7%), there was a small but significant reduction in LVEF at 6 months (58.4%, 95%CI 56.7-60.0%, p = 0.034) and 12 months (57.9%, 95%CI 56.4-59.7%, p = 0.012), but not at 18 months (60.2%, 95%CI 58.2-62.2%, p = 0.93). Similarly, compared to baseline, GLS and GCS decreased significantly at 6 months (p = 0.024 and < 0.001, respectively) and 12 months (p = 0.002 and < 0.001, respectively) with an increase in LV end-diastolic volume, but not at 18 months. There were significant correlations between the temporal (6 month-baseline) changes in LVEF, and all global strain measurements (Pearson's r = -0.60 and r = -0.75 for GLS and GCS, respectively, all p < 0.001). CONCLUSION There was a significant reduction in LV strain during trastuzumab treatment, which correlated with a concurrent subtle decline in LVEF and was associated with an increase in LV end-diastolic volume. LV strain assessment by CMR may be a promising method to monitor for subclinical myocardial dysfunction in breast cancer patients receiving chemotherapy. Future studies are needed to determine its prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Geraldine Ong
- Terrence Donnelly Heart Centre, St. Michael's Hospital, Toronto, ON, Canada
| | - Christine Brezden-Masley
- Terrence Donnelly Heart Centre, St. Michael's Hospital, Toronto, ON, Canada; Division of Hematology/Oncology, St Michael's Hospital, Toronto, ON, Canada.
| | - Vinita Dhir
- Division of Hematology/Oncology, St Michael's Hospital, Toronto, ON, Canada.
| | - Djeven P Deva
- University of Toronto, Toronto, ON, Canada; Department of Medical Imaging, St. Michael's Hospital, Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada.
| | - Kelvin K W Chan
- University of Toronto, Toronto, ON, Canada; Sunnybrook Health Sciences Centre, University of Toronto, Cancer Care Ontario, Canadian Center for Applied Research in Cancer Control, Toronto, ON, Canada.
| | - Chi-Ming Chow
- Terrence Donnelly Heart Centre, St. Michael's Hospital, Toronto, ON, Canada; University of Toronto, Toronto, ON, Canada.
| | - Dinesh Thavendiranathan
- University of Toronto, Toronto, ON, Canada; Toronto General Hospital, University Health Network, Toronto, ON, Canada.
| | - Rashida Haq
- Division of Hematology/Oncology, St Michael's Hospital, Toronto, ON, Canada; University of Toronto, Toronto, ON, Canada.
| | - Joseph J Barfett
- University of Toronto, Toronto, ON, Canada; Department of Medical Imaging, St. Michael's Hospital, Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada.
| | - Teresa M Petrella
- University of Toronto, Toronto, ON, Canada; Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
| | - Kim A Connelly
- Terrence Donnelly Heart Centre, St. Michael's Hospital, Toronto, ON, Canada; University of Toronto, Toronto, ON, Canada; Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada.
| | - Andrew T Yan
- Terrence Donnelly Heart Centre, St. Michael's Hospital, Toronto, ON, Canada; University of Toronto, Toronto, ON, Canada; Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada.
| |
Collapse
|
8
|
Lu Q, Wang L, Zhang Y, Yu X, Wang C, Wang H, Yang Y, Chong X, Xia T, Meng Y, Wang Y, Lu C, Zhou L, Li B. An anti-ErbB2 fully human antibody circumvents trastuzumab resistance. Oncotarget 2018; 7:67129-67141. [PMID: 27564098 PMCID: PMC5341862 DOI: 10.18632/oncotarget.11562] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/11/2016] [Indexed: 02/04/2023] Open
Abstract
Trastuzumab, an anti-HER2/ErbB2 humanized antibody, has shown great clinical benefits in ErbB2-positive breast cancer treatment. Despite of its effectiveness, response rate to trastuzumab is limited and resistance is common. Here, we developed a new anti-ErbB2 antibody, denoted as H2-18, which was isolated from a phage display human antibody library. Previous studies have demonstrated that trastuzumab recognizes the juxtamembrane region of domain IV, and pertuzumab, another humanized ErbB2-specific antibody, binds to ErbB2 near the center of domain II. Our crystallographic analysis showed that the epitope recognized by H2-18 is within domain I of the ErbB2 molecule. H2-18 potently induced programmed cell death (PCD) in both trastuzumab-sensitive and -resistant breast cancer cell lines, while trastuzumab and pertuzumab, either used alone or in combination, only exhibits very weak PCD-inducing activity. More importantly, H2-18 could inhibit the growth of trastuzumab-resistant breast cancer cells far more effectively than trastuzumab plus pertuzumab, both in vitro and in vivo. In conclusion, H2-18 shows a unique ability to overcome trastuzumab resistance, suggesting that it has the great potential to be translated to the clinic.
Collapse
Affiliation(s)
- Qiong Lu
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Lingfei Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Yajun Zhang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Xiaojie Yu
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Chao Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Huajing Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Yang Yang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Xiaodan Chong
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Tian Xia
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| | - Yanchun Meng
- School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Yuxiao Wang
- Central Laboratory, Navy General Hospital, Beijing People's Republic of China
| | - Cuihua Lu
- Department of Gastroenterology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Lijun Zhou
- Central Laboratory, Navy General Hospital, Beijing People's Republic of China
| | - Bohua Li
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, People's Republic of China
| |
Collapse
|
9
|
Yu X, Wang L, Shen Y, Wang C, Zhang Y, Meng Y, Yang Y, Liang B, Zhou B, Wang H, Wei H, Lei C, Hu S, Li B. Targeting EGFR/HER2 heterodimerization with a novel anti-HER2 domain II/III antibody. Mol Immunol 2017; 87:300-307. [PMID: 28531814 DOI: 10.1016/j.molimm.2017.05.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 05/04/2017] [Accepted: 05/14/2017] [Indexed: 11/25/2022]
Abstract
HER2, a ligand-free tyrosine kinase receptor of the HER family, is frequently overexpressed in breast cancer. The anti-HER2 antibody trastuzumab has shown significant clinical benefits in metastatic breast cancer. Despite the effectiveness of trastuzumab, its efficacy remains variable and often modest. Thus, there is an urgent need to improve ErbB2-targeting therapy. Here, we describe a novel anti-HER2 antibody, 7C3, which was developed using hybridoma technique. Structural analysis confirms that the epitope of this antibody is in domain II/III of HER2. Moreover, a structural conformation change was observed in HER2 in complex with 7C3. Interestingly, this novel anti-HER2 antibody exhibits efficacy in blocking HER2/EGFR heterodimerization and signaling. The results highlight the different function role of HER2 domains and the unique potential of 7C3 to inhibit the HER2/EGFR heterodimer, which may complement current anti-HER2 treatments.
Collapse
Affiliation(s)
- Xiaojie Yu
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, People's Republic of China; International Joint Cancer Institute, the Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Lingfei Wang
- International Joint Cancer Institute, the Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Yafeng Shen
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, People's Republic of China
| | - Chao Wang
- International Joint Cancer Institute, the Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Yajun Zhang
- International Joint Cancer Institute, the Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Yanchun Meng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Yang Yang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, People's Republic of China; International Joint Cancer Institute, the Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Beibei Liang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, People's Republic of China
| | - Bo Zhou
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, People's Republic of China
| | - Huajing Wang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, People's Republic of China; International Joint Cancer Institute, the Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Huafeng Wei
- International Joint Cancer Institute, the Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Changhai Lei
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, People's Republic of China
| | - Shi Hu
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, People's Republic of China.
| | - Bohua Li
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, People's Republic of China; International Joint Cancer Institute, the Second Military Medical University, Shanghai 200433, People's Republic of China.
| |
Collapse
|
10
|
Wang L, Yu X, Wang C, Pan S, Liang B, Zhang Y, Chong X, Meng Y, Dong J, Zhao Y, Yang Y, Wang H, Gao J, Wei H, Zhao J, Wang H, Hu C, Xiao W, Li B. The anti-ErbB2 antibody H2-18 and the pan-PI3K inhibitor GDC-0941 effectively inhibit trastuzumab-resistant ErbB2-overexpressing breast cancer. Oncotarget 2017; 8:52877-52888. [PMID: 28881779 PMCID: PMC5581078 DOI: 10.18632/oncotarget.17907] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/05/2017] [Indexed: 11/25/2022] Open
Abstract
Trastuzumab, an anti-ErbB2 humanized antibody, brings benefit to patients with ErbB2-amplified metastatic breast cancers. However, the resistance to trastuzumab is common. Our previously reported H2-18, an anti-ErbB2 antibody, potently induced programmed cell death in trastuzumab-resistant breast cancer cells. Here, we aim to investigate the antitumor efficacy of H2-18 in combination with the pan-PI3K inhibitor GDC-0941 in trastuzumab-resistant breast cancer cell lines. The results showed that H2-18 and GDC-0941 synergistically inhibited the in vitro proliferation of BT-474, SKBR-3, HCC-1954 and HCC-1419 breast cancer cells. H2-18 plus GDC-0941 showed significantly enhanced programmed cell death-inducing activity compared with each drug used alone. The combination of H2-18 and GDC-0941 did not increase the effect of single agent on ROS production, cell cycle and ErbB2 signaling. Importantly, the in vivo antitumor efficacy of H2-18 plus GDC-0941 was superior to that of single agent. Thus, the enhanced in vivo antitumor efficacy of H2-18 plus GDC-0941 may mainly be attributable to its increased programmed cell death-inducing activity. Collectively, H2-18 plus GDC-0941 could effectively inhibit tumor growth, suggesting the potential to be translated into clinic as an efficient strategy for ErbB2-overexpressing breast cancers.
Collapse
Affiliation(s)
- Lingfei Wang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Xiaojie Yu
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Chao Wang
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Shujun Pan
- Hangzhou Sanatorium of People's Liberation Army, Hangzhou 310007, China
| | - Beibei Liang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Yajun Zhang
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Xiaodan Chong
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Yanchun Meng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Jian Dong
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Yirong Zhao
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Yang Yang
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Huajing Wang
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Jie Gao
- Department of Pharmaceutical Sciences, Second Military Medical University, Shanghai 200433, China
| | - Huafeng Wei
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Jian Zhao
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Hao Wang
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Chaohua Hu
- Department of General Surgery, Xiaogan Central Hospital Affiliated to Wuhan University of Science and Technology, Wuhan 432000, China
| | - Wenze Xiao
- Department of Rheumatology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Bohua Li
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
11
|
Shukla S, Myers JT, Woods SE, Gong X, Czapar AE, Commandeur U, Huang AY, Levine AD, Steinmetz NF. Plant viral nanoparticles-based HER2 vaccine: Immune response influenced by differential transport, localization and cellular interactions of particulate carriers. Biomaterials 2016; 121:15-27. [PMID: 28063980 DOI: 10.1016/j.biomaterials.2016.12.030] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/18/2016] [Accepted: 12/27/2016] [Indexed: 12/13/2022]
Abstract
Cancer vaccines are designed to elicit an endogenous adaptive immune response that can successfully recognize and eliminate residual or recurring tumors. Such approaches can potentially overcome shortcomings of passive immunotherapies by generating long-lived therapeutic effects and immune memory while limiting systemic toxicities. A critical determinant of vaccine efficacy is efficient transport and delivery of tumor-associated antigens to professional antigen presenting cells (APCs). Plant viral nanoparticles (VNPs) with natural tropism for APCs and a high payload carrying capacity may be particularly effective vaccine carriers. The applicability of VNP platform technologies is governed by stringent structure-function relationships. We compare two distinct VNP platforms: icosahedral cowpea mosaic virus (CPMV) and filamentous potato virus X (PVX). Specifically, we evaluate in vivo capabilities of engineered VNPs delivering human epidermal growth factor receptor 2 (HER2) epitopes for therapy and prophylaxis of HER2+ malignancies. Our results corroborate the structure-function relationship where icosahedral CPMV particles showed significantly enhanced lymph node transport and retention, and greater uptake by/activation of APCs compared to filamentous PVX particles. These enhanced immune cell interactions and transport properties resulted in elevated HER2-specific antibody titers raised by CPMV- vs. PVX-based peptide vaccine. The 'synthetic virology' field is rapidly expanding with numerous platforms undergoing development and preclinical testing; our studies highlight the need for systematic studies to define rules guiding the design and rational choice of platform, in the context of peptide-vaccine display technologies.
Collapse
Affiliation(s)
- Sourabh Shukla
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Jay T Myers
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sarah E Woods
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Xingjian Gong
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Anna E Czapar
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ulrich Commandeur
- Department of Molecular Biotechnology, RWTH-Aachen University, 52064 Aachen, Germany
| | - Alex Y Huang
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Alan D Levine
- Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nicole F Steinmetz
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Radiology, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Materials Science and Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Macromolecular Science and Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA; Division of General Medical Sciences-Oncology, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
12
|
Wang L, Yu X, Dong J, Meng Y, Yang Y, Wang H, Wang C, Zhang Y, Zhao Y, Zhao J, Wang H, Lu C, Li B. Combined SRC inhibitor saracatinib and anti-ErbB2 antibody H2-18 produces a synergistic antitumor effect on trastuzumab-resistant breast cancer. Biochem Biophys Res Commun 2016; 479:563-570. [PMID: 27666484 DOI: 10.1016/j.bbrc.2016.09.111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 09/21/2016] [Indexed: 10/21/2022]
Abstract
Despite of the effectiveness of the anti-ErbB2 humanized antibody trastuzumab, trastuzumab resistance emerges as a major and common clinical problem. Thus, circumventing trastuzumab resistance has become an urgent need. Recently, Src inhibitor saracatinib has drawn great attention for its key role in trastuzumab response. As shown in our previous study, H2-18, an anti-ErbB2 antibody, could potently induce programmed cell death (PCD) in trastuzumab-resistant breast cancer cells. Here we combined H2-18 and a Src inhibitor, saracatinib, and studied the antitumor activity of this drug combination in trastuzumab-resistant breast cancer cell lines. The results showed that H2-18 and saracatinib could synergistically inhibit cell proliferation of BT-474, SKBR-3, HCC-1954 and HCC-1419 breast cancer cell lines in vitro. H2-18 plus saracatinib could also inhibit the HCC-1954 tumor growth more effectively in vivo than each drug alone. H2-18 plus saracatinib showed a significantly more potent PCD-inducing activity compared with either H2-18 or saracatinib alone. We conclude that enhanced PCD may contribute to the superior antitumor efficacy of this combination therapy. The combination of H2-18 and SRC inhibitor has the potential to be translated into clinic.
Collapse
Affiliation(s)
- Lingfei Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Xiaojie Yu
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Jian Dong
- Department of Vascular Surgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Yanchun Meng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Yang Yang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Huajing Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Chao Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Yajun Zhang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Yirong Zhao
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Jian Zhao
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Hao Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China.
| | - Cuihua Lu
- Department of Gastroenterology, The Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, People's Republic of China.
| | - Bohua Li
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China.
| |
Collapse
|
13
|
Verma S, Goyal S, Jamal S, Singh A, Grover A. Hsp90: Friends, clients and natural foes. Biochimie 2016; 127:227-40. [DOI: 10.1016/j.biochi.2016.05.018] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/29/2016] [Indexed: 12/13/2022]
|
14
|
Bharti R, Dey G, Ojha PK, Rajput S, Jaganathan SK, Sen R, Mandal M. Diacerein-mediated inhibition of IL-6/IL-6R signaling induces apoptotic effects on breast cancer. Oncogene 2015; 35:3965-75. [PMID: 26616855 DOI: 10.1038/onc.2015.466] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 09/29/2015] [Accepted: 10/05/2015] [Indexed: 02/08/2023]
Abstract
Interleukin-6 (IL-6) signaling network has been implicated in oncogenic transformations making it attractive target for the discovery of novel cancer therapeutics. In this study, potent antiproliferative and apoptotic effect of diacerein were observed against breast cancer. In vitro apoptosis was induced by this drug in breast cancer cells as verified by increased sub-G1 population, LIVE/DEAD assay, cell cytotoxicity and presence of terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells, as well as downregulation of antiapoptotic proteins Bcl-2 and Bcl-xL and upregulation of apoptotic protein Bax. In addition, apoptosis induction was found to be caspase dependent. Further molecular investigations indicated that diacerein instigated apoptosis was associated with inhibition of IL-6/IL-6R autocrine signaling axis. Suppression of STAT3, MAPK and Akt pathways were also observed as a consequence of diacerein-mediated upstream inhibition of IL-6/IL-6R. Fluorescence study and western blot analysis revealed cytosolic accumulation of STAT3 in diacerein-treated cells. The docking study showed diacerein/IL-6R interaction that was further validated by competitive binding assay and isothermal titration calorimetry. Most interestingly, it was found that diacerein considerably suppressed tumor growth in MDA-MB-231 xenograft model. The in vivo antitumor effect was correlated with decreased proliferation (Ki-67), increased apoptosis (TUNEL) and inhibition of IL-6/IL-6R-mediated STAT3, MAPK and Akt pathway in tumor remnants. Taken together, diacerein offered a novel blueprint for cancer therapy by hampering IL-6/IL-6R/STAT3/MAPK/Akt network.
Collapse
Affiliation(s)
- R Bharti
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - G Dey
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - P K Ojha
- Drug Theoretics and Cheminformatics Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - S Rajput
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - S K Jaganathan
- IJN-UTM Cardiovascular Engineering Centre, Faculty of Biosciences and Medical Engineering, Universiti Teknologi, Malaysia
| | - R Sen
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - M Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
15
|
Price KAR, Cohen EEW. Mechanisms of and therapeutic approaches for overcoming resistance to epidermal growth factor receptor (EGFR)-targeted therapy in squamous cell carcinoma of the head and neck (SCCHN). Oral Oncol 2015; 51:399-408. [PMID: 25725588 DOI: 10.1016/j.oraloncology.2015.01.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 01/19/2015] [Accepted: 01/28/2015] [Indexed: 01/07/2023]
Abstract
The majority of squamous cell carcinoma of the head and neck (SCCHN) overexpress epidermal growth factor receptor (EGFR), which has been associated with poor treatment response and survival. However, only modest success has been achieved with the use of single agents that target EGFR, possibly due to primary and acquired resistance. This review will discuss key mechanisms of and therapeutic approaches to overcoming resistance to EGFR-targeted therapy in SCCHN. Recent preclinical and clinical investigations have demonstrated that other ErbB family receptors (eg, HER2 and HER3) and other horizontal resistance mechanisms, as well as activation of downstream signaling pathways, epigenetic events, and nuclear EGFR, are possible mediators of resistance to anti-EGFR therapeutics. Key downstream pathways that may be implicated in EGFR resistance include phosphatidylinositol-3-kinase/protein kinase B, vascular endothelial growth factor (VEGF), and mammalian target of rapamycin (mTOR). Multiple agents that target EGFR and other ErbB family members (ie, lapatinib, afatinib, and dacomitinib), as well as combination therapies that target EGFR and selected other pathways (eg, VEGF, mTOR, and c-Met) are being investigated clinically. In addition, several phase II and III trials continue to investigate strategies to enhance the efficacy of EGFR-targeted therapy in SCCHN.
Collapse
Affiliation(s)
| | - Ezra E W Cohen
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
16
|
Yallowitz AR, Li D, Lobko A, Mott D, Nemajerova A, Marchenko N. Mutant p53 Amplifies Epidermal Growth Factor Receptor Family Signaling to Promote Mammary Tumorigenesis. Mol Cancer Res 2015; 13:743-54. [PMID: 25573952 DOI: 10.1158/1541-7786.mcr-14-0360] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 12/17/2014] [Indexed: 12/14/2022]
Abstract
UNLABELLED The EGFR family (ErbB2/Her2 and EGFR/ErbB1/Her1) often modulates the transcriptional program involved in promoting mammary tumorigenesis. In humans, the majority of ErbB2-positive sporadic breast cancers harbor p53 mutations, which correlate with poor prognosis. Also, the extremely high incidence of ErbB2-positive breast cancer in women with p53 germline mutations (Li-Fraumeni syndrome) suggests a key role of mutant p53 specifically in ErbB2-mediated mammary tumorigenesis. To examine the role of mutant p53 during ErbB2-mediated mammary tumorigenesis, a mutant p53 allele (R172H) was introduced into the (MMTV)-ErbB2/Neu mouse model system. Interestingly, we show in heterozygous p53 mice that mutant p53 R172H is a more potent activator of ErbB2-mediated mammary tumorigenesis than simple loss of p53. The more aggressive disease in mutant p53 animals was reflected by earlier tumor onset, increased mammary tumor multiplicity, and shorter survival. These in vivo and in vitro data provide mechanistic evidence that mutant p53 amplifies ErbB2 and EGFR signaling to promote the expansion of mammary stem cells and induce cell proliferation. IMPLICATIONS This study identifies mutant p53 as an essential player in ErbB2 and EGFR-mediated mammary tumorigenesis and indicates the potential translational importance of targeting mutant p53 in this subset of patients with breast cancer.
Collapse
Affiliation(s)
- Alisha R Yallowitz
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, New York
| | - Dun Li
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, New York
| | - Anthony Lobko
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, New York
| | - Daniel Mott
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, New York
| | - Alice Nemajerova
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, New York
| | - Natalia Marchenko
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, New York.
| |
Collapse
|
17
|
Wang HM, Xu YF, Ning SL, Yang DX, Li Y, Du YJ, Yang F, Zhang Y, Liang N, Yao W, Zhang LL, Gu LC, Gao CJ, Pang Q, Chen YX, Xiao KH, Ma R, Yu X, Sun JP. The catalytic region and PEST domain of PTPN18 distinctly regulate the HER2 phosphorylation and ubiquitination barcodes. Cell Res 2014; 24:1067-90. [PMID: 25081058 PMCID: PMC4152746 DOI: 10.1038/cr.2014.99] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 04/27/2014] [Accepted: 05/26/2014] [Indexed: 12/23/2022] Open
Abstract
The tyrosine phosphorylation barcode encoded in C-terminus of HER2 and its ubiquitination regulate diverse HER2 functions. PTPN18 was reported as a HER2 phosphatase; however, the exact mechanism by which it defines HER2 signaling is not fully understood. Here, we demonstrate that PTPN18 regulates HER2-mediated cellular functions through defining both its phosphorylation and ubiquitination barcodes. Enzymologic characterization and three crystal structures of PTPN18 in complex with HER2 phospho-peptides revealed the molecular basis for the recognition between PTPN18 and specific HER2 phosphorylation sites, which assumes two distinct conformations. Unique structural properties of PTPN18 contribute to the regulation of sub-cellular phosphorylation networks downstream of HER2, which are required for inhibition of HER2-mediated cell growth and migration. Whereas the catalytic domain of PTPN18 blocks lysosomal routing and delays the degradation of HER2 by dephosphorylation of HER2 on pY(1112), the PEST domain of PTPN18 promotes K48-linked HER2 ubiquitination and its rapid destruction via the proteasome pathway and an HER2 negative feedback loop. In agreement with the negative regulatory role of PTPN18 in HER2 signaling, the HER2/PTPN18 ratio was correlated with breast cancer stage. Taken together, our study presents a structural basis for selective HER2 dephosphorylation, a previously uncharacterized mechanism for HER2 degradation and a novel function for the PTPN18 PEST domain. The new regulatory role of the PEST domain in the ubiquitination pathway will broaden our understanding of the functions of other important PEST domain-containing phosphatases, such as LYP and PTPN12.
Collapse
Affiliation(s)
- Hong-Mei Wang
- 1] Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong 250012, China [2] Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Yun-Fei Xu
- 1] Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong 250012, China [2] Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Shang-Lei Ning
- Qilu Hospital, Shandong University, Jinan, Shandong 250012, China
| | - Du-Xiao Yang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Yi Li
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Yu-Jie Du
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Fan Yang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Ya Zhang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Nan Liang
- 1] Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong 250012, China [2] Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Wei Yao
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Ling-Li Zhang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Li-Chuan Gu
- Shandong University, School of Life Science, Jinan, Shandong 250012, China
| | - Cheng-Jiang Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Qi Pang
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Yu-Xin Chen
- Qilu Hospital, Shandong University, Jinan, Shandong 250012, China
| | - Kun-Hong Xiao
- Duke University, School of Medicine, Durham, 27705, USA
| | - Rong Ma
- Qilu Hospital, Shandong University, Jinan, Shandong 250012, China
| | - Xiao Yu
- 1] Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong 250012, China [2] Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China [3] Qilu Hospital, Shandong University, Jinan, Shandong 250012, China
| | - Jin-Peng Sun
- 1] Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong 250012, China [2] Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| |
Collapse
|
18
|
Abstract
Hsp90 is a major molecular chaperone that is expressed abundantly and plays a pivotal role in assisting correct folding and functionality of its client proteins in cells. The Hsp90 client proteins include a wide variety of signal transducing molecules such as protein kinases and steroid hormone receptors. Cancer is a complex disease, but most types of human cancer share common hallmarks, including self-sufficiency in growth signals, insensitivity to growth-inhibitory mechanism, evasion of programmed cell death, limitless replicative potential, sustained angiogenesis, and tissue invasion and metastasis. A surprisingly large number of Hsp90-client proteins play crucial roles in establishing cancer cell hallmarks. We start the review by describing the structure and function of Hsp90 since conformational changes during the ATPase cycle of Hsp90 are closely related to its function. Many co-chaperones, including Hop, p23, Cdc37, Aha1, and PP5, work together with Hsp90 by modulating the chaperone machinery. Post-translational modifications of Hsp90 and its cochaperones are vital for their function. Many tumor-related Hsp90-client proteins, including signaling kinases, steroid hormone receptors, p53, and telomerase, are described. Hsp90 and its co-chaperones are required for the function of these tumor-promoting client proteins; therefore, inhibition of Hsp90 by specific inhibitors such as geldanamycin and its derivatives attenuates the tumor progression. Hsp90 inhibitors can be potential and effective cancer chemotherapeutic drugs with a unique profile and have been examined in clinical trials. We describe possible mechanisms why Hsp90 inhibitors show selectivity to cancer cells even though Hsp90 is essential also for normal cells. Finally, we discuss the "Hsp90-addiction" of cancer cells, and suggest a role for Hsp90 in tumor evolution.
Collapse
Affiliation(s)
- Yoshihiko Miyata
- Department of Cell & Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan.
| | | | | |
Collapse
|
19
|
Miyata Y, Nakamoto H, Neckers L. The therapeutic target Hsp90 and cancer hallmarks. Curr Pharm Des 2013; 19:347-65. [PMID: 22920906 DOI: 10.2174/138161213804143725] [Citation(s) in RCA: 244] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 08/15/2012] [Indexed: 01/22/2023]
Abstract
Hsp90 is a major molecular chaperone that is expressed abundantly and plays a pivotal role in assisting correct folding and functionality of its client proteins in cells. The Hsp90 client proteins include a wide variety of signal transducing molecules such as protein kinases and steroid hormone receptors. Cancer is a complex disease, but most types of human cancer share common hallmarks, including self-sufficiency in growth signals, insensitivity to growth-inhibitory mechanism, evasion of programmed cell death, limitless replicative potential, sustained angiogenesis, and tissue invasion and metastasis. A surprisingly large number of Hsp90-client proteins play crucial roles in establishing cancer cell hallmarks. We start the review by describing the structure and function of Hsp90 since conformational changes during the ATPase cycle of Hsp90 are closely related to its function. Many co-chaperones, including Hop, p23, Cdc37, Aha1, and PP5, work together with Hsp90 by modulating the chaperone machinery. Post-translational modifications of Hsp90 and its cochaperones are vital for their function. Many tumor-related Hsp90-client proteins, including signaling kinases, steroid hormone receptors, p53, and telomerase, are described. Hsp90 and its co-chaperones are required for the function of these tumor-promoting client proteins; therefore, inhibition of Hsp90 by specific inhibitors such as geldanamycin and its derivatives attenuates the tumor progression. Hsp90 inhibitors can be potential and effective cancer chemotherapeutic drugs with a unique profile and have been examined in clinical trials. We describe possible mechanisms why Hsp90 inhibitors show selectivity to cancer cells even though Hsp90 is essential also for normal cells. Finally, we discuss the "Hsp90-addiction" of cancer cells, and suggest a role for Hsp90 in tumor evolution.
Collapse
Affiliation(s)
- Yoshihiko Miyata
- Department of Cell & Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan.
| | | | | |
Collapse
|
20
|
Tumor protein p53 (TP53) testing and Li-Fraumeni syndrome : current status of clinical applications and future directions. Mol Diagn Ther 2013; 17:31-47. [PMID: 23355100 DOI: 10.1007/s40291-013-0020-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Prevalent as an acquired abnormality in cancer, the role of tumor protein p53 (TP53) as a germline mutation continues to evolve. The clinical impact of a germline TP53 mutation is often dramatic and affects the full life course, with a propensity to develop rare tumors in childhood and multiple common cancers of unexpectedly early onset in adulthood. In this article, we review the clinical relevance of germline mutations in the TP53 tumor suppressor gene to current healthcare practice, including the optimal ways to identify patients with Li-Fraumeni syndrome (LFS), to recognize the core cancers associated with LFS, and to develop strategies for early detection of LFS-associated tumors. Several TP53-targeted approaches to improve outcomes in LFS patients are also reviewed. A case report is used to highlight special TP53 testing dilemmas and unique challenges associated with genetic testing decisions in the current age of rapidly advancing genomic technologies.
Collapse
|
21
|
Robinson WH, Lindstrom TM, Cheung RK, Sokolove J. Mechanistic biomarkers for clinical decision making in rheumatic diseases. Nat Rev Rheumatol 2013; 9:267-76. [PMID: 23419428 DOI: 10.1038/nrrheum.2013.14] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The use of biomarkers is becoming increasingly intrinsic to the practice of medicine and holds great promise for transforming the practice of rheumatology. Biomarkers have the potential to aid clinical diagnosis when symptoms are present or to provide a means of detecting early signs of disease when they are not. Some biomarkers can serve as early surrogates of eventual clinical outcomes or guide therapeutic decision making by enabling identification of individuals likely to respond to a specific therapy. Using biomarkers might reduce the costs of drug development by enabling individuals most likely to respond to be enrolled in clinical trials, thereby minimizing the number of participants required. In this Review, we discuss the current use and the potential of biomarkers in rheumatology and in select fields at the forefront of biomarker research. We emphasize the value of different types of biomarkers, addressing the concept of 'actionable' biomarkers, which can be used to guide clinical decision making, and 'mechanistic' biomarkers, a subtype of actionable biomarker that is embedded in disease pathogenesis and, therefore, represents a potentially superior biomarker. We provide examples of actionable and mechanistic biomarkers currently available, and discuss how development of such biomarkers could revolutionize clinical practice and drug development.
Collapse
Affiliation(s)
- William H Robinson
- VA Palo Alto Health Care System, 3801 Miranda Ave, Palo Alto, CA 94304, USA.
| | | | | | | |
Collapse
|
22
|
Current World Literature. Curr Opin Obstet Gynecol 2013; 25:81-9. [DOI: 10.1097/gco.0b013e32835cc6b6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
23
|
El-Sayed MI, Maximous DW, Zakhary MM, Mikhail NNH. Biological markers and response to neoadjuvant taxane-based chemotherapy in patients with locally advanced breast cancer. ISRN ONCOLOGY 2013; 2012:245891. [PMID: 23316390 PMCID: PMC3536061 DOI: 10.5402/2012/245891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 10/24/2012] [Indexed: 01/25/2023]
Abstract
Introduction. Biological markers as Her2/neu, p53, and hormonal receptors (HmRs) may be reliable parameters for prognostic assessment of patients of locally advanced breast cancer (LABC). This work aims at assessing the potential value of these biological markers for the prediction of disease outcome after neoadjuvant taxane-based chemotherapy and its implication on the surgical role. Patients and Methods. From March 2006 to September 2011, 95 patients with LABC were treated by neoadjuvant taxane-based chemotherapy given at intervals of 3 weeks. Expression of Her2/neu and p53 was examined in the initial tissue biopsy by using ELISA technique. Status of HmRs was determined using a commercial enzyme immunoassay. Three weeks after the third cycle, patients underwent surgical resection followed by 3 more cycles of taxane-based chemotherapy and radiotherapy as an adjuvant therapy. Relations of Her2/neu overexpression to p53, HmRs, and conventional prognostic factors were analyzed. Results. Median followup was 61 months. The 5-year DFS and OAS rates were significantly higher in patients with positive HmRs than in those with negative HmRs, patients with Her2- than those with Her2+ breast cancer, and patients with intact p53 breast cancer than those with inactive p53. HER-2 overexpression was statistically significant associated with loss of HmR positive immunostaining (P < 0.0001), grade III breast cancer (P < 0.0001), advanced nodal status (P = 0.0039), and younger (<50 years) age (P = 0.0108). Conclusion. Her2/neu overexpression was associated with poor DFS and OAS rates, as it was significantly associated with negative HmR and high grade.
Collapse
Affiliation(s)
- Mohamed I El-Sayed
- Department of Radiation Oncology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | | | | | | |
Collapse
|
24
|
Update on clinical trials: genetic targets in breast cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 779:35-54. [PMID: 23288634 DOI: 10.1007/978-1-4614-6176-0_2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Breast cancer is the most commonly diagnosed cancer in women in United States. From data of American Cancer Society from 2007 reported total of 178,480 women diagnosed with breast cancer. The death rate from breast cancer has decreased in North America over time, but still accounts for second highest cancer death, following lung cancer. Breast cancer is staged based on tumor size, nodal involvement, and distant metastasis like any other solid tumors. However clinical staging is not the only important factor in management of breast cancer. Various molecular features divides breast cancer into many subgroups - that act differently, and respond differently from therapy. Thus the focus of breast cancer treatment has evolved focusing on specific targets. The most important biologic markers in subtyping of breast cancer so far are hormone receptor positivity and HER2/neu protein expression. Five molecular subtypes using intrinsic gene set include Basal mRNA, HER2 + mRNA, Luminal AmRNA, Luminal B mRNA, and Normal-like mRNA. In addition, better understanding of genetic target of breast cancer has given us arsenal of personalized, and more effective treatment approach.This review will focus on examples that highlight several mechanism of tumorigenesis, giving us not just understanding of gene pathways and the molecular biology, that could lead us to therapeutic target. Several important molecular targets have been investigated in preclinical and clinical trials, others are yet to be explored. We will also describe genetic mechanisms discovery related to overcoming resistance to current targeted therapies in breast cancer, including hormone receptor expression and HER 2- neu amplification. We will also review other exciting developments in understanding of breast cancer, the tumor microenvironment and cancer stem cells, and targeting agents in that area.
Collapse
|
25
|
Ramsey DM, McAlpine SR. Halting metastasis through CXCR4 inhibition. Bioorg Med Chem Lett 2012; 23:20-5. [PMID: 23211868 DOI: 10.1016/j.bmcl.2012.10.138] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 10/31/2012] [Indexed: 12/14/2022]
Abstract
Metastasis occurs when cancer cells leave the primary tumor site and migrate to distant parts of the body. The CXCR4-SDF-1 pathway facilitates this migration, and its expression has become the hallmark of several metastatic cancers. Targeted approaches are currently being developed to inhibit this pathway, and several candidates are now in clinical trials. Continued exploration of CXCR4 inhibitors will generate compounds that have improved activity over current candidates.
Collapse
Affiliation(s)
- Deborah M Ramsey
- Department of Chemistry, University of New South Wales, Sydney NSW 2052, Australia.
| | | |
Collapse
|
26
|
Marzaro G, Guiotto A, Chilin A. Quinazoline derivatives as potential anticancer agents: a patent review (2007 - 2010). Expert Opin Ther Pat 2012; 22:223-52. [PMID: 22404097 DOI: 10.1517/13543776.2012.665876] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Due to the increase in knowledge about cancer pathways, there is a growing interest in finding novel potential drugs. Quinazoline is one of the most widespread scaffolds amongst bioactive compounds. A number of patents and papers appear in the literature regarding the discovery and development of novel promising quinazoline compounds for cancer chemotherapy. Although there is a progressive decrease in the number of patents filed, there is an increasing number of biochemical targets for quinazoline compounds. AREAS COVERED This paper provides a comprehensive review of the quinazolines patented in 2007 - 2010 as potential anticancer agents. Information from articles published in international peer-reviewed journals was also included, to give a more exhaustive overview. EXPERT OPINION From about 1995 to 2006, the anticancer quinazolines panorama has been dominated by the 4-anilinoquinazolines as tyrosine kinase inhibitors. The extensive researches conducted in this period could have caused the progressive reduction in the ability to file novel patents as shown in the 2007 - 2010 period. However, the growing knowledge of cancer-related pathways has recently highlighted some novel potential targets for therapy, with quinazolines receiving increasing attention. This is well demonstrated by the number of different targets of the patents considered in this review. The structural heterogeneity in the patented compounds makes it difficult to derive general pharmacophores and make comparisons among claimed compounds. On the other hand, the identification of multi-target compounds seems a reliable goal. Thus, it is reasonable that quinazoline compounds will be studied and developed for multi-target therapies.
Collapse
Affiliation(s)
- Giovanni Marzaro
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Padova, via Marzolo 5, 35131 Padova, Italy.
| | | | | |
Collapse
|
27
|
Nanocarriers as Nanomedicines. NANOBIOTECHNOLOGY - INORGANIC NANOPARTICLES VS ORGANIC NANOPARTICLES 2012. [DOI: 10.1016/b978-0-12-415769-9.00014-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|