1
|
Darmadi D, Aminov Z, Hjazi A, R R, Kazmi SW, Mustafa YF, Hosseen B, Sharma A, Alubiady MHS, Al-Abdeen SHZ. Investigation of the regulation of EGF signaling by miRNAs, delving into the underlying mechanism and signaling pathways in cancer. Exp Cell Res 2024; 442:114267. [PMID: 39313176 DOI: 10.1016/j.yexcr.2024.114267] [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: 07/20/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 09/25/2024]
Abstract
The EGF receptors (EGFRs) signaling pathway is essential for tumorigenesis and progression of cancer. Emerging evidence suggests that miRNAs are essential regulators of EGF signaling, influencing various pathway components and tumor behavior. This article discusses the underlying mechanisms and clinical implications of miRNA-mediated regulation of EGF signaling in cancer. miRNAs utilize multiple mechanisms to exert their regulatory effects on EGF signaling. They can target EGF ligands, including EGF and TGF-directly, inhibiting their expression and secretion. In addition, miRNAs can modulate EGF signaling indirectly by targeting EGF receptors, downstream signaling molecules, and transcription factors implicated in regulating the EGF pathway. These miRNAs can disrupt the delicate equilibrium of EGF signaling, resulting in aberrant activation and fostering tumor cell proliferation, survival, angiogenesis, and metastasis. The dysregulation of the expression of specific miRNAs has been linked to clinical outcomes in numerous types of cancer. Specific profiles of miRNA expression have been identified as prognostic markers, reflecting tumor characteristics, invasiveness, metastatic potential, and therapeutic response. These miRNAs can serve as potential therapeutic targets for interventions that modulate EGF signaling and improve patient outcomes. Understanding the intricate relationship between miRNAs and EGF signaling in cancer can transform cancer diagnosis, prognosis, and treatment. The identification of specific miRNAs involved in the regulation of the EGF pathway opens the door to the development of targeted therapies and personalized medicine approaches. In addition, miRNA-based interventions promise to overcome therapeutic resistance and improve the efficacy of existing treatments. miRNAs are crucial regulators of EGF signaling in cancer, affecting tumor behavior and clinical outcomes. Further research is required to decipher the complex network of miRNA-mediated EGF signaling regulation and translate these findings into clinically applicable strategies for enhanced cancer treatment.
Collapse
Affiliation(s)
- Darmadi Darmadi
- Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia.
| | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan.
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia.
| | - Roopashree R
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Syeda Wajida Kazmi
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, 140307, Punjab, India.
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq.
| | - Beneen Hosseen
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq; Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq.
| | - Abhishek Sharma
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India.
| | | | | |
Collapse
|
2
|
Trembath HE, Yeh JJ, Lopez NE. Gastrointestinal Malignancy: Genetic Implications to Clinical Applications. Cancer Treat Res 2024; 192:305-418. [PMID: 39212927 DOI: 10.1007/978-3-031-61238-1_15] [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: 09/04/2024]
Abstract
Advances in molecular genetics have revolutionized our understanding of the pathogenesis, progression, and therapeutic options for treating gastrointestinal (GI) cancers. This chapter provides a comprehensive overview of the molecular landscape of GI cancers, focusing on key genetic alterations implicated in tumorigenesis across various anatomical sites including GIST, colon and rectum, and pancreas. Emphasis is placed on critical oncogenic pathways, such as mutations in tumor suppressor genes, oncogenes, chromosomal instability, microsatellite instability, and epigenetic modifications. The role of molecular biomarkers in predicting prognosis, guiding treatment decisions, and monitoring therapeutic response is discussed, highlighting the integration of genomic profiling into clinical practice. Finally, we address the evolving landscape of precision oncology in GI cancers, considering targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Hannah E Trembath
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA
| | - Jen Jen Yeh
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA
| | - Nicole E Lopez
- Division of Colon and Rectal Surgery, Department of Surgery, University of California San Diego, 4303 La Jolla Village Drive Suite 2110, San Diego, CA, 92122, USA.
- Division of Surgical Oncology, Department of Surgery, University of North Carolina, 170 Manning Drive, CB#7213, 1150 Physician's Office Building, Chapel Hill, NC, 27599-7213, USA.
| |
Collapse
|
3
|
Unadkat V, Rohit S, Parikh P, Sanna V, Singh S. Rational design-aided discovery of novel 1,2,4-oxadiazole derivatives as potential EGFR inhibitors. Bioorg Chem 2021; 114:105124. [PMID: 34328857 DOI: 10.1016/j.bioorg.2021.105124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022]
Abstract
A molecular dynamics-based sampling of epidermal growth factor receptor tyrosine kinase (EGFR-TK) was carried out to search for energetically more stable protein, which was then used for molecular docking of a series of 1,2,4-oxadiazole derivatives previously reported from our laboratory. A total of 14 compounds were docked, where compounds 6a and 6b showed better binding to EGFR in silico. Further, physicochemical properties of all the compounds were calculated, which suggested that all the molecules obeyed Lipinski's rule of 5 and had favorable polar surface area and CaCO2 permeability along with the low potential for HERG inhibition. All the compounds were then screened for their ability to produce cytotoxicity in four different cell lines overexpressing EGFR (A549, HCT-116, HEPG2, MCF-7) and one EGFR negative cancer cell line (SW620); at three concentrations: 10, 1, and 0.1 µM. None of the compounds showed activity against SW620, which suggested that the compounds show cytotoxicity through inhibition of EGFR. Compounds that showed promise in this 3-concentration screen were further subjected to multiple dose-response curves to identify the IC50 values for the shortlisted eight compounds. It was encouraging to see 6a and 6b showing the best IC50 values against almost all the cell-lines which further suggests that our design protocol can be applied to optimize this lead (which are currently in the low micromolar range) to design the homologous compounds to achieve the desired potency in the nanomolar range and also to achieve selectivity across a range of kinases.
Collapse
Affiliation(s)
- Vishal Unadkat
- Kashiv Biosciences Pvt Ltd, 27-2 & 43 Building Block B Paiki, Mauje Sarkhej, Opp Applewoods Township, Sarkhej, Ahmedabad 382210, Gujarat, India.
| | - Shishir Rohit
- Kashiv Biosciences Pvt Ltd, 27-2 & 43 Building Block B Paiki, Mauje Sarkhej, Opp Applewoods Township, Sarkhej, Ahmedabad 382210, Gujarat, India
| | - Paranjay Parikh
- Piramal Pharma Solutions, Plot 18, PHARMEZ, Matoda, Sarkhej-Bavla NH 8A, Taluka Sanand, Ahmedabad 382213, Gujarat, India
| | - Vinod Sanna
- Piramal Pharma Solutions, Plot 18, PHARMEZ, Matoda, Sarkhej-Bavla NH 8A, Taluka Sanand, Ahmedabad 382213, Gujarat, India
| | - Sanjay Singh
- Division of Biological & Life Sciences (Formerly Institute of Life Sciences), School of Arts & Sciences, Ahmedabad University, Navaragnpura, Ahmedabad 380009, Gujarat, India; National Institute of Animal Biotechnology, Near Gowlidoddy, Extended Q City Road, Gachibowli, Hyderabad 500032, Telangana, India
| |
Collapse
|
4
|
Yang F, Yang YH, Zeng WH. The Inhibition of Cell Growth Through the EGFR/ERK/MMP-2 Pathway Induced by Ampelopsin in the Human Malignant Melanoma A375 Cell Line. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20912864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Malignant melanoma is one of the most aggressive skin cancers, having a very high mortality rate. However, its effective treatment is not clear. Ampelopsin, a plant flavonoid, has been reported to inhibit cell growth and/or induce apoptosis in various types of tumor. In this study, it was shown that ampelopsin significantly inhibits melanoma A375 cell line proliferation in a concentration-dependent/time-dependent manner. The flow cytometric data clearly demonstrated that ampelopsin causes cell cycle arrest in the G2/M phase. Moreover, it also confirmed that growth inhibition mediated by treatment with ampelopsin is related to the decreased expression of Cdc2, Cdc25c, cyclin B1, and activation of caspase-3 and Bax, purportedly by epidermal growth factor receptor (EGFR), extracellular regulated protein kinases, and matrix metalloproteinase-2 (MMP-2) downregulation. As a result of this work, these findings suggest that ampelopsin inhibits human malignant melanoma A375 cell line proliferation by suppressing the EGFR/ERK/MMP-2 pathway.
Collapse
Affiliation(s)
- Fan Yang
- Department of Plastic Surgery and Burns, Affiliated Taihe Hospital of Xi’an Jiaotong University Health Science Center, Shiyan, China
| | - Yin-hui Yang
- Department of Plastic Surgery and Burns, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Wei-hui Zeng
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
5
|
Lynce F, Wang H, Petricoin EF, Pohlmann PR, Smaglo B, Hwang J, He AR, Subramaniam DS, Deeken J, Marshall J, Pishvaian MJ. A phase I study of HER1, HER2 dual kinase inhibitor lapatinib plus the proteasome inhibitor bortezomib in patients with advanced malignancies. Cancer Chemother Pharmacol 2019; 84:1145-1151. [PMID: 31538230 DOI: 10.1007/s00280-019-03947-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 08/28/2019] [Indexed: 11/29/2022]
Abstract
PURPOSE This phase I trial evaluated the maximum tolerated dose, safety and preliminary efficacy of lapatinib, a HER1, HER2 dual kinase inhibitor plus bortezomib, a proteasome inhibitor, in adult patients with advanced malignancies. METHODS Patients were enrolled in a standard 3 + 3 design with lapatinib (L) 750, 1000, 1250 or 1500 mg daily, and bortezomib (B) 0.7, 1.0, 1.3 or 1.6 mg/m2 for 3 weeks with 1 week off. Dose-limiting toxicities (DLT) were assessed during the first 28 days RESULTS: Fifteen patients received the combination of lapatinib and bortezomib in three different cohorts and ten were evaluable for DLT. There were no DLTs. Anorexia was the most common adverse event. Biomarker analysis showed upregulation of p27 expression with lapatinib and the combination. No tumor response was observed and thus the study was closed early. CONCLUSION The combination of lapatinib and bortezomib was well tolerated but no complete or partial tumor responses were observed at the dose levels tested. CLINICALTRIALS. GOV IDENTIFIER NCT01497626.
Collapse
Affiliation(s)
- Filipa Lynce
- Lombardi Comprehensive Cancer Center, 3800 Reservoir Road NW, Washington, DC, 20007, USA.,Georgetown University Medical Center, Washington, DC, USA
| | - Hongkun Wang
- Department of Biostatistics, Bioinformatics, and Biomathematics, Georgetown University, Washington, DC, USA
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Paula R Pohlmann
- Lombardi Comprehensive Cancer Center, 3800 Reservoir Road NW, Washington, DC, 20007, USA.,Georgetown University Medical Center, Washington, DC, USA
| | - Brandon Smaglo
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jimmy Hwang
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC, USA
| | - Aiwu R He
- Lombardi Comprehensive Cancer Center, 3800 Reservoir Road NW, Washington, DC, 20007, USA.,Georgetown University Medical Center, Washington, DC, USA
| | - Deepa S Subramaniam
- Lombardi Comprehensive Cancer Center, 3800 Reservoir Road NW, Washington, DC, 20007, USA.,Georgetown University Medical Center, Washington, DC, USA.,AstraZeneca plc, Gaithersburg, Maryland, USA
| | - John Deeken
- Inova Schar Cancer Institute, Inova Health System, Falls Church, Fairfax, VA, USA
| | - John Marshall
- Lombardi Comprehensive Cancer Center, 3800 Reservoir Road NW, Washington, DC, 20007, USA.,Georgetown University Medical Center, Washington, DC, USA
| | - Michael J Pishvaian
- Lombardi Comprehensive Cancer Center, 3800 Reservoir Road NW, Washington, DC, 20007, USA. .,Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
6
|
Jiménez Aguilar E, Zugazagoitia Fraile J, Paz-Ares Rodríguez L. Necitumumab: a new option for first-line treatment of squamous cell lung cancer. Expert Opin Drug Metab Toxicol 2018; 14:765-772. [PMID: 30025476 DOI: 10.1080/17425255.2018.1498839] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION First-line treatment with platinum-based chemotherapy has been the standard treatment for non-small-cell lung cancer (NSCLC) during the past decades. The development of new targeted drugs based on molecular alterations (EGFR, ALK, and ROS1) has led to important outcome benefits, but not for squamous cell carcinoma (SCC). However, the aberrant function of the EGFR pathway in SCC may be important in the development of the tumor and has been explored in preclinical and clinical studies as a potential target. Areas covered: Necitumumab is a human IgG1 anti-EGFR antibody that binds to the receptor and inhibits further pathway activation, thereby inhibiting cell differentiation, proliferation and migration. The phase III SQUIRE trial was a randomized study of gemcitabine-cisplatin plus necitumumab versus gemcitabine-cisplatin alone for first-line stage IV squamous NSCLC, showing a higher overall survival and better disease control with the addition of necitumumab. Despite the good results, the lack of robust predictive biomarkers makes the selection of the patients who will benefit the most complex. Expert opinion: Necitumumab plus cisplatin-gemcitabine is a first-line treatment option in SCC that improves overall survival and preserves the patient's quality of life with a manageable toxicity profile.
Collapse
Affiliation(s)
- Elizabeth Jiménez Aguilar
- a Medical Oncology Department , Hospital Universitario 12 de Octubre and Instituto de Investigación i+12 , Madrid , Spain.,b Lung Cancer Group, Clinical Research Program , CNIO (Centro Nacional de Investigaciones Oncológicas) and Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) , Madrid , Spain
| | - Jon Zugazagoitia Fraile
- a Medical Oncology Department , Hospital Universitario 12 de Octubre and Instituto de Investigación i+12 , Madrid , Spain.,b Lung Cancer Group, Clinical Research Program , CNIO (Centro Nacional de Investigaciones Oncológicas) and Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) , Madrid , Spain
| | - Luis Paz-Ares Rodríguez
- a Medical Oncology Department , Hospital Universitario 12 de Octubre and Instituto de Investigación i+12 , Madrid , Spain.,b Lung Cancer Group, Clinical Research Program , CNIO (Centro Nacional de Investigaciones Oncológicas) and Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) , Madrid , Spain.,c Medical Oncology department , Complutense University , Madrid , Spain
| |
Collapse
|
7
|
Volatile oil from Saussurea lappa exerts antitumor efficacy by inhibiting epithelial growth factor receptor tyrosine kinase-mediated signaling pathway in hepatocellular carcinoma. Oncotarget 2018; 7:79761-79773. [PMID: 27806329 PMCID: PMC5346749 DOI: 10.18632/oncotarget.12962] [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: 06/20/2016] [Accepted: 10/19/2016] [Indexed: 12/25/2022] Open
Abstract
Hepatocellular carcinoma (HCC) treatment remains lack of effective chemotherapeutic drugs, therefore, discovering novel anti-HCC drugs is a very attractive and urgent task. In this study, we reported VOSL (volatile oil from Saussurea lappa root) exhibits potent therapeutic effect on SMMC-7721 xenografts without obvious side effects. In the in vitro experiments, VOSL inhibited HCC cell proliferation by arresting cell cycle at S and G2/M phases, and induced HCC cell apoptosis by activating the Caspase3 pathway. VOSL also decreased the capability of HCC cell migration and invasion through MMP-9 depression. Moreover, mechanistic study indicated that VOSL can act as an epithelial growth factor receptor (EGFR) inhibitor to suppress EGFR activation and then to suppress its downstream MEK/P38 and PI3-K/Akt pathways. These results suggested that VOSL may be a novel anti-HCC drug candidate.
Collapse
|
8
|
Ding HW, Deng CL, Li DD, Liu DD, Chai SM, Wang W, Zhang Y, Chen K, Li X, Wang J, Song SJ, Song HR. Design, synthesis and biological evaluation of novel 4-aminoquinazolines as dual target inhibitors of EGFR-PI3Kα. Eur J Med Chem 2018; 146:460-470. [DOI: 10.1016/j.ejmech.2018.01.081] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 01/22/2018] [Accepted: 01/24/2018] [Indexed: 02/08/2023]
|
9
|
Development and validation of an ELISA to study panitumumab pharmacokinetics. Bioanalysis 2018; 10:205-214. [DOI: 10.4155/bio-2016-0292] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: Panitumumab is a monoclonal antibody directed against EGFR that is approved for the treatment of metastatic colorectal cancer. To investigate its pharmacokinetics and concentration–response relationship, a validated assay is required. Results: An ELISA assay was developed and validated according to international recommendations. Six calibrators (ranging from 0.1 to 20 mg/l) plus one anchor point (50 mg/l) and three quality controls (0.45, 2 and 8 mg/l) were defined. The limit of detection, lower limit of quantification and upper limit of quantification were 0.033, 0.112 and 10 mg/l, respectively. Conclusion: This method is validated and can be used to study pharmacokinetics of panitumumab or to perform therapeutic drug monitoring.
Collapse
|
10
|
Hirsh V. Turning EGFR mutation-positive non-small-cell lung cancer into a chronic disease: optimal sequential therapy with EGFR tyrosine kinase inhibitors. Ther Adv Med Oncol 2018; 10:1758834017753338. [PMID: 29383041 PMCID: PMC5784552 DOI: 10.1177/1758834017753338] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 12/19/2017] [Indexed: 01/24/2023] Open
Abstract
Four epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs),
erlotinib, gefitinib, afatinib and osimertinib, are currently available for the
management of EGFR mutation-positive non-small-cell lung cancer
(NSCLC), with others in development. Although tumors are exquisitely sensitive
to these agents, acquired resistance is inevitable. Furthermore, emerging data
indicate that first- (erlotinib and gefitinib), second- (afatinib) and
third-generation (osimertinib) EGFR TKIs differ in terms of efficacy and
tolerability profiles. Therefore, there is a strong imperative to optimize the
sequence of TKIs in order to maximize their clinical benefit. Osimertinib has
demonstrated striking efficacy as a second-line treatment option in patients
with T790M-positive tumors, and also confers efficacy and tolerability
advantages over first-generation TKIs in the first-line setting. However, while
accrual of T790M is the most predominant mechanism of resistance to erlotinib,
gefitinib and afatinib, resistance mechanisms to osimertinib have not been
clearly elucidated, meaning that possible therapy options after osimertinib
failure are not clear. At present, few data comparing sequential regimens in
patients with EGFR mutation-positive NSCLC are available and
prospective clinical trials are required. This article reviews the similarities
and differences between EGFR TKIs, and discusses key considerations when
assessing optimal sequential therapy with these agents for the treatment of
EGFR mutation-positive NSCLC.
Collapse
Affiliation(s)
- Vera Hirsh
- McGill Department of Oncology, Royal Victoria Hospital, Glen Site, 1001 Décarie Blvd, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
11
|
Girard N. Optimizing outcomes in EGFR mutation-positive NSCLC: which tyrosine kinase inhibitor and when? Future Oncol 2018; 14:1117-1132. [PMID: 29336166 DOI: 10.2217/fon-2017-0636] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Despite the efficacy of standard-of-care EGFR tyrosine kinase inhibitors (TKIs), erlotinib, gefitinib and afatinib, in EGFR mutation-positive non-small-cell lung cancer, resistance develops, most commonly due to the T790M mutation. Osimertinib showed clinical activity in the treatment of T790M-positive disease following progression on a first-line TKI, and is approved in this setting. Recently, osimertinib improved efficacy versus first-generation TKIs (erlotinib and gefitinib) in the first-line setting. Multiple factors can influence first-line treatment decisions, including subsequent therapy options, presence of brain metastases and tolerability, all of which should be considered in the long-term treatment plan. Further research into treatment sequencing is also needed, to optimize outcomes in EGFR mutation-positive non-small-cell lung cancer.
Collapse
Affiliation(s)
- Nicolas Girard
- Thoracic Oncology, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, 69622, France.,Thoracic Surgery, Institut Curie, Institut du Thorax Curie-Montsouris, Paris, 75248, France
| |
Collapse
|
12
|
Papanastasiou AD, Sirinian C, Plakoula E, Zolota V, Zarkadis IK, Kalofonos HP. RANK and EGFR in invasive breast carcinoma. Cancer Genet 2017; 216-217:61-66. [PMID: 29025596 DOI: 10.1016/j.cancergen.2017.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 01/16/2023]
Abstract
Breast cancer is the most common malignancy, affecting one in eight women in North America and Europe. The human epidermal growth factor receptor (EGFR) protein comprises a major determinant of normal development but also cancer. RANK receptor (Receptor Activator of Nuclear factor-κB) is a tumor necrosis superfamily member and a binding partner for RANKL, which was recently implicated in breast cancer initiation, progression and metastasis. Here we provide preliminary evidence of a possible interplay between RANK and EGFR signaling in breast cancer. TCGA (cancergenome.nih.gov) publicly available data for EGFR and TNFRSF11A (RANK) genes from breast cancer patients and breast cancer cell lines were retrieved and analyzed. RANK mRNA showed a statistically significant positive correlation (p <0.001) with the mRNA and protein expression of EGFR, but not with ERBB2/3/4. Further analyses of survival data of a group of breast cancer patients (n = 248) from TCGA, revealed an EGFRhi/RANKhi subpopulation that showed a statistically significant (p = 0.001) reduced overall survival when compared to EGFRlow/RANKlow group of patients. Finally, EGFR and RANK combinatorial in vitro analyses revealed a significant upregulation of AKT and ERK signaling after EGF stimulation in cell lines and also an increase of breast cancer cell invasiveness.
Collapse
Affiliation(s)
- Anastasios D Papanastasiou
- Clinical and Molecular Oncology Laboratory, Division of Oncology, School of Medicine, University of Patras, 26504, Greece.
| | - Chaido Sirinian
- Clinical and Molecular Oncology Laboratory, Division of Oncology, School of Medicine, University of Patras, 26504, Greece
| | - Eva Plakoula
- Clinical and Molecular Oncology Laboratory, Division of Oncology, School of Medicine, University of Patras, 26504, Greece; Department of Biology, School of Medicine, University of Patras, 26504, Greece
| | - Vassiliki Zolota
- Department of Pathology, School of Medicine, University of Patras, 26504, Greece
| | - Ioannis K Zarkadis
- Department of Biology, School of Medicine, University of Patras, 26504, Greece
| | - Haralabos P Kalofonos
- Clinical and Molecular Oncology Laboratory, Division of Oncology, School of Medicine, University of Patras, 26504, Greece
| |
Collapse
|
13
|
In vivo evaluation of a novel format of a bivalent HER3-targeting and albumin-binding therapeutic affibody construct. Sci Rep 2017; 7:43118. [PMID: 28230065 PMCID: PMC5322329 DOI: 10.1038/srep43118] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/19/2017] [Indexed: 01/21/2023] Open
Abstract
Overexpression of human epidermal growth factor receptor 3 (HER3) is involved in resistance to several therapies for malignant tumours. Currently, several anti-HER3 monoclonal antibodies are under clinical development. We introduce an alternative approach to HER3-targeted therapy based on engineered scaffold proteins, i.e. affibody molecules. We designed a small construct (22.5 kDa, denoted 3A3), consisting of two high-affinity anti-HER3 affibody molecules flanking an albumin-binding domain ABD, which was introduced for prolonged residence in circulation. In vitro, 3A3 efficiently inhibited growth of HER3-expressing BxPC-3 cells. Biodistribution in mice was measured using 3A3 that was site-specifically labelled with 111In via a DOTA chelator. The residence time of 111In-DOTA-3A3 in blood was extended when compared with the monomeric affibody molecule. 111In-DOTA-3A3 accumulated specifically in HER3-expressing BxPC-3 xenografts in mice. However, 111In-DOTA-3A3 cleared more rapidly from blood than a size-matched control construct 111In-DOTA-TAT, most likely due to sequestering of 3A3 by mErbB3, the murine counterpart of HER3. Repeated dosing and increase of injected protein dose decreased uptake of 111In-DOTA-3A3 in mErbB3-expressing tissues. Encouragingly, growth of BxPC-3 xenografts in mice was delayed in an experimental (pilot-scale) therapy study using 3A3. We conclude that the 3A3 affibody format seems promising for treatment of HER3-overexpressing tumours.
Collapse
|
14
|
Mu K, Zhang S, Ai T, Jiang J, Yao Y, Jiang L, Zhou Q, Xiang H, Zhu Y, Yang X, Zhu W. Monoclonal antibody-conjugated superparamagnetic iron oxide nanoparticles for imaging of epidermal growth factor receptor-targeted cells and gliomas. Mol Imaging 2016; 14. [PMID: 26044549 DOI: 10.2310/7290.2015.00002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The objective of this study was to successfully synthesize epidermal growth factor receptor monoclonal antibody-conjugated superparamagnetic iron oxide nanoparticles (EGFRmAb-SPIONs) and explore their biocompatibility and potential applications as a targeted magnetic resonance imaging (MRI) contrast agent for the EGFR-specific detection of brain glioma in vivo. After conjugation of EGFRmAb with SPIONs, the magnetic characteristics of EGFRmAb-SPIONs were investigated. Thereafter, the targeting abilities of EGFRmAb-SPIONs with MRI were qualitatively and quantitatively assessed in EGFR-positive C6 glioma cells in vitro and in a Wistar rat model bearing C6 glioma in vivo. Furthermore, the preliminary biocompatibility and toxicity of EGFRmAb-SPIONs were evaluated in normal rats through hematology assays and histopathologic analyses. Statistical analysis was performed using one-way analysis of variance and Student t-test, with a significance level of p < .05. From the results of EGFRmAb-SPION characterizations, the average particle size was 10.21 nm and the hydrodynamic diameter was 161.5 ± 2.12 nm. The saturation magnetization was 55 emu/g·Fe, and T2 relaxivity was 92.73 s-1mM-1 in distilled water. The preferential accumulation of the EGFRmAb-SPIONs within glioma and subsequent MRI contrast enhancement were demonstrated both in vitro in C6 cells and in vivo in rats bearing C6 glioma. After intravenous administration of EGFRmAb-SPIONs, T2-weighted MRI of the rat model with brain glioma exhibited an apparent hypointense region within glioma from 2 to 48 hours. The maximal image contrast was reached at 24 hours, where the signal intensity decreased and the R2 value increased by 30% compared to baseline. However, T2-weighted imaging of the rat model administered with SPIONs showed no visible signal changes within the tumor over the same time period. Moreover, no evident toxicities in vitro and in vivo with EGFRmAb-SPIONs were clearly identified based on the laboratory examinations. EGFRmAb-SPIONs could potentially be employed as a targeted contrast agent in the molecule-specific diagnosis of brain glioma in MRI.
Collapse
|
15
|
Murray BS, Babak MV, Hartinger CG, Dyson PJ. The development of RAPTA compounds for the treatment of tumors. Coord Chem Rev 2016. [DOI: 10.1016/j.ccr.2015.06.014] [Citation(s) in RCA: 266] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
16
|
Achell Nava L, Hierro Orozco S, Maya Talamantes EI. Manejo y prevención de efectos secundarios al empleo de un anticuerpo monoclonal inhibidor de la señal de los receptores del factor de crecimiento epidérmico. GACETA MEXICANA DE ONCOLOGÍA 2016. [DOI: 10.1016/j.gamo.2015.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
17
|
Komposch K, Sibilia M. EGFR Signaling in Liver Diseases. Int J Mol Sci 2015; 17:E30. [PMID: 26729094 PMCID: PMC4730276 DOI: 10.3390/ijms17010030] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 12/17/2015] [Accepted: 12/21/2015] [Indexed: 02/07/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) is a transmembrane receptor tyrosine kinase that is activated by several ligands leading to the activation of diverse signaling pathways controlling mainly proliferation, differentiation, and survival. The EGFR signaling axis has been shown to play a key role during liver regeneration following acute and chronic liver damage, as well as in cirrhosis and hepatocellular carcinoma (HCC) highlighting the importance of the EGFR in the development of liver diseases. Despite the frequent overexpression of EGFR in human HCC, clinical studies with EGFR inhibitors have so far shown only modest results. Interestingly, a recent study has shown that in human HCC and in mouse HCC models the EGFR is upregulated in liver macrophages where it plays a tumor-promoting function. Thus, the role of EGFR in liver diseases appears to be more complex than what anticipated. Further studies are needed to improve the molecular understanding of the cell-specific signaling pathways that control disease development and progression to be able to develop better therapies targeting major components of the EGFR signaling network in selected cell types. In this review, we compiled the current knowledge of EGFR signaling in different models of liver damage and diseases, mainly derived from the analysis of HCC cell lines and genetically engineered mouse models (GEMMs).
Collapse
Affiliation(s)
- Karin Komposch
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria.
| | - Maria Sibilia
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria.
| |
Collapse
|
18
|
Qian L, Liu Y, Xu Y, Ji W, Wu Q, Liu Y, Gao Q, Su C. Matrine derivative WM130 inhibits hepatocellular carcinoma by suppressing EGFR/ERK/MMP-2 and PTEN/AKT signaling pathways. Cancer Lett 2015; 368:126-134. [PMID: 26259512 DOI: 10.1016/j.canlet.2015.07.035] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 07/27/2015] [Accepted: 07/31/2015] [Indexed: 12/14/2022]
Abstract
Matrine, a sophora alkaloid, has been demonstrated to exert antitumor effects on many types of cancer. However, its bioactivity is weak and its potential druggability is low. We modified the structure of matrine and obtained a new matrine derivative, WM130 (C30N4H40SO5F), which exhibited better pharmacological activities than matrine. In this study, we investigated the antitumor activity and the underlying mechanisms of WM130 on hepatocellular carcinoma (HCC) cells in vitro and in vivo, and found that WM130 inhibited the proliferation, invasion, migration and induced apoptosis of HCC cells in a dose-dependent manner. Furthermore, after treatment with WM130, the expressions of p-EGFR, p-ERK, p-AKT, MMP-2 and the ratio of Bcl-2/Bax were significantly down-regulated, whereas the expression of PTEN was increased in HCC cells. Moreover, WM130 inhibited Huh-7 xenograft tumor growth in a dose-dependent manner after intravenous administration. Immunohistochemistry results demonstrated that WM130 treatment resulted in down-regulation of p-EGFR, MMP-2, and Ki67 and up-regulation of PTEN. The findings indicated that WM130 could inhibit cell proliferation, invasion, migration and induced apoptosis in HCC cells by suppressing EGFR/ERK/MMP-2 and PTEN/AKT signaling pathways and may be a novel effective candidate for HCC treatment.
Collapse
Affiliation(s)
- Liqiang Qian
- Department of General Surgery, Wujiang No. 1 People's Hospital, Suzhou 215200, China; Department of Molecular Oncology, Eastern Hepatobiliary Surgery Hospital & National Center of Liver Cancer, Second Military Medical University, Shanghai 200438, China
| | - Yan Liu
- Department of General Surgery, Wujiang No. 1 People's Hospital, Suzhou 215200, China
| | - Yang Xu
- Department of Molecular Oncology, Eastern Hepatobiliary Surgery Hospital & National Center of Liver Cancer, Second Military Medical University, Shanghai 200438, China
| | - Weidan Ji
- Department of Molecular Oncology, Eastern Hepatobiliary Surgery Hospital & National Center of Liver Cancer, Second Military Medical University, Shanghai 200438, China
| | - Qiuye Wu
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Yongjing Liu
- Deparment of Cardiothoracic Surgery, No. 105 Hospital of PLA, Hefei 230031, China
| | - Quangen Gao
- Department of General Surgery, Wujiang No. 1 People's Hospital, Suzhou 215200, China.
| | - Changqing Su
- Department of Molecular Oncology, Eastern Hepatobiliary Surgery Hospital & National Center of Liver Cancer, Second Military Medical University, Shanghai 200438, China.
| |
Collapse
|
19
|
Dahle-Smith A, Petty RD. Biomarkers and novel agents in esophago-gastric cancer: are we making progress? Expert Rev Anticancer Ther 2015; 15:1103-19. [DOI: 10.1586/14737140.2015.1071669] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
20
|
Amiloride sensitizes human pancreatic cancer cells to erlotinib in vitro through inhibition of the PI3K/AKT signaling pathway. Acta Pharmacol Sin 2015; 36:614-26. [PMID: 25864651 DOI: 10.1038/aps.2015.4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 01/08/2015] [Indexed: 01/05/2023] Open
Abstract
AIM Blockade of EGFR by EGFR tyrosine kinase inhibitors such as erlotinib is insufficient for effective treatment of human pancreatic cancer due to independent activation of the Akt pathway, while amiloride, a potassium-sparing diuretic, has been found as a potential Akt inhibitor. The aim of this study was to investigate the anticancer effects of combined amiloride with erlotinib against human pancreatic cancer cells in vitro. METHODS Cell proliferation, colony formation, cell cycle and apoptosis were analyzed in 4 human pancreatic cancer cell lines Bxpc-3, PANC-1, Aspc-1 and CFPAC-1 treated with erlotinib or amiloride alone, or in their combination. The synergistic analysis for the effects of combinations of amiloride and erlotinib was performed using Chou-Talalay's combination index isobolographic method. RESULTS Amiloride (10, 30, and 100 μmol/L) concentration-dependently potentiated erlotinib-induced inhibition of cell proliferation and colony formation in the 4 pancreatic cancer cell lines. Isobolographic analysis confirmed that combinations of amiloride and erlotinib produced synergistic cytotoxic effects. Amiloride significantly potentiated erlotinib-induced G0/G1 cell-cycle arrest and apoptosis in Bxpc-3 and PANC-1 cells. Amiloride inhibited EGF-stimulated phorsphorylation of AKT, and significantly enhanced erlotinib-induced downregulation of phorsphorylation of EGFR, AKT, PI3K P85 and GSK 3β in Bxpc-3 and PANC-1 cells. CONCLUSION Amiloride sensitizes human pancreatic cancer cells to erlotinib in vitro through inhibition of the PI3K/AKT signaling pathway. Treatment of pancreatic cancer patients with combination of erlotinib and amiloride merits further investigation.
Collapse
|
21
|
Hypersensitivity and tumor lysis syndrome associated with cetuximab treatment: should we be afraid? TUMORI JOURNAL 2015; 101:e40-5. [PMID: 25721679 DOI: 10.5301/tj.5000246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2014] [Indexed: 11/20/2022]
Abstract
The majority of the chemotherapy agents in use today cause various infusion reactions, from mild flushing to life-threatening events. The frequency of the reported hypersensitivity reactions induced by cetuximab varies between 3% and 22%. It is recommended in the literature to stop the infusion and replace cetuximab with panitumumab in case of hypersensitivity reactions observed during the treatment of colon cancer. Tumor lysis syndrome (TLS) may occur in colorectal cancers with heavy tumor load. Tumor lysis syndrome may be life-threatening. In our patient with widespread bone and liver metastases, treatment continued with cetuximab as a combination therapy with irinotecan in spite of the hypersensitivity and TLS led to a complete treatment response. The complete response observed after 3 months through continued therapy in our patient may present an example supporting treatment with cetuximab in spite of severe reactions.
Collapse
|
22
|
Weiss A, Ding X, van Beijnum JR, Wong I, Wong TJ, Berndsen RH, Dormond O, Dallinga M, Shen L, Schlingemann RO, Pili R, Ho CM, Dyson PJ, van den Bergh H, Griffioen AW, Nowak-Sliwinska P. Rapid optimization of drug combinations for the optimal angiostatic treatment of cancer. Angiogenesis 2015; 18:233-44. [PMID: 25824484 PMCID: PMC4473022 DOI: 10.1007/s10456-015-9462-9] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 03/13/2015] [Indexed: 01/13/2023]
Abstract
Drug combinations can improve angiostatic cancer treatment efficacy and enable the reduction of side effects and drug resistance. Combining drugs is non-trivial due to the high number of possibilities. We applied a feedback system control (FSC) technique with a population-based stochastic search algorithm to navigate through the large parametric space of nine angiostatic drugs at four concentrations to identify optimal low-dose drug combinations. This implied an iterative approach of in vitro testing of endothelial cell viability and algorithm-based analysis. The optimal synergistic drug combination, containing erlotinib, BEZ-235 and RAPTA-C, was reached in a small number of iterations. Final drug combinations showed enhanced endothelial cell specificity and synergistically inhibited proliferation (p < 0.001), but not migration of endothelial cells, and forced enhanced numbers of endothelial cells to undergo apoptosis (p < 0.01). Successful translation of this drug combination was achieved in two preclinical in vivo tumor models. Tumor growth was inhibited synergistically and significantly (p < 0.05 and p < 0.01, respectively) using reduced drug doses as compared to optimal single-drug concentrations. At the applied conditions, single-drug monotherapies had no or negligible activity in these models. We suggest that FSC can be used for rapid identification of effective, reduced dose, multi-drug combinations for the treatment of cancer and other diseases.
Collapse
Affiliation(s)
- Andrea Weiss
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), 1015, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Liu Y, Bi T, Shen G, Li Z, Wu G, Wang Z, Qian L, Gao Q. Lupeol induces apoptosis and inhibits invasion in gallbladder carcinoma GBC-SD cells by suppression of EGFR/MMP-9 signaling pathway. Cytotechnology 2014; 68:123-133. [PMID: 25037728 DOI: 10.1007/s10616-014-9763-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 06/25/2014] [Indexed: 12/13/2022] Open
Abstract
The cytostatic drug from fruits and other plant derived products have acted as a chemotherapeutic agent used in treatment of a wide variety of cancers. Lupeol, a dietary triterpene, present in many fruits and medicinal plants, has been shown to possess many pharmacological properties including anti-cancer effect in both in vitro and in vivo assay systems. However, the cancer proliferative and invasive inhibitory effects and molecular mechanisms on gallbladder carcinoma GBC-SD cells have not been studied. In the present study, GBC-SD cells were treated by lupeol and subjected to methyl thiazolyl tetrazolium analysis, Hoechst 33342 staining, annexin V/propidium iodide double-staining, transwell chamber assay and Western blot analysis. In addition, GBC-SD xenograft tumors were established in male nude BALB/c mice, and lupeol was intravenously administered to evaluate the anti-cancer capacity in vivo. Our results showed that lupeol inhibited the proliferation, migration, invasion and induced apoptosis of GBC-SD cells in a dose-dependent manner in vitro. Furthermore, the expression of p-EGFR, p-AKT and MMP-9 levels were significantly down-regulated. These protein interactions may play a pivotal role in the regulation of apoptosis and invasion. More importantly, our in vivo studies showed that administration of lupeol decreased tumor growth in a dose-dependent manner. Immunohistochemistry analysis demonstrated the down-regulation of p-EGFR and MMP-9 in tumor tissues following lupeol treatment, consistent with the in vitro results. Taken together, our findings indicated that lupeol can induce apoptotic cell death and inhibit the migration as well as invasion of GBC-SD cells. The mechanism may be associated with the suppression of EGFR/MMP-9 signaling. These results might offer a therapeutic potential advantage for human gallbladder carcinoma chemoprevention or chemotherapy.
Collapse
Affiliation(s)
- Yan Liu
- Department of General Surgery, Wujiang No.1 People's Hospital, Suzhou, 215200, China
- Graduate School, Xuzhou Medical College, Xuzhou, 221004, China
| | - Tingting Bi
- Department of General Surgery, Wujiang No.1 People's Hospital, Suzhou, 215200, China
- Graduate School, Xuzhou Medical College, Xuzhou, 221004, China
| | - Genhai Shen
- Department of General Surgery, Wujiang No.1 People's Hospital, Suzhou, 215200, China
| | - Zhimin Li
- Department of General Surgery, Wujiang No.1 People's Hospital, Suzhou, 215200, China
| | - Guoliang Wu
- Department of General Surgery, Wujiang No.1 People's Hospital, Suzhou, 215200, China
| | - Zheng Wang
- Department of General Surgery, Wujiang No.1 People's Hospital, Suzhou, 215200, China
| | - Liqiang Qian
- Department of General Surgery, Wujiang No.1 People's Hospital, Suzhou, 215200, China
| | - Quangen Gao
- Department of General Surgery, Wujiang No.1 People's Hospital, Suzhou, 215200, China.
| |
Collapse
|
24
|
Lee CC, Shiao HY, Wang WC, Hsieh HP. Small-molecule EGFR tyrosine kinase inhibitors for the treatment of cancer. Expert Opin Investig Drugs 2014; 23:1333-48. [DOI: 10.1517/13543784.2014.928283] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
25
|
CKAP4 inhibited growth and metastasis of hepatocellular carcinoma through regulating EGFR signaling. Tumour Biol 2014; 35:7999-8005. [PMID: 24838946 DOI: 10.1007/s13277-014-2000-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Accepted: 04/22/2014] [Indexed: 12/22/2022] Open
Abstract
CKAP4, one kind of type II trans-membrane protein, plays an important role to maintain endoplasmic reticulum structure and inhibits the proliferation of bladder cancer cells by combining its ligand anti-proliferative factor (APF). However, the biological function of CKAP4 in the progression of liver cancer has not been clearly demonstrated. In the present study, we knocked down or overexpressed CKAP4 in hepatocellular carcinoma (HCC) cells and cell proliferation, invasion, and migration capacities were investigated by CCK-8 and transwell assays. In vivo tumor model in mice was used to evaluate the role of CKAP4 on growth and metastasis of HCC. The data documented that HCC cells with high CKAP4 levels were featured by low proliferation capability as well as low invasion potential. Interestingly, we found that CKAP4 suppressed the activation of epithelial growth factor receptor (EGFR) signaling, which may partly explain the role of CKAP4 in cell biological behavior of HCC. Further study revealed that CKAP4 could associate with EGFR at basal status and the complex was reduced upon EGF stimulation, leading to release EGFR into cytoplasm. Thus, we demonstrate the novel mechanism, for the first time, expression of CKAP4 regulates progression and metastasis of HCC and it may provide therapeutic values in this tumor.
Collapse
|
26
|
Golfier S, Kopitz C, Kahnert A, Heisler I, Schatz CA, Stelte-Ludwig B, Mayer-Bartschmid A, Unterschemmann K, Bruder S, Linden L, Harrenga A, Hauff P, Scholle FD, Müller-Tiemann B, Kreft B, Ziegelbauer K. Anetumab ravtansine: a novel mesothelin-targeting antibody-drug conjugate cures tumors with heterogeneous target expression favored by bystander effect. Mol Cancer Ther 2014; 13:1537-48. [PMID: 24714131 DOI: 10.1158/1535-7163.mct-13-0926] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mesothelin is a tumor differentiation antigen frequently overexpressed in tumors such as mesothelioma, ovarian, pancreatic, and lung adenocarcinomas while showing limited expression in nonmalignant tissues. Mesothelin is therefore an attractive target for cancer therapy using antibody-drug conjugates (ADC). This study describes the detailed characterization of anetumab ravtansine, here referred to as BAY 94-9343, a novel ADC consisting of a human anti-mesothelin antibody conjugated to the maytansinoid tubulin inhibitor DM4 via a disulfide-containing linker. Binding properties of the anti-mesothelin antibody were analyzed using surface plasmon resonance, immunohistochemistry, flow cytometry, and fluorescence microscopy. Effects of BAY 94-9343 on cell proliferation were first studied in vitro and subsequently in vivo using subcutaneous, orthotopic, and patient-derived xenograft tumor models. The antibody binds to human mesothelin with high affinity and selectivity, thereby inducing efficient antigen internalization. In vitro, BAY 94-9343 demonstrated potent and selective cytotoxicity of mesothelin-expressing cells with an IC(50) of 0.72 nmol/L, without affecting mesothelin-negative or nonproliferating cells. In vivo, BAY 94-9343 localized specifically to mesothelin-positive tumors and inhibited tumor growth in both subcutaneous and orthotopic xenograft models. In addition, BAY 94-9343 was able to induce a bystander effect on neighboring mesothelin-negative tumor cells. Antitumor efficacy of BAY 94-9343 correlated with the amount of mesothelin expressed and was generally superior to that of standard-of-care regimen resulting in complete tumor eradication in most of the models. BAY 94-9343 is a selective and highly potent ADC, and our data support its development for the treatment of patients with mesothelin-expressing tumors.
Collapse
Affiliation(s)
- Sven Golfier
- Authors' Affiliation: Bayer HealthCare Pharmaceuticals, Berlin/Wuppertal, Germany
| | - Charlotte Kopitz
- Authors' Affiliation: Bayer HealthCare Pharmaceuticals, Berlin/Wuppertal, Germany
| | - Antje Kahnert
- Authors' Affiliation: Bayer HealthCare Pharmaceuticals, Berlin/Wuppertal, Germany
| | - Iring Heisler
- Authors' Affiliation: Bayer HealthCare Pharmaceuticals, Berlin/Wuppertal, Germany
| | - Christoph A Schatz
- Authors' Affiliation: Bayer HealthCare Pharmaceuticals, Berlin/Wuppertal, Germany
| | | | | | | | - Sandra Bruder
- Authors' Affiliation: Bayer HealthCare Pharmaceuticals, Berlin/Wuppertal, Germany
| | - Lars Linden
- Authors' Affiliation: Bayer HealthCare Pharmaceuticals, Berlin/Wuppertal, Germany
| | - Axel Harrenga
- Authors' Affiliation: Bayer HealthCare Pharmaceuticals, Berlin/Wuppertal, Germany
| | - Peter Hauff
- Authors' Affiliation: Bayer HealthCare Pharmaceuticals, Berlin/Wuppertal, Germany
| | - Frank-Detlef Scholle
- Authors' Affiliation: Bayer HealthCare Pharmaceuticals, Berlin/Wuppertal, Germany
| | - Beate Müller-Tiemann
- Authors' Affiliation: Bayer HealthCare Pharmaceuticals, Berlin/Wuppertal, Germany
| | - Bertolt Kreft
- Authors' Affiliation: Bayer HealthCare Pharmaceuticals, Berlin/Wuppertal, Germany
| | - Karl Ziegelbauer
- Authors' Affiliation: Bayer HealthCare Pharmaceuticals, Berlin/Wuppertal, Germany
| |
Collapse
|
27
|
Chemotherapeutic Strategies in Advanced or Metastatic Pancreatic Adenocarcinoma. Am J Clin Oncol 2014; 37:194-200. [DOI: 10.1097/coc.0b013e318248d8bb] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Liu D, Jiang J, Zhang L, Tan F, Wang Y, Zhang D, Hu P. Clinical pharmacokinetics of Icotinib, an anti-cancer drug: evaluation of dose proportionality, food effect, and tolerability in healthy subjects. Cancer Chemother Pharmacol 2014; 73:721-7. [PMID: 24488324 DOI: 10.1007/s00280-014-2398-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 01/21/2014] [Indexed: 11/27/2022]
Abstract
PURPOSE Icotinib, an oral epidermal growth factor receptor tyrosine kinase inhibitor, has proved effectiveness in xenografted nude mice. Purpose of the present studies was to investigate tolerability and pharmacokinetics of Icotinib in healthy subjects for the first time, including dose proportionality, food effect, and tolerability. METHODS Two studies were conducted in total of 22 healthy subjects: a randomized, two-Latin-square crossover, dose proportional study (n = 12) and a randomized two-way crossover food-effect study (n = 10). RESULTS Plasma concentration of Icotinib reached peak at a median Tmax of 0.75-3.5 h after single dose and then declined with a mean t1/2β of 6.02-7.83 h. Over the dose range of 100-600 mg, AUC values were proportional to dose and Cmax showed a slight saturation when dose increases. Only 0.2 % of the dose was excreted through kidney in unchanged Icotinib. After dosing 400 mg of Icotinib with high-fat and high-calorie meal, mean Cmax and AUC were significantly increased by 59 and 79 %, respectively. Three subjects experienced four adverse events (rash, increase in AST and ALT, and external injury). Rash and increased levels of AST and ALT were considered as drug-related. No serious adverse events were reported. CONCLUSION The current work demonstrated that Icotinib was well tolerated in healthy male subjects (n = 22) over the dose range of 100-600 mg with or without food. Icotinib exposure, expressed in AUC, was proportionally increased with dose over the above dose range. Food intake significantly increased the absorption and exposure of Icotinib in healthy subjects.
Collapse
Affiliation(s)
- Dongyang Liu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100032, China,
| | | | | | | | | | | | | |
Collapse
|
29
|
Specenier PM, Vermorken JB. Recurrent head and neck cancer: current treatment and future prospects. Expert Rev Anticancer Ther 2014; 8:375-91. [DOI: 10.1586/14737140.8.3.375] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
30
|
Yoshimoto M, Hirata M, Kanai Y, Naka S, Nishii R, Kagawa S, Kawai K, Ohmomo Y. Monitoring of Gefitinib Sensitivity with Radioiodinated PHY Based on EGFR Expression. Biol Pharm Bull 2014; 37:355-60. [DOI: 10.1248/bpb.b13-00559] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Mitsuyoshi Yoshimoto
- Division of Functional Imaging, National Cancer Center Hospital East
- Faculty of Health Sciences, Kanazawa University
| | | | - Yasukazu Kanai
- Department of Molecular Imaging in Medicine, Osaka University Graduate School of Medicine
| | - Sadahiro Naka
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine
| | - Ryuichi Nishii
- Department of Radiology, Faculty of Medicine, University of Miyazaki
| | | | | | | |
Collapse
|
31
|
Wang RY, Chen L, Chen HY, Hu L, Li L, Sun HY, Jiang F, Zhao J, Liu GMY, Tang J, Chen CY, Yang YC, Chang YX, Liu H, Zhang J, Yang Y, Huang G, Shen F, Wu MC, Zhou WP, Wang HY. MUC15 inhibits dimerization of EGFR and PI3K-AKT signaling and is associated with aggressive hepatocellular carcinomas in patients. Gastroenterology 2013; 145:1436-48.e1-12. [PMID: 23933603 DOI: 10.1053/j.gastro.2013.08.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Revised: 07/30/2013] [Accepted: 08/04/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Aberrant expression of MUC15 correlates with development of colorectal adenocarcinoma, and MUC15 has been reported to prevent trophoblast invasion of human placenta. However, little is known about the role of MUC15 in pathogenesis of hepatocellular carcinoma (HCC). METHODS We analyzed HCC samples and matched nontumor liver tissues (controls) collected from 313 patients who underwent hepatectomy in Shanghai, China, from January 2006 through September 2009. Levels of messenger RNAs and proteins were determined by immunohistochemical, quantitative reverse transcription polymerase chain reaction, and immunoblot analyses. Statistical analyses were used to associate levels of MUC15 with tumor features and patient outcomes. RESULTS Levels of MUC15 messenger RNA and protein were reduced in a greater percentage of HCC samples than control tissues. Tumors with reduced levels of MUC15 were more likely to have aggressive characteristics (eg, high levels of α-fetoprotein, vascular invasion, lack of encapsulation, and poor differentiation) than those with low levels. Patients whose tumors had reduced levels of MUC15 had shorter overall survival times (24 months vs 46 months for patients with tumors with high levels of MUC15) and time to disease recurrence. Stable expression of MUC15 in HCC cell lines (SMMC-7721 and HCC-LM3) reduced their proliferation and invasive features in vitro, and ability to form metastatic tumors in mice. MUC15 reduced transcription of the matrix metalloproteinases 2 and 7 increased expression of tissue inhibitor of metalloproteinase-2, which required phosphoinositide 3-kinase-v-akt murine thymoma viral oncogene homolog signaling. Physical interaction between MUC15 and epidermal growth factor receptor led to its relocation and degradation within early endosomes and was required for inactivation of phosphoinositide 3-kinase-v-akt murine thymoma viral oncogene homolog signaling. CONCLUSIONS Reduced levels of MUC15 in HCCs are associated with shorter survival times of patients and reduced time to disease recurrence. Expression of MUC15 in HCC cells reduces their aggressive behavior in vitro and in mice by inducing dimerization of epidermal growth factor receptor and decreasing phosphoinositide 3-kinase signaling via v-akt murine thymoma viral oncogene homolog.
Collapse
Affiliation(s)
- Ruo-Yu Wang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Shanghai, China; The Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Di Franco S, Todaro M, Dieli F, Stassi G. Colorectal cancer defeating? Challenge accepted! Mol Aspects Med 2013; 39:61-81. [PMID: 23927966 DOI: 10.1016/j.mam.2013.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 07/01/2013] [Accepted: 07/23/2013] [Indexed: 02/07/2023]
Abstract
Colorectal tumours are actually considered as aberrant organs, within it is possible to notice a different stage of cell growth and differentiation. Their origin is reported to arise from a subpopulation of tumour cells endowed with, just like the healthy stem cells, self-renewal and aberrant multi-lineage differentiation capacity likely to be called colorectal cancer stem cells (CCSCs). Cancer stem cells (CSCs) fate, since their origin, reflects the influences from their microenvironment (or niche) both in the maintenance of stemness, in promoting their differentiation, and in inducing epithelial-mesenchymal transition, responsible of CSCs dissemination and subsequent formation of metastatic lesions. The tumour cells heterogeneity and their immuno-response resistance nowadays probably responsible of the failure of the conventional therapies, make this research field an open issue. Even more importantly, our increasing understanding of the cellular and molecular mechanisms that regulate CSC quiescence and cell cycle regulation, self-renewal, chemotaxis and resistance to cytotoxic agents, is expected to eventually result in tailor-made therapies with a significant impact on the morbidity and overall survival of colorectal cancer patients.
Collapse
Affiliation(s)
- S Di Franco
- Department of Surgical and Oncological Sciences, University of Palermo, Via Liborio Giuffre' 5, 90127 Palermo, Italy
| | - M Todaro
- Department of Surgical and Oncological Sciences, University of Palermo, Via Liborio Giuffre' 5, 90127 Palermo, Italy
| | - F Dieli
- Division of Immunology and Immunogenetics, Department of Biotechnology and Medical and Forensic Biopathological (DIBIMEF), Palermo, Italy
| | - G Stassi
- Department of Surgical and Oncological Sciences, University of Palermo, Via Liborio Giuffre' 5, 90127 Palermo, Italy.
| |
Collapse
|
33
|
Development of an epidermal growth factor derivative with EGFR blocking activity. PLoS One 2013; 8:e69325. [PMID: 23935985 PMCID: PMC3728333 DOI: 10.1371/journal.pone.0069325] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 06/09/2013] [Indexed: 11/22/2022] Open
Abstract
The members of the epidermal growth factor (EGF)/ErbB family are prime targets for cancer therapy. However, the therapeutic efficiency of the existing anti-ErbB agents is limited. Thus, identifying new molecules that inactivate the ErbB receptors through novel strategies is an important goal on cancer research. In this study we have developed a shorter form of human EGF (EGFt) with a truncated C-terminal as a novel EGFR inhibitor. EGFt was designed based on the superimposition of the three-dimensional structures of EGF and the Potato Carboxypeptidase Inhibitor (PCI), an EGFR blocker previously described by our group. The peptide was produced in E. coli with a high yield of the correctly folded peptide. EGFt showed specificity and high affinity for EGFR but induced poor EGFR homodimerization and phosphorylation. Interestingly, EGFt promoted EGFR internalization and translocation to the cell nucleus although it did not stimulate the cell growth. In addition, EGFt competed with EGFR native ligands, inhibiting the proliferation of cancer cells. These data indicate that EGFt may be a potential EGFR blocker for cancer therapy. In addition, the lack of EGFR-mediated growth-stimulatory activity makes EGFt an excellent delivery agent to target toxins to tumours over-expressing EGFR.
Collapse
|
34
|
Ito Y, Yamada Y, Asada K, Ushijima T, Iwasa S, Kato K, Hamaguchi T, Shimada Y. EGFR L2 domain mutation is not correlated with resistance to cetuximab in metastatic colorectal cancer patients. J Cancer Res Clin Oncol 2013; 139:1391-6. [PMID: 23722667 DOI: 10.1007/s00432-013-1454-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 05/17/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND The KRAS mutation has been associated with resistance to cetuximab, an anti-epidermal growth factor receptor (EGFR) monoclonal antibody, in metastatic colorectal cancer (mCRC). However, the predictive biomarkers of cetuximab resistance in KRAS wild-type mCRC remain unknown except BRAF, NRAS, and PIK3CA exon 20. The objective of the study is to study the impact of EGFR L2 mutations on resistance to cetuximab in KRAS wild-type patients. PATIENTS AND METHODS A total of 247 mCRC patients were screened for KRAS status at the National Cancer Center Hospital between September 2008 and April 2010. We analyzed the EGFR L2 domain mutation status in KRAS wild type and in the patients treated with cetuximab-based therapy. RESULTS There were 136 patients with wild-type KRAS (55%). Sixty-five patients were analyzed for the L2 domain mutation status, and all patients received cetuximab-based therapy. One patient who had a mutation at exon 9 showed a partial response to cetuximab plus irinotecan. CONCLUSION Mutation of the EGFR L2 domain was analyzed in mCRC patients. Our findings do not provide sufficient evidence that EGFR L2 domain mutation is correlated with resistance to cetuximab.
Collapse
Affiliation(s)
- Yuriko Ito
- Department of Clinical Oncology, Yamagata University, 2-2-2 Iidanishi, Yamagata, Yamagata, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Dynamic dual-tracer MRI-guided fluorescence tomography to quantify receptor density in vivo. Proc Natl Acad Sci U S A 2013; 110:9025-30. [PMID: 23671066 DOI: 10.1073/pnas.1213490110] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The up-regulation of cell surface receptors has become a central focus in personalized cancer treatment; however, because of the complex nature of contrast agent pharmacokinetics in tumor tissue, methods to quantify receptor binding in vivo remain elusive. Here, we present a dual-tracer optical technique for noninvasive estimation of specific receptor binding in cancer. A multispectral MRI-coupled fluorescence molecular tomography system was used to image the uptake kinetics of two fluorescent tracers injected simultaneously, one tracer targeted to the receptor of interest and the other tracer a nontargeted reference. These dynamic tracer data were then fit to a dual-tracer compartmental model to estimate the density of receptors available for binding in the tissue. Applying this approach to mice with deep-seated gliomas that overexpress the EGF receptor produced an estimate of available receptor density of 2.3 ± 0.5 nM (n = 5), consistent with values estimated in comparative invasive imaging and ex vivo studies.
Collapse
|
36
|
Staberg M, Riemer C, Xu R, Dmytriyeva O, Bock E, Berezin V. Identification of a novel antagonist of the ErbB1 receptor capable of inhibiting migration of human glioblastoma cells. Cell Oncol (Dordr) 2013; 36:201-11. [PMID: 23580313 DOI: 10.1007/s13402-013-0128-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2013] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Receptors of the ErbB family are involved in the development of various cancers, and the inhibition of these receptors represents an attractive therapeutic concept. Upon ligand binding, ErbB receptors become activated as homo- or heterodimers, leading to the activation of downstream signaling cascades that result in the facilitation of cell proliferation and migration. A region of the extracellular part of the receptor, termed the 'dimerization arm', is important for the formation of receptor dimers and represents an attractive target for the design of ErbB inhibitors. METHODS An ErbB1 targeting peptide, termed Herfin-1, was designed based on a model of the tertiary structure of the EGF-EGFR ternary complex. The binding kinetics of this peptide were determined employing surface plasmon resonance analyses. ErbB1-4 expression and phosphorylation in human glioblastoma cell lines U87 and U118 were determined by Western blotting using specific antibodies. Cell proliferation was determined by MTS staining. Cell migration was examined using a Chemotaxis Migration Kit. Neurite outgrowth from primary cerebellar granule neurons was evaluated by fluorescence microscopy and image processing. RESULTS The present study shows that Herfin-1 functions as an ErbB1 antagonist. It binds to the extracellular domain of ErbB1 with a KD value of 361 nM. In U87 and U118 cells, both expressing high levels of ErbB1, Herfin-1 inhibits EGF-induced ErbB1 phosphorylation and cell migration. Additionally, Herfin-1 was found to increase neurite outgrowth in cerebellar granule neurons, likely through the inhibition of a sustained weak ErbB1 activation. CONCLUSIONS Targeting the ErbB1 receptor dimerization interface is a promising strategy to inhibit receptor activation in ErbB1-expressing glioma cells.
Collapse
Affiliation(s)
- Mikkel Staberg
- Department of Neuroscience and Pharmacology, Protein Laboratory, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
37
|
Chen CH, Wang SW, Chen CW, Huang MR, Hung JS, Huang HC, Lin HH, Chen RJ, Shyu MK, Huang MC. MUC20 overexpression predicts poor prognosis and enhances EGF-induced malignant phenotypes via activation of the EGFR-STAT3 pathway in endometrial cancer. Gynecol Oncol 2012; 128:560-7. [PMID: 23262208 DOI: 10.1016/j.ygyno.2012.12.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 12/07/2012] [Accepted: 12/08/2012] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Mucins play a critical role in the malignancy of various tumors and have been identified as diagnostic markers and as attractive therapeutic targets. However, the role of mucin (MUC) 20 in endometrial cancer (EC) is still unknown. METHODS The relationship between MUC20 expression and clinical characteristics of EC was analyzed in 97 EC tumors and 16 normal tissues by immunohistochemistry. Effects of MUC20 on EC cells, HEC-1A and RL95-2, were examined by in vitro cell growth, migration, and invasion assays, as well as in vivo tumor growth in SCID mouse model. Western blotting was performed to analyze signaling pathways modulated by MUC20. RESULTS MUC20 expression was significantly higher in EC tumors compared with the normal tissue. High levels of MUC20 expression in EC tumors were correlated with an unfavorable histologic subtype. Furthermore, MUC20 was an independent prognostic factor for poor survival as evaluated by multivariate analyses. Overexpression of MUC20 in EC cells significantly enhanced cell growth, migration, and invasion, as well as tumor growth in vivo. The MUC20-enhanced invasive behavior was significantly blocked by erlotinib, an EGFR inhibitor. Moreover, MUC20 overexpression enhanced EGF-mediated migration and invasion, suggesting a critical role of EGFR in MUC20-mediated effects. We found that MUC20 overexpression could enhance EGF-induced phosphorylation of EGFR and STAT3. Inhibition of the STAT3 activity by its inhibitor Stattic significantly suppressed the MUC20-enhanced invasive behavior. CONCLUSIONS MUC20 is novel prognostic factor for EC and its overexpression enhances EGF-triggered invasive behavior through activation of EGFR-STAT3 pathway.
Collapse
Affiliation(s)
- Chi-Hau Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Ge H, Liu H, Fu Z, Sun Z. Therapeutic and preventive effects of an epidermal growth factor receptor inhibitor on oral squamous cell carcinoma. J Int Med Res 2012; 40:455-66. [PMID: 22613406 DOI: 10.1177/147323001204000207] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Oral squamous cell carcinoma (OSCC) is the sixth most common human neoplasm worldwide. Epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase that is highly expressed in human OSCC and is a target for cancer therapy and prevention. This study investigated the therapeutic and preventive effects of an inhibitor of EGFR (PD153035) on OSCC. METHODS The effects of PD153035 were examined in human cancer cell lines in vitro, in an athymic nude mouse xenograft model in vivo, and in the 7,12-dimethyl benz[a]anthracene (DMBA)-induced hamster cheek pouch tumour model in vivo. RESULTS PD153035 significantly inhibited cell growth, delayed cell cycle progression and induced apoptosis in human OSCC cells in vitro. In vivo, PD153035 inhibited xenograft tumour growth in nude mice in a dose-dependent manner and prevented the development of OSCC at the postinitiation stage in the DMBA-induced hamster cheek pouch tumour model. PD153035 inhibited the DMBA-induced increases in cell proliferation and in levels of phosphorylated EGFR and phosphorylated signal transducer and activator of transcription 3 (STAT3) protein in the hamster cheek pouch. CONCLUSIONS Inhibitors of EGFR, such as PD153035, have potential value in the treatment and prevention of OSCC.
Collapse
Affiliation(s)
- H Ge
- Department of Oral Medicine, Capital Medical University, School of Stomatology, Beijing, China
| | | | | | | |
Collapse
|
39
|
Burotto M, Hartley ML, Marshall JL, Pishvaian MJ. Future of targeted agents in metastatic colorectal cancer. COLORECTAL CANCER 2012; 1:10.2217/crc.12.52. [PMID: 24273599 PMCID: PMC3834580 DOI: 10.2217/crc.12.52] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Great strides have been made in improving the outcome of patients with metastatic colorectal cancer and targeted agents are an important part of the treatment arsenal. The approved monoclonal antibodies, bevacizumab, cetuximab and panitumumab, are part of the standard of care, yet only recently have we begun to define which patients benefit from these therapies using predictive tumor biomarkers. More recently, novel agents including aflibercept and regorafenib have had promising results and may become approved therapies. In addition, agents targeting the mTOR pathway and the TNF pathway have demonstrated early evidence of benefit. In the coming years, we may experience an influx of new therapies, possibly leading to further prolongation of patient survival or even, for some, a cure.
Collapse
Affiliation(s)
- Mauricio Burotto
- Medical Oncology Branch, National Cancer Institute, NIH, MD, USA
| | - Marion L Hartley
- The Lombardi Comprehensive Cancer Center, Georgetown University, Podium B, 3800 Reservoir Road, NW, WA 20007, USA
| | - John L Marshall
- The Lombardi Comprehensive Cancer Center, Georgetown University, Podium B, 3800 Reservoir Road, NW, WA 20007, USA
| | - Michael J Pishvaian
- The Lombardi Comprehensive Cancer Center, Georgetown University, Podium B, 3800 Reservoir Road, NW, WA 20007, USA
| |
Collapse
|
40
|
Pratap A, Singh S, Mundra V, Yang N, Panakanti R, Eason JD, Mahato RI. Attenuation of early liver fibrosis by pharmacological inhibition of smoothened receptor signaling. J Drug Target 2012; 20:770-82. [PMID: 22994359 DOI: 10.3109/1061186x.2012.719900] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hedgehog (Hh) signaling is involved in the pathogenesis of liver fibrosis. It has been previously shown that Hh-inhibitor cyclopamine (CYA) can reduce liver fibrosis in rats. However, CYA is not stable in vivo, which limits its clinical application. This study compares the antifibrotic potential of two known Hh antagonists, vismodegib (GDC-0449, abbreviated to GDC) and CYA. GDC is a synthetic molecule presently in clinical cancer trials and has been reported to be safe and efficacious. These drugs attenuated early liver fibrosis in common bile duct ligated rats, improved liver function, and prevented hepatic stellate cell (HSC) activation, thereby suppressing epithelial to mesenchymal transition (EMT). While both CYA and GDC increased the number of proliferating cell nuclear antigen positive liver cells in vivo, only CYA increased Caspase-3 expression in HSCs in rat livers, suggesting that while GDC and CYA effectively attenuate early liver fibrosis, their hepatoprotective effects may be mediated through different modes of action. Thus, GDC has the potential to serve as a new therapeutic agent for treating early liver fibrosis.
Collapse
Affiliation(s)
- Akshay Pratap
- Division of Solid Organ Transplantation, Methodist University Hospital, Memphis, TN, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Abdullah SE, Haigentz M, Piperdi B. Dermatologic Toxicities from Monoclonal Antibodies and Tyrosine Kinase Inhibitors against EGFR: Pathophysiology and Management. CHEMOTHERAPY RESEARCH AND PRACTICE 2012; 2012:351210. [PMID: 22997576 PMCID: PMC3446637 DOI: 10.1155/2012/351210] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 02/08/2023]
Abstract
Epidermal growth factor receptor (EGFR) inhibition has now been well established as an effective treatment for various cancers. The EGFR belongs to the ErbB family of tyrosine kinase receptors which regulate tumor cell differentiation, survival and proliferation. Activation of EGFR drives tumorigenesis in lung, head and neck, colorectal and pancreatic cancers. Irrespective of the type of cancer being treated and the mechanism by which tumor EGFR drives tumorigenesis, the major side effect of EGFR inhibition is a papulopustular (also described as maculopapular or acneiform) rash which occurs in about two thirds of treated patients. Interestingly, this rash has been commonly correlated with better clinical outcomes (objective tumor response and patient survival). The pathophysiology of dermatological toxicity from EGFR inhibitors is an important area of clinical research, and the proper management of the rash is essential to increase the therapeutic index from this class of drugs. In this paper, we review the dermatologic toxicities associated with EGFR inhibitors with an emphasis on its pathophysiology and clinical management.
Collapse
Affiliation(s)
- Shaad E. Abdullah
- Division of Oncology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New york, NY 10461, USA
| | - Missak Haigentz
- Division of Oncology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New york, NY 10461, USA
| | - Bilal Piperdi
- Division of Oncology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New york, NY 10461, USA
| |
Collapse
|
42
|
Twu C, Han ES. Clinical utility of targeted treatments in the management of epithelial ovarian cancer. Biologics 2012; 6:233-44. [PMID: 22904615 PMCID: PMC3421473 DOI: 10.2147/btt.s29356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Indexed: 11/23/2022]
Abstract
Epithelial ovarian cancer is typically found in its advanced stages, where a combination of surgical debulking and platinum/taxane-based chemotherapy is recommended. Although over 70%-80% of patients achieve remission, a significant proportion develop recurrence of their disease. Additional cytotoxic chemotherapy, as well as surgery, is typically used to manage disease recurrence. Therapies that target specific pathways in cancer cells are rapidly developing in the laboratory and are increasingly being studied in patients with ovarian cancer. We review the current status of novel therapies in the management of epithelial ovarian cancer.
Collapse
Affiliation(s)
- Cheryl Twu
- Department of Women’s Health, Arrowhead Regional Medical Center, Colton, CA, USA
| | - Ernest S Han
- Division of Gynecologic Oncology, City of Hope, Duarte, CA, USA
| |
Collapse
|
43
|
Specenier P, Vermorken JB. Biologic therapy in head and neck cancer: a road with hurdles. ISRN ONCOLOGY 2012; 2012:163752. [PMID: 22745915 PMCID: PMC3382358 DOI: 10.5402/2012/163752] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Accepted: 04/11/2012] [Indexed: 12/28/2022]
Abstract
The epidermal growth factor receptor (EGFR) is overexpressed in the vast majority of cases of squamous cell carcinoma of the head and neck (SCCHN). A high EGFR expression is associated with an unfavorable prognosis. Cetuximab is a chimeric human/murine IgG1 antibody which binds with high affinity to the EGFR. It is the only targeted agent which got approval for the treatment of SCCHN from the regulatory agencies of Europe and the United States, both in locoregionally advanced disease, in association with radiation, and in recurrent/metastatic disease. The outcome of trials involving other EGFR-directed monoclonal antibodies, that is, zalutumumab and panitumumab, was consistent with the results with cetuximab. However these trials failed to meet their primary endpoint. The results with EGFR-directed tyrosine kinase inhibitors have been disappointing. Other potential targets for treatment in SCCHN include the entire ErbB family, the vascular endothelial growth factor (VEGF) and its receptor (VEGFR), the insulin-like growth factor 1 receptor (IGF-1R), the insulin receptor (IR), histone deacetylases (HDAC), the mammalian target of rapamycin (mTOR), the platelet-derived growth factor receptor (PDGFR), heat-shock protein 90 (HSP90), nuclear factor-kappa B (NF-κB), aurora A or B, and phosphatidylinositol 3-kinase (PIK3CA).
Collapse
Affiliation(s)
- Pol Specenier
- Department of Medical Oncology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Jan B. Vermorken
- Department of Medical Oncology, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| |
Collapse
|
44
|
Carrión-Salip D, Panosa C, Menendez JA, Puig T, Oliveras G, Pandiella A, De Llorens R, Massaguer A. Androgen-independent prostate cancer cells circumvent EGFR inhibition by overexpression of alternative HER receptors and ligands. Int J Oncol 2012; 41:1128-38. [PMID: 22684500 DOI: 10.3892/ijo.2012.1509] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 05/04/2012] [Indexed: 11/05/2022] Open
Abstract
The deregulation of the epidermal growth factor receptor (EGFR) pathway plays a major role in the pathogenesis of prostate cancer (PCa). However, therapies targeting EGFR have demonstrated limited effectiveness in PCa. A potential mechanism to overcome EGFR blockade in cancer cells is the autocrine activation of alternative receptors of the human EGFR (HER) family through the overexpression of the HER receptors and ligands. In the present study, we were interested in analyzing if this intrinsic resistance mechanism might contribute to the inefficacy of EGFR inhibitors in PCa. To this end, we selected two androgen-independent human prostate carcinoma cell lines (DU145 and PC3) and established DU145 erlotinib-resistant cells (DUErR). Cells were treated with three EGFR inhibitors (cetuximab, gefinitib and erlotinib) and the sensitivity to each treatment was assessed. The gene expression of the four EGFR/HER receptors and seven ligands of the HER family was analyzed by real-time PCR prior to and after each treatment. The receptors expression and activation were further characterized by flow cytometry and western blot analysis. EGFR inhibition rapidly induced enhanced gene expression of the EGF, betacellulin and neuregulin-1 ligands along with HER2, HER3 and HER4 receptors in the DU145 cells. In contrast, slight changes were observed in the PC3 cells, which are defective in the phosphatase and tensin homolog (PTEN) tumor suppressor gene. In the erlotinib-resistant DUErR cells, the expression of HER2 and HER3 was increased at mRNA and protein levels together with neuregulin-1, leading to enhanced HER3 phosphorylation and the activation of the downstream PI3K/Akt survival pathway. HER3 blockage by a monoclonal antibody restored the cytostatic activity of erlotinib in DUErR cells. Our results confirm that the overexpression and autocrine activation of HER3 play a key role in mediating the resistance to EGFR inhibitors in androgen-independent PCa cells.
Collapse
Affiliation(s)
- Dolors Carrión-Salip
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, Girona 17071, Spain
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Suppression of the epidermal growth factor receptor inhibits epithelial-mesenchymal transition in human pancreatic cancer PANC-1 cells. Dig Dis Sci 2012; 57:1181-9. [PMID: 22271412 DOI: 10.1007/s10620-012-2036-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 01/04/2012] [Indexed: 02/06/2023]
Abstract
BACKGROUND Aberrant expression of epidermal growth factor receptor (EGFR) has been detected in pancreatic cancer; however, the mechanisms of EGFR in inducing pancreatic cancer development have not been adequately elucidated. The objective of this study was to determine the role of EGFR in mediating epithelial-mesenchymal transition (EMT) in pancreatic cancer cells. METHODS Pancreatic cancer cell line PANC-1 was transfected with small interfering RNA of EGFR by use of a lentiviral expression vector to establish an EGFR-knockdown cell line (si-PANC-1). PANC-1 cells transfected with lentiviral vector expressing negative control sequence were used as negative control (NC-PANC-1). Scratch assay and transwell study were used to analyze cell migration and invasion. Real-time PCR and Western blotting were used to detect the expression of EMT markers E-cadherin, N-cadherin, vimentin, and fibronectin and transcription factors snail, slug, twist1, and sip1 in PANC-1, NC-PANC-1, and si-PANC-1 cells. Immunofluorescent staining with these antibodies and confocal microscopy were used to observe their cellular location and morphologic changes. RESULTS After RNA interference of EGFR, the migration and invasion ability of si-PANC-1 cells decreased significantly. The expression of epithelial phenotype marker E-cadherin increased and the expression of mesenchymal phenotype markers N-cadherin, vimentin, and fibronectin decreased, indicating reversion of EMT. We also observed intracellular translocation of E-cadherin. Expression of transcription factors snail and slug in si-PANC-1 cells decreased significantly. CONCLUSION Suppression of EGFR expression can significantly inhibit EMT of pancreatic cancer PANC-1 cells. The mechanism may be related with the down-regulation of the expression of transcription factors snail and slug.
Collapse
|
46
|
Patel AN, Mehnert JM, Kim S. Treatment of recurrent metastatic head and neck cancer: focus on cetuximab. CLINICAL MEDICINE INSIGHTS. EAR, NOSE AND THROAT 2012; 5:1-16. [PMID: 24179404 PMCID: PMC3791949 DOI: 10.4137/cment.s5129] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
EGFR belongs to the ErbB family of receptor tyrosine kinases and is associated with worse prognosis in head and neck squamous cell carcinoma (HNSCC). Cetuximab is a monoclonal antibody to the extracellular domain of EGFR and inhibits its downstream actions via multiple mechanisms. Besides its proven efficacy in locally advanced and incurable HNSCC, cetuximab has the distinct advantage of having a relatively tolerable side effect profile and not potentiating radiation toxicity. Though therapies for advanced HNSCC are evolving, locoregional recurrence and/or distant metastases occur in a large percentage of patients. Though some patients can be salvaged with surgery or radiation therapy, the majority are incurable, and are treated palliatively with systemic therapy. In the setting of first line therapy for recurrent/metastatic HNSCC, the EXTREME trial provided level 1 evidence that cetuximab improves overall survival when combined with cisplatinum and 5 FU. Following progression on first line chemotherapy, several phase II trials suggest that cetuximab monotherapy is a reasonable choice in this setting. Future studies should concentrate on clinical and molecular markers that may allow more personalized approaches to treating HNSCC, and combining EGFR inhibitors with other agents in a synergistic approach.
Collapse
Affiliation(s)
- Akshar N Patel
- Department of Radiation Oncology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ
| | | | | |
Collapse
|
47
|
Chen G, Kronenberger P, Teugels E, Umelo IA, De Grève J. Targeting the epidermal growth factor receptor in non-small cell lung cancer cells: the effect of combining RNA interference with tyrosine kinase inhibitors or cetuximab. BMC Med 2012; 10:28. [PMID: 22436374 PMCID: PMC3334713 DOI: 10.1186/1741-7015-10-28] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Accepted: 03/21/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The epidermal growth factor receptor (EGFR) is a validated therapeutic target in non-small cell lung cancer (NSCLC). However, current single agent receptor targeting does not achieve a maximal therapeutic effect, and some mutations confer resistance to current available agents. In the current study we have examined, in different NSCLC cell lines, the combined effect of RNA interference targeting the EGFR mRNA, and inactivation of EGFR signaling using different receptor tyrosine kinase inhibitors (TKIs) or a monoclonal antibody cetuximab. METHODS NSCLC cells (cell lines HCC827, H292, H358, H1650, and H1975) were transfected with EGFR siRNA and/or treated with the TKIs gefitinib, erlotinib, and afatinib, and/or with the monoclonal antibody cetuximab. The reduction of EGFR mRNA expression was measured by real-time quantitative RT-PCR. The down-regulation of EGFR protein expression was measured by western blot, and the proliferation, viability, caspase3/7 activity, and apoptotic morphology were monitored by spectrophotometry, fluorimetry, and fluorescence microscopy. The combined effect of EGFR siRNA and different drugs was evaluated using a combination index. RESULTS EGFR-specific siRNA strongly inhibited EGFR protein expression almost equally in all cell lines and inhibited cell growth and induced cell apoptosis in all NSCLC cell lines studied, albeit with a different magnitude. The effects on growth obtained with siRNA was strikingly different from the effects obtained with TKIs. The effects of siRNA probably correlate with the overall oncogenic significance of the receptor, which is only partly inhibited by the TKIs. The cells which showed weak response to TKIs, such as the H1975 cell line containing the T790M resistance mutation, were found to be responsive to siRNA knockdown of EGFR, as were cell lines with downstream TKI resistance mutations. The cell line HCC827, harboring an exon 19 deletion mutation, was more than 10-fold more sensitive to TKI proliferation inhibition and apoptosis induction than any of the other cell lines. Cetuximab alone had no relevant in vitro activity at concentrations obtainable in the clinic. The addition of EGFR siRNA to either TKIs or cetuximab additively enhanced growth inhibition and induction of apoptosis in all five cell lines, independent of the EGFR mutation status (wild-type or sensitizing mutation or resistant mutation). The strongest biological effect was observed when afatinib was combined with an EGFR-specific siRNA. CONCLUSIONS EGFR knockdown by siRNA further decreases the cell growth of lung cancer cells that are treated with TKIs or cetuximab alone, confirming that single agent drug targeting does not achieve a maximal biological effect. The siRNA inhibits EGFR oncogenic activity that bypasses downstream "resistance" mutations such as KRAS and PTEN. The combined treatment of siRNA and EGFR inhibitory agents is additive. The combination of a potent, irreversible kinase inhibitor such as afatinib, with EGFR-specific siRNAs should be further investigated as a new strategy in the treatment of lung cancer and other EGFR dependent cancers, including those with downstream resistance mutations.
Collapse
Affiliation(s)
- Gang Chen
- Laboratory of Medical and Molecular Oncology and Department of Medical Oncology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | | | | | | | | |
Collapse
|
48
|
Correlation between epidermal growth factor receptor-specific nanobody uptake and tumor burden: a tool for noninvasive monitoring of tumor response to therapy. Mol Imaging Biol 2012; 13:940-8. [PMID: 20865332 DOI: 10.1007/s11307-010-0428-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE Nanobodies represent an interesting class of probes for the generic development of molecular imaging agents. We studied the relationship between tumor uptake of the epidermal growth factor receptor (EGFR)-specific nanobody (99m)Tc-7C12 and tumor burden and evaluated the possibility of using this probe to monitor tumor response to erlotinib. PROCEDURES The specificity and affinity of (99m)Tc-7C12 was determined on A431 cells. Cells expressing firefly luciferase were used to evaluate tumor burden using bioluminescence imaging. We evaluated the effect of erlotinib on tumor burden and (99m)Tc-7C12 uptake in vitro as well as in vivo. In vivo bioluminescence imaging was performed followed by pinhole single-photon emission computed tomography/micro-computed tomography. RESULTS (99m)Tc-7C12 binds specifically to the receptor with high affinity (3.67 ± 0.59 nM). Erlotinib reduced tumor uptake and cell viability in a concentration-dependent manner. Tumor uptake of (99m)Tc-7C12 showed good correlation with tumor burden. Erlotinib treatment resulted in a progressive reduction of tumor burden and tumor uptake of (99m)Tc-7C12. CONCLUSION (99m)Tc-7C12 binds to EGFR with high affinity and specificity. Tumor uptake is correlated with tumor burden. Quantification of (99m)Tc-7C12 uptake is promising for monitoring therapy response of EGFR-expressing tumors.
Collapse
|
49
|
Hamzeh-Mivehroud M, Mahmoudpour A, Dastmalchi S. Identification of new peptide ligands for epidermal growth factor receptor using phage display and computationally modeling their mode of binding. Chem Biol Drug Des 2012; 79:246-59. [PMID: 22136656 DOI: 10.1111/j.1747-0285.2011.01282.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Peptide phage display, a powerful method for ligand identification, was used to identify new peptide ligands for epidermal growth factor receptor. A-431 cells expressing epidermal growth factor receptor were used as the matrix in a cell-based subtractive biopanning approach using a 7-mer peptide displaying phage library. Two novel peptide ligands were identified and tested for their affinities and functional effects on epidermal growth factor receptor. The identified peptides were able to inhibit the epidermal growth factor-induced phosphorylation of epidermal growth factor receptor in a concentration-dependent manner. The results of affinity binding experiments showed that the natural ligand, that is epidermal growth factor, was able to inhibit competitively the binding of peptide-bearing phage to epidermal growth factor receptor expressing A-431 cells. Molecular modeling studies were used to calculate the free energies for the binding of peptides to the receptor-binding site as well as proposing the interaction modes for this binding. The calculated values for the binding energies were found to be similar to our experimental data and those of previously reported studies.
Collapse
|
50
|
Kreutzer JN, Salvador A, Diana P, Cirrincione G, Vedaldi D, Litchfield DW, Issinger OG, Guerra B. 2-Triazenoazaindoles: α novel class of triazenes inducing transcriptional down-regulation of EGFR and HER-2 in human pancreatic cancer cells. Int J Oncol 2011; 40:914-22. [PMID: 22134789 PMCID: PMC3584806 DOI: 10.3892/ijo.2011.1272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 10/25/2011] [Indexed: 01/29/2023] Open
Abstract
Pancreatic cancer is a complex malignancy arising from the accumulation of genetic and epigenetic defects in the affected cells. Standard chemotherapy for patients with advanced disease shows only modest effects and is associated with considerable toxicity. Overexpression or aberrant activation of members of the epidermal growth factor receptor tyrosine kinase family, which includes EGFR and HER-2, occurs frequently and is associated with multiple drug resistance and decreased patient survival. In this study, we have investigated the therapeutic potential of AS104, a novel compound of the triazene class, with potential inhibitory effects on EGFR. We found that treatment of cells with AS104 causes significant reduction of cell growth and metabolic activity in four human pancreatic cancer cell lines. Furthermore, we show that the AS104-mediated induction of apoptotic cell death is associated with stimulation of autophagy in a dose-dependent manner. Treatment of cells with AS104 results in significant down-regulation of EGFR and HER-2 expression and activity and subsequent inhibition of downstream signaling proteins. Quantitative RT-PCR analysis and assays with proteasome inhibitors revealed that AS104 regulates the expression of EGFR and HER-2 at the transcriptional level. These findings provide for the first time experimental evidence for efficacy of AS104 in the simultaneous transcriptional repression of EGFR and HER-2 genes and suggest that AS104 may have therapeutic potential in the treatment of pancreatic cancers that express high levels of the aforementioned receptor tyrosine kinases.
Collapse
Affiliation(s)
- Jan N Kreutzer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | | | | | | | | | | | | | | |
Collapse
|